View Article

  • Advancements in HPLC Techniques for Detecting and identification of Process-Related and Degradants Impurities in Pharmaceutical Compounds

  • 1,4Department of Chemistry, Anurag University, Hyderabad, India.

    2,3Analytical Research and Development, Aurore Life Sciences Private Limited, Hyderabad, India.

Abstract

Advancements in High-Performance Liquid Chromatography (HPLC) have significantly improved the detection, identification and quantification of process-related impurities and degradants in pharmaceuticals. Key innovations include Ultra-High-Performance Liquid Chromatography (UHPLC), which provides enhanced resolution and sensitivity, and multi-dimensional chromatography for better separation of complex mixtures. The integration of HPLC with Mass Spectrometry, particularly LC-MS/MS and High-Resolution Mass Spectrometry (HRMS), enables precise identification and quantification of impurities at trace levels. Advanced detectors, such as Diode-Array Detection (DAD), fluorescence detection, and evaporative light scattering detection (ELSD), further enhance sensitivity and selectivity for various impurities. Chiral chromatography is employed to resolve enantiomeric impurities. Additionally, the use of greener solvents and inline HPLC for real-time monitoring contribute to more sustainable and efficient practices. These advancements collectively ensure the safety, purity, and regulatory compliance of pharmaceutical products.

Keywords

High-Performance Liquid Chromatography (HPLC), Ultra-High-Performance Liquid Chromatography (UHPLC), LC-MS/MS, High-Resolution Mass Spectrometry (HRMS) degradation impurities. pharmaceutical applications, method development and validation, ICH guidelines.

Introduction

The pharmaceutical industry faces increasing regulatory scrutiny and the demand for high-quality drug products, which requires the development of advanced analytical techniques for detecting and quantifying impurities and degradation products that may compromise drug safety and efficacy [1,2] High-Performance Liquid Chromatography (HPLC) has been the standard method for such analyses, offering robust separation and quantification of complex pharmaceutical matrices [3]. However, with growing formulation complexities and stringent regulations, there is a need for more sensitive, efficient, and reliable methods. Recent advancements in chromatographic technologies, such as Ultra-High-Performance Liquid Chromatography (UHPLC), have enhanced resolution, throughput, and sensitivity [4]. The integration of HPLC with Mass Spectrometry (LC-MS/MS) [5] and High-Resolution Mass Spectrometry (HRMS) has revolutionized impurity detection, enabling the identification and quantification of trace impurities in complex matrices. Additionally, novel detection technologies, such as Diode-Array Detection (DAD) and Evaporative Light Scattering Detection (ELSD), offer improved sensitivity for a broader range of pharmaceutical analytes, including those that do not absorb UV light [6].

This review explores recent advancements in HPLC techniques, highlighting their role in detecting and quantifying process-related impurities and degradants, thereby ensuring the safety, quality, and regulatory compliance of pharmaceutical products throughout their lifecycle [3].

  1. Significance of Detecting and identification Process-Related Impurities and Degradants in Pharmaceuticals.

Detecting and identification process-related impurities and degradants in pharmaceuticals is critical for several reasons, particularly in terms of drug safety, efficacy, regulatory compliance, and product quality. Here's a breakdown of why this is so important [1-2].

    1. Patient Safety and Health:
  • Toxicity Risk: Some impurities and degradants can be toxic or have adverse effects on patient health. Detecting these substances ensures that only safe, high-quality drugs reach patients.
  • Long-Term Effects: Certain impurities might not cause immediate harm but could have long-term cumulative effects. For example, they could contribute to carcinogenic, mutagenic, or teratogenic risks, potentially leading to serious health problems after prolonged exposure.
  • Allergic Reactions: Even trace amounts of impurities can trigger allergic reactions in sensitive patients. Identifying and removing these contaminants helps protect vulnerable populations, such as those with sensitivities or preexisting health conditions.
    1. Drug Efficacy and Therapeutic Performance:
  • Impact on Efficacy: Some impurities or degradants may interfere with the active pharmaceutical ingredient (API) or alter its therapeutic performance. This could result in reduced effectiveness of the drug or failure to achieve the desired clinical outcome.
  • Bioavailability Issues: Impurities can affect the solubility or absorption of a drug, potentially compromising bioavailability. Accurate detection helps ensure that the drug is delivered effectively to the target site in the body.
    1.  Regulatory Compliance:
  • Adherence to Guidelines: Regulatory bodies like the FDA, EMA, and ICH require rigorous testing of impurities and degradants to ensure pharmaceuticals meet safety and quality standards. Failure to detect and control impurities can lead to regulatory sanctions, including product recalls, fines, or market withdrawal.
  • Good Manufacturing Practices (GMP): Detecting impurities is part of maintaining GMP standards. GMP-compliant processes require consistent monitoring and analysis of all components, including raw materials, intermediates, and finished products, to ensure they meet established specifications.
  • Validation and Stability Studies: Regular testing for impurities and degradants is also part of stability studies required for new drug approvals. These studies help determine the shelf life of a product and the conditions under which it remains safe and effective.
    1.   Product Quality and Consistency:
  • Uniformity Across Batches: Impurities and degradants can vary from batch to batch, so routine monitoring is essential to ensure consistency in pharmaceutical products. This consistency ensures that patients receive the correct dose, and the drug performs reliably over time.
  • Contamination Control: Process-related impurities often arise from manufacturing conditions (e.g., equipment residues, solvents, or byproducts), packaging materials, or storage conditions. Detecting and measuring them ensures that contamination is controlled, and the product is free from harmful levels of foreign substances.
    1.   Risk Management in Pharmaceutical Development:
  • Understanding Impurity Profiles: Analyzing the impurity profile of a drug allows for a deeper understanding of the manufacturing process, helps identify potential risks, and guides process optimization.
  • Early Detection of Degradation Products: Degradation can occur due to heat, light, moisture, or chemical interactions during manufacturing or storage. Identifying these degradation products early in the development process allows for formulation adjustments to prevent their formation or reduce their levels in the final product.
    1.   Economic and Market Considerations:
  • Cost of Recalls or Litigation: If harmful impurities are not detected and the drug reaches the market, the financial cost of product recalls, legal action, or damage to brand reputation can be enormous.
  • Market Access and Acceptance: Drugs that meet high-quality standards and demonstrate rigorous impurity control are more likely to be accepted in global markets. Meeting or exceeding regulatory impurity limits is a key factor in obtaining market approval across regions.
    1. Analytical and Process Optimization:
  • Improved Manufacturing Efficiency: Monitoring impurities allows manufacturers to identify potential inefficiencies in the production process. By understanding the root causes of impurities or degradation, they can optimize conditions (e.g., temperature, pH, reaction times) to reduce unwanted byproducts.
  • Optimizing Formulation Stability: By understanding how impurities and degradants impact the stability of the drug, pharmaceutical developers can create more stable formulations, extending shelf life and ensuring the product remains effective throughout its intended use period.
    1. Environmental Impact:
  • Safe Disposal: Process-related impurities or degradants that remain in waste products or solvents need to be carefully measured and controlled. Proper identification ensures that these substances are disposed of safely, in compliance with environmental regulations, and that no harmful byproducts contaminate the surrounding environment.
    1. Supporting Product Lifecycle Management:
  • Post-Launch Monitoring: Even after a pharmaceutical product is launched, ongoing monitoring for process-related impurities and degradants is essential to ensure long-term product safety. Impurities might emerge or increase in concentration over time due to changes in manufacturing or storage conditions.
    1. . Innovation in Formulation Development:
  • Better Drug Design: Continuous monitoring of impurities and degradants encourages innovative drug formulations that minimize degradation pathways or impurity formation, leading to more robust drug products that have improved shelf stability and patient compliance.
  1. High-Performance Liquid Chromatography Key concepts:

High-Performance Liquid Chromatography (HPLC) is a widely used analytical technique for separating, identifying, and quantifying components in a mixture based on their differential interactions with the stationary and mobile phases [7]. The stationary phase is typically a column packed with a solid material, such as silica, while the mobile phase is a liquid solvent or mixture that carries the sample through the column. The components in the sample are separated due to differences in their retention times, which depend on their affinity for the stationary phase. This separation is followed by detection using various techniques, including UV-Vis absorption, fluorescence, and mass spectrometry. The high resolution and sensitivity of HPLC make it

Instrumentation: Essential components are involved in the structure of HPLC instrumentation:

Solvent Reservoir

Degasser

Pump

Injection System

Columns Oven

Detectors

Data Acquisition System

 

 

Figure-1 Schematic Representation of High-Performance liquid chromatography

Solvent Reservoir:

The HPLC solvent reservoir is a critical component that stores the mobile phase, typically a mixture of organic solvents and aqueous buffers, which are necessary for the chromatographic separation. The solvent is continuously pumped through the system, ensuring a stable flow rate and consistent solvent composition. The reservoir is equipped with a venting mechanism to equalize pressure and prevent air ingress, which could lead to baseline instability. To maintain solvent integrity, the reservoir is often fitted with filters to remove particulate matter and prevent contamination. The careful management of solvent purity and the reservoir's design are vital for reproducible chromatographic analysis [7].

    1. Degasser:

The HPLC degasser is a crucial component of the High-Performance Liquid Chromatography system that removes dissolved gases (such as air) from the mobile phase. This helps improve the accuracy and reproducibility of the analysis by preventing bubbles that can interfere with detector signals and cause pressure fluctuations. Here are some alternative ways to describe the HPLC degasser [7].

    1. Pump:

The HPLC pump is responsible for delivering the mobile phase through the chromatographic system at a controlled flow rate and pressure, typically ranging from 500 psi to 6000 psi. The pump ensures the mobile phase is introduced into the column with precise flow, which is critical for reproducible retention times and chromatographic resolution. Pumps commonly used in HPLC include reciprocating and syringe pumps, which provide constant flow and minimize pulsations that can affect detector stability. The pump mechanism may be equipped with a piston and check valves to maintain unidirectional solvent flow and prevent backflow. Proper pump maintenance is essential for preventing air bubbles, leaks, or fluctuations in pressure, which could degrade chromatographic performance. Accurate pump operation is crucial for the efficiency of separations and the quality of analytical results [7,9].

    1. Injection System:

The HPLC injector is a critical component responsible for introducing the sample into the mobile phase stream with precision and reproducibility. It typically utilizes either a manual syringe or an automated injection valve, which ensures that the sample volume is accurately injected into the flow path. The most common type of automated injector is the loop injector, where the sample is loaded into a fixed-loop volume and then injected into the system via a rotating valve. This method minimizes sample loss and ensures that the injection volume remains constant. Proper sample introduction is essential for maintaining chromatographic resolution and avoiding sample contamination. The injector must also be carefully cleaned to prevent carryover and ensure consistent results across injections [7,9].

    1. Columns Oven:

The HPLC column compartment is a temperature-controlled area where the chromatographic column is placed during the separation process. Temperature regulation is crucial, as variations can lead to changes in retention times, resolution, and peak shape. The compartment is typically equipped with a thermostat or Peltier cooler to maintain a constant temperature, which enhances the reproducibility of the chromatographic analysis. The column itself contains a stationary phase, which interacts with the analytes in the mobile phase, resulting in their separation based on their differential affinities for the stationary phase. Consistent temperature control within the column compartment ensures that the separation conditions remain stable, minimizing variability in results. Column maintenance and proper handling within this compartment are essential for preserving column life and ensuring optimal performance [7,9].

    1. Detectors:

High-Performance Liquid Chromatography (HPLC) employs a range of sophisticated detection systems to facilitate both qualitative and quantitative analyses, each tailored to the specific physicochemical properties of target analytes. These detectors convert various analyte characteristics into measurable signals, enabling precise identification and quantification. Key detection modalities include [7,9].

2.7 UV-Visible Absorption Detectors:

UV/Vis absorbance detectors are essential components in High-Performance Liquid Chromatography (HPLC) systems, Diode-array detectors (DAD) enhance the identification and quantification of analytes based on their absorption of ultraviolet and visible light. These detectors operate by passing light through a flow cell containing the eluted sample and measuring the intensity of transmitted light across specific wavelengths. The resulting data, typically presented as a chromatogram, allows for the determination of analyte concentration and identification [9].

    1. Fluorescent Spectrometers:

Fluorescence detectors are integral to High-Performance Liquid Chromatography (HPLC) systems, offering exceptional sensitivity and selectivity by detecting the emitted light from analytes upon excitation. These detectors are particularly effective for trace-level quantification in complex matrices, as they measure the intensity of light emitted when specific compounds are excited at wavelengths. Applications include the detection of compounds that are inherently fluorescent or have been derivatized to exhibit fluorescence [9].

    1. LC coupled with Mass Spectrometry (LC-MS/MS).

Integrating High-Performance Liquid Chromatography (HPLC) with Mass Spectrometry (MS) enables thorough structural elucidation and accurate quantification of analytes. This combination provides molecular mass determination, structural fragmentation patterns, and isotopic distribution analysis, which are essential for characterizing complex mixtures, impurities, metabolites, and genotoxic compounds with high specificity and sensitivity. The incorporation of tandem mass spectrometry (LC-MS/MS) further enhances detection limits and resolution, supporting robust analysis in pharmaceutical, biological, and environmental applications [5].

    1.  LC coupled with High-Resolution Mass Spectrometry (LC-HRMS).

Integrating Liquid Chromatography with High-Resolution Mass Spectrometry (LC-HRMS) enables comprehensive structural elucidation and precise quantification of analytes. This combination provides accurate molecular mass determination, detailed structural fragmentation patterns, and high-resolution isotopic distribution analysis, which are crucial for characterizing complex mixtures, impurities, metabolites, and genotoxic compounds with exceptional specificity and sensitivity. The incorporation of high-resolution and accurate mass capabilities further enhances detection limits and mass accuracy, supporting robust analysis in pharmaceutical, biological, and environmental applications. [52].

2.10 Monitoring and Data Collection System:

A Data Acquisition System (DAS) is essential in High-Performance Liquid Chromatography (HPLC), enabling the collection, processing, and analysis of detector signals to generate chromatographic data. It interfaces with the detector to record variables such as peak area, retention time, and absorbance. Typically, the DAS converts the analog signals from the detector into digital format using an analog-to-digital converter (ADC), ensuring accurate and consistent data. The system often includes specialized software that enables the visualization of chromatograms, peak integration, and quantitative analysis. The DAS also allows for Instantaneous observation and adjustments of system parameters, Assuring high-quality data collection. Integration with databases and reporting systems allows for seamless analysis and documentation of results. [9].

  1. Systematic Procedure:
    1. Sample Treatment:

Prior to injection into the HPLC system, proper sample preparation is essential for ensuring accurate and reproducible analytical results. The process begins with filtration, where particulate matter is removed using filters with pore sizes typically ranging from 0.45 µm to 0.2 µm. This step is critical to avoid any potential blockage or damage to the chromatographic column and to maintain consistent flow rates within the system. Following filtration, the sample undergoes dissolution, ensuring complete solubilization of the analytes in an appropriate solvent. The solvent choice is typically aligned with the mobile phase composition to prevent phase separation or any solubility-related issues that could affect the stability and consistency of the sample. Lastly, the sample may require concentration adjustment to ensure that the analyte concentrations fall within the detectable range of the HPLC system’s detector. This dilution step is essential for optimizing sensitivity and ensuring the accurate quantification of the analytes without overloading the system. [10].

    1. System Configuration / HPLC system setup:

The HPLC system setup comprises several integral components that work in unison to facilitate efficient chromatographic separation. The pump delivers the mobile phase at a constant flow rate and pressure, ensuring a stable and reproducible elution process. The injector introduces the sample into the mobile phase stream, either manually or via an autosampler, ensuring accurate and consistent injection volumes. The column, housing the stationary phase, facilitates separation based on differential interactions between the analytes and the stationary phase. Following separation, the detector (such as UV-Vis, fluorescence, or mass spectrometry) identifies and quantifies the analytes by measuring their interaction with the mobile phase as they elute from the column. Each component plays a critical role in achieving precise separation, detection, and quantification of the analytes. It involves selecting and connecting hardware, software, and peripherals, adjusting parameters, and calibrating instruments. Proper configuration ensures accurate, reproducible results while preventing errors. Effective system configuration is crucial for maintaining system integrity and achieving reliable analytical outcomes [7].

    1. Column equilibration:

is a critical step in HPLC to ensure consistent and reproducible separation. Prior to sample injection, the column is flushed with the mobile phase at the desired flow rate to saturate the stationary phase with the mobile phase, ensuring stable baseline conditions. This process helps to stabilize the column temperature, pressure, and composition, reducing the risk of sample retention anomalies and ensuring the stationary phase is fully equilibrated. Equilibration also prevents phase separation and ensures optimal interaction between the analytes and the stationary phase during the separation process [7].

    1. Loading the Sample:

Loading the sample in HPLC or LC-MS involves introducing the prepared sample into the analytical system for analysis. This step is critical to ensure that the sample is properly injected into the column or the ion source for separation and detection. Careful loading prevents contamination, ensures consistent sample volume, and optimizes system performance. Proper technique during sample loading is essential for achieving accurate, reproducible results [11].

    1. Chromatographic Separation:

Chromatographic separation in HPLC involves the process of separating components of a sample based on their interactions with the stationary phase and the mobile phase. The different affinities of compounds for these phases cause them to travel at different speeds, resulting in their separation. This technique is fundamental for isolating individual analytes in complex mixtures. Efficient chromatographic separation ensures clear, distinct peaks in the chromatogram, leading to accurate identification and quantification [12,13].

    1. Analyte Identification:

Analytes are identified as they emerge from the column. The detection method (e.g., UV-Vis, fluorescence, or mass spectrometry) is selected based on the analyte’s chemical properties, allowing for specific and sensitive detection of the compounds of interest. Analyte identification in HPLC involves determining the chemical identity of compounds based on their retention times, which are compared with known standards. The use of a detector, such as UV-Vis, helps confirm the presence of specific analytes by measuring their absorbance at characteristic wavelengths. Coupling retention time data with other techniques, like mass spectrometry, enhances accuracy. This step is essential for confirming the identity of compounds in complex mixtures and ensuring reliable analytical results [4].

    1. Analytical Data Evaluation:

Analytical data evaluation involves interpreting chromatograms to extract meaningful results, such as peak areas, retention times, and resolution. It includes assessing the quality of data, verifying system suitability, and applying calibration curves for quantification. Proper evaluation ensures accuracy, precision, and compliance with analytical standards. This step is critical for drawing reliable conclusions from experimental results. [14].

  1. Significance of HPLC in the Pharmaceutical analysis:

HPLC plays a crucial role in pharmaceutical analysis by ensuring the quality, safety, and efficacy of drugs. It is widely used for quantifying active pharmaceutical ingredients (APIs), identifying impurities, and validating formulations. The technique provides high precision, accuracy, and sensitivity, making it indispensable for compliance with regulatory standards [8]. HPLC supports all stages of drug development, from research to final product testing.

    1. Assessment of Active Pharmaceutical Ingredients (APIs):

HPLC is widely used to quantify the concentration of APIs in raw materials and finished products. Accurate measurement ensures that formulations meet specified potency requirements [1].

    1. Impurity Profiling and Quantification: 

In pharmaceutical analysis, HPLC and LC-MS/MS are both essential techniques for impurity profiling and quantification. HPLC separates and identifies impurities based on their interaction with the stationary phase, while LC-MS/MS offers enhanced sensitivity and specificity by combining separation with mass spectrometry for precise detection of trace impurities. Together, these methods provide accurate quantification of impurities and degradation products, ensuring compliance with regulatory standards. They play a critical role in maintaining the safety, efficacy, and quality of pharmaceutical products [15].

    1. Stability studies:

Stability studies in pharmaceuticals evaluate how environmental factors like temperature, humidity, and light affect a drug's quality over time. These studies determine the shelf life, storage conditions, and expiration date of pharmaceutical products. By identifying degradation pathways, stability studies ensure the safety, efficacy, and integrity of medications throughout their lifecycle. They are essential for regulatory compliance and product reliability [16,17].

    1. Bioavailability and bioequivalence studies

Bioavailability and bioequivalence studies are critical in pharmaceutical analysis to evaluate how drugs are absorbed, distributed, and utilized in the body. Bioavailability measures the rate and extent of active ingredient absorption, while bioequivalence compares two formulations to ensure they produce similar effects. These studies are essential for generic drug development and regulatory approval. They ensure therapeutic equivalence and consistent clinical outcomes across different drug formulations [18].

    1. Dissolution testing

Dissolution testing is a key aspect of pharmaceutical analysis used to evaluate the rate and extent at which an active pharmaceutical ingredient (API) is released from its dosage form. It helps predict drug behaviour in the gastrointestinal tract and ensures consistent bioavailability. This test is critical for quality control, formulation development, and regulatory compliance. Dissolution profiles guide product optimization and ensure batch-to-batch uniformity [19].

    1. Method validation:

in pharmaceutical analysis ensures that analytical methods are reliable, accurate, and suitable for their intended purpose. It involves evaluating parameters like precision, accuracy, linearity, specificity, and robustness. Validation is essential for regulatory compliance and ensures consistent and reproducible results. This process guarantees the quality and integrity of data used in drug development and quality control [20,21,22].

    1. Quality Control (QC) testing

Quality Control (QC) testing in pharmaceutical analysis ensures that drugs meet predefined quality standards for safety, efficacy, and consistency. It involves testing raw materials, intermediates, and finished products for parameters like potency, purity, and stability. QC testing helps detect defects, maintain batch-to-batch uniformity, and comply with regulatory guidelines. This process is essential for delivering reliable and high-quality medications to patients [23, 24].

    1. Separation of Chiral Compounds:

HPLC is widely used for the separation of chiral compounds in pharmaceutical analysis, enabling the resolution of enantiomers with distinct biological properties. By employing chiral stationary phases, HPLC differentiates between the two optical isomers based on their interactions with the column. This technique is vital for producing enantiomerically pure drugs, ensuring safety and efficacy. It plays a critical role in drug development, quality control, and regulatory compliance. [13,25].

  1. Advantages of HPLC in the Pharmaceutical Industry:
    1. High Sensitivity and Accuracy:

HPLC provides precise and reliable results for the quantification and detection of trace amounts of active pharmaceutical ingredients (APIs) and impurities, ensuring accurate formulation and quality control [7].

    1. Versatility:

HPLC can be used to analyze a wide range of substances, including small molecules, biologics, and complex formulations, making it a versatile technique for pharmaceutical testing [27,28].

    1. Regulatory Compliance:

HPLC is an established and validated technique that meets regulatory requirements from agencies like the FDA and EMA, ensuring that pharmaceutical products meet

pharmaceutical products meet stringent quality standards [1-2-28].

    1.  Repeatability and Reliability

HPLC ensures reproducible results across different batches, essential for maintaining  consistent product quality during manufacturing and ensuring batch-to-batch uniformity [28].

    1. Swift Performance:

With automated systems, HPLC provides rapid analysis, which speeds up the process of drug development, stability testing, and quality control [7].

    1. Adaptability for Increased Demand:

HPLC (High-Performance Liquid Chromatography) demonstrates strong adaptability for increased demand in pharmaceutical and industrial applications. As production volumes rise, HPLC systems can be adjusted to handle larger sample sizes and higher throughput without compromising accuracy or precision. This scalability ensures that the method remains effective for both small-scale research and large-scale manufacturing. Its versatility allows for the optimization of parameters, making it suitable for evolving analytical needs. [29].

    1. Minimal Sample Preparation: HPLC requires relatively minimal sample preparation compared to other techniques, reducing analysis time and avoiding potential errors introduced during sample handling [11].
  1. Impurity Categories and Their Sources in Pharmaceutical Manufacturing:

Impurity Categories and Their Sources in Pharmaceutical Manufacturing refer to the various types of impurities, such as degradation products, residual solvents, or by-products, that can arise during drug production. These impurities can originate from raw materials, chemical reactions, or environmental contamination, potentially affecting drug safety and efficacy. The impurities can be classified into the following types:

    1. Impurities generated during process development

 in pharmaceutical manufacturing can arise from various sources such as chemical reactions, side reactions, degradation, and contamination from raw materials or equipment. These impurities may include residual solvents, by-products, and degradation products that could affect the safety, efficacy, and quality of the final product [28]. HPLC is commonly used for separating and quantifying process-related impurities and degradation products, while GC is suitable for analyzing volatile impurities such as residual solvents. LC-MS/MS, combining the separation capabilities of liquid chromatography with the sensitivity of mass spectrometry, offers enhanced detection and identification of trace impurities. These methods play a critical role in ensuring compliance with regulatory standards and maintaining product quality [29].

    1. Impurities from Degradation

Degradation products are formed when APIs or formulations undergo chemical changes due to environmental factors such as light, heat, moisture, or oxidative conditions. These products can adversely affect the safety, efficacy, and shelf life of the drug. Stability studies using GC or HPLC allow for the identification and quantification of degradation products, supporting the development of robust formulations and proper storage guidelines [8,20].

    1. Raw Material Impurities: These are contaminants that originate from the starting materials used in drug production, such as active pharmaceutical ingredients (APIs), excipients, or reagents. Impurities may include residual solvents, impurities from the synthesis process, or contaminants introduced during the production of raw materials.
    2. Extraneous Substances

Extraneous Substances refer to materials or compounds that are not part of the intended formulation or composition of a pharmaceutical product. These substances may be introduced during the manufacturing process, handling, or storage, and can include contaminants, by-products, or unintended chemicals that may compromise the quality, safety, or efficacy of the drug. Extraneous substances can be identified and quantified during quality control testing, often through methods like HPLC, GC, or LC-MS/MS, to ensure that the final product meets regulatory standards and is free from unwanted components. ensuring adherence to Good Manufacturing Practices (GMP) [1,2].

  1. Classification in Compliance with ICH Standards:

International Council for Harmonisation (ICH). The ICH guidelines provide a structured framework for identifying, quantifying, and controlling impurities to ensure the safety, efficacy, and quality of pharmaceutical products. Impurities can be classified into categories such as process-related impurities, degradation products, inorganic impurities and residual solvents, among others, and their origin can be traced to factors like raw materials, chemical reactions, environmental conditions, or the drug's degradation over time. Compliance with ICH standards ensures that these impurities are controlled within acceptable limits, promoting global regulatory consistency and protecting patient safety. This classification and control process is crucial for maintaining the integrity of pharmaceutical formulations and ensuring that the products meet stringent regulatory requirements.

particularly when applying analytical techniques such as High-Performance Liquid Chromatography (HPLC), which is instrumental in identifying and quantifying these impurities to ensure the safety and quality of pharmaceutical products [1,2,30].

    1. Organic Impurities

Organic Impurities as per ICH guidelines are defined as unwanted organic compounds that may be present in drug substances and drug products. These impurities typically arise from the manufacturing process or as degradation products during storage [22]. According to ICH Q3A (R2) and Q3B (R2), organic impurities are categorized into process-related impurities and degradation products. Process-related impurities result from raw materials, reagents, or by-products formed during chemical synthesis, while degradation products occur due to environmental factors such as light, heat, or moisture. The guidelines set limits for these impurities to ensure that their levels do not compromise the safety, quality, and efficacy of pharmaceutical products, helping maintain patient safety and regulatory compliance. HPLC is particularly effective in separating and quantifying these organic impurities, using methods such as reversed-phase chromatography and UV-Vis detection to monitor their concentration in complex formulations [1,2].

    1. Inorganic Impurities

Inorganic Impurities as per ICH guidelines refer to non-organic substances, such as metals, salts, and other inorganic contaminants, which may be present in pharmaceutical substances or products. These impurities can originate from raw materials, catalysts, or environmental contamination during the manufacturing process. According to ICH Q3A (R2) and Q3B (R2), inorganic impurities must be identified and quantified to ensure that they do not exceed acceptable levels that could affect the drug's safety and efficacy. The precise quantification of these impurities often requires HPLC coupled with inductively coupled plasma mass spectrometry (ICP-MS) or atomic absorption spectroscopy (AAS), and ion chromatography (IC), which are highly sensitive for detecting trace inorganic substances. These methods play a critical role in ensuring that pharmaceutical products meet regulatory standards for impurity levels and overall quality [1,2].

    1. Residual Solvents

Residual Solvents as per ICH guidelines are organic solvents used during the manufacturing process that may remain in the final pharmaceutical product. These solvents can arise from reactions, purification steps, or storage conditions. According to ICH Q3C guidelines (International Council for Harmonisation ICH), residual solvents must be identified and quantified to ensure they are within safe and acceptable limits to avoid toxicological risks. Common identification techniques include gas chromatography (GC), which is highly effective for volatile solvents, and headspace gas chromatography for the analysis of trace solvents [31]. These methods ensure that residual solvent levels are compliant with ICH standards, safeguarding product quality and patient safety [32].

    1. Mutagenic (genotoxic) impurities:

Mutagenic (genotoxic) impurities are chemical compounds that can cause genetic mutations, leading to potential carcinogenic risks in pharmaceutical products. These impurities may originate from raw materials, synthesis byproducts, degradation reactions, or contamination from packaging materials. Regulatory agencies, including the [30], set strict limits on their permissible levels, often based on the Threshold of Toxicological Concern (TTC). Advanced analytical techniques like LC-MS/MS, GC-MS, and the Ames test are used to detect and quantify these impurities. Controlling and minimizing their presence is crucial to ensuring drug safety, efficacy, and regulatory compliance.

In summary, The identification, quantification, and control of impurities in pharmaceutical products are critical to ensuring their safety, efficacy, and quality. The International Council for Harmonisation (ICH) guidelines provide a structured framework for classifying impurities, including organic impurities, inorganic impurities, residual solvents, and mutagenic (genotoxic) impurities, and setting stringent regulatory limits to minimize risks to patients. High-Performance Liquid Chromatography (HPLC), along with other advanced analytical techniques such as LC-MS/MS, GC-MS, ICP-MS, and AAS, plays a vital role in detecting and quantifying these impurities, ensuring compliance with regulatory standards. By adhering to ICH Q3A, Q3B, Q3C and ICH M7 guidelines, pharmaceutical manufacturers can maintain product integrity, meet global regulatory requirements, and safeguard public health. Effective impurity control strategies not only enhance drug quality but also reinforce trust in pharmaceutical formulations, emphasizing the industry's commitment to patient safety.

  1. Regulatory Approach to Analytical Method Development:

The development of methods in High-Performance Liquid Chromatography (HPLC) is a meticulous and structured process essential for achieving consistent, reproducible, and efficient analytical results, especially in regulated industries like pharmaceuticals. Successful method development hinges on the careful selection and optimization of both the stationary phase and mobile phase, as these factors significantly impact the separation efficiency, resolution, and retention times of analytes [33].

    1. Choice of Stationary Phase:

Stationary Phase selection is a crucial step in method development, as it determines the separation efficiency and resolution of analytes. The choice of column depends on factors such as the chemical properties of the analytes, including polarity, size, and charge, and the nature of the mobile phase. Common stationary phases include C18 (octadecylsilane) for reversed-phase chromatography, C8 for slightly less hydrophobic separations, and phenyl or aminopropyl phases for specific interactions [34]. Column length, diameter, and particle size also affect resolution, pressure, and analysis time, with smaller particles (e.g., 3-5 ?m) providing higher resolution but requiring higher pressure Additionally, column pore size is selected based on the molecular weight of the analytes, with larger pores suitable for larger molecules. The appropriate selection ensures optimal separation, reproducibility, and reliability of the chromatographic method [35].

    1. Refining Mobile Phase Parameters:

Refining mobile phase in HPLC method development is essential for achieving optimal separation and resolution of analytes. The choice of mobile phase composition, including solvent type, polarity, and pH, directly influences the interaction between the analyte and stationary phase [7]. Water and organic solvents (e.g., methanol, acetonitrile) are commonly used, with buffered solutions adjusting pH to enhance selectivity for ionizable compounds. The modification of mobile phase ionic strength and the use of additives such as acidic or basic modifiers can improve peak shape and resolution [36]. Gradient elution can be employed for complex mixtures, allowing better separation of compounds with varying polarities. These refinements ensure reproducibility, sensitivity, and compliance with regulatory standards for pharmaceutical analysis.

    1. Buffer Selection Criteria

Buffer selection in HPLC method development is crucial for optimizing separation efficiency, as it influences the pH, ionic strength, and solubility of analytes. The primary role of buffers is to maintain a stable pH environment, which ensures that analytes are in their appropriate ionic forms for interaction with the stationary phase [7]. Phosphate buffers are commonly used due to their excellent buffering capacity and ability to maintain consistent pH across a wide range. Acetate buffers are often employed for separating weakly acidic or basic compounds, while formic acid and ammonium acetate are utilized in reversed-phase HPLC to improve peak shape and resolution [36]. The buffer's ionic strength can also affect the analyte's migration rate and resolution, and adjustments to the buffer concentration are often made for optimal performance. Buffer selection, therefore, plays a vital role in achieving reproducible, accurate, and efficient separations in pharmaceutical analysis.

    1. pH Fine-Tuning:

pH optimization in HPLC method development is essential for maximizing analyte retention and resolution by ensuring that the analyte exists in its optimal ionic or non-ionic form for interaction with the stationary phase. The pH of the mobile phase directly affects the solubility, ionization, and polarity of the analyte, which can significantly impact the separation [7]. Acidic compounds generally ionize at higher pH values, while basic compounds ionize at lower pH values, thus the pH range must be selected based on the nature of the analytes to control their charge state. Typically, phosphate buffers are used for maintaining a stable pH range of 2-8, suitable for most pharmaceutical analytes, while acetic acid buffers may be preferred for separating weakly basic compounds [36]. pH optimization ensures that analytes have the appropriate retention characteristics for efficient separation and enhances reproducibility in method development. The pH also influences peak shape, resolution, and the overall efficiency of the separation.

    1. Significance of organic modifiers

Organic modifiers play a crucial role in HPLC method development by enhancing the solubility of non-polar analytes and improving peak shape and resolution. Acetonitrile and methanol are commonly used organic solvents in reversed-phase chromatography, where they help reduce the polarity of the mobile phase and control analyte retention [37]. These modifiers can alter the hydrophobic interaction between the analyte and stationary phase, allowing for more efficient separation of complex mixture. The concentration of organic modifiers is carefully optimized to achieve the desired separation, minimizing tailing and improving reproducibility.

    1. Detector selection in chromatographic analysis :

Detector selection in HPLC analysis is crucial for achieving accurate, sensitive, and reliable results, as it directly influences the detection of analytes at low concentrations. Common detectors, such as UV-Vis and fluorescence detectors, are chosen based on the analyte's optical properties, while refractive index detectors are used for compounds without a chromophore [38]. Mass spectrometric (MS) detectors, often coupled with HPLC as LC-MS, provide enhanced sensitivity and selectivity for complex mixtures by identifying analytes based on their mass-to-charge ratio. The detector must be carefully selected to match the analyte's characteristics and ensure high specificity, linearity, and reproducibility for optimal chromatographic analysis.

    1. Thermal control and its influence:

Thermal control in HPLC method development is essential for ensuring consistent retention times, reproducibility, and accurate separation of analytes. Temperature influences the viscosity of the mobile phase, the diffusion rate of analytes, and the interaction between the analytes and stationary phase [39]. Maintaining a stable column temperature helps prevent changes in retention time, ensuring that analytes are consistently separated across different runs. For high-resolution separations, temperature is controlled using column ovens to minimize thermal gradients and maintain chromatographic performance.

In summary, developing HPLC methods for the pharmaceutical industry demands a thorough and scientifically driven strategy to choose and fine-tune system components and operating parameters. Following regulatory standards and conducting comprehensive validation are essential to ensure analytical methods are dependable, precise, and compliant, ultimately protecting the quality and safety of pharmaceutical products.


Table 1: Details of method development various pharmaceutical compounds

 

Name of the product

HPLC Mode

Column Details

Detector

Citation

Brexpiprazole

Reverse Phase

Hypurity C18 (100 mm × 4.6 mm, 5 µm)

UV-Visible

[17]

Amlodipine besylate

Reverse Phase

Core shell C18 (100 mm × 4.6 mm, 2.6 µm)

UV-Visible

[21]

Empagliflozin

Reverse Phase

Poroshell 120 EC-C18, 4.6×100  mm, 4  µm

UV-Visible

[23]

Dabigatran Etexilate Mesylate

Reverse Phase

Poroshell 120 EC -18 (150 mm × 4.6 mm, 2.7µ)

UV-Visible

[24]

Etoricoxib

Reverse Phase

C18 column (4.6 mm x 100 mm, 2.5 µm)

UV-Visible

[26]

Dapagliflozin

Reverse Phase

Xbridge Phenyl C18 column, 250×4.6  mm, 5  ?m

UV-Visible

[40]


Table 2: Details of identification and characterization of  process-related  and degradation impurities in various pharmaceutical compounds.

 

Name of the product

Objective

Findings & Results

Citation

Zuclopenthixol

Study focused to develop a simple and sensitive HPLC method for resolution and estimation process-related impurities of zuclopenthixol and further assessment of forced degradation behaviourof zuclopenthixol.

The analysis involved a comparison of collision-induced dissociation mass spectrometry data between the degradation products and zuclopenthixol Achieved the mechanism of stress degradation in zuclopenthixol. potential structures for six degradation compounds were suggested.

 

[41]

Ambrisentan

Characterization of degradation products of ambrisentan by liquid chromatography-tandem mass spectrometry

 

The degradation behaviour of ambrisentan was studied under various stress conditions. In total, six degradation products were formed and five of them were characterized by liquid chromatography and tandem mass spectrometry and accurate mass measurements.

[42]

Gilteritinib

Investigation entails the characterization of seven degradation products (DPs) formed in different stress conditions of Gilteritinib employing liquid chromatography tandem mass spectrometry (LC-MS/MS)

characterized degradation products include seven compounds (DP 1 to DP 7), each with a distinct chemical structure. study effectively elucidated the fragmentation pathways and characterized the degradation products of gilteritinib

 

[43]

Rifampicin

The study aims to identify and quantify genotoxic impurities (GTIs) in rifampicin, an essential first-line tuberculosis drug, using advanced analytical techniques by using HPLC-HRMS.

the study successfully identified and quantified the genotoxic impurity (25-deacetyl-23-acetyl-rifampicin) The impurity is likely formed due to transesterification under high humidity conditions

[44]

Netarsudil

The aim of this study is to examine resolution, identification, and characterization of forced degradation products of Netarsudil by liquid chromatography-tandem mass spectrometry

Considerable degradation was observed in stress studies, and the degradation products were well resolved from process-related impurities. The characterization of degradation products was performed on the basis of collision-induced dissociation mass spectral data, and the possible structures of the six degradation compounds of Netarsudil were proposed.

[45]


  1. Regulatory Compliance and Validation

Regulatory authorities, such as the FDA, EMA, and ICH, establish strict guidelines for method development, especially in pharmaceutical applications. They require that all HPLC methods undergo thorough validation,Validation assures that the methods comply with regulatory standards and meet quality assurance protocols, guaranteeing dependable results for regulatory review [28,46]

    1. Guidelines for Method Validation in High-Performance Liquid Chromatography

Method validation is a crucial step in ensuring that analytical methods consistently deliver accurate and reliable results, especially for routine analyses in the tightly regulated pharmaceutical industry. Key validation parameters play a vital role in verifying that HPLC methods comply with both scientific principles and regulatory requirements [28,46].

    1. Specificity:

Specificity is the capability of an analytical method to accurately identify and quantify the target analyte without interference from other substances present in the sample matrix. This ensures that the measurement reflects only the compound of interest, unaffected by contributions from impurities or co-existing components. Achieving high specificity in HPLC involves meticulous selection of the stationary and mobile phases, as well as fine-tuning chromatographic conditions to ensure effective separation of the analyte from any potential co-eluting substances [28].

    1. Accuracy:

Accuracy refers to the closeness of the measured value to the true or known value. In HPLC, it is assessed by comparing the method's results against those obtained from a reference value.  A validated method must consistently produce results that closely match the true analyte concentration, ensuring the reliability of the analytical data for precise quantitative analysis [47].

    1. Precision:

Precision measures the reproducibility of an analytical method when performed under consistent conditions. It is evaluated by conducting multiple analyses of the same sample and analyzing the variation in the results. Precision is usually expressed as the relative standard deviation (RSD) and is classified into repeatability (within a single day) and intermediate precision (across different days or analysts). High precision guarantees that the HPLC method yields consistent and reliable results, which is crucial for its application in routine quality control processes [3].

    1. Linearity:

Linearity indicates the direct relationship between the analyte concentration and the detector response within a defined concentration range. A linear relationship enables precise quantification of different analyte levels in the sample. The method should exhibit a correlation coefficient (r?2;) near 1 throughout the expected concentration range, ensuring that the detector’s response is consistent and dependable for accurately measuring both low and high analyte concentrations [46].

    1. Limit of Detection (LOD) and Limit of Quantification (LOQ):

The LOD (Limit of Detection) and LOQ (Limit of Quantification) are key parameters that determine the sensitivity of the HPLC method.

  • The LOD Stands for the lowest concentration of the analyte that can be detected with trustworthiness, though it may not be quantifiable.
  • The LOQ is the smallest concentration at which the analyte can be accurately and precisely quantified with acceptable reliability.
  • These values are crucial for assessing the method's ability to detect trace-level impurities or analytes at low concentrations within complex sample matrices [48].
    1. Robustness

Robustness assesses the method’s capability to maintain consistent performance despite small changes in experimental conditions, such as slight variations in temperature, mobile phase composition, or column batch. A robust method ensures dependable results even when minor deviations from standard procedures occur, making it adaptable to various laboratory environments and sustainable over time [3].


Table 3: Details of method validation parameters and their importance.

 

Validation attribute

Description

The role of HPLC is essential

Citation

Specificity

The method's capability to differentiate the analyte from other components or potential interferences present in the sample.

Assures the measurement of only the target analyte, unaffected by co-eluting compounds.

[46,40]

Accuracy

The extent to which the measured value aligns with the true or Established value.

Verifies that the method delivers accurate and dependable quantitative results for the substance under study

[28, 46,40]

Precision

The consistency of results when the same sample is analyzed repeatedly under identical conditions.

Confirms uniformity of results across various runs, analysts, or laboratory conditions.

[28-49]

Linearity

The relationship between analyte concentration and detector response across a specified range of concentrations.

Certifies the method's ability to accurately quantify different amounts of analyte across a wide concentration range.

[47-46-50]

Limit of Detection (LOD)

The lowest concentration of the analyte that can be detected with  adequate level of certainty, though not necessarily quantified.

Assesses the method's sensitivity in detecting trace amounts of an analyte.

[28-46-48]

Limit of Quantification (LOQ)

The minimum concentration at which the analyte can be consistently quantified with suitable precision and accuracy.

Assures accurate quantification of trace concentrations of the analyte, essential for identifying impurities or low-level drugs.

[28-46-51]

Robustness

The method's capacity to maintain consistent performance despite small changes in experimental conditions, such as slight variations in temperature or mobile phase composition.

Verifies that the method will yield consistent results despite minor deviations from optimal conditions.

[46]


These validation parameters, in accordance with regulatory standards set by organizations such as the FDA, EMA, and ICH, confirm that the HPLC method is suitable for its intended use, accurate, dependable, and in compliance with rigorous quality control standards. Method validation ensures confidence in the analytical data, ensuring the safety, efficacy, and quality of pharmaceutical products

CONCLUSION:

The advancements in High-Performance Liquid Chromatography (HPLC) have significantly improved the detection, identification and quantification of process-related impurities and degradants in pharmaceuticals. Innovations such as Ultra-High-Performance Liquid Chromatography (UHPLC), LC-MS/MS, High-Resolution Mass Spectrometry (HRMS), and advanced detection techniques enhance sensitivity, accuracy, and efficiency. Additionally, and the adoption of greener solvents contribute to better impurity profiling and regulatory compliance. These advancements ensure pharmaceutical safety, efficacy, and quality while streamlining analytical processes. Moving forward, continuous improvements in HPLC technology will further optimize drug analysis, supporting global healthcare and regulatory standards.

REFERENCES

        1. International Council for Harmonisation (ICH). (2006). Q3A(R2): Impurities in new drug substances (ICH harmonised tripartite guideline). ICH. https://www.ich.org
        2. International Council for Harmonisation (ICH). (2006). Q3B(R2): Impurities in new drug products (ICH harmonised tripartite guideline). ICH. https://www.ich.org
        3. Garg, S., & Gupta, M. (2017). Degradation studies on pharmaceutical compounds: Identification and quantification of degradation products. Pharmaceutical Chemistry Journal, 51(4), 305-311. https://doi.org/10.1007/s11094-017-1677-x
        4. Gritti, F., & Guiochon, G. (2011). HPLC detection techniques: Principles, applications, and recent developments. Journal of Chromatography A, 1218(3), 391-406. https://doi.org/10.1016/j.chroma.2010.12.048
        5. Santos, F., & Rodrigues, J. (2018). Application of LC-MS/MS in impurity profiling of drug substances: Current trends and challenges. Analytica Chimica Acta, 1000, 1-10. https://doi.org/10.1016/j.aca.2018.02.019
        6. Ashrani S, Goyal A, Vaishnav R. HPLC Detectors, Their Types and Use: A Review. Organic & Medicinal Chem IJ. 2018; 6(5): 555700. DOI: 10.19080/OMCIJ.2018.06.555700 In place of 6th references
        7. Snyder, L. R., Kirkland, J. J., & Dolan, J. W. (2023). Introduction to modern liquid chromatography (4th ed.). Wiley.
        8. Mehtab, Y., & Sharma, R. (2016). Analysis of pharmaceutical impurities and degradation products: Challenges and techniques. Pharmaceutical Sciences & Research, 8(3), 178-185. https://doi.org/10.1016/j.pscr.2016.02.002
        9. Harris, D. C. (2015). Quantitative Chemical Analysis (9th ed.). W.H. Freeman and Company.
        10. Klee, M., & Hempel, D. (2015). Sample preparation for chromatography: Overview and critical aspects. Journal of Chromatography A, 1409, 43-56. https://doi.org/10.1016/j.chroma.2014.12.024
        11. Hill, J. R., & West, C. (2017). Sample injection techniques for HPLC: An overview. Chromatographic Science Series, 143, 1-28.
        12. Baertschi, S. W., Alsante, K. M., & Reed, R. A. (2016). Pharmaceutical stress testing: Predicting drug degradation (2nd ed.). CRC Press.
        13. Ahuja, S., & Dong, M. W. (2017). Chiral separation techniques: A practical approach. Wiley-VCH.
        14. Swartz, M. E. (2010). Data analysis and acquisition in HPLC. Analytical Chemistry, 82(24), 9802-9812. https://doi.org/10.1021/ac101925w
        15. Chaudhuri, S., & Srivastava, A. (2015). Impurity profiling of pharmaceutical substances: A review. Journal of Pharmaceutical and Biomedical Analysis, 105, 211-224. https://doi.org/10.1016/j.jpba.2014.12.014
        16. International Council for Harmonisation (ICH). (2003). Q1A(R2): Stability testing of new drug substances and products (ICH harmonised tripartite guideline). ICH. https://www.ich.org
        17. Jagdale, A.S., Pendbhaje, N.S., Nirmal, R.V. et al. Development and validation of RP-HPLC method for estimation of Brexpiprazole in its bulk and tablet dosage form using Quality by Design approach. Futur J Pharm Sci 7, 142 (2021).
        18. Emara LH, Emam MF, Taha NF, Raslan HM, El-Ashmawy AA. A Simple and Sensitive HPLC/UV Method for Determination of Meloxicam in Human Plasma for Bioavailability and Bioequivalence Studies. J App Pharm Sci, 2016; 6 (07): 012-019. DOI: 10.7324/JAPS.2016.60702555700
        19. Upadhyay, Y., Sharma, N., Sarma, G. S., & Rawal, R. K. (2015). Application of RP–HPLC method in dissolution testing and statistical evaluation by NASSAM for simultaneous estimation of tertiary combined dosage forms. Journal of Pharmaceutical Analysis, 5(5), 307–315. https://doi.org/10.1016/j.jpha.2014.11.001
        20. Blessy, M., Ruchi, D. P., Prajesh, N. P., & Agrawal, Y. K. (2014). Development of forced degradation and stability indicating studies of drugs—A review. Journal of Pharmaceutical Analysis, 4(3), 159–165. https://doi.org/10.1016/j.jpha.2013.09.003.
        21. Jeelani, S., & Kouznetsova, N. (2023). A new stability-indicating HPLC-UV method for determination of amlodipine besylate and its impurities in drug substance. Heliyon, 9, e19993. https://doi.org/10.1016/j.heliyon.2023.e19993.
        22. Pathak, S., & Mishra, P. (2021). Stability-indicating HPLC-DAD method for the determination of empagliflozin. Future Journal of Pharmaceutical Sciences, 7:181. https://doi.org/10.1186/s43094-021-00329-w.
        23. Pathak, S., & Mishra, P. (2021). Stability-indicating HPLC-DAD method for the determination of empagliflozin. Future Journal of Pharmaceutical Sciences, 7:181. https://doi.org/10.1186/s43094-021-00329-w.
        24. Dare M, Jain R, Pandey A (2015) Method Validation for Stability Indicating Method of Related Substance in Active Pharmaceutical Ingredients Dabigatran Etexilate Mesylate by Reverse Phase Chromatography. J Chromatogr Sep Tech 6: 263.  doi:10.4172/2157-7064.1000263.
        25. Melloni, M. P., & Basso, M. D. (2009). Chiral HPLC in pharmaceutical analysis: From method development to routine analysis. Journal of Pharmaceutical and Biomedical Analysis, 49(1), 42-56. https://doi.org/10.1016/j.jpba.2008.11.007
        26. Patil K, Narkhede S, Nemade M, Rane S, Chaudhari R, Dhobale G, Tare H. A Validated Sensitive Stability Indicating HPLC Method for the Determination of Etoricoxib in Bulk and Formulation. International Journal of Pharmaceutical Quality Assurance. 2023;14(2):352-357. DOI: 10.25258/ijpqa.14.2.19
        27. Ahuja, S., & Dong, M. W. (2005). Handbook of modern pharmaceutical analysis (Vol. 1). Elsevier. https://doi.org/10.1016/B978-012597420-3/50004-7
        28. U.S. Food and Drug Administration (FDA). (2018). Guidance for industry: Analytical procedures and methods validation. FDA Guidelines.
        29. European Medicines Agency (EMA). (2018). Guideline on the limits of genotoxic impurities. EMA/CHMP/QWP/251344/2006. Retrieved from https://www.ema.europa.eu
        30. International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH). (2017). ICH M7(R1): Assessment and control of DNA reactive (mutagenic) impurities in pharmaceuticals to limit potential carcinogenic risk. ICH Guidelines. Retrieved from https://www.ich.org.
        31. Singh, R., & Bansal, A. K. (2010). Identification and quantification of residual solvents in pharmaceutical formulations using gas chromatography. Journal of Pharmaceutical and Biomedical Analysis, 52(5), 693-701. https://doi.org/10.1016/j.jpba.2010.02.004
        32. McAuliffe, C. A., & Bylund, D. A. (2009). Method development for residual solvent analysis using GC-MS. Journal of Chromatography A, 1216(13), 2305-2312. https://doi.org/10.1016/j.chroma.2009.01.016
        33.   International Council for Harmonisation. (2022). ICH Q14: Analytical procedure development.
        34. Williams, A. R., & Lee, D. (2019). Optimization of chromatographic columns for the analysis of complex pharmaceutical compounds. Journal of Pharmaceutical Sciences, 108(3), 679-689. https://doi.org/10.1016/j.xphs.2019.01.017.
        35. Smith, R. M., & Xie, F. (2009). High-performance liquid chromatography: Applications to impurity analysis. Elsevier.
        36. Dong, M. W. (2006). Modern HPLC for practice: A guide for beginners. Wiley-VCH.
        37. Zhang, Y., & Li, X. (2019). Effect of organic modifiers on the retention behaviour of pharmaceutical compounds in HPLC. Journal of Chromatography A, 1600, 185-193. https://doi.org/10.1016/j.chroma.2019.02.017.
        38. Zhang, J., & Lee, K. (2019). Evaluation of detector performance for the analysis of complex pharmaceutical mixtures. In Proceedings of the International Conference on Analytical Chemistry (pp. 45-52). Springer. https://doi.org/10.1007/s00216-019-02075-z
        39. Huang, Y., & Chen, X. (2019). Influence of thermal control on chromatographic separation in HPLC systems. Journal of Chromatography A, 1602, 112-119. https://doi.org/10.1016/j.chroma.2019.03.011
        40. Sunkara, B., & Tummalapalli, N. V. (2023). Development of novel gradient RP-HPLC method for separation of dapagliflozin and its process-related impurities. Future Journal of Pharmaceutical Sciences, 9(107). https://doi.org/10.1186/s43094-023-00556-3.
        41. Sesharamsingh, B., Kumar, J.S., Viswanath, I.V.K. et al. Development of stability-indicating HPLC method for quantification of pharmacopeia impurities of Zuclopenthixol and characterization of its stress degradation products by LCMS/MS. Futur J Pharm Sci 9, 101 (2023). https://doi.org/10.1186/s43094-023-00554-5
        42. Ramisetti, N. R., & Kuntamukkala, R. (2014). LC-MS/MS characterization of forced degradation products of Ambrisentan: Development and validation of a stability-indicating RP-HPLC method. New Journal of Chemistry, 38(7), 3050–3061. https://doi.org/10.1039/C4NJ00075G.
        43.  Katta, S. R., Ramachandra Rao, I. ., Punitha, P., & Siva prasad, T. (2024). LC-MS/MS characterization of stress degradation products of Gilteritinib and establishment of HPLC method for analysis of process related impurities of Gilteritinib. Journal of Applied Pharmaceutical Research, 12(2), 109-123. https://doi.org/10.18231/j.joapr.2024.12.2.109.123
        44.  Jiang, Y., Zhou, F., Yao, H., Wang, H., Wu, H., Huang, Y., & Gu, M. (2025). Identification and quantitative analysis of genotoxic impurities in rifampicin: Development and validation of a targeted LC-MS/MS method for 1-amino-4-methylpiperazine. Journal of Pharmaceutical and Biomedical Analysis, 252, 116459. https://doi.org/10.1016/j.jpba.2024.116459
        45. ANNA VR, KUMAR BS, HARISH J, TATAVARTI BK, ESWARLAL T. Characterization of Forced Degradation Products of Netarsudil: Optimization and Validation of a Stability-Indicating RP-HPLC Method for Simultaneous Quantification of Process-Related Impurities. Turk J Pharm Sci. 2024 Jun;21(3):224-233. doi:10.4274/tjps.galenos.2023.99148.
        46. International Council for Harmonisation. (2022). ICH Q2(R2): Validation of analytical procedures. https://www.ich.or
        47. Araujo, M. C. U., & de Souza, A. R. (2016). HPLC method validation and applications in pharmaceutical analysis. Journal of Pharmaceutical and Biomedical Analysis, 118, 46-58. https://doi.org/10.1016/j.jpba.2015.10.015
        48. Johnson, R., & Taylor, A. (2019). Approaches to determine LOD and LOQ in complex pharmaceutical analysis. In Proceedings of the International Conference on Analytical Chemistry (pp. 67-73). Springer. https://doi.org/10.1007/s00357-019-0047-8.
        49. international Council for Harmonisation (ICH). (2009). Guideline for residual solvents Q3C(R6). Retrieved from https://www.ich.org
        50. Zhang, Q., & Liu, J. (2017). Evaluation of precision and linearity in high-performance liquid chromatography for pharmaceutical analysis. Journal of Pharmaceutical and Biomedical
        51. FDA (2017). Guidance for Industry: Bioanalytical Method Validation. U.S. Food and Drug Administration.
        52. Géhin C, Holman SW. (2021). Advances in high-resolution mass spectrometry applied to pharmaceuticals in 2020: a whole new age of information. Anal Sci Adv; 2:142–156. https://doi.org/10.1002/ansa.202000149..

Reference

  1. International Council for Harmonisation (ICH). (2006). Q3A(R2): Impurities in new drug substances (ICH harmonised tripartite guideline). ICH. https://www.ich.org
  2. International Council for Harmonisation (ICH). (2006). Q3B(R2): Impurities in new drug products (ICH harmonised tripartite guideline). ICH. https://www.ich.org
  3. Garg, S., & Gupta, M. (2017). Degradation studies on pharmaceutical compounds: Identification and quantification of degradation products. Pharmaceutical Chemistry Journal, 51(4), 305-311. https://doi.org/10.1007/s11094-017-1677-x
  4. Gritti, F., & Guiochon, G. (2011). HPLC detection techniques: Principles, applications, and recent developments. Journal of Chromatography A, 1218(3), 391-406. https://doi.org/10.1016/j.chroma.2010.12.048
  5. Santos, F., & Rodrigues, J. (2018). Application of LC-MS/MS in impurity profiling of drug substances: Current trends and challenges. Analytica Chimica Acta, 1000, 1-10. https://doi.org/10.1016/j.aca.2018.02.019
  6. Ashrani S, Goyal A, Vaishnav R. HPLC Detectors, Their Types and Use: A Review. Organic & Medicinal Chem IJ. 2018; 6(5): 555700. DOI: 10.19080/OMCIJ.2018.06.555700 In place of 6th references
  7. Snyder, L. R., Kirkland, J. J., & Dolan, J. W. (2023). Introduction to modern liquid chromatography (4th ed.). Wiley.
  8. Mehtab, Y., & Sharma, R. (2016). Analysis of pharmaceutical impurities and degradation products: Challenges and techniques. Pharmaceutical Sciences & Research, 8(3), 178-185. https://doi.org/10.1016/j.pscr.2016.02.002
  9. Harris, D. C. (2015). Quantitative Chemical Analysis (9th ed.). W.H. Freeman and Company.
  10. Klee, M., & Hempel, D. (2015). Sample preparation for chromatography: Overview and critical aspects. Journal of Chromatography A, 1409, 43-56. https://doi.org/10.1016/j.chroma.2014.12.024
  11. Hill, J. R., & West, C. (2017). Sample injection techniques for HPLC: An overview. Chromatographic Science Series, 143, 1-28.
  12. Baertschi, S. W., Alsante, K. M., & Reed, R. A. (2016). Pharmaceutical stress testing: Predicting drug degradation (2nd ed.). CRC Press.
  13. Ahuja, S., & Dong, M. W. (2017). Chiral separation techniques: A practical approach. Wiley-VCH.
  14. Swartz, M. E. (2010). Data analysis and acquisition in HPLC. Analytical Chemistry, 82(24), 9802-9812. https://doi.org/10.1021/ac101925w
  15. Chaudhuri, S., & Srivastava, A. (2015). Impurity profiling of pharmaceutical substances: A review. Journal of Pharmaceutical and Biomedical Analysis, 105, 211-224. https://doi.org/10.1016/j.jpba.2014.12.014
  16. International Council for Harmonisation (ICH). (2003). Q1A(R2): Stability testing of new drug substances and products (ICH harmonised tripartite guideline). ICH. https://www.ich.org
  17. Jagdale, A.S., Pendbhaje, N.S., Nirmal, R.V. et al. Development and validation of RP-HPLC method for estimation of Brexpiprazole in its bulk and tablet dosage form using Quality by Design approach. Futur J Pharm Sci 7, 142 (2021).
  18. Emara LH, Emam MF, Taha NF, Raslan HM, El-Ashmawy AA. A Simple and Sensitive HPLC/UV Method for Determination of Meloxicam in Human Plasma for Bioavailability and Bioequivalence Studies. J App Pharm Sci, 2016; 6 (07): 012-019. DOI: 10.7324/JAPS.2016.60702555700
  19. Upadhyay, Y., Sharma, N., Sarma, G. S., & Rawal, R. K. (2015). Application of RP–HPLC method in dissolution testing and statistical evaluation by NASSAM for simultaneous estimation of tertiary combined dosage forms. Journal of Pharmaceutical Analysis, 5(5), 307–315. https://doi.org/10.1016/j.jpha.2014.11.001
  20. Blessy, M., Ruchi, D. P., Prajesh, N. P., & Agrawal, Y. K. (2014). Development of forced degradation and stability indicating studies of drugs—A review. Journal of Pharmaceutical Analysis, 4(3), 159–165. https://doi.org/10.1016/j.jpha.2013.09.003.
  21. Jeelani, S., & Kouznetsova, N. (2023). A new stability-indicating HPLC-UV method for determination of amlodipine besylate and its impurities in drug substance. Heliyon, 9, e19993. https://doi.org/10.1016/j.heliyon.2023.e19993.
  22. Pathak, S., & Mishra, P. (2021). Stability-indicating HPLC-DAD method for the determination of empagliflozin. Future Journal of Pharmaceutical Sciences, 7:181. https://doi.org/10.1186/s43094-021-00329-w.
  23. Pathak, S., & Mishra, P. (2021). Stability-indicating HPLC-DAD method for the determination of empagliflozin. Future Journal of Pharmaceutical Sciences, 7:181. https://doi.org/10.1186/s43094-021-00329-w.
  24. Dare M, Jain R, Pandey A (2015) Method Validation for Stability Indicating Method of Related Substance in Active Pharmaceutical Ingredients Dabigatran Etexilate Mesylate by Reverse Phase Chromatography. J Chromatogr Sep Tech 6: 263.  doi:10.4172/2157-7064.1000263.
  25. Melloni, M. P., & Basso, M. D. (2009). Chiral HPLC in pharmaceutical analysis: From method development to routine analysis. Journal of Pharmaceutical and Biomedical Analysis, 49(1), 42-56. https://doi.org/10.1016/j.jpba.2008.11.007
  26. Patil K, Narkhede S, Nemade M, Rane S, Chaudhari R, Dhobale G, Tare H. A Validated Sensitive Stability Indicating HPLC Method for the Determination of Etoricoxib in Bulk and Formulation. International Journal of Pharmaceutical Quality Assurance. 2023;14(2):352-357. DOI: 10.25258/ijpqa.14.2.19
  27. Ahuja, S., & Dong, M. W. (2005). Handbook of modern pharmaceutical analysis (Vol. 1). Elsevier. https://doi.org/10.1016/B978-012597420-3/50004-7
  28. U.S. Food and Drug Administration (FDA). (2018). Guidance for industry: Analytical procedures and methods validation. FDA Guidelines.
  29. European Medicines Agency (EMA). (2018). Guideline on the limits of genotoxic impurities. EMA/CHMP/QWP/251344/2006. Retrieved from https://www.ema.europa.eu
  30. International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH). (2017). ICH M7(R1): Assessment and control of DNA reactive (mutagenic) impurities in pharmaceuticals to limit potential carcinogenic risk. ICH Guidelines. Retrieved from https://www.ich.org.
  31. Singh, R., & Bansal, A. K. (2010). Identification and quantification of residual solvents in pharmaceutical formulations using gas chromatography. Journal of Pharmaceutical and Biomedical Analysis, 52(5), 693-701. https://doi.org/10.1016/j.jpba.2010.02.004
  32. McAuliffe, C. A., & Bylund, D. A. (2009). Method development for residual solvent analysis using GC-MS. Journal of Chromatography A, 1216(13), 2305-2312. https://doi.org/10.1016/j.chroma.2009.01.016
  33.   International Council for Harmonisation. (2022). ICH Q14: Analytical procedure development.
  34. Williams, A. R., & Lee, D. (2019). Optimization of chromatographic columns for the analysis of complex pharmaceutical compounds. Journal of Pharmaceutical Sciences, 108(3), 679-689. https://doi.org/10.1016/j.xphs.2019.01.017.
  35. Smith, R. M., & Xie, F. (2009). High-performance liquid chromatography: Applications to impurity analysis. Elsevier.
  36. Dong, M. W. (2006). Modern HPLC for practice: A guide for beginners. Wiley-VCH.
  37. Zhang, Y., & Li, X. (2019). Effect of organic modifiers on the retention behaviour of pharmaceutical compounds in HPLC. Journal of Chromatography A, 1600, 185-193. https://doi.org/10.1016/j.chroma.2019.02.017.
  38. Zhang, J., & Lee, K. (2019). Evaluation of detector performance for the analysis of complex pharmaceutical mixtures. In Proceedings of the International Conference on Analytical Chemistry (pp. 45-52). Springer. https://doi.org/10.1007/s00216-019-02075-z
  39. Huang, Y., & Chen, X. (2019). Influence of thermal control on chromatographic separation in HPLC systems. Journal of Chromatography A, 1602, 112-119. https://doi.org/10.1016/j.chroma.2019.03.011
  40. Sunkara, B., & Tummalapalli, N. V. (2023). Development of novel gradient RP-HPLC method for separation of dapagliflozin and its process-related impurities. Future Journal of Pharmaceutical Sciences, 9(107). https://doi.org/10.1186/s43094-023-00556-3.
  41. Sesharamsingh, B., Kumar, J.S., Viswanath, I.V.K. et al. Development of stability-indicating HPLC method for quantification of pharmacopeia impurities of Zuclopenthixol and characterization of its stress degradation products by LCMS/MS. Futur J Pharm Sci 9, 101 (2023). https://doi.org/10.1186/s43094-023-00554-5
  42. Ramisetti, N. R., & Kuntamukkala, R. (2014). LC-MS/MS characterization of forced degradation products of Ambrisentan: Development and validation of a stability-indicating RP-HPLC method. New Journal of Chemistry, 38(7), 3050–3061. https://doi.org/10.1039/C4NJ00075G.
  43.  Katta, S. R., Ramachandra Rao, I. ., Punitha, P., & Siva prasad, T. (2024). LC-MS/MS characterization of stress degradation products of Gilteritinib and establishment of HPLC method for analysis of process related impurities of Gilteritinib. Journal of Applied Pharmaceutical Research, 12(2), 109-123. https://doi.org/10.18231/j.joapr.2024.12.2.109.123
  44.  Jiang, Y., Zhou, F., Yao, H., Wang, H., Wu, H., Huang, Y., & Gu, M. (2025). Identification and quantitative analysis of genotoxic impurities in rifampicin: Development and validation of a targeted LC-MS/MS method for 1-amino-4-methylpiperazine. Journal of Pharmaceutical and Biomedical Analysis, 252, 116459. https://doi.org/10.1016/j.jpba.2024.116459
  45. ANNA VR, KUMAR BS, HARISH J, TATAVARTI BK, ESWARLAL T. Characterization of Forced Degradation Products of Netarsudil: Optimization and Validation of a Stability-Indicating RP-HPLC Method for Simultaneous Quantification of Process-Related Impurities. Turk J Pharm Sci. 2024 Jun;21(3):224-233. doi:10.4274/tjps.galenos.2023.99148.
  46. International Council for Harmonisation. (2022). ICH Q2(R2): Validation of analytical procedures. https://www.ich.or
  47. Araujo, M. C. U., & de Souza, A. R. (2016). HPLC method validation and applications in pharmaceutical analysis. Journal of Pharmaceutical and Biomedical Analysis, 118, 46-58. https://doi.org/10.1016/j.jpba.2015.10.015
  48. Johnson, R., & Taylor, A. (2019). Approaches to determine LOD and LOQ in complex pharmaceutical analysis. In Proceedings of the International Conference on Analytical Chemistry (pp. 67-73). Springer. https://doi.org/10.1007/s00357-019-0047-8.
  49. international Council for Harmonisation (ICH). (2009). Guideline for residual solvents Q3C(R6). Retrieved from https://www.ich.org
  50. Zhang, Q., & Liu, J. (2017). Evaluation of precision and linearity in high-performance liquid chromatography for pharmaceutical analysis. Journal of Pharmaceutical and Biomedical
  51. FDA (2017). Guidance for Industry: Bioanalytical Method Validation. U.S. Food and Drug Administration.
  52. Géhin C, Holman SW. (2021). Advances in high-resolution mass spectrometry applied to pharmaceuticals in 2020: a whole new age of information. Anal Sci Adv; 2:142–156. https://doi.org/10.1002/ansa.202000149..

Photo
P. Chandrashekhar Reddy
Corresponding author

Department of Chemistry, Anurag University, Hyderabad, India. Analytical Research and Development, Aurore Life Sciences Private Limited, Hyderabad, India

Photo
G. Sampath Kumar Reddy
Co-author

Analytical Research and Development, Aurore Life Sciences Private Limited, Hyderabad, India

Photo
B. Jainendra Kumar
Co-author

Department of Chemistry, Anurag University, Hyderabad, India

P. Chandrashekhar Reddy*, G. Sampath Kumar Reddy, B. Jainendra Kumar, Advancements in HPLC Techniques for Detecting and identification of Process-Related and Degradants Impurities in Pharmaceutical Compounds, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 2, 527-546. https://doi.org/10.5281/zenodo.14834264

More related articles
Method Development, Validation and Forced Degradat...
S. Yasotha, Dr. R. Manivannan, Dr. D. Kamalakannan, Vanmathi. R. ...
An Effective Quantitative Gas Chromatography-Head ...
Radharavi K , P.D. Gokulana, K. L. Senthil Kumara, R. Ramyaa, M. ...
Comprehensive Insights into High-Performance Liqui...
M. Manivardhan Reddy, G. Sampath Kumar Reddy, K. Ramadevi, ...
Simultaneous Estimation of Dapagliflozin and Saxagliptin: Analytical Method Deve...
Dr. Vivek B. Panchabhai, Komal Chavan, Moein S Attar, Dr. Ram S. Sakhare, ...
Stability Indicating Rp-Hplc Method Development and Validation for Estimation of...
Vishal Chudasama, Megha Patel, Kiran Suthar, Bharat Rajpurohit, ...
An Effective Quantitative Gas Chromatography-Head Space Method Development And V...
Radharavi K , P.D. Gokulana, K. L. Senthil Kumara, R. Ramyaa, M. Karthik, ...
Related Articles
Stability Indicating Rp-Hplc Method Development and Validation for Estimation of...
Vishal Chudasama, Megha Patel, Kiran Suthar, Bharat Rajpurohit, ...
Extensive Review of Gas Chromatography in Pharmaceutical Applications: Emphasizi...
Deshoju Srinu, G. Sampath Kumar Reddy, B. Jainendra Kumar, ...
Method Development, Validation and Forced Degradation Study of Lasmiditan by HPL...
S. Yasotha, Dr. R. Manivannan, Dr. D. Kamalakannan, Vanmathi. R. L., Gowtham T., Praveen Kumar S., ...
More related articles
Method Development, Validation and Forced Degradation Study of Lasmiditan by HPL...
S. Yasotha, Dr. R. Manivannan, Dr. D. Kamalakannan, Vanmathi. R. L., Gowtham T., Praveen Kumar S., ...
An Effective Quantitative Gas Chromatography-Head Space Method Development And V...
Radharavi K , P.D. Gokulana, K. L. Senthil Kumara, R. Ramyaa, M. Karthik, ...
Comprehensive Insights into High-Performance Liquid Chromatography for Pharmaceu...
M. Manivardhan Reddy, G. Sampath Kumar Reddy, K. Ramadevi, ...
Method Development, Validation and Forced Degradation Study of Lasmiditan by HPL...
S. Yasotha, Dr. R. Manivannan, Dr. D. Kamalakannan, Vanmathi. R. L., Gowtham T., Praveen Kumar S., ...
An Effective Quantitative Gas Chromatography-Head Space Method Development And V...
Radharavi K , P.D. Gokulana, K. L. Senthil Kumara, R. Ramyaa, M. Karthik, ...
Comprehensive Insights into High-Performance Liquid Chromatography for Pharmaceu...
M. Manivardhan Reddy, G. Sampath Kumar Reddy, K. Ramadevi, ...