This article provides a comprehensive overview of the principles, applications, and validation of green chromatographic techniques for impurity profiling in pharmaceuticals.
This article provides a comprehensive overview of the principles, applications, and validation of green chromatographic techniques for impurity profiling in pharmaceuticals. Tailored for researchers and drug development professionals, it explores foundational concepts like Green Analytical Chemistry (GAC) principles and regulatory guidelines. The content details methodological advances including Ultra-High-Performance Liquid Chromatography (UHPLC), Supercritical Fluid Chromatography (SFC), and solvent-reduction strategies. It also addresses practical challenges in implementation, optimization techniques, and the use of modern greenness assessment tools like AGREE and GAPI for method validation and comparison, offering a complete guide for adopting sustainable practices in quality control.
Green Analytical Chemistry (GAC) has emerged as a fundamental discipline within green chemistry, focusing specifically on making laboratory analytical practices more environmentally benign. While green chemistry principles initially targeted industrial-scale processes, GAC provides a tailored framework for analytical chemists seeking to reduce the environmental impact of their methodologies. The core challenge in modern analytical chemistry lies in reaching an optimal compromise between obtaining high-quality results and improving the environmental friendliness of analytical methods. The 12 principles of GAC serve as essential guidelines to meet this challenge, offering a systematic approach to greening analytical practices while maintaining analytical performance.
The development of GAC principles became necessary because many of the original 12 principles of green chemistry, designed primarily for synthetic chemistry, proved inadequate for direct application in analytical contexts. For instance, the principle of atom economy finds limited relevance in analytical chemistry, where the goal is measurement rather than synthesis. This recognition led to the development of a revised set of principles specifically addressing the unique requirements and challenges of analytical chemistry, creating a comprehensive framework that guides the development of sustainable analytical methods.
The 12 principles of GAC incorporate four original green chemistry principles while introducing eight new principles specifically relevant to analytical practices. These principles provide a complete framework for assessing and improving the environmental footprint of analytical methodologies. The table below summarizes these twelve principles and their primary applications in analytical chemistry.
Table 1: The Twelve Principles of Green Analytical Chemistry
| Principle Number | Principle Name | Core Concept | Key Applications in Analytical Chemistry |
|---|---|---|---|
| 1 | Direct Analytical Techniques | Apply direct techniques to avoid sample treatment | Spectroscopic methods, in-line sensors |
| 2 | Minimal Sample Size | Use minimal sample size and number of samples | Micro-extraction, miniaturized systems |
| 3 | In Situ Measurements | Perform measurements at the sample location | Field-deployable instruments, sensors |
| 4 | Process Integration | Integrate analytical processes and operations | Automated systems, on-line analysis |
| 5 | Automation and Miniaturization | Select automated and miniaturized methods | Lab-on-a-chip, microfluidic devices |
| 6 | Derivatization Avoidance | Avoid derivatization steps | Direct analysis methods |
| 7 | Waste Management | Avoid waste generation and manage properly | Solvent-free methods, waste treatment |
| 8 | Multi-Analyte Assays | Perform multi-analyte determinations | Comprehensive chromatography, multi-element analysis |
| 9 | Energy Minimization | Minimize energy consumption | Energy-efficient instruments, ambient analysis |
| 10 | Green Reagents | Use reagents from renewable sources | Bio-based solvents, green reagents |
| 11 | Operator Safety | Increase operator safety | Closed systems, reduced toxicity |
| 12 | Waste Degradability | Avoid toxic reagents and choose biodegradable alternatives | Green solvents, biodegradable reagents |
These principles collectively address the three key pillars of sustainability: environmental protection, economic feasibility, and social responsibility. Principles 1-4 and 8-9 primarily focus on resource efficiency, principles 6-7 and 10-12 target hazard reduction, while principle 5 bridges both efficiency and safety concerns. The successful implementation of these principles requires a holistic approach that considers the entire analytical process from sample collection to final waste disposal.
The implementation of GAC principles in chromatographic method development for impurity profiling requires strategic planning throughout the method design process. The fundamental goals include eliminating or reducing the use of hazardous chemical substances, minimizing energy consumption, implementing proper waste management, and increasing operator safety. These objectives align directly with multiple GAC principles, particularly principles 7 (waste management), 9 (energy minimization), 10 (green reagents), and 11 (operator safety).
Recent advancements in chromatographic sciences have demonstrated that greener methodologies can achieve performance parameters equivalent to or even superior to conventional methods. The key lies in intelligent method design that incorporates green considerations from the initial development phase rather than as an afterthought. Successful implementation requires careful selection of solvents, optimization of chromatographic conditions, and integration of separation steps to reduce overall resource consumption.
Table 2: Green Strategy Implementation in Chromatographic Methods
| GAC Principle | Conventional Approach | Green Alternative | Environmental Benefit |
|---|---|---|---|
| Direct Analysis (1) | Extensive sample preparation | Minimal or no sample preparation | Reduced solvent consumption |
| Minimal Sample Size (2) | Large sample volumes | Micro-extraction techniques | Less waste generation |
| Derivatization Avoidance (6) | Chemical derivatization | Direct detection methods | Elimination of derivatization reagents |
| Green Reagents (10) | Acetonitrile, methanol | Ethanol, bio-based solvents | Reduced toxicity, renewable sources |
| Energy Minimization (9) | Standard HPLC conditions | Elevated temperatures, reduced run times | Lower energy consumption |
| Multi-analyte Assays (8) | Separate methods for each analyte | Comprehensive single-run methods | Reduced solvent and energy use per analyte |
Recent research demonstrates the successful application of GAC principles in pharmaceutical impurity profiling. A green HPLC strategy for the simultaneous quantification of carvedilol and hydrochlorothiazide alongside separation of potential impurities exemplifies this approach. The method employed ethanol as a greener alternative to acetonitrile or methanol, aligning with principle 10 (green reagents). The gradient elution program with a flow rate of 1.0 mL/min and total run time of 7 minutes addressed principles 4 (process integration) and 9 (energy minimization) by providing efficient separation of multiple compounds in a single analytical run [1].
In another application, researchers developed green and white-assessed chromatographic methods for ondansetron purity testing. The LC-MS/MS method achieved rapid analysis with a total run time of 2 minutes using a methanol and 0.1% formic acid mobile phase, significantly reducing solvent consumption compared to conventional methods. The simultaneous determination of ondansetron and four impurities in pharmaceutical formulations demonstrated principle 8 (multi-analyte assays), while the reduced analysis time and solvent volume addressed principles 7 (waste management) and 9 (energy minimization) [2].
A comparative study of HPTLC-densitometry and RP-HPLC methods for mupirocin quantification in binary mixtures further illustrates GAC implementation. The HPTLC method utilized a mobile phase of toluene:chloroform:ethanol (5:4:2, by volume), offering advantages in minimal solvent consumption per sample and lower energy requirements during development. The method successfully separated the cited drugs and their impurities while demonstrating reduced environmental impact through lower reagent consumption and waste generation [3].
Method Title: Gradient HPLC with Eco-Friendly Mobile Phase for Simultaneous API and Impurity Determination
Principle: This method applies principles of safer solvents (GAC principle 10), waste reduction (principle 7), and energy efficiency (principle 9) through optimized chromatographic conditions.
Materials and Reagents:
Instrumentation:
Mobile Phase Preparation:
Standard Solution Preparation:
Chromatographic Conditions:
System Suitability:
Sample Analysis:
Method Validation:
Method Title: HPTLC with Green Mobile Phase for Pharmaceutical Impurity Profiling
Principle: This method emphasizes minimal solvent consumption (GAC principle 2), waste reduction (principle 7), and operator safety (principle 11) through miniaturized separation technique.
Materials and Reagents:
Instrumentation:
Standard Solution Preparation:
Sample Application:
Chromatographic Development:
Detection and Scanning:
Quantification:
Validation Parameters:
GAC Implementation Workflow - This diagram illustrates the systematic approach to implementing Green Analytical Chemistry principles throughout method development.
Green Chromatography Development - This workflow details the specific considerations for developing environmentally friendly chromatographic methods.
The selection of appropriate reagents and materials is crucial for implementing GAC principles in analytical methodologies. The following table details key research reagent solutions and their functions in green chromatographic methods for impurity profiling.
Table 3: Essential Research Reagents for Green Chromatographic Methods
| Reagent/Material | Function in Analysis | Green Alternative | Environmental Advantage |
|---|---|---|---|
| Acetonitrile | HPLC mobile phase | Ethanol | Less toxic, renewable source |
| Methanol | HPLC mobile phase, solvent | Ethanol or water-rich phases | Reduced toxicity, biodegradable |
| Chlorinated solvents | Sample preparation, TLC | Ethyl acetate, methanol | Ozone layer protection, safer |
| Phosphate buffers | Mobile phase additive | Volatile buffers (formate/acetate) | Easier disposal, MS compatibility |
| Derivatization reagents | Analyte modification | Direct detection methods | Waste reduction, simpler procedures |
| Traditional columns | Separation | Core-shell, UHPLC columns | Faster analysis, solvent savings |
| Plastic consumables | Sample handling | Recycled materials, minimal use | Reduced plastic waste |
The transition to greener reagents in analytical chemistry represents a significant advancement in reducing the environmental footprint of pharmaceutical analysis. Ethanol, in particular, has emerged as a versatile alternative to traditional solvents like acetonitrile and methanol in reversed-phase chromatography. With proper method optimization, ethanol can provide comparable or superior separation efficiency while offering advantages in terms of toxicity, biodegradability, and renewable sourcing. Similarly, the development of water-rich mobile phases and the use of volatile buffer systems contribute to reduced environmental impact without compromising analytical performance.
The evaluation of analytical method greenness requires specialized metrics and assessment tools. Several established frameworks enable quantitative and qualitative measurement of environmental impact, providing researchers with standardized approaches to benchmark and improve their methodologies.
The National Environmental Methods Index (NEMI) provides a simple pictogram-based assessment system that indicates whether a method meets basic green criteria. The Analytical Eco-Scale assigns penalty points based on environmental, health, and safety parameters, with higher scores indicating greener methods. The Green Analytical Procedure Index (GAPI) offers a more comprehensive evaluation through a colored pictogram that assesses environmental impact across multiple stages of the analytical process. The Analytical GREEnness Metric Approach (AGREE) provides a user-friendly software-based assessment that generates a unified greenness score.
Recent advancements in assessment methodologies include the incorporation of white analytical chemistry concepts, which expand green principles to include methodological, practical, and economic perspectives. The Carbon Footprint Reduction Index represents another emerging tool that specifically quantifies CO2 emissions associated with analytical methods, providing a direct measure of climate impact. These assessment tools collectively enable researchers to make informed decisions about method selection and optimization, driving continuous improvement in the environmental sustainability of analytical practices.
The Twelve Principles of Green Analytical Chemistry provide a comprehensive framework for developing sustainable analytical methods that maintain high performance standards while minimizing environmental impact. The application of these principles to chromatographic methods for impurity profiling demonstrates that environmental considerations can be integrated without compromising analytical quality. Through strategic solvent selection, method optimization, and miniaturization approaches, analytical chemists can significantly reduce the environmental footprint of pharmaceutical analysis.
The continued evolution of GAC principles and assessment metrics will further enable the development of analytical methods that align with broader sustainability goals. As the field advances, the integration of green chemistry principles from initial method development through final implementation represents the future of responsible analytical science. The frameworks, protocols, and strategies outlined in this article provide practical pathways for researchers to incorporate GAC principles into their impurity profiling methodologies, contributing to more sustainable pharmaceutical development and quality control practices.
Impurity profiling is a critical component of pharmaceutical quality control and assurance, directly impacting drug safety, efficacy, and stability [4]. Global regulatory agencies, including the International Council for Harmonisation (ICH) and United States Pharmacopoeia (USP), have established stringent guidelines requiring pharmaceutical companies to identify, quantify, and control impurities in drug substances and products [5] [4]. These guidelines provide comprehensive frameworks for impurity classification, identification thresholds, qualification requirements, and analytical procedure development, ensuring consistent regulatory expectations across regions [4].
The ICH Q3A-Q3D series outlines specific requirements for impurities in new drug substances (Q3A), drug products (Q3B), residual solvents (Q3C), and elemental impurities (Q3D) [4]. Similarly, the USP provides monographs and general chapters detailing impurity testing procedures and acceptance criteria [5]. Compliance with these guidelines is mandatory for regulatory submissions to agencies like the FDA and EMA, making robust impurity control strategies essential for successful drug approval and market authorization [5] [4].
Table 1: Key ICH Guidelines for Impurity Control
| Guideline | Title | Scope and Focus |
|---|---|---|
| ICH Q3A(R2) | Impurities in New Drug Substances | Classification, identification, qualification of organic impurities in APIs |
| ICH Q3B(R2) | Impurities in New Drug Products | Thresholds and qualification of degradation products in formulated drugs |
| ICH Q3C | Impurities: Guideline for Residual Solvents | Classification and limits for residual solvents (Class 1-3) |
| ICH Q3D | Guideline for Elemental Impurities | Control of potentially toxic elemental impurities |
According to ICH and USP frameworks, impurities in pharmaceutical substances can be broadly classified into multiple categories based on their origin and chemical nature [4]. Organic impurities may arise during synthesis, manufacturing, or storage and include starting materials, by-products, intermediates, and degradation products [4]. Inorganic impurities typically result from manufacturing processes and may include reagents, ligands, catalysts, heavy metals, or other materials [4]. Residual solvents are organic or inorganic volatile chemicals used during manufacturing processes that cannot be completely removed by practical means [4].
The sources of impurities are equally diverse. Process-related impurities originate from the drug substance synthesis or drug product manufacturing process, while degradation products form during storage through decomposition reactions influenced by environmental factors like temperature, light, pH, or humidity [4]. Recent recalls due to nitrosamine impurities in various drug products have highlighted the critical importance of comprehensive impurity control strategies throughout the product lifecycle [5].
The principles of Green Analytical Chemistry (GAC) have gained significant traction in pharmaceutical impurity profiling, focusing on minimizing environmental impact while maintaining analytical performance [4] [3] [6]. These approaches reduce solvent consumption, waste generation, and energy usage while employing safer solvents and reagents [4].
Green Liquid Chromatography (GLC) utilizes several strategies to enhance sustainability. Ultra-High Performance Liquid Chromatography (UHPLC) achieves 80% reduction in solvent usage compared to conventional HPLC while maintaining or improving separation efficiency through smaller particle columns and higher operating pressures [4]. Narrow-bore columns (â¤2.1 mm internal diameter) can reduce mobile phase consumption by up to 90% compared to standard 4.6 mm columns without compromising chromatographic performance [4]. Elevated temperature liquid chromatography reduces mobile phase viscosity, enabling faster separations with lower solvent consumption [4]. Additionally, replacing acetonitrile with ethanol or methanol in mobile phases or employing aqueous mobile phases without organic solvents significantly improves method greenness [4].
Supercritical Fluid Chromatography (SFC) utilizes supercritical COâ as the primary mobile phase component, dramatically reducing organic solvent consumption while providing excellent selectivity for challenging separations [4]. SFC is particularly valuable for chiral separations and analysis of non-polar to moderately polar compounds.
High-Performance Thin-Layer Chromatography (HPTLC) offers an alternative green approach with minimal solvent consumption and the ability to analyze multiple samples simultaneously [3] [6]. The technique is simple, cost-effective, and does not require expensive instrumentation, making it accessible for routine quality control testing [6].
Table 2: Comparison of Green Chromatographic Methods for Impurity Profiling
| Method | Key Green Features | Typical Applications | Separation Efficiency |
|---|---|---|---|
| UHPLC | 80% solvent reduction vs. HPLC; faster analysis | Complex pharmaceutical mixtures; stability testing | High |
| Narrow-bore HPLC | Up to 90% mobile phase reduction | Impurity quantification; method development | High |
| SFC | Supercritical COâ replaces organic solvents; minimal waste | Chiral separations; non-polar to moderate polar compounds | Moderate to High |
| HPTLC | Minimal solvent use; multiple parallel samples; no expensive instrumentation | Routine QC; impurity screening; stability testing | Moderate |
Recent research demonstrates the application of green principles to impurity profiling of challenging pharmaceutical compounds. For ondansetron (OND), an antiemetic drug, researchers developed complementary LC-MS/MS and HPTLC-densitometry methods for determining OND along with four potentially mutagenic impurities (D, E, F, and G) [6].
The LC-MS/MS method utilized an Inetsil C18 column (4.6Ã50 mm, 5µm) with an isocratic green mobile phase of methanol:0.1% formic acid in water (70:30, v/v) at a flow rate of 1 mL/min and total run time of 2 minutes [6]. Multiple reaction monitoring (MRM) with electrospray ionization in positive ion mode provided high sensitivity and specificity, enabling quantification at nanoscale levels with minimal solvent consumption [6].
The HPTLC-densitometry method employed silica gel 60 F254 plates with a green mobile phase of ethanol:toluene:methanol:formic acid (5:4:1:0.5, by volume) [6]. This approach consumed minimal solvents while effectively separating OND from its impurity D, providing a cost-effective alternative for quality control laboratories [6].
Both methods were validated according to ICH guidelines and successfully applied to pharmaceutical formulations, demonstrating compliance with green chemistry principles through solvent selection, waste minimization, and operational safety [6].
This protocol describes a reversed-phase HPLC method for the simultaneous determination of mupirocin (MUP) in binary mixtures with fluticasone propionate (FLU) or mometasone furoate (MF), along with their impurities (Pseudomonic acid-D and Fluticasone impurity C) [3].
Materials and Reagents:
Chromatographic Conditions:
Sample Preparation:
Validation Parameters:
This protocol outlines an HPTLC-densitometry method for impurity profiling of pharmaceutical compounds, applicable to drugs like mupirocin and its impurities [3].
Materials and Reagents:
Chromatographic Conditions:
Sample Preparation:
Validation Parameters:
Table 3: Essential Research Reagents and Materials for Impurity Profiling
| Item | Function/Application | Examples/Specifications |
|---|---|---|
| Certified Impurity Standards | Reference materials for identification and quantification | ISO 17034 certified; with Certificate of Analysis (COA); traceable to USP/BP standards [5] |
| Stable Isotope-Labeled Standards | Internal standards for LC-MS quantification; improves accuracy | Deuterated analogs for exact compensation of matrix effects [5] |
| Green Mobile Phase Solvents | Environmentally friendly alternatives to traditional solvents | Ethanol, methanol, aqueous phases, supercritical COâ [4] |
| UHPLC Columns | High-efficiency separations with reduced solvent consumption | Small particle size (sub-2μm); 1.0-2.1 mm internal diameter [4] |
| HPTLC Plates | Simultaneous analysis of multiple samples with minimal solvent | Silica gel 60 F254; various sizes (10Ã10 cm, 20Ã20 cm) [3] [6] |
| Solid Phase Microextraction (SPME) | Green sample preparation; minimal solvent use | Molecularly Imprinted Polymers (MIPs) for selective extraction [4] |
| kaikasaponin III | kaikasaponin III, CAS:115330-90-0, MF:C48H78O17, MW:927.1 g/mol | Chemical Reagent |
| (+)-Kavain | (+)-Kavain, CAS:500-64-1, MF:C14H14O3, MW:230.26 g/mol | Chemical Reagent |
The evaluation of method environmental impact is essential in modern impurity profiling. Several assessment tools have been developed to quantify and compare the greenness of analytical methods [3] [6] [7].
The National Environmental Methods Index (NEMI) provides a simple pictogram indicating whether a method meets green criteria in four categories: persistent, bioaccumulative, and toxic (PBT) chemicals; hazardous chemicals; corrosivity (pH <2 or >12); and waste generation [6] [7].
The Analytical Eco-Scale assigns penalty points to hazardous reagents, energy consumption, and waste generation, with higher scores indicating greener methods [3] [6]. Methods scoring above 75 are considered excellent green methods, while scores below 50 indicate inadequate greenness [3].
The Green Analytical Procedure Index (GAPI) evaluates the environmental impact throughout the method lifecycle using a multi-criteria assessment with a five-element pictogram [6] [7]. This tool provides a more comprehensive evaluation than NEMI, covering sample collection, preparation, transportation, detection, and final disposition [6].
The Analytical GREEnness metric (AGREE) is a recently developed tool that uses a unified scoring system from 0 to 1, considering all twelve principles of green chemistry [6] [7]. This approach provides a comprehensive, user-friendly assessment of method environmental performance [6].
Figure 1: Impurity Control Method Development Workflow. This diagram outlines the systematic approach for developing impurity control methods that comply with both regulatory requirements and green chemistry principles. The process begins with clearly defined analytical objectives and proceeds through technique selection, optimization for sustainability, greenness assessment, and formal validation before implementation.
Figure 2: Impurity Control Decision Pathway. This diagram illustrates the decision-making process for impurity control according to ICH guidelines, highlighting the sequential thresholds for reporting, identification, and qualification. The pathway includes special considerations for genotoxic impurities and outlines the procedure for handling out-of-specification results.
The integration of ICH and USP guidelines with green chromatographic methods represents the current standard for impurity identification and control in pharmaceutical development. By adopting green analytical techniques that reduce environmental impact while maintaining regulatory compliance, pharmaceutical scientists can achieve sustainable impurity profiling without compromising data quality or patient safety. The continued development and application of green assessment tools will further advance the pharmaceutical industry's commitment to environmental responsibility while ensuring the quality, safety, and efficacy of drug products.
The pharmaceutical industry faces increasing pressure to balance its critical role in global health with the urgent need to minimize its environmental footprint. Sustainable pharmaceutical analysis represents a paradigm shift, integrating the principles of green chemistry directly into analytical control methods to ensure drug safety, quality, and efficacy. This approach is particularly crucial for impurity profiling, a mandatory component of pharmaceutical quality control that traditionally relies on solvent-intensive techniques like high-performance liquid chromatography (HPLC). The environmental imperative is clear: pharmaceutical analysis laboratories generate substantial volumes of chemical waste, primarily from organic solvents like acetonitrile and methanol, which pose both environmental and health hazards [8]. Simultaneously, compelling economic driversâincluding rising solvent disposal costs, stringent regulatory expectations, and operational efficiency goalsâare accelerating industry adoption of greener methodologies. This document details the application of sustainable chromatographic protocols within a research program focused on green impurity profiling, providing actionable frameworks for implementation by researchers, scientists, and drug development professionals.
The traditional reliance on hazardous solvents in pharmaceutical analysis presents multifaceted environmental challenges. Acetonitrile and methanol, the most common reversed-phase LC solvents, are derived from petrochemicals and carry significant toxicity. Methanol exposure can cause severe acidosis and retinal damage, while acetonitrile is metabolized in the body to cyanide, causing cytotoxic anoxia [8]. The cumulative environmental impact of manufacturing, using, and disposing of these solvents contributes substantially to the pharmaceutical industry's carbon footprint, which some reports indicate exceeds that of the automotive sector [9]. Furthermore, pharmaceutical pollution from production and waste disposal undermines progress toward multiple United Nations Sustainable Development Goals (SDGs), creating a pressing need for integrated management solutions [9].
Beyond environmental responsibility, powerful economic and regulatory factors drive the transition to sustainable analysis.
Table 1: Quantitative Environmental and Economic Benefits of Green Chromatographic Techniques
| Technique | Solvent Reduction vs. HPLC | Key Economic Benefit | Analysis Time Impact |
|---|---|---|---|
| UHPLC | Up to 80% [4] | Lower solvent purchase & disposal costs | Significant reduction |
| Narrow-Bore Columns (â¤2.1 mm ID) | Up to 90% [4] | Lower solvent purchase & disposal costs | Comparable or slightly faster |
| Supercritical Fluid Chromatography (SFC) | >90% (replaces organic with COâ) [4] | Eliminates organic solvent waste | Typically faster |
| Elevated Temperature LC | 40-60% (via higher water content) [4] | Lower solvent costs & faster throughput | Significant reduction |
The implementation of sustainable pharmaceutical analysis is operationalized through defined frameworks and standardized protocols. The core principles of Green Analytical Chemistry (GAC), encapsulated by the SIGNIFICANCE mnemonic, provide the foundation for these methods, emphasizing the minimization of hazardous waste and enhanced safety for analysts and the environment [7].
GLC focuses on modifying conventional HPLC to reduce its environmental impact without compromising performance [4]. Key strategies include:
SFC uses supercritical COâ as the primary mobile phase constituent, replacing up to 90% of the organic solvents used in normal-phase chromatography. COâ is non-toxic, non-flammable, and can be sourced from renewable processes, making SFC an inherently green technology ideal for chiral separations and impurity profiling of non-polar to moderately polar compounds [4].
Objective: To separate, identify, and quantify the active ingredients (Lidocaine HCl and Miconazole Nitrate), preservatives (Methyl Paraben, Saccharin Sodium), and a potential genotoxic impurity (Dimethylaniline, DMA) in an oral gel formulation using a green RP-HPLC method [7].
Principles of Greenness: This protocol replaces traditional acetonitrile-rich mobile phases with a methanol-buffer system and employs a gradient elution for efficient separation of a complex mixture, aligning with GAC principles of waste and hazard reduction [7].
1. Materials and Reagents (The Scientist's Toolkit) Table 2: Essential Research Reagents and Materials
| Item | Specification/Function |
|---|---|
| HPLC System | Agilent 1260 Infinity Series (or equivalent) with DAD detector and quaternary pump [7]. |
| Analytical Column | Waters XSelect CSH C18 (250 mm à 4.6 mm, 5 µm). Provides the stationary phase for separation [7]. |
| Mobile Phase A | Phosphate Buffer (pH 6.0). Aqueous component, adjusted with phosphoric acid/potassium dihydrogen phosphate. |
| Mobile Phase B | HPLC-Grade Methanol. Less toxic alternative to acetonitrile [7]. |
| Analytical Standards | Lidocaine HCl, Miconazole Nitrate, Methyl Paraben, Saccharin Sodium, Dimethylaniline (DMA). For identification and quantification. |
| Sample Solvent | Methanol-Water mixture. For dissolving the gel formulation and standards. |
2. Instrumental Parameters and Chromatographic Conditions
3. Sample Preparation
4. Validation and Data Analysis
The choice of solvent is central to greening chromatographic methods. While water is the ideal green solvent, its chromatographic properties are often insufficient alone. A hierarchy of greener alternatives exists, each with specific advantages and limitations [8].
Table 3: Evaluation of Green Solvent Alternatives for Liquid Chromatography
| Solvent | Greenness Advantages | Key Limitations in HPLC | Recommended Application |
|---|---|---|---|
| Ethanol | Renewable, biodegradable, low toxicity [8]. | High UV cut-off (~210 nm), higher viscosity [8]. | Replacing acetonitrile in methods using detection >220-230 nm. |
| Methanol | Less toxic than acetonitrile [8]. | Toxic alcohol, higher UV cut-off (~205 nm) than ACN [8]. | Common, less-hazardous substitute for ACN; requires UV caution. |
| Dimethyl Carbonate | Biodegradable, low toxicity [8]. | Low elution strength, immiscibility with water at high proportions [8]. | Modifier in normal-phase or for specific reversed-phase applications. |
| Propylene Carbonate | Low volatility, low toxicity [8]. | High viscosity, high UV cut-off [8]. | Suitable for preparative chromatography where viscosity is less critical. |
| Glycerol-Water Mixes | Non-toxic, renewable [8]. | Very high viscosity, leading to extreme backpressure [8]. | Low-percentage modifier for challenging separations; requires high temp. |
A critical consideration in implementing these methods is the trade-off between greenness and practical performance. For instance, substituting acetonitrile with ethanol is chromatographically feasible but may require method re-development to adjust selectivity and could be limited by ethanol's higher viscosity and UV cut-off [8]. The strategic approach is to select the greenest solvent that maintains the required analytical performance for the specific impurity profiling application.
The transition to sustainable pharmaceutical analysis, particularly in the critical area of impurity profiling, is no longer optional but a necessity driven by compelling environmental, economic, and regulatory forces. The application notes and protocols detailed herein provide a practical roadmap for implementing green chromatographic methods, demonstrating that it is possible to maintain the highest standards of analytical rigor while significantly reducing the environmental footprint. The continued adoption of these practices, coupled with ongoing research into even greener solvents and technologies like machine learning-assisted method development, will be pivotal in steering the pharmaceutical industry toward a more sustainable and responsible future.
Impurity profiling is a critical component of pharmaceutical quality control, directly impacting drug safety, efficacy, and stability [4]. The systematic classification and control of impurities are mandated by regulatory authorities worldwide to ensure patient safety and product quality [10]. According to current regulatory standards from the International Council for Harmonisation (ICH) and United States Pharmacopoeia (USP), impurities in pharmaceutical substances and products are comprehensively categorized into three primary classes: organic impurities, inorganic impurities, and residual solvents [4] [10].
The growing emphasis on environmental sustainability has driven the adoption of Green Analytical Chemistry (GAC) principles within impurity profiling. These principles focus on reducing the environmental impact of analytical methods by minimizing solvent consumption, reducing waste generation, and improving energy efficiency, without compromising analytical performance [4] [11]. This application note details the classification of impurities and provides validated, green chromatographic protocols for their analysis, supporting the broader thesis of implementing sustainable practices in pharmaceutical research and development.
Impurities can be introduced at various stages of the drug development process, including synthesis, formulation, and storage [10]. A clear understanding of their classification, sources, and associated regulatory guidelines is fundamental to effective control strategies.
Table 1: Classification of Pharmaceutical Impurities and Regulatory Guidelines
| Impurity Class | Definition and Examples | Common Sources | Primary ICH Guidelines |
|---|---|---|---|
| Organic Impurities | Starting materials, intermediates, by-products, degradation products, reagents, ligands, catalysts [10]. | Synthesis, formulation, or degradation of the drug substance or product during storage [4] [10]. | Q3A (R2): Impurities in New Drug Substances [4]Q3B (R2): Impurities in New Drug Products [4] |
| Inorganic Impurities | Heavy metals, catalysts, inorganic salts, filter aids, charcoal [10]. | Raw materials, reagents, manufacturing equipment, water, and the environment [10]. | Q3D (R2): Guideline for Elemental Impurities [4] |
| Residual Solvents | Volatile organic chemicals used or produced in the manufacture of drug substances or excipients [10]. | Synthesis or purification processes (e.g., heptane, DMF, THF) [12]. | Q3C (R8): Impurities: Guidelines for Residual Solvents [4] |
The ICH guidelines establish thresholds for reporting, identifying, and qualifying impurities based on the maximum daily dose of the drug product, ensuring a standardized global approach to impurity control [10]. Regulatory agencies, including the US FDA and EMA, require comprehensive documentation and validated analytical methods for impurity testing as part of current Good Manufacturing Practices (cGMP) [10].
The integration of Green Analytical Chemistry (GAC) principles aims to make impurity profiling more sustainable. Key green techniques include:
The greenness of analytical methods can be evaluated using dedicated assessment tools such as the Analytical Eco-Scale, the Green Analytical Procedure Index (GAPI), and the AGREE metric [11].
This protocol, adapted from a study on suvorexant, provides a green approach for the simultaneous determination of eight residual solvents, including n-heptane, dimethyl sulfoxide (DMSO), and N,N-dimethylformamide (DMF) [12].
Table 2: Key Reagents and Materials for HS-GC Analysis
| Item | Function/Description | Specification/Example |
|---|---|---|
| Gas Chromatograph | Instrument separation and detection. | Equipped with Flame Ionization Detector (FID) [12]. |
| Headspace Autosampler | Automated sampling of the vapor phase. | For static headspace analysis to introduce solvent vapors [12]. |
| Capillary Column | Stationary phase for chromatographic separation. | DB-624 (30 m à 0.53 mm, 3.0 μm) or equivalent [12]. |
| High-Purity Gases | Carrier and detector gases. | Helium or Nitrogen as carrier gas; Hydrogen and air for FID [12]. |
| Reference Standards | For identification and quantification. | Certified reference materials of target solvents (e.g., n-heptane, DMF) [12]. |
| Dimethyl sulfoxide (DMSO) | Sample solvent. | High-purity grade for dissolving the API [12]. |
This protocol outlines a green HPLC method for the simultaneous quantification of active ingredients and related organic impurities, as demonstrated for carvedilol and hydrochlorothiazide and its impurities [1].
Table 3: Key Reagents and Materials for Green HPLC Analysis
| Item | Function/Description | Specification/Example |
|---|---|---|
| HPLC System with PDA | Instrument for separation and detection. | System capable of gradient elution and photodiode array detection [1]. |
| Analytical Column | Stationary phase for separation. | YMC Triart-Phenyl column (150 x 4.6 mm, 5 μm) or equivalent [1]. |
| Green Solvents | Mobile phase components. | Ethanol (HPLC-grade), water, 0.1% formic acid [1]. |
| Reference Standards | For identification and quantification. | Certified standards of API and known impurities (e.g., Salamide, Chlorothiazide) [1]. |
| Time (min) | % Solvent B |
|---|---|
| 0 - 5 | 20% |
| 5 - 7 | 20% â 80% |
| 7 - 12 | 80% |
| 12 - 13 | 80% â 20% |
| 13 - 17 | 20% (Re-equilibration) [1] |
The precise classification of impurities into organic, inorganic, and residual solvents, as defined by ICH guidelines, is the foundation of a robust pharmaceutical quality control system. The experimental protocols detailed herein for HS-GC and Green HPLC not only comply with regulatory requirements but also align with the principles of Green Analytical Chemistry. By adopting these green chromatographic methodsâwhich utilize ethanol/water mobile phases, UHPLC, and miniaturized techniquesâresearchers and drug development professionals can significantly reduce the environmental footprint of impurity profiling. This application note provides a practical framework for implementing these sustainable practices, thereby contributing to the development of safer pharmaceuticals and a healthier environment.
The drive towards sustainable laboratory practices has made green analytical chemistry a central pillar of modern pharmaceutical analysis. Within this framework, impurity profilingâa critical activity for ensuring drug safety and efficacyâis undergoing a significant transformation. The adoption of Ultra-High Performance Liquid Chromatography (UHPLC) coupled with narrow-bore and capillary-scale columns represents a powerful strategy to drastically reduce solvent consumption without compromising analytical performance [4] [1]. This approach aligns perfectly with the principles of Green Analytical Chemistry (GAC) by minimizing waste generation, reducing the use of hazardous chemicals, and lowering the environmental footprint of analytical procedures [4] [13].
The technical foundation of this strategy lies in moving from traditional 4.6 mm internal diameter (i.d.) columns to columns with smaller internal diameters. This shift enables a substantial reduction in mobile phase volumetric flow rates while maintaining optimal linear velocity, leading to a direct and often dramatic decrease in solvent usage [14] [4]. This application note details the practical implementation, benefits, and specific protocols for employing UHPLC with narrow-bore columns to achieve greener impurity profiling methods.
The theoretical advantages of scaling down column dimensions translate into measurable, significant benefits in the laboratory. The following table summarizes the typical solvent consumption and key operational parameters for different column formats, illustrating the progressive savings achievable through miniaturization.
Table 1: Solvent Consumption and Operational Parameters for Different LC Column Scales
| Column Scale | Typical Internal Diameter (mm) | Typical Flow Rate (mL/min) | Approximate Solvent Use per Run* | Primary Application Context |
|---|---|---|---|---|
| Conventional Analytical | 4.6 | 1.0 - 1.5 | 20 - 30 mL | Standard HPLC methods |
| Narrow-Bore | 2.0 - 3.0 | 0.2 - 0.6 | 4 - 9 mL | Greener UHPLC methods [4] |
| Micro-Bore | 1.0 | ~0.05 | < 1 mL | High-sensitivity LC-MS [4] |
| Capillary-Scale | 0.15 - 0.50 | 1 - 20 µL | ~0.3 mL | 'Omics, limited samples [14] |
*Estimate based on a 20-minute gradient run.
The environmental and practical impacts of this reduction are substantial. A direct comparison cited in the literature confirms that analyzing pharmaceutical contaminants using 1.0 mm i.d. columns can reduce mobile phase consumption by up to 90% compared to conventional 4.6 mm i.d. columns [4]. This leads to a proportional reduction in solvent waste, lowering purchasing and disposal costs. Furthermore, the reduced flow rates are ideal for electrospray ionization mass spectrometry (ESI-MS), as they improve ionization efficiency, which can increase sensitivityâa crucial factor for detecting low-level impurities [14].
This section provides a detailed procedure for translating an existing HPLC impurity method to a greener UHPLC narrow-bore method and applying it for impurity profiling.
The successful implementation of this strategy relies on specific materials and instruments.
Table 2: Essential Materials and Equipment for UHPLC Solvent Reduction
| Item | Specification/Function |
|---|---|
| UHPLC System | Capable of operating at pressures up to 1000-1500 bar and delivering precise, low-flow gradients. |
| Narrow-Bore Column | e.g., 2.1 mm i.d., particle size 1.7 - 1.8 µm. Select a stationary phase (e.g., C18, phenyl-hexyl) equivalent to the original method [15]. |
| Green Mobile Phase | Replace acetonitrile with ethanol or methanol where possible [4] [1]. Use aqueous mobile phases or ionic liquids as additives to reduce organic solvent impact [4]. |
| Low-Dispersion Accessories | Use small i.d. connection tubing (e.g., 0.005") and "zero dead volume" fittings to minimize extra-column volume and preserve separation efficiency [14]. |
| Autosampler | Calibrated for accurate injection of small volumes (1-2 µL). |
The following diagram outlines the core workflow for transitioning a method to a solvent-reduced format and applying it to impurity profiling.
Step 1: Method Translation
Flow_new = Flow_old à (id_new² / id_old²). For example, translating from a 4.6 mm column running at 1.0 mL/min to a 2.1 mm column: Flow_new = 1.0 à (2.1² / 4.6²) â 0.21 mL/min.Step 2: Method Optimization and Validation
Step 3: Application to Impurity Profiling and Stability Studies
Step 4: Greenness Assessment
The move toward miniaturization extends beyond narrow-bore columns to capillary-scale LC (0.075â0.5 mm i.d.), which operates at flow rates of 1â20 µL/min [14]. This offers even greater reductions in solvent consumption and is especially valuable in 'omics' fields and high-sensitivity bioanalysis where sample volumes are limited [14] [18].
The integration of Artificial Intelligence (AI) and Machine Learning (ML) is also emerging as a powerful tool for accelerating method development. AI can help predict optimal separation conditions and automate the translation of methods to different scales, further enhancing laboratory efficiency and sustainability [18].
The integration of UHPLC with narrow-bore columns provides a robust, practical, and immediately accessible pathway for pharmaceutical laboratories to advance their sustainability goals. This approach directly addresses the core tenets of Green Analytical Chemistry by drastically reducing solvent consumption and waste generation. Moreover, it does so without sacrificingâand often while enhancingâthe chromatographic resolution, speed, and sensitivity required for rigorous impurity profiling and stability-indicating analyses. Adopting these strategies is a definitive step toward more environmentally responsible quality control and drug development.
The drive towards Green Analytical Chemistry (GAC) has intensified the search for sustainable alternatives to traditional reversed-phase liquid chromatography solvents. Acetonitrile, while historically preferred for its performance, is classified as a Class 2 solvent with inherent toxicity, creating significant environmental and safety concerns [19]. This application note details practical strategies for replacing acetonitrile with ethanol-water mobile phases, providing validated methodologies for pharmaceutical impurity profiling that align with green chemistry principles while maintaining robust analytical performance.
Replacing acetonitrile addresses critical issues in laboratory sustainability. Conventional HPLC methods consume approximately 750 mL of organic solvent daily per instrument when operated continuously, creating substantial waste disposal challenges and environmental impact [20]. Ethanol offers a safer toxicological profile, better biodegradability, and reduced environmental footprint while remaining cost-effective for routine analytical methods.
A validated green HPLC method for impurity profiling of Ivosidenib demonstrates the effective application of ethanol-based mobile phases in pharmaceutical analysis [21].
Chromatographic Conditions:
Method Performance Characteristics:
This method successfully characterized degradation products formed under various stress conditions (acidic, UV, and oxidative) using LC-MS/MS techniques, with in silico toxicity prediction indicating neurotoxicity and respiratory toxicity for all degradation products [21].
Table: Systematic Approach for Acetonitrile to Ethanol Method Conversion
| Development Step | Key Parameters | Optimization Strategy |
|---|---|---|
| Initial Scouting | Organic modifier type, column chemistry | Test ethanol, isopropanol, and acetonitrile for comparison |
| Selectivity Optimization | Mobile phase pH, column temperature | Adjust pH (2-4) with formic acid; temperature (25-40°C) |
| Efficiency Enhancement | Flow rate, gradient profile | Reduce flow rate to manage backpressure; extend gradient |
| Final Validation | System suitability, robustness | Verify resolution, peak symmetry, and reproducibility |
Protocol Details:
Column Selection: Begin with C18 columns known for compatibility with ethanol-based mobile phases. The Zorbax Eclipse Plus series has demonstrated excellent performance with ethanol-water systems [21].
Mobile Phase Preparation: Prepare the aqueous phase containing 0.1% formic acid in purified water. Mix with HPLC-grade ethanol in the ratio 55:45 (v/v aqueous:ethanol). Filter through a 0.45 µm membrane and degass before use.
System Equilibration: Allow sufficient equilibration time (5-10 column volumes) due to the higher viscosity of ethanol-water mixtures compared to acetonitrile-based mobile phases.
Backpressure Management: Anticipate approximately 2-fold higher backpressure with ethanol-water mixtures compared to acetonitrile-water at equivalent percentages. Adjust flow rates accordingly (typically 0.8 mL/min vs. 1.0 mL/min for standard 4.6 mm ID columns) [22].
Green HPLC Method Development Workflow
Table: Physicochemical Properties of HPLC Organic Modifiers
| Solvent | Eluotropic Strength (ε°) | Viscosity (cP) | UV Cutoff (nm) | GREEN Assessment | Safety Profile |
|---|---|---|---|---|---|
| Acetonitrile | 0.65 | 0.34 | 190 | Problematic (Class 2) | Toxic, environmental concerns |
| Methanol | 0.73 | 0.55 | 205 | Preferred | Toxic, biodegradable |
| Ethanol | 0.68 | 1.08 | 210 | Preferred | Low toxicity, renewable |
| Isopropanol | 0.82 | 1.96 | 210 | Preferred | Low toxicity, biodegradable |
Data compiled from multiple sources [20] [22] [19]
The selectivity differences between ethanol and acetonitrile can be advantageous for challenging separations. Ethanol's higher viscosity necessitates adjustments to flow rates but provides alternative selectivity for structurally similar impurities [19].
The Ivosidenib impurity profiling method demonstrated high compliance with green analytical chemistry principles when using ethanol-water mobile phases [21]. Assessment using AGREE and GAPI tools confirmed significantly reduced environmental impact compared to conventional acetonitrile-based methods.
Key green advantages include:
Aqueous Mobile Phase Best Practices:
Ethanol-Wobile Phase Considerations:
Common Challenges and Solutions:
Table: Essential Reagents and Materials for Ethanol-Based HPLC Methods
| Item | Specification | Application Purpose |
|---|---|---|
| HPLC-Grade Ethanol | â¥99.9% purity, low UV absorbance | Primary organic modifier replacing acetonitrile |
| Acid Additives | Formic acid, trifluoroacetic acid (0.05-0.1%) | pH modification for ionizable compounds |
| C18 Columns | High purity silica, endcapped | Stationary phase compatible with ethanol modifiers |
| In-line Filters | 0.5 µm porosity | Protection against particulate matter |
| Pre-column Filters | 0.2-0.5 µm porosity | Guard column protection for impurity profiling |
| Forchlorfenuron | Forchlorfenuron, CAS:68157-60-8, MF:C12H10ClN3O, MW:247.68 g/mol | Chemical Reagent |
| Fortunellin | Fortunellin, CAS:20633-93-6, MF:C28H32O14, MW:592.5 g/mol | Chemical Reagent |
The replacement of acetonitrile with ethanol in reversed-phase HPLC mobile phases represents a viable, greener alternative for pharmaceutical impurity profiling. The documented method for Ivosidenib demonstrates that ethanol-water mobile phases can provide the required selectivity, resolution, and sensitivity for rigorous impurity analysis while aligning with green chemistry principles.
With proper method development and optimization, ethanol-based methods can deliver equivalent chromatographic performance to acetonitrile-based systems while offering significant advantages in safety, environmental impact, and sustainability. The continued adoption of these approaches supports the pharmaceutical industry's transition toward more environmentally responsible analytical practices without compromising data quality or regulatory compliance.
Supercritical Fluid Chromatography (SFC) is a separation technique that utilizes supercritical fluids as the mobile phase. It functions similarly to other chromatography methods, where a mixture is injected into a flow of supercritical fluid and passed through a stationary phase. The individual components separate based on their differing partitioning behaviors between the mobile and stationary phases [24].
Carbon dioxide (COâ) is the most commonly used supercritical fluid due to its advantageous properties: it has a low critical temperature (31 °C) and pressure (73.8 bar), is inert, non-flammable, non-toxic, and available in high purity at low cost [25] [24]. A supercritical fluid is a state of matter achieved above its critical temperature and pressure, where it exhibits properties intermediate between those of gases and liquidsâwith density similar to liquids and viscosity and diffusivity akin to gases. These properties contribute to SFC's high efficiency and rapid separations [25] [24].
Table 1: Advantages of SFC as a Green Chromatographic Technique
| Advantage | Comparison to Traditional HPLC | Environmental and Economic Impact |
|---|---|---|
| Reduced Solvent Consumption | Uses up to 8 times less organic solvent [24] | Less hazardous waste generation and lower solvent purchase/disposal costs |
| Faster Analysis | Generally a 3 to 4 times faster process [24] | Lower energy consumption during analysis |
| Easier Post-Process Isolation | Up to 7 times lower energy consumption for solvent removal in preparative applications [24] | Significant reduction in overall process energy footprint |
| Greener Mobile Phase | Primary mobile phase is COâ, often reused from other processes [24] | Nontoxic, non-flammable, and offers a COâ-neutral alternative to hazardous solvents |
Impurity profiling is a critical component of pharmaceutical quality control, essential for ensuring drug safety, efficacy, and stability. Regulatory guidelines from the International Council for Harmonisation (ICH) mandate the identification, qualification, and control of impurities in active pharmaceutical ingredients (APIs) and drug products [4] [3]. The principles of Green Analytical Chemistry (GAC) are gaining prominence in this field, aiming to minimize the environmental impact of analytical methods by reducing solvent consumption, avoiding waste generation, and using less hazardous chemicals [4] [2].
SFC represents a versatile and greener alternative to traditional reversed-phase liquid chromatography [26]. Its application is particularly suitable for impurity profiling due to its ability to provide different selectivity, which can help resolve complex mixtures of APIs and their structurally related impurities. The technique is especially valuable for separating polar and chiral compounds, where conventional LC methods often show poor retention and resolution [26]. Furthermore, the coupling of SFC with mass spectrometry (MS) enhances its capability for the identification and characterization of unknown impurities [26].
The analysis of polyphenolsâa large group of plant metabolites including flavonoids, stilbenes, and lignansâis challenging due to the complexity of plant extracts and the limited availability of authentic standards [26]. While reversed-phase LC is commonly used, it often results in poor retention and resolution for very polar polyphenols. This application note summarizes the use of SFC-MS for the characterization of these compounds.
Instrumentation and Materials:
Method Details:
SFC has demonstrated excellent potential for the separation of polar and chiral polyphenols, filling a gap left by traditional LC methods [26]. However, a survey of recent literature (2019-2024) indicates that SFC is still underutilized in this field, particularly for large-scale metabolomics studies. Most applications are quantitative targeted studies, and the technique's adoption has been limited by the need for extensive optimization of critical parameters like the stationary phase and modifier composition [26]. Despite this, SFC-MS is a powerful tool for better characterization of complex plant extracts.
Table 2: Essential Materials for SFC Analysis
| Item | Function | Specific Examples & Notes |
|---|---|---|
| Liquid COâ Supply | Primary component of the mobile phase | High-purity grade; requires a chiller unit to maintain liquid state [25]. |
| Organic Modifiers | Adjusts polarity and strength of the mobile phase to control elution and selectivity | Methanol, Ethanol, Isopropanol; ethanol is a preferred green solvent [4] [28]. |
| Additives | Improves peak shape for acidic or basic analytes | Formic acid, ammonium hydroxide, ammonium acetate [24]. |
| Analytical Columns | Stationary phase for chromatographic separation | Silica, Cyano, Diol, Amide, and chiral phases; 4.6 mm x 150-250 mm, 3-5 µm particle size [24]. |
| Reference Standards | For method development and identification of impurities | API and available impurity standards [3]. |
System Setup and Conditioning:
Mobile Phase Preparation:
Sample Preparation:
Method Development and Optimization:
Detection:
System Shutdown:
The following diagram illustrates the logical workflow for developing an SFC method for impurity profiling.
A significant advancement is the on-line coupling of Supercritical Fluid Extraction (SFE) with SFC. This integrated approach allows for the direct extraction and analysis of compounds from solid samples, such as active ingredients from formulations, without extensive sample pretreatment. A 2025 study by Shimadzu scientists successfully used on-line SFE-SFC for the solid-state injection and purification of ibuprofen, separating it from structurally related compounds in under 3 minutes [27]. This method is particularly useful for samples that are challenging to solubilize and for rapidly loading a sample to facilitate swift analysis.
In drug development, preparative SFC is the method of choice for the high-throughput purification of chiral and achiral compounds. Its advantages over preparative HPLC include faster run times, significantly lower consumption of organic solvents, and easier isolation of purified compounds due to the easy evaporation of the COâ-based mobile phase [24]. Optimization for maximum yield involves careful attention to sample loading techniques, choice of co-solvent, and precise control of temperature and pressure [28].
Supercritical Fluid Chromatography, with COâ as its primary mobile phase, stands as a powerful, green, and efficient technique for impurity profiling in pharmaceutical research and other fields. Its superior speed, reduced environmental impact, and unique selectivity, especially for polar and chiral molecules, make it a compelling complement or alternative to traditional HPLC. While challenges such as the need for method optimization and limited familiarity remain, ongoing technological advancementsâsuch as UHPSFC, improved stationary phases, and on-line SFE-SFC systemsâare solidifying SFC's role as a cornerstone of modern, sustainable analytical chemistry.
In the context of impurity profiling research for pharmaceutical quality control, sample preparation has traditionally been a resource-intensive process, characterized by high solvent consumption and significant waste generation. The integration of green analytical chemistry principles is transforming this foundational step, aiming to minimize the environmental impact of analytical procedures while maintaining or enhancing performance [4]. This application note details practical implementations of miniaturized, automated, and solvent-free sample preparation techniques, specifically Solid-Phase Microextraction (SPME) and related methods, which align with the twelve principles of green chemistry by reducing solvent use, preventing pollution, and promoting safer procedures [4] [29].
These green sample-preparation approaches are particularly crucial for impurity profiling, a process essential for ensuring drug safety, efficacy, and stability [4]. The International Council for Harmonisation (ICH) guidelines Q3A-Q3D provide a framework for classifying and controlling impurities, requiring highly sensitive and reliable analytical methods [4]. The techniques described herein support these regulatory requirements while advancing the sustainability of pharmaceutical analysis.
The following table catalogues essential materials and reagents for implementing the green sample preparation protocols discussed in this note.
Table 1: Key Research Reagent Solutions for Green Sample Preparation
| Item | Function/Description | Application Example |
|---|---|---|
| SPME Fibers (PAL Smart) | Automated, solvent-free extraction of analytes from various matrices. Smart chips store usage history and parameters [30]. | General microextraction for volatile/semi-volatile compounds. |
| DVB/CAR/PDMS Fiber | A common triple-coating (Divinylbenzene/Carboxen/Polydimethylsiloxane) for a broad analyte range [31]. | Headspace analysis of Biogenic Volatile Organic Compounds (BVOCs) [31]. |
| Mixed-Mode SPME Coatings (e.g., C8-SCX, HLB) | Enhance direct immersion (DI) mode sampling of non-volatile analytes with diverse polarities [32]. | Analysis of phytocannabinoids and endocannabinoids in complex biological matrices [32]. |
| YMC Triart-Phenyl Column | Analytical column for HPLC using green mobile phases like ethanol-water mixtures [33]. | Green HPLC quantification of carvedilol and hydrochlorothiazide with impurity profiling [33]. |
| Green Solvents (Ethanol, Ionic Liquids) | Replace traditional, more hazardous solvents like acetonitrile in mobile phases [4]. | Green Liquid Chromatography (GLC) for pharmaceutical separation [4]. |
| AGREEprep & ComplexGAPI Software | Automated green assessment tools for evaluating the environmental impact and practicality of analytical methods [31]. | Sustainability assessment of HS-SPME-GC-QTOF-MS methods [31]. |
This protocol describes a miniaturized, solvent-free method for analyzing Biogenic Volatile Organic Compounds (BVOCs) using Headspace Solid-Phase Microextraction coupled to Gas ChromatographyâQuadrupole Time-of-Flight Mass Spectrometry (HS-SPME-GC-QTOF-MS) [31]. The method exemplifies the core principles of green analytical chemistry through miniaturization, using only 0.20 g of plant material, and by eliminating hazardous reagents. It has been rigorously assessed with green metrics (AGREE, AGREEprep, ComplexGAPI) and demonstrated applicability for complex biological matrices, making it a valuable model for impurity profiling and metabolomic studies [31].
This protocol utilizes SPME in Direct Immersion (DI) mode coupled to Liquid Chromatography-Mass Spectrometry (LC-MS) for the precise monitoring of phytocannabinoids and endocannabinoids in complex biological matrices like plasma and brain tissue [34] [32]. SPME is an advanced, non-exhaustive sample-preparation technique that facilitates the precise and efficient isolation of trace analyte amounts [34]. Its primary green advantage is minimal organic solvent consumption. Furthermore, the coating can filter out macromolecules, reducing matrix effects and protecting labile analytes from degradation, which is crucial for accurate impurity and metabolite profiling [32].
This protocol outlines a green High-Performance Liquid Chromatography (HPLC) strategy for the simultaneous quantification of active pharmaceutical ingredients and their impurities, with a focus on reducing solvent consumption and waste [4] [33]. The method leverages miniaturization strategies including Ultra-High Performance Liquid Chromatography (UHPLC) and narrow-bore columns, which can reduce mobile phase consumption by up to 80-90% compared to conventional HPLC [4]. Furthermore, it incorporates green solvents like ethanol to replace toxic acetonitrile in mobile phases, aligning with the principles of green chemistry and improving the method's environmental footprint [4] [33].
The field of green sample preparation is rapidly evolving towards higher integration and efficiency. Two key trends are shaping its future:
Table 2: Quantitative Comparison of Green Sample Preparation Techniques
| Technique | Sample Volume | Solvent Consumption | Key Green Advantage | Reported Performance |
|---|---|---|---|---|
| HS-SPME-GC-QTOF-MS [31] | 0.20 g | Solvent-free (extraction) | Eliminates solvent use in sample prep; minimal waste. | Successfully profiled BVOCs from trees; high reproducibility. |
| SPME-LC-MS [32] | Low µL-mL range | A few µL (for desorption) | Drastic solvent reduction vs. exhaustive methods. | Precise analysis of trace-level endocannabinoids in brain tissue. |
| Green UHPLC (Narrow-Bore) [4] | Standard injection | 80-90% reduction vs. conventional HPLC | Massive solvent savings and reduced waste. | Maintains or improves separation efficiency. |
| Direct SPME-MS [35] | Minimal | A few µL (for desorption) | Bypasses LC, saving time, solvent, and energy. | Enables high-throughput, rapid screening. |
Impurity profiling is a critical component of pharmaceutical quality control and assurance, directly impacting drug safety, efficacy, and stability [4]. In recent years, the paradigm has shifted toward embracing Green Analytical Chemistry (GAC) principles to minimize the environmental footprint of analytical methods while maintaining rigorous performance standards [4] [11]. This application note details practical implementations of green chromatographic techniques for impurity profiling in pharmaceutical substances and products. It provides validated protocols and case studies demonstrating how sustainable chromatography achieves regulatory compliance while reducing solvent consumption, waste generation, and energy usage.
The development of green chromatographic methods is guided by the 12 principles of Green Analytical Chemistry, which include direct analytical techniques, waste minimization, safer solvents/reagents, and energy efficiency [37]. To quantitatively evaluate the environmental impact of analytical methods, several greenness assessment tools have been developed and widely adopted.
Table 1: Greenness Assessment Tools for Analytical Methods
| Tool Name | Graphical Representation | Main Focus | Output Type | Notable Features |
|---|---|---|---|---|
| Analytical Eco-Scale | Not applicable | Solvent toxicity, energy consumption, waste generation | Penalty point system | Simple semi-quantitative evaluation [11] |
| GAPI (Green Analytical Procedure Index) | Color-coded pictogram | Entire analytical workflow | Visual pictogram | Easy visualization of environmental impact across all stages [11] [37] |
| AGREE (Analytical GREEnness) | Radial chart (0-1) | All 12 principles of GAC | Single score (0-1) + visual | Holistic assessment with comprehensive output [11] [37] |
| AGREEprep | Pictogram + score | Sample preparation | Dedicated score | First metric specifically for sample preparation greenness [37] |
| BAGI (Blue Applicability Grade Index) | Asteroid pictogram | Practical applicability | Numeric score + visual | Evaluates practical viability in real-world settings [37] |
The AGREE metric, which evaluates all 12 GAC principles, is particularly valuable for providing a comprehensive environmental assessment [37]. Additionally, the emerging concept of White Analytical Chemistry (WAC) seeks to balance analytical performance (red), environmental sustainability (green), and practical applicability (blue), with an ideal "white" method harmonizing all three dimensions [37].
Figure 1: Green Chromatographic Method Development Workflow. This diagram illustrates the iterative process of developing and optimizing green chromatographic methods, incorporating Green Analytical Chemistry principles, validation, and multi-metric assessment to achieve a balanced "white" method.
This case study focuses on developing green chromatographic methods for the analysis of Mupirocin (MUP) in combination with corticosteroids Fluticasone propionate (FLU) or Mometasone furoate (MF), along with their specified impurities [3]. MUP is a topical antibacterial agent, while FLU and MF are corticosteroids used in various dermatological conditions [3]. The challenge involved simultaneous determination of these drug substances along with their impurities (Pseudomonic acid-D for MUP and Fluticasone impurity C for FLU) in pharmaceutical formulations, requiring methods capable of resolving all components while adhering to green chemistry principles [3].
Both developed methods successfully separated the cited drugs and their impurities and were validated according to ICH guidelines [3]. The greenness of the proposed methods was evaluated using the National Environmental Method Index, Analytical Eco-Scale, Green Analytical Procedure Index, and Analytical Greenness metric approaches, demonstrating superior environmental performance compared to conventional methods [3].
Table 2: Method Validation Parameters for Mupirocin and Impurity Analysis
| Parameter | HPTLC-Densitometry Method | HPLC Method |
|---|---|---|
| Linearity Range | 0.1-500 µg/band (depending on analyte) | 0.1-100 µg/mL (depending on analyte) |
| Precision (% RSD) | <1.5% | <1.2% |
| Accuracy (% Recovery) | 98.5-101.2% | 98.8-101.5% |
| Detection Limit | 0.03-0.05 µg/band | 0.02-0.04 µg/mL |
| Quantification Limit | 0.1-0.15 µg/band | 0.05-0.1 µg/mL |
| Analysis Time | ~20 minutes | ~15 minutes |
| Solvent Consumption | ~13 mL per analysis | ~15 mL per analysis |
The greenness assessment confirmed that both methods significantly reduced environmental impact compared to reported conventional methods, with the HPTLC method showing particularly favorable performance in terms of solvent consumption and waste generation [3].
This case study addresses the development of a green HPLC method for the simultaneous determination of Methocarbamol (MET), Aspirin (ASP), and their related impurities Guaifenesin (GUF) and Salicylic acid (SA) [38]. MET is a muscle relaxant, while ASP is an analgesic, and they are commonly co-formulated for musculoskeletal pain [38]. The method was designed to separate all four compounds in a single chromatographic run with low retention times, suitable for quality control laboratories, and applicable to pharmaceutical formulations and spiked human plasma [38].
The method successfully achieved separation of all four compounds within 10 minutes, with MET, ASP, GUF, and SA eluting at approximately 2.5, 4.5, 6.5, and 8.5 minutes, respectively [38]. Molecular docking studies were employed to understand the interaction mechanisms between the analytes and the stationary phase, which corroborated the elution order [38].
Table 3: Analytical Performance Characteristics for MET/ASP Method
| Analyte | Linearity Range (µg/mL) | Regression Equation | Correlation Coefficient (r²) | LOD (µg/mL) | LOQ (µg/mL) |
|---|---|---|---|---|---|
| GUF | 0.30-50.00 | y = 12.45x + 5.23 | 0.9998 | 0.09 | 0.30 |
| MET | 1.00-300.00 | y = 8.76x + 15.32 | 0.9999 | 0.30 | 1.00 |
| ASP | 10.00-500.00 | y = 10.21x + 25.67 | 0.9997 | 3.00 | 10.00 |
| SA | 0.10-50.00 | y = 15.43x + 3.45 | 0.9999 | 0.03 | 0.10 |
The greenness of the method was evaluated using whiteness assessment, Analytical Eco-Scale, and AGREE metrics, confirming its environmental superiority over conventional methods [38]. The method was successfully applied to pharmaceutical formulations and spiked human plasma, with recovery rates of 98.5-101.3% for tablet analysis and 97.8-99.5% for plasma analysis [38].
Table 4: Essential Materials for Green Chromatographic Analysis of Pharmaceutical Impurities
| Item | Specification/Example | Function/Application |
|---|---|---|
| HPLC Columns | C~18~ or C~8~ columns (e.g., X-Bridge, Eclipse XDB) | Stationary phase for reversed-phase separation [3] [38] |
| HPTLC Plates | Silica gel 60 F~254~ pre-coated plates | Stationary phase for planar chromatography [3] |
| Green Solvents | Ethanol, methanol, water, supercritical CO~2~ | Eco-friendly mobile phase components [4] [11] |
| Buffer Salts | Sodium dihydrogen phosphate, ammonium acetate | Mobile phase modifiers for pH control [3] [39] |
| pH Adjusters | o-Phosphoric acid, triethylamine | Mobile phase pH optimization [38] |
| Reference Standards | USP/BP reference standards | Method validation and quantification [3] |
| Sample Preparation Materials | Microsyringes, volumetric flasks, sonicator | Accurate sample preparation and dissolution [3] [38] |
| Kinamycin C | Kinamycin C, CAS:35303-08-3, MF:C24H20N2O10, MW:496.4 g/mol | Chemical Reagent |
| Kulinone | Kulinone, CAS:21688-61-9, MF:C30H48O2, MW:440.7 g/mol | Chemical Reagent |
Figure 2: Green Analytical Workflow for Pharmaceutical Impurity Profiling. This diagram outlines the key stages in the analytical process, from sample preparation to greenness assessment, highlighting the integration of sustainability metrics at the conclusion of the workflow.
Regulatory compliance is paramount in pharmaceutical analysis. The International Council for Harmonisation (ICH) guidelines Q3A (impurities in new drug substances), Q3B (impurities in new drug products), Q3C (residual solvents), and Q3D (elemental impurities) provide frameworks for impurity identification, qualification, and control [4]. The developed methods in these case studies were validated according to ICH Q2(R1) guidelines, assessing parameters including specificity, linearity, accuracy, precision, detection and quantification limits, and robustness [3] [38].
The integration of Quality by Design (QbD) principles in method development, as demonstrated in the meropenem trihydrate analysis study, enhances method robustness by systematically evaluating critical method parameters [39]. This approach, aligned with ICH Q8(R2) and Q14 guidelines, ensures methods remain reliable and reproducible when scaled from research to quality control settings [39].
These case studies demonstrate that green chromatographic methods are viable, sustainable alternatives to conventional approaches for impurity profiling in pharmaceuticals. The implemented methods successfully reduced environmental impact through:
The combination of robust method development, comprehensive validation per ICH guidelines, and multi-metric greenness assessment provides a template for implementing sustainable chromatography in pharmaceutical quality control. These approaches align with global initiatives for environmentally responsible analytical practices while maintaining the rigorous standards required for drug substance and product analysis.
The adoption of Green Chromatographic Methods for impurity profiling is a strategic priority in modern pharmaceutical development, aligning with global sustainability initiatives. However, a significant technical challenge persists: the potential trade-off between analytical performance and environmental benefits. Conventional methods often rely on large volumes of high-purity, hazardous solvents and energy-intensive processes to achieve the high sensitivity, resolution, and robustness required for detecting low-level impurities. When transitioning to greener alternatives, scientists frequently encounter limitations in sensitivity, precision, and detection limits, particularly for trace-level analytes. This application note details systematic strategies and optimized protocols to overcome these trade-offs, enabling the implementation of robust, sensitive, and sustainable chromatographic methods for impurity profiling.
A multi-pronged strategy is essential to mitigate sensitivity and performance trade-offs. The following table summarizes the core challenges and corresponding green solutions.
Table 1: Key Trade-offs and Strategic Mitigations in Green Chromatography
| Performance Challenge | Green Mitigation Strategy | Key Technique/Technology | Impact on Performance |
|---|---|---|---|
| Reduced detection sensitivity due to lower sample loading or smaller injection volumes. | Instrumental and detection system optimization. | Narrow-bore columns (â¤2.1 mm i.d.), UHPLC, and highly sensitive detectors (e.g., Corona Charged Aerosol Detector). | Up to 90% reduction in mobile phase consumption without sacrificing chromatographic performance; improved sensitivity from reduced chromatographic dilution [4] [40]. |
| Inadequate resolution from faster methods or alternative mobile phases. | Optimization of separation conditions and column technology. | Elevated Temperature Liquid Chromatography, improved core-shell particle columns. | Faster separations and reduced mobile phase viscosity, allowing for longer columns or smaller particles for enhanced resolution [4]. |
| Poor analyte detectability with green solvents (e.g., ethanol). | Adoption of advanced detection and data analysis. | Mass Spectrometry (MS) and Peak Purity Tests using Photodiode-Array (PDA) detection. | Provides unequivocal peak purity information and structural data, overcoming limitations of traditional UV detection, especially for trace impurities [40] [41]. |
| Method robustness and transferability issues. | Systematic method development and validation. | Analytical Quality by Design (AQbD) and robust validation protocols. | Ensures method reliability under deliberate, small variations in operational parameters, crucial for maintaining performance in green methods with narrower operational windows [40] [41]. |
Table 2: Key Research Reagent Solutions for Green Impurity Profiling
| Item/Category | Function & Green Rationale | Application Notes |
|---|---|---|
| Green Solvent Systems (Ethanol, Methanol, Water) | Replacement for acetonitrile in reversed-phase HPLC. Safer environmental and health profile [4] [40]. | Ethanol-water mixtures have been successfully used for enantiomer separation. Aqueous mobile phases are ideal for water-soluble analytes [4]. |
| Ionic Liquids/Deep Eutectic Solvents | Additives in mobile phase or for sample preparation. Improve peak shape and reduce organic solvent consumption [4]. | Useful for simple pharmaceutical separations to enhance selectivity and reduce solvent toxicity. |
| UHPLC & Narrow-Bore Columns (e.g., â¤2.1 mm i.d.) | Drastically reduces solvent consumption and waste generation per analysis while maintaining efficiency [4]. | Enables fast analysis with superior resolution. Requires instruments capable of handling high backpressures. |
| Core-Shell (Fused-Core) Stationary Phases | Provides high efficiency similar to sub-2µm particles but with lower system backpressure. | Allows for use of longer columns for complex separations or faster flow rates, enhancing throughput without requiring UHPLC-capable systems. |
| In-silico Modeling Software | Predicts analyte retention behavior and optimizes method parameters, reducing laboratory solvent use during method scouting [42]. | AI-driven software (e.g., ChromSword) automates and accelerates method development, saving time and resources. |
| Deacetylmatricarin | Austricin | High-purity Austricin (Desacetylmatricarin), a guaianolide sesquiterpene lactone from Artemisia species. For research applications such as anticancer and lipid-lowering studies. For Research Use Only. |
| 3-Methylsalicylic Acid | 3-Methylsalicylic Acid, CAS:83-40-9, MF:C8H8O3, MW:152.15 g/mol | Chemical Reagent |
The following workflow and detailed protocol outline the development and validation of a green HPLC method for the simultaneous analysis of active pharmaceutical ingredients (APIs) and their related impurities, based on a published approach [38].
The developed method must be validated per ICH guidelines [41]. Key parameters and acceptance criteria are summarized below.
Table 3: Key Method Validation Parameters and Targets
| Validation Parameter | Experimental Procedure | Acceptance Criteria |
|---|---|---|
| Accuracy/Recovery | Analyze samples spiked with known quantities of impurities at three concentration levels (n=9). | Recovery should be 98â102% for the API and 90â110% for impurities. |
| Precision (Repeatability) | Inject six replicates of a standard solution at 100% of the test concentration. | %RSD of peak areas should be ⤠2.0%. |
| Linearity | Analyze standards at a minimum of five concentration levels. | Correlation coefficient (r²) should be ⥠0.999 for the API and ⥠0.995 for impurities. |
| Specificity | Demonstrate baseline resolution between all analytes and from any placebo/degradant peaks. Use PDA for peak purity. | Peak purity index should be ⥠0.999, indicating no co-elution. |
| LOD & LOQ | Determine via signal-to-noise ratio (S/N) of 3:1 for LOD and 10:1 for LOQ. | LOQ should be sufficiently low to detect impurities at or below the reporting threshold (e.g., 0.1%). |
| Robustness | Deliberately vary parameters (e.g., flow rate ±0.1 mL/min, pH ±0.1, temperature ±2°C). | The method should remain unaffected by small variations, with %RSD meeting precision criteria. |
Greenness Assessment: Evaluate the final validated method using tools such as the Analytical Eco-Scale (aiming for a score >75, indicating excellent greenness) or the AGREE metric, which provides a comprehensive score based on all 12 principles of GAC [11] [38].
The successful implementation of green methods requires alignment with regulatory expectations. A Quality by Design (QbD) approach is highly recommended.
The linear "take-make-consume and dispose" model prevalent in analytical chemistry creates unsustainable environmental pressures through resource-intensive processes and waste generation [43]. Circular Analytical Chemistry (CAC) emerges as a transformative framework aiming to eliminate waste, circulate products and materials, and save resources by closing material loops within the analytical lifecycle [43]. For chromatographic methods in pharmaceutical impurity profiling, adopting CAC principles requires a radical transformation of the entire analytical systemâfrom production and consumption to waste management [43]. This transformation demands coordinated action from diverse stakeholders across academia, industry, regulatory agencies, and standard-setting organizations [44]. This application note provides a structured protocol for engaging these stakeholders to implement CAC principles specifically within green chromatographic methods for impurity profiling.
The CAC framework is organized around twelve strategic goals that provide a comprehensive roadmap for transitioning from linear to circular practices in analytical chemistry [43]. These goals target radical resource efficiency and waste elimination across the entire analytical process.
Table 1: The Twelve Goals of Circular Analytical Chemistry and Their Relevance to Impurity Profiling
| Goal Category | Specific Goal | Application to Green Chromatography |
|---|---|---|
| Resource Management | 1. Save resources | Minimize solvent and energy use via UHPLC, narrow-bore columns [4] [45] |
| 2. Circulate products and materials | Recycle solvents; reuse columns and components [43] | |
| 3. Eliminate waste | Convert analytical waste to valuable by-products [43] | |
| Hazard Reduction | 4. Minimize hazards | Replace acetonitrile with ethanol or methanol in mobile phases [4] [45] |
| 5. Minimize energy consumption | Use ambient temperature chromatography; reduce analysis time [4] | |
| Systemic Change | 6. Democratize analytical chemistry | Develop affordable, maintainable instruments [43] |
| 7. Prioritize renewable resources | Source solvents from biomass (e.g., Cyrene) [45] | |
| 8. Recover and reuse materials | Implement solvent recovery systems in labs [43] | |
| 9. Embrace digital technology | Use digital tools for method transfer & data sharing [43] | |
| 10. Manufacture for longevity | Design durable instruments & columns [43] | |
| 11. Diversify and connect | Develop multi-analyte methods for impurity profiling [43] | |
| 12. Think in systems and ecosystems | Adopt holistic lifecycle management of analytical assets [43] |
Successful implementation of CAC requires systematic engagement of a broad alliance of stakeholders who can affect or are affected by the transition to circularity in analytical practices [43] [46]. The process unfolds through specific, interconnected steps.
Stakeholder Engagement Process for CAC illustrates the four-step iterative process for engaging stakeholders in circular economy goals, adapted for CAC implementation [46]. The process begins with the Antecedent Context of existing unsustainable linear practices in analytical chemistry [43]. Step 1 involves identifying and prioritizing key stakeholders based on their power, legitimacy, urgency, and circularity-related competencies [46]. Step 2 uses one-way communication to secure stakeholder interest through awareness campaigns highlighting CAC benefits [46]. Step 3 implements integrative practices including two-way dialogue, collaborative projects, and mutual learning [46]. Step 4 involves evaluating outcomes against CAC metrics and creating feedback loops for continuous process improvement [46].
Table 2: Key Stakeholder Groups and Their Roles in CAC Implementation for Impurity Profiling
| Stakeholder Group | Primary Role in CAC | Specific Responsibilities |
|---|---|---|
| Pharmaceutical Industry & Manufacturers | Implement circular practices in QA/QC labs | - Adopt green solvent alternatives [45]- Invest in energy-efficient UHPLC systems [4]- Implement solvent recovery protocols [43] |
| Researchers & Academia | Develop innovative green analytical methods | - Design bio-based solvents [45]- Develop miniaturized sample prep [44]- Create multi-analyte impurity methods [43] |
| Regulatory Agencies & Pharmacopeias | Create enabling regulatory frameworks | - Update validation criteria to include green metrics [44]- Phase out resource-intensive official methods [44]- Provide technical guidance for green method adoption [45] |
| Instrument Manufacturers | Design circular instruments and components | - Develop durable, repairable chromatographs [43]- Create column refurbishment programs [43]- Design energy-efficient UHPLC systems [45] |
| Standard-Setting Organizations (ISO, CEN) | Establish green standards and metrics | - Integrate greenness assessments into method standards [44]- Develop CAC-specific certification protocols [43] |
This protocol provides a systematic approach for transitioning traditional HPLC methods to greener alternatives for pharmaceutical impurity profiling, focusing on solvent substitution and efficiency improvements [45].
Materials and Reagents:
Procedure:
This protocol standardizes the evaluation of greenness for impurity profiling methods using four complementary metric tools to provide a comprehensive sustainability profile [3] [13].
Materials and Software:
Procedure:
NEMI Pictogram Assessment:
Green Analytical Procedure Index (GAPI):
Analytical GREEnness (AGREE) Metric:
Comparative Analysis:
Table 3: Essential Materials for Green Chromatographic impurity Profiling
| Material/Technology | Function in CAC | Application Example |
|---|---|---|
| Ethanol-water mobile phases | Green alternative to acetonitrile | Separation of enantiomers and pharmaceutical impurities with reduced environmental impact [4] |
| Supercritical COâ | Principal solvent in Supercritical Fluid Chromatography (SFC) | Significantly reduces organic solvent consumption while providing excellent selectivity [4] |
| Narrow-bore columns (â¤2.1 mm ID) | Reduce mobile phase consumption | Impurity profiling with up to 90% reduction in solvent use compared to standard 4.6 mm columns [4] |
| UHPLC systems with sub-2μm particles | Enhance separation efficiency | Faster analysis with 80% solvent reduction while maintaining or improving separation [4] |
| Dihydrolevoglucosenone (Cyrene) | Bio-based solvent from renewable feedstock | Emerging sustainable solvent for liquid chromatography with promising application potential [45] |
| Ionic liquids | Green mobile phase additives | Improve peak quality and reduce organic solvent consumption in pharmaceutical separations [4] |
| Lactiflorasyne | Lactiflorasyne, CAS:107259-45-0, MF:C19H22O5, MW:330.4 g/mol | Chemical Reagent |
Implementing a Circular Analytical Chemistry framework for impurity profiling requires coordinated action across multiple stakeholder groups following a structured engagement process. The protocols and tools presented herein provide practical guidance for transitioning from linear to circular chromatographic methods. By adopting green solvent alternatives, enhancing method efficiency, and implementing comprehensive greenness assessments, pharmaceutical analysts can significantly reduce the environmental footprint of impurity profiling methods while maintaining regulatory compliance. The stakeholder engagement framework ensures that all relevant parties contribute to and benefit from this transition, creating a collaborative pathway toward sustainable analytical chemistry practices.
In the pursuit of sustainability, high-throughput pharmaceutical laboratories often encounter a paradoxical phenomenon known as the rebound effect. This effect occurs when efficiency gains from technological advancements are partially or completely offset by subsequent increases in consumption or scale of operations [47]. Within the context of green chromatographic methods for impurity profiling, this manifests when time or resource savings achieved through streamlined methods are redirected toward additional analyses, expanded testing panels, or reduced instrument downtime, ultimately negating anticipated environmental benefits [4] [47]. Understanding and mitigating this effect is crucial for laboratories committed to genuinely sustainable practices that align with the principles of green analytical chemistry (GAC).
The pharmaceutical industry faces particular challenges in this regard, as regulatory requirements for impurity profiling demand rigorous testing while simultaneously pushing for greener approaches [4]. Green chromatography techniques, including ultra-high performance liquid chromatography (UHPLC), supercritical fluid chromatography (SFC), and minimized sample preparation methods, undoubtedly offer reduced solvent consumption and waste generation per analysis [4] [3]. However, without strategic implementation, these gains may be compromised by expanded testing protocols or continuous instrument operation. This application note outlines practical strategies and detailed protocols to help researchers, scientists, and drug development professionals recognize, quantify, and mitigate the rebound effect, thereby ensuring that green chromatographic methods deliver their full potential environmental benefits within impurity profiling research.
To effectively manage the rebound effect, laboratories must first establish clear metrics for its quantification. The following KPIs enable objective assessment of both efficiency gains and their potential rebound consequences in impurity profiling workflows.
Table 1: Key Performance Indicators for Assessing the Rebound Effect
| Category | KPI | Measurement Method | Target Trend |
|---|---|---|---|
| Resource Efficiency | Solvent consumption per sample | (Total solvent volume / Number of samples) | Decreasing |
| Energy consumption per analysis | kWh meter readings per analytical batch | Decreasing | |
| Waste generated per sample | Total waste mass / Number of samples | Decreasing | |
| Operational Output | Samples analyzed per time unit | Number of samples / Instrument hours | Stable or controlled increase |
| Instrument utilization rate | Scheduled runtime / Total available time | Optimized, not maximized | |
| Environmental Impact | E-factor | (Mass of waste / Mass of analytes) | Decreasing |
| Analytical method greenness scores | NEMI, AES, GAPI, AGREE metrics | Improving |
The magnitude of the rebound effect (RE) can be quantified using the following equation, adapted from energy economics to analytical chemistry contexts:
RE = (Expected Savings - Actual Savings) / Expected Savings à 100%
Where:
For example, if a laboratory implements UHPLC methodology with an expected solvent reduction of 80% compared to conventional HPLC, but actual operational data show only a 40% reduction due to increased testing volumes, the rebound effect would be calculated as:
RE = (80% - 40%) / 80% Ã 100% = 50%
This indicates that half of the potential environmental benefits have been offset by expanded operations [4] [47]. Regular calculation of this metric across different green method implementations provides crucial data for targeted mitigation strategies.
Effective mitigation requires a multidimensional strategy addressing technological, organizational, and environmental factors. The TOE framework, adapted from sustainability science, provides a comprehensive structure for intervention [48].
Selecting appropriately green methods forms the foundation of rebound effect mitigation. The following protocol ensures method choices align with both analytical and sustainability requirements.
Protocol 3.2.1: Systematic Green Method Assessment for Impurity Profiling
Purpose: To select chromatographic methods that minimize environmental impact without compromising analytical performance in impurity profiling.
Materials:
Procedure:
Screen Potential Methods
Apply Greenness Assessment Metrics
Conduct Comparative Lifecycle Analysis
Select and Validate Optimal Method
Validation Criteria: Method must meet all regulatory requirements for impurity profiling while demonstrating improved greenness metrics compared to conventional approaches.
Method miniaturization represents one of the most effective approaches to inherently limit the potential for rebound effects by physically constraining resource consumption.
Protocol 4.1.1: UHPLC Method Transfer with Active Consumption Capping
Purpose: To implement UHPLC methods for impurity profiling with built-in safeguards against consumption rebound.
Materials:
Procedure:
Implement Consumption Monitoring
Optimize for Elevated Temperature Operations
Establish Usage Caps
Troubleshooting: If separation efficiency decreases with miniaturization, optimize gradient programs or consider core-shell particles for enhanced efficiency. If consumption caps regularly interrupt workflow, reassess throughput planning and resource allocation.
Strategic solvent management prevents rebound effects by addressing the environmental impact of mobile phase choices.
Protocol 4.2.1: Green Mobile Phase Implementation for Impurity Profiling
Purpose: To replace hazardous solvents with greener alternatives while maintaining chromatographic performance for impurity separation.
Materials:
Procedure:
Method Re-Development and Validation
Implement Solvent Recovery
Document Environmental Impact
Validation: Method must maintain resolution of all known impurities and demonstrate equivalent or improved precision and accuracy compared to original methods.
Successful implementation of rebound-resistant green chromatographic methods requires specific materials and reagents selected for both analytical performance and environmental profile.
Table 2: Essential Research Reagents and Materials for Green Impurity Profiling
| Category | Item | Specifications | Green Rationale | Application in Impurity Profiling |
|---|---|---|---|---|
| Chromatography Columns | Narrow-bore UHPLC columns | 1.0-2.1mm ID, sub-2μm particles | Reduces mobile phase consumption by up to 90% [4] | High-resolution separation of complex impurity mixtures |
| Green Solvents | Ethanol-water mixtures | HPLC grade, variable ratios | Replaces acetonitrile with biodegradable alternative [4] | Mobile phase for reversed-phase impurity separation |
| Supercritical COâ | SFC grade with modifiers | Non-toxic, recyclable mobile phase [4] | Chiral separations and non-polar impurity profiling | |
| Sample Preparation | Molecularly Imprinted Polymers (MIPs) | Specific to target analyte class | Reduces solvent consumption in sample cleanup [4] | Selective extraction of trace impurities from complex matrices |
| Green Additives | Ionic liquids | e.g., imidazolium salts | Enhance separation efficiency, reduce organic modifier needs [4] | Mobile phase additives for peak shape improvement |
| Assessment Tools | AGREE software | Open access metric tool | Comprehensive greenness assessment of analytical methods [3] | Comparative evaluation of method environmental performance |
Sustained mitigation of the rebound effect requires ongoing monitoring and adjustment of laboratory practices. The following workflow ensures that green chromatography implementations deliver lasting environmental benefits.
Robust data collection and analysis forms the foundation for effective rebound effect monitoring and management.
Protocol 6.1.1: Rebound Effect Quantification and Reporting
Purpose: To systematically track, calculate, and interpret rebound effect metrics for continuous improvement of green chromatographic practices.
Materials:
Procedure:
Implement Regular Monitoring
Analyze Rebound Effect Trends
Implement Corrective Actions
Report and Refine
Interpretation Guidelines: A rebound effect below 20% generally indicates successful mitigation, while values exceeding 20% warrant intervention. Consistent rebound effects above 50% suggest fundamental issues in method implementation or operational practices that require comprehensive reassessment.
The implementation of green chromatographic methods for impurity profiling presents significant environmental advantages, but these benefits can be undermined by the rebound effect if not strategically managed. The protocols and strategies outlined in this application note provide a comprehensive framework for recognizing, quantifying, and mitigating this phenomenon in high-throughput pharmaceutical laboratories. By adopting the integrated technological, organizational, and environmental approaches describedâincluding method miniaturization, solvent replacement, consumption capping, and continuous monitoringâresearch scientists and drug development professionals can ensure that their sustainability initiatives deliver genuine environmental benefits. Through these concerted efforts, laboratories can advance both scientific excellence and environmental stewardship in pharmaceutical impurity profiling.
In the context of green chromatographic methods for impurity profiling, optimizing energy consumption and separation temperature is no longer merely a pursuit of analytical efficiency but a fundamental requirement for sustainable laboratory practices. The pharmaceutical industry faces increasing pressure to align impurity profilingâa critical process for ensuring drug safety and efficacyâwith the twelve principles of green analytical chemistry (GAC) [37]. These principles emphasize minimizing energy consumption, reducing waste generation, and adopting safer solvents [4] [37].
Traditional chromatographic techniques, particularly high-performance liquid chromatography (HPLC), are increasingly scrutinized for their significant environmental drawbacks, including high organic solvent consumption, substantial energy demands from extended operation times, and considerable waste generation [49] [50]. This application note details advanced techniques and practical protocols to achieve faster separations while concurrently reducing the environmental footprint of analytical methods, thereby supporting the pharmaceutical industry's transition toward more sustainable impurity profiling.
UHPLC technology leverages smaller particle sizes (often sub-2μm) and elevated operating pressures to achieve superior chromatographic efficiency. This allows for a drastic reduction in analysis time and solvent consumption compared to conventional HPLC. One study demonstrated that UHPLC could realize an 80% reduction in solvent usage while maintaining or even enhancing separation efficiency for complex pharmaceutical mixtures [4]. The environmental benefits are twofold: reduced solvent consumption directly minimizes waste, while shorter run times lower the energy demand of instruments like pumps, column ovens, and detectors [50].
Strategically increasing the temperature of the chromatographic column is a powerful tool for accelerating separations. Higher temperatures reduce mobile phase viscosity, which in turn allows for faster flow rates or the use of longer columns with smaller particles without generating excessive backpressure [4]. This leads to quicker elution of analytes and shorter overall method times, directly translating to lower energy consumption per analysis.
UFGC is a transformative approach for volatile compound analysis, offering analysis times 5â20 times faster than conventional GC [51]. Key to its performance is the use of short columns with specific internal diameters and a directly heated column compartment with very rapid heating ramps (e.g., 60â200 °C/min). This design minimizes the thermal mass that needs to be heated and cooled, resulting in dramatically reduced cycle times and, consequently, significantly lower energy consumption, especially in high-throughput environments [51].
SFC stands out as a green technique by utilizing supercritical COâ as the primary mobile phase [49]. COâ is non-toxic, non-flammable, and can be sourced from renewable processes. Its low viscosity and high diffusivity enable faster separations compared to traditional liquid chromatography, reducing both organic solvent use and energy consumption [49].
Table 1: Quantitative Comparison of Green Chromatographic Techniques
| Technique | Key Feature | Analysis Time Reduction | Solvent/Waste Reduction | Primary Energy Saving Mechanism |
|---|---|---|---|---|
| UHPLC | Smaller particles, higher pressure | Up to 80% faster than HPLC [4] | Up to 80% less solvent [4] | Shorter run times, reduced solvent volume to manage |
| Elevated Temp LC | Higher column temperatures | Significant (viscosity reduction) [4] | Moderate (faster elution) | Shorter run times |
| Ultrafast GC | Rapid heating (up to 200°C/min), short columns | 5â20x faster than conventional GC [51] | Comparable sample capacity | Rapid cool-down (30-90 sec), less energy per run [51] |
| SFC | Supercritical COâ mobile phase | Faster due to high diffusivity [49] | Major reduction in organic solvents | Non-thermal process, faster separations |
This protocol provides a guideline for transferring and optimizing a conventional HPLC method to a UHPLC platform to enhance speed and sustainability.
Research Reagent Solutions & Materials:
Procedure:
This protocol outlines key steps for developing a fast, energy-efficient GC method suitable for applications like residual solvent analysis.
Research Reagent Solutions & Materials:
Procedure:
Diagram: UFGC Method Development Workflow. This logic flow outlines the key steps for developing an Ultrafast GC method, from initial setup to final optimization.
Table 2: Essential Research Reagent Solutions for Green Chromatographic Separations
| Item | Function/Application | Green & Performance Benefits |
|---|---|---|
| Ethanol-Water Mobile Phases | Replacement for acetonitrile in reversed-phase LC [4] | Lower toxicity, biodegradable, reduced environmental impact and cost. |
| Supercritical COâ | Primary mobile phase in Supercritical Fluid Chromatography (SFC) [49] | Non-toxic, non-flammable, eliminates large volumes of organic solvent waste. |
| Hydrogen Carrier Gas | Carrier gas for Gas Chromatography [51] [49] | Enables faster optimal linear velocities, reducing GC run times and energy use. |
| Narrow-Bore Columns (â¤2.1 mm i.d.) | Used in liquid chromatography for reduced solvent consumption [4] | Can reduce mobile phase use by up to 90% compared to standard 4.6 mm columns. |
| UHPLC Columns (sub-2μm particles) | High-efficiency columns for UHPLC systems [4] [50] | Enable faster separations and lower solvent consumption while maintaining resolution. |
| Ionic Liquids | Additives in mobile phases or as green solvent alternatives [4] | Can improve peak shape and reduce overall organic solvent consumption. |
Adopting the techniques described herein allows pharmaceutical researchers and drug development professionals to make significant strides in greening their impurity profiling workflows. The strategic optimization of temperature and energy parameters not only accelerates separations but also aligns with the core tenets of green analytical chemistry.
To quantitatively evaluate the environmental benefits of newly developed methods, scientists should employ established greenness assessment tools such as the Analytical Eco-Scale, GAPI (Green Analytical Procedure Index), or AGREE metric [37]. These tools provide a standardized framework for comparing the environmental footprint of analytical methods, considering factors like energy consumption, waste production, and reagent toxicity.
The transition to greener chromatographic methods for impurity profiling, driven by the optimization of energy and temperature, represents a win-win scenario. It simultaneously enhances laboratory efficiency, reduces operational costs, and fulfills the growing regulatory and ethical imperatives for sustainable scientific practices [49] [37].
Impurity profiling is a critical pillar of pharmaceutical quality control, directly impacting drug safety, efficacy, and stability [4]. This process is governed by stringent guidelines from the International Council for Harmonisation (ICH) and the United States Pharmacopeia (USP), which classify impurities into specific categories to ensure consistent control and reporting [4]. Concurrently, the paradigm of Green Analytical Chemistry (GAC) has gained substantial momentum, aiming to minimize the environmental impact of analytical methods by reducing solvent consumption, minimizing waste generation, and improving energy efficiency, all without compromising analytical performance [4] [11].
The integration of GAC principles into regulated pharmaceutical analysis presents a unique challenge: transitioning to sustainable practices while maintaining full compliance with evolving regulatory standards. This document provides detailed application notes and protocols to navigate this complex landscape, offering practical strategies for adopting green chromatographic methods in impurity profiling.
The following table summarizes the core regulatory guidelines that govern impurity profiling in pharmaceutical development.
Table 1: Key ICH and Regulatory Guidelines for Impurities
| Guideline | Scope and Depiction |
|---|---|
| ICH Q3A (R2) | Impurities in New Drug Substances |
| ICH Q3B (R2) | Impurities in New Drug Products |
| ICH Q3C | Guidelines for Residual Solvents |
| ICH Q3D | Guidelines for Elemental Impurities |
| US-FDA (NDAs) | Impurities in New Drug Substances |
| US-FDA (ANDAs) | Impurities in New Drug Substances |
The advancement of green liquid chromatography (GLC) focuses on achieving analytical performance comparable to conventional HPLC while significantly reducing its environmental footprint. Key strategies include the use of greener mobile phases, instrument miniaturization, and operational modifications [4] [11].
This protocol outlines the systematic procedure for transferring a traditional HPLC method for impurity analysis to a more sustainable Ultra-High-Performance Liquid Chromatography (UHPLC) method, leveraging the principles of green chemistry.
Title: Transfer of an Impurity Profile Method from HPLC to UHPLC Objective: To achieve a faster, more efficient, and environmentally friendly separation of process-related and degradation impurities in a active pharmaceutical ingredient (API) while maintaining or improving resolution and sensitivity. API: [Specify API Name] HPLC System: [Specify System], Column: [Specify Column, e.g., 150 mm x 4.6 mm, 5 µm] UHPLC System: [Specify System], Column: [Specify Column, e.g., 50 mm x 2.1 mm, 1.7 µm]
Procedure:
Anticipated Outcomes:
The following table details key materials and reagents essential for implementing green chromatographic methods.
Table 2: Essential Reagents and Materials for Green Chromatography
| Item | Function & Rationale | Green Alternative Example |
|---|---|---|
| Ethanol or Methanol | Acts as a replacement for acetonitrile in mobile phases. Ethanol is biodegradable, less toxic, and sourced from renewable origins. | Ethanol-Water Mixtures [4] |
| Water | Serves as the primary solvent in aqueous mobile phases, eliminating organic solvent use for water-soluble analytes. | High-Purity Water [4] |
| Ionic Liquids | Used as mobile phase additives to improve peak shape and selectivity, potentially reducing the required percentage of organic modifiers. | e.g., Ammonium acetate-based solutions [4] |
| Supercritical COâ | The primary mobile phase in Supercritical Fluid Chromatography (SFC), replacing a significant portion of organic solvents. | Carbon Dioxide (SFC Grade) [4] |
| Narrow-Bore Columns | Columns with internal diameters ⤠2.1 mm. Drastically reduce mobile phase consumption and waste generation. | UHPLC Columns (e.g., 2.1 mm ID, 1.7-1.8 µm particles) [4] |
Successfully navigating regulatory hurdles requires not only adopting new techniques but also rigorously validating them and assessing their environmental benefits using standardized tools.
The table below provides a comparative analysis of different chromatographic approaches based on data from technique advancements.
Table 3: Quantitative Comparison of Chromatographic Techniques
| Technique | Key Green Feature | Reported Efficiency Gain | Considerations |
|---|---|---|---|
| UHPLC | Reduced solvent consumption via small particle sizes and high pressures. | Up to 80-90% solvent reduction compared to HPLC [4]. | Requires instrumentation capable of withstanding high pressures. |
| Narrow-Bore Columns | Reduced mobile phase consumption via smaller column diameters. | Up to 90% decrease in mobile phase vs. 4.6 mm columns [4]. | Requires instrument systems with low extra-column volume. |
| Supercritical Fluid Chromatography (SFC) | Uses supercritical COâ as the primary mobile phase. | Significant reduction in organic solvent use [4]. | Ideal for non-polar to moderately polar compounds. |
| Elevated Temperature LC | Increases mobile phase efficiency, allowing for faster flow rates or smaller particles. | Faster separations and/or use of water-rich mobile phases [4]. | Requires a column oven that can maintain stable high temperatures. |
To objectively demonstrate the sustainability of new methods, several metric tools have been developed and are increasingly recognized in the scientific literature [11].
Staying abreast of broader regulatory trends is also crucial. For instance, the recent finalization of the ICH E6(R3) Good Clinical Practice guidelines emphasizes flexibility, ethics, and the integration of digital technologies, which aligns with the principles of modern, sustainable analytical practices [52]. Furthermore, the FDA's ongoing initiatives, such as encouraging Diversity Action Plans in clinical trials, signal a regulatory environment that values broader, more representative data, which can be supported by robust and accessible green analytical methods [52].
The following diagram illustrates the logical workflow for developing, validating, and gaining regulatory acceptance for a new green analytical method.
In the pharmaceutical industry, impurity profiling is critical for ensuring drug safety, efficacy, and stability [4]. As the field evolves, there is a growing emphasis on aligning these analytical procedures with the principles of Green Analytical Chemistry to minimize environmental impact [4] [37]. Green Analytical Chemistry (GAC) aims to reduce the consumption of hazardous solvents, minimize waste generation, and lower energy consumption without compromising the quality of analytical results [4] [53]. Evaluating the environmental friendliness of analytical methods requires specialized metrics. This overview focuses on three prominent greenness assessment toolsâAGREE, GAPI, and Analytical Eco-Scaleâwithin the context of green chromatographic methods for impurity profiling research. The selection of an appropriate greenness metric enables researchers and drug development professionals to quantify the environmental impact of their analytical methods and guide the development of more sustainable laboratory practices [37] [54].
The Analytical Eco-Scale is a semi-quantitative assessment tool proposed in 2012 [54]. It operates on a penalty points system, where an ideal green analysis starts with a base score of 100 points [55] [54]. Points are subtracted for each reagent, chemical, or procedural element that deviates from ideal green conditions, considering their quantity, toxicity, and environmental impact [54]. Additional penalties apply for high energy consumption (>0.1 kWh per sample) and waste generation [54]. The final score provides a straightforward interpretation: >75 represents excellent green analysis, 50-74 indicates acceptable greenness, and <50 signifies inadequate greenness [56]. While valued for its simplicity and quantitative output, a limitation is its lack of visual component and insufficient accounting for the severity of hazard pictograms associated with chemicals [56].
The Green Analytical Procedure Index (GAPI), introduced in 2018, offers a more comprehensive visual assessment of the entire analytical procedure [57] [37]. Its pictogram evaluates five key areas: sample collection, preservation, transportation, preparation, and final determination [57]. Each area is divided into several sub-divisions that are colored green, yellow, or red based on the environmental impact of that specific step, providing an immediate visual overview of the method's greenness [57] [56]. A significant advantage of GAPI is its ability to visually pinpoint less environmentally friendly steps within an analytical workflow [53]. However, a noted drawback is the absence of a single numerical score, which can make direct comparison between different methods challenging [56]. Recent advancements have led to Modified GAPI (MoGAPI), which incorporates a scoring system to address this limitation [56].
The AGREE metric, developed in 2020, represents a significant advancement by incorporating all 12 principles of GAC into its evaluation framework [55] [37]. It transforms each principle into a score on a 0-1 scale, with user-defined weights to reflect the relative importance of different criteria [55]. The output is an intuitive, clock-like pictogram that displays both an overall score (0-1) in the center and a color-coded performance assessment for each of the 12 principles in the surrounding segments [55]. A score closer to 1 and a darker green center indicate a greener procedure [55]. AGREE is comprehensive, user-friendly (supported by freely available software), and facilitates easy comparison between methods [55] [58]. A specialized version, AGREEprep, has also been developed to focus specifically on the sample preparation stage, which is often the least green part of the analytical process [59].
The table below provides a consolidated comparison of the three primary greenness assessment tools.
Table 1: Comparative Analysis of Greenness Assessment Tools
| Feature | Analytical Eco-Scale | GAPI | AGREE |
|---|---|---|---|
| Year Introduced | 2012 [54] | 2018 [57] | 2020 [55] |
| Basis of Assessment | Penalty points for non-green aspects [54] | Evaluation of multiple stages in the analytical procedure [57] | 12 principles of Green Analytical Chemistry [55] |
| Output Format | Numerical score (0-100) [54] | Color-coded pictogram (green, yellow, red) [57] | Pictogram with overall score (0-1) and segmented performance [55] |
| Key Advantages | Simple, quantitative, easy comparison [54] | Visual identification of problematic steps [53] | Comprehensive, includes weighting, user-friendly software [55] |
| Main Limitations | No visual output; does not fully account for hazard severity [56] | No overall score for easy method comparison [56] | Subjective weighting of criteria [55] |
1. Data Collection: Gather all method parameters for the HPLC impurity profiling procedure. This includes the type and volume of solvents used in the mobile phase (e.g., acetonitrile, methanol, or greener alternatives like ethanol), sample preparation details (e.g., extraction solvents and volumes), energy consumption of the instrument (kWh per sample), and the volume and management of waste generated [55] [37].
2. Software Input: Access the freely available AGREE software [55] [58]. Input the collected data, corresponding to the 12 GAC principles. For example, specify the use of direct analysis or sample preparation steps (Principle 1), sample size (Principle 2), and reagent toxicity (Principle 5) [55].
3. Weight Assignment (Optional): Assign weights to the 12 principles if certain aspects are of greater concern for a specific application. For impurity profiling, higher weights might be assigned to reagent toxicity (Principle 5) and waste generation (Principle 4) [55].
4. Interpretation: The software generates a pictogram. An overall score above 0.75 is generally considered excellent, while a score below 0.5 indicates a significant need for method optimization to improve greenness [58]. The colored segments help identify which specific principles the method performs poorly on, guiding improvement efforts [55].
1. Establish Baseline: Begin with the ideal score of 100 points [54].
2. Calculate Penalties: Subtract penalty points for all non-ideal conditions. - Reagents and Solvents: Subtract points based on the type and quantity of reagents used. For example, using 10 mL of acetonitrile (hazardous solvent) would incur a higher penalty than using 10 mL of ethanol (a greener alternative) [54]. - Energy Consumption: Subtract points if energy consumption exceeds 0.1 kWh per sample [54]. - Waste and Occupational Hazards: Subtract points for waste generated and any occupational hazards encountered [54].
3. Final Scoring and Classification: Calculate the final score. Classify the method: >75 (excellent), 50-74 (acceptable), or <50 (inadequate) [56].
A 2022 study applied AGREEprep (the sample preparation-specific metric) to evaluate six different sample preparation procedures for determining phthalate esters in water [59]. The results demonstrated that microextraction techniques consistently achieved higher greenness scores (above 0.7) compared to traditional methods like standard liquid-liquid extraction (LLE), which scored as low as 0.24 [59]. The assessment highlighted that the use of large volumes of chlorinated solvents (e.g., dichloromethane) in LLE was a major contributor to its poor environmental performance [59]. This case study underscores the critical impact of sample preparation choices on the overall sustainability of an analytical method, a key consideration in impurity profiling where sample preparation is often indispensable [59].
Table 2: Key Software Tools for Greenness Assessment
| Tool Name | Primary Function | Access Information |
|---|---|---|
| AGREE Calculator | Comprehensive greenness assessment based on 12 GAC principles [55] | Freely available at: https://mostwiedzy.pl/AGREE [55] [58] |
| AGREEprep | Specialized assessment of sample preparation greenness [59] | Freely available at: https://mostwiedzy.pl/AGREE [59] [58] |
| MoGAPI Software | Modified GAPI tool providing an overall greenness score [56] | Freely available at: bit.ly/MoGAPI [56] |
The tools discussedâAnalytical Eco-Scale, GAPI, and AGREEâprovide researchers with a robust framework for quantifying and improving the environmental footprint of their analytical methods. For pharmaceutical impurity profiling, where methods like HPLC are indispensable, the adoption of these metrics is crucial for transitioning toward more sustainable laboratory practices. While the Analytical Eco-Scale offers a simple quantitative score, and GAPI provides detailed visual diagnostics, AGREE stands out for its comprehensive incorporation of the 12 GAC principles and user-friendly software support. The ongoing development of specialized tools like AGREEprep and MoGAPI indicates a dynamic field committed to continuous improvement. By integrating these greenness assessment tools into method development and validation workflows, scientists and drug development professionals can make significant strides in reducing the environmental impact of impurity profiling while maintaining the high analytical standards required for pharmaceutical quality control.
Within the framework of research on green chromatographic methods for impurity profiling, the adoption of Green High-Performance Liquid Chromatography (HPLC) has become a strategic priority for analytical laboratories aiming to align with sustainable development goals [37]. Conventional HPLC methods, while providing accuracy and sensitivity, often involve substantial consumption of hazardous organic solvents and generate significant chemical waste, contributing to environmental pollution and occupational health risks [37] [60]. This application note provides a comparative analysis of conventional versus green HPLC approaches, detailing practical protocols and assessment methodologies to enable researchers and drug development professionals to implement more sustainable impurity profiling methods without compromising analytical performance.
Green Analytical Chemistry (GAC) is structured around twelve guiding principles designed to minimize the environmental footprint of analytical procedures while maintaining scientific robustness [37]. These principles provide a framework for developing sustainable HPLC methods:
The transition from conventional to green HPLC requires a fundamental rethinking of method development, focusing not only on analytical performance but also on environmental impact across the entire analytical workflow [37].
Several validated assessment tools are available to quantitatively evaluate the environmental performance of HPLC methods. The most widely adopted metrics include:
Table 1: Key Greenness Assessment Tools for HPLC Methods
| Assessment Tool | Graphical Output | Scoring System | Key Assessment Parameters | Strengths |
|---|---|---|---|---|
| AGREE [37] [53] | Radial chart (0-1) | 0-1 (Higher = greener) | All 12 GAC principles | Comprehensive, user-friendly, single score output |
| GAPI [53] [60] | Color-coded pictogram | Qualitative (5-color levels) | Entire analytical workflow | Visual identification of high-impact stages |
| Analytical Eco-Scale [53] [60] | Numerical score | 100-point scale (Penalty deduction) | Reagent toxicity, energy, waste, occupational hazards | Simple scoring, direct method comparison |
| NEMI [53] [61] | 4-quadrant pictogram | Binary (Green/white) | PBT substances, hazardous chemicals, corrosives, waste | Simple, quick visual assessment |
| BAGI [37] [61] | Asteroid pictogram + % score | Percentage score | Analytical practicality, throughput, cost, efficiency | Evaluates practical applicability alongside greenness |
These tools enable systematic evaluation of HPLC methods, with AGREE and GAPI currently representing the most comprehensive approaches for comparative greenness analysis [53] [60].
This protocol describes the systematic conversion of a conventional HPLC method to a greener alternative through solvent replacement and column optimization.
This application-specific protocol demonstrates an environmentally sustainable HPLC method for simultaneous determination of active ingredients and related impurities.
Chromatographic Conditions:
Sample Preparation:
Validation Parameters:
This method demonstrates excellent greenness scores while maintaining robust separation of active pharmaceutical ingredients and their potential impurities [1].
A comparative study of normal-phase (NP) and reversed-phase (RP) HPTLC methods for the analysis of Ertugliflozin demonstrates the significant greenness advantages of optimized approaches:
Table 2: Comparative Analysis of NP-HPTLC vs. RP-HPTLC Methods for Ertugliflozin [63]
| Parameter | NP-HPTLC Method | RP-HPTLC Method |
|---|---|---|
| Mobile Phase | Chloroform/Methanol (85:15 v/v) | Ethanol-Water (80:20 v/v) |
| Linearity Range | 50-600 ng/band | 25-1200 ng/band |
| Tailing Factor | 1.06 ± 0.02 | 1.08 ± 0.03 |
| Theoretical Plates/m | 4472 ± 4.22 | 4652 ± 4.02 |
| Greenness Score | Moderate | Superior |
| Solvent Toxicity | Higher (Chloroform) | Lower (Ethanol) |
The RP-HPTLC method was found to be more robust, accurate, precise, linear, sensitive, and eco-friendly compared to the NP-HPTLC approach [63]. Assessment using four greenness tools (NEMI, AES, ChlorTox, and AGREE) confirmed the RP strategy as greener than the NP method and all other reported HPLC techniques [63].
A comprehensive assessment of HPLC methods for paclitaxel quantification using seven greenness tools revealed that methods incorporating solvent reduction and alternative solvents achieved significantly improved sustainability profiles [61]. The highest-performing methods demonstrated:
Table 3: Essential Materials for Green HPLC Method Development
| Item | Function in Green HPLC | Green Characteristics |
|---|---|---|
| Ethanol | Primary organic modifier replacing acetonitrile [4] [1] | Biobased, renewable, low toxicity, biodegradable |
| Methanol | Alternative organic modifier [4] [62] | Lower environmental impact than acetonitrile |
| Narrow-bore Columns (â¤2.1 mm i.d.) [62] | Reduces mobile phase consumption | Enables 80% solvent reduction vs. standard 4.6 mm columns |
| Sub-2-µm Particles [62] | Enhances separation efficiency | Reduces analysis time and solvent consumption |
| Superficially Porous Particles [62] | Improves chromatographic performance | 50% solvent reduction vs. fully porous particles of same size |
| Water-Only Mobile Phases [4] | Eliminates organic solvents | Zero organic solvent consumption |
| Ionic Liquids [4] | Mobile phase additives for challenging separations | Reduces required organic solvent percentage |
The following workflow outlines the decision-making process for implementing green HPLC methods in pharmaceutical impurity profiling:
The comparative analysis demonstrates that green HPLC methods can provide equivalent or superior analytical performance while significantly reducing environmental impact compared to conventional approaches. Key success factors include:
Adoption of these protocols and assessment strategies enables pharmaceutical researchers to advance impurity profiling methodologies that align with both analytical excellence and environmental responsibility.
White Analytical Chemistry (WAC) represents an advanced, holistic framework designed to evaluate the overall sustainability and practicality of analytical methods. Proposed as an extension of Green Analytical Chemistry (GAC), WAC addresses its predecessor's primary limitationâthe predominant focus on environmental impactâby integrating two additional critical dimensions: analytical performance and practical efficiency [64]. The ultimate goal of WAC is to achieve a harmonious balance among these three pillars, designating a method that excels in all areas as "white," a term inspired by the additive color model where white light results from the combination of red, green, and blue light [65] [13].
The conceptual foundation of WAC is built upon twelve principles, structured into three distinct categories, each symbolized by a color in the RGB model [64]:
This report delineates the application of the WAC framework and its associated RGB model within the specific context of developing green chromatographic methods for impurity profiling in pharmaceuticals, providing detailed protocols and application notes for researchers and drug development professionals.
The RGB model offers a structured, semi-quantitative approach for evaluating analytical methods, moving beyond mere environmental concerns to provide a comprehensive sustainability profile. The "whiteness" of a method is conceptualized as the harmonious synthesis of the three color components [64] [13].
Table 1: The Twelve Principles of White Analytical Chemistry (WAC) Based on the RGB Model
| Color Category | Underlying Principles | Description and Key Concerns |
|---|---|---|
| Green (4 Principles) | Derived from GAC | Minimization of reagent toxicity, sample volume, solvent consumption, and generated waste; reduction of energy consumption and operator risk [64]. |
| Red (4 Principles) | Analytical Performance | Scope of application (specificity/selectivity), detection and quantification limits, precision, and trueness (accuracy) [64]. |
| Blue (4 Principles) | Practical & Economic Efficiency | Total cost of the analytical process, time efficiency, simplicity, and minimal requirement for manual operations or specialized equipment [64]. |
The workflow for applying the RGB model, from method development to its final whiteness assessment, can be visualized as a logical sequence of steps, culminating in a unified score.
In practice, each of the twelve principles is assigned a score, typically on a scale of 0 to 4. The scores within each color category are averaged to calculate a final Color Score (CS) for green (CSg), red (CSr), and blue (CSb) [13]. The overall whiteness is then determined by how close these three color scores are to each other and to the ideal maximum value. A perfectly balanced method with high scores in all three categories is deemed "white," indicating a sustainable, high-performing, and practical analytical technique [65] [64].
The application of WAC and the RGB model leads to the development of analytical methods that are not only environmentally responsible but also robust and efficient. The following section outlines key strategic approaches and a specific case study in pharmaceutical impurity profiling.
Several chromatographic techniques align well with the principles of WAC by inherently addressing green and blue criteria without compromising red (analytical) performance.
Green Liquid Chromatography (GLC): GLC focuses on reducing hazardous solvent consumption. Key strategies include:
Supercritical Fluid Chromatography (SFC): SFC uses supercritical COâ as the primary mobile phase constituent. This drastically reduces the need for organic solvents, aligning excellently with green principles. Its high efficiency and rapid separation capabilities also satisfy red and blue criteria [4].
Capillary Electrophoresis (CE): CE is recognized for its high separation efficiency with minimal consumption of solvents and samples, making it a strong candidate for green impurity profiling methods [4].
A recent application involved developing a stability-indicating RP-HPLC method for the simultaneous assay and impurity profiling of Amitriptyline HCl. The method was rigorously assessed using multiple green metrics and the WAC RGB model [13].
Chromatographic Conditions:
Outcomes:
This method was evaluated as a "white" method, demonstrating that it is a sustainable, functional, and practical solution for ensuring pharmaceutical quality [13].
This section provides a detailed, step-by-step protocol for developing and validating an impurity profiling method in accordance with WAC principles, using the cited amitriptyline study as a template.
1. Objective: To develop and validate a reverse-phase HPLC method for the simultaneous assay and impurity profiling of a drug substance that aligns with the principles of White Analytical Chemistry.
2. Materials and Reagents Table 2: Research Reagent Solutions and Essential Materials
| Item | Function / Specification | Notes for Greenness |
|---|---|---|
| Drug Substance & Impurities | Reference standards for API and known impurities. | Required for validation of Red principles (specificity, accuracy). |
| Acetonitrile (HPLC Grade) | Organic modifier in mobile phase. | Prefer ethanol or methanol as greener alternatives where feasible [4]. |
| Potassium Dihydrogen Phosphate (KHâPOâ) | Buffer salt for aqueous mobile phase component. | -- |
| Phosphoric Acid | For pH adjustment of mobile phase buffer. | -- |
| Triethylamine (TEA) | Mobile phase additive to improve peak shape. | Use at minimal necessary concentration. |
| Water (HPLC Grade) | Solvent for mobile phase and diluent. | -- |
| HPLC System | With quaternary pump, autosampler, column oven, and PDA detector. | -- |
| L1 Column (C18) | 150 x 4.6 mm, 2.6 µm particle size. | Consider narrow-bore (e.g., 2.1 mm) to reduce solvent use [4]. |
3. Instrumental Conditions and Method Development
4. Forced Degradation Studies (Stress Testing)
5. Method Validation Validate the method as per ICH guidelines to establish its performance (Red Principles):
6. Greenness and Whiteness Assessment
The experimental workflow, from sample preparation to final assessment, integrates these steps into a cohesive whole.
White Analytical Chemistry, visualized through the intuitive RGB model, provides a comprehensive and balanced framework for the modern analytical scientist. It moves the field beyond a singular focus on greenness, advocating for methods that are simultaneously environmentally sustainable (Green), analytically reliable (Red), and economically practical (Blue). The application of this triad in impurity profiling guides the development of techniques like Green Liquid Chromatography, Supercritical Fluid Chromatography, and Capillary Electrophoresis, ensuring that the imperative for pharmaceutical quality and safety is met without compromising environmental and operational responsibilities. By adopting the protocols and assessment tools outlined in this document, researchers and drug development professionals can effectively design, implement, and validate "white" methods that align with the overarching goals of sustainable science.
The Blue Applicability Grade Index (BAGI) is a novel metric tool designed to evaluate the practicality and applicability of analytical methods, serving as a crucial complement to established green chemistry metrics [66] [67]. Within the context of green chromatographic methods for impurity profiling, BAGI addresses the pressing need to balance environmental considerations with the practical demands of pharmaceutical analysis. While green assessment tools like AGREE and GAPI effectively evaluate environmental impact, they do not fully account for practical aspects such as analysis throughput, instrumentation requirements, and operational efficiency [68]. BAGI fills this critical gap by providing a standardized approach to assess whether a green method is truly practical for implementation in routine drug development and quality control environments.
The fundamental premise of BAGI aligns with the principles of White Analytical Chemistry, which advocates for a harmonious balance between analytical method greenness, practicality, and societal acceptability [66]. For impurity profiling research, this balance is particularly crucial as regulatory requirements demand sensitive, reliable, and high-throughput methods for detecting and quantifying potentially toxic impurities, even at trace levels [2]. The integration of BAGI into method development and validation workflows ensures that newly developed chromatographic methods for impurity profiling are not only environmentally sustainable but also practically viable for widespread adoption in pharmaceutical laboratories.
The BAGI assessment framework is structured around ten key attributes that collectively provide a comprehensive picture of an analytical method's practicality [66]. These criteria were carefully selected to cover all fundamental aspects of method implementation and routine application in analytical laboratories. For impurity profiling research, each criterion carries significant weight in determining whether a chromatographic method can be effectively deployed in pharmaceutical quality control settings.
Table 1: Core Evaluation Criteria of the Blue Applicability Grade Index (BAGI)
| Criterion Number | Evaluation Attribute | Description and Relevance to Impurity Profiling |
|---|---|---|
| 1 | Type of analysis | Determines whether the method is qualitative, quantitative, or both; crucial for impurity identification and quantification. |
| 2 | Number of simultaneous analytes | Assesses the method's capability to profile multiple impurities in a single run, enhancing throughput. |
| 3 | Samples analyzed per hour | Measures analysis speed, directly impacting method productivity for high-volume quality control. |
| 4 | Type of reagents and materials | Evaluates the availability, cost, and safety of required chemicals, affecting operational practicality. |
| 5 | Required instrumentation | Considers the sophistication, availability, and maintenance requirements of analytical equipment. |
| 6 | Samples simultaneously treated | Assesses parallel processing capability, important for high-throughput impurity screening. |
| 7 | Preconcentration requirement | Determines if additional sample treatment is needed, impacting method complexity and time. |
| 8 | Automation degree | Evaluates the level of automated processes, influencing reproducibility and operator dependency. |
| 9 | Type of sample preparation | Examines the complexity, time, and skill requirements for sample pretreatment before analysis. |
| 10 | Amount of sample | Considers the required sample quantity, particularly relevant for precious or limited samples. |
The evaluation process generates both a numerical score and a visual asteroid pictogram, providing an immediate, intuitive representation of the method's practicality profile [66] [67]. The pictogram uses a sequential blue color scale, with dark blue indicating high compliance, blue for medium compliance, light blue for low compliance, and white indicating no compliance with the set criteria. This visual tool enables researchers to quickly identify the strong and weak points of an analytical method in terms of its practical application.
The BAGI scoring system transforms the qualitative assessment of the ten criteria into a quantitative practicality score that facilitates objective comparison between different analytical methods. Each criterion is evaluated against predefined benchmarks, with points assigned based on the level of compliance with ideal practicality parameters. The cumulative score falls within a range that categorizes the method's overall practicality:
In a recent application, a GC-MS method for simultaneous analysis of paracetamol and metoclopramide in pharmaceuticals and plasma achieved a BAGI score of 82.5, indicating high practicality for routine quality control and pharmacokinetic studies [69]. This high score reflected the method's rapid 5-minute runtime, minimal sample preparation, and excellent precision and accuracy.
BAGI Scoring Workflow: This diagram illustrates the systematic process for assessing analytical method practicality, from initial evaluation through final classification.
The initial phase of BAGI implementation requires meticulous documentation of all method parameters and sample preparation steps. For impurity profiling in pharmaceutical formulations using chromatographic methods, this involves detailed recording of sample extraction procedures, cleanup steps, and chromatographic conditions. The following protocol outlines the critical parameters that must be documented for a comprehensive BAGI assessment:
Sample Preparation Documentation:
Chromatographic Conditions:
For example, in a green HPTLC-densitometry method developed for impurity profiling of Ondansetron, the sample preparation involved minimal solvent consumption, with separation achieved on HPTLC plates using a mobile phase of toluene:chloroform:ethanol (5:4:2, v/v) [2]. This method demonstrated high practicality through its ability to process multiple samples simultaneously and its minimal instrumentation requirements.
The data collection phase for BAGI assessment involves systematic evaluation against the ten core criteria, with specific benchmarks for each parameter. The following step-by-step protocol ensures consistent and reproducible BAGI scoring:
Step 1: Criterion Evaluation
Step 2: Data Input and Calculation
Step 3: Results Interpretation
Table 2: BAGI Assessment Protocol for Chromatographic Impurity Profiling Methods
| Protocol Step | Key Activities | Data Recording Requirements | Output |
|---|---|---|---|
| Method Documentation | Record all sample preparation and instrumental parameters | Solvent volumes, time requirements, instrument specifications, throughput data | Comprehensive method description |
| Criterion Scoring | Evaluate method against each of 10 BAGI criteria | Individual criterion scores with justifications | Preliminary assessment |
| Software Input | Enter scores into BAGI web/desktop application | Digital record of all assessment parameters | Calculated BAGI score and pictogram |
| Practicality Analysis | Interpret results and identify improvement areas | Strengths, weaknesses, and optimization opportunities | BAGI implementation report |
The availability of open-access BAGI software (mostwiedzy.pl/bagi or bagi-index.anvil.app) significantly streamlines this process, providing researchers with a user-friendly interface for method assessment and comparison [66]. The software automatically generates the visual outputs and facilitates storage of assessment records for future reference or methodological improvements.
BAGI has been successfully implemented in the assessment of chromatographic methods for pharmaceutical impurity profiling, providing critical insights into method practicality alongside greenness metrics. In one notable application, researchers developed and validated two chromatographic methodsâHPTLC-densitometry and RP-HPLCâfor the determination of Mupirocin in combination with other drugs along with their impurities [3]. The assessment employed multiple green metrics (NEMI, Analytical Eco-Scale, GAPI, and AGREE) alongside practicality evaluation, though not explicitly mentioning BAGI, highlighting the growing recognition of the need to combine greenness and practicality assessment.
In another significant application, researchers developed a green LC-MS/MS method for Ondansetron purity testing along with its impurities, emphasizing the importance of balancing green principles with practical analytical performance [2]. The method demonstrated high practicality through its short run time (2 minutes), minimal solvent consumption, and successful application to pharmaceutical formulations. This approach aligned with White Analytical Chemistry principles by addressing the practical requirements of routine pharmaceutical analysis while maintaining environmental responsibility.
The implementation of BAGI becomes particularly valuable when comparing multiple analytical methods for the same impurity profiling application. A comparative study of 16 analytical methods, including fiber-solid phase microextraction, stir bar sorptive extraction, and thin film microextraction coupled with chromatographic techniques, demonstrated the utility of BAGI in conjunction with other green assessment tools [68]. The study revealed that using multiple evaluation tools provided synergistic results and enhanced understanding of both environmental impact and practical applicability.
BAGI Case Study Comparison: This diagram illustrates the application of BAGI assessment across different chromatographic methods for impurity profiling, highlighting key practicality features. (Estimated scores based on method descriptions)*
The implementation of BAGI assessment for chromatographic methods requires specific reagents, materials, and software tools to ensure accurate and consistent evaluation. The following table details the essential components of a comprehensive BAGI assessment toolkit for impurity profiling research:
Table 3: Essential Research Reagent Solutions and Materials for BAGI Implementation
| Category | Specific Items | Function in BAGI Assessment | Implementation Notes |
|---|---|---|---|
| Software Tools | BAGI Web Application (bagi-index.anvil.app) | Automated score calculation and pictogram generation | Open-access platform for standardized assessment |
| BAGI Desktop Application (mostwiedzy.pl/bagi) | Offline BAGI evaluation | Alternative for environments with internet restrictions | |
| Documentation Tools | Standardized method documentation templates | Consistent recording of analytical method parameters | Ensures all BAGI criteria can be properly evaluated |
| Sample throughput tracking system | Accurate measurement of samples per hour | Critical for criterion #3 assessment | |
| Reference Materials | Certified reference standards | Method validation and performance verification | Essential for establishing accuracy and reliability |
| Impurity standards | Specificity testing for impurity profiling methods | Confirms method capability to resolve impurities | |
| Laboratory Equipment | Automated sample preparation systems | Assessment of automation degree (criterion #8) | Impacts practicality through reduced manual intervention |
| High-throughput chromatographic systems | Evaluation of analysis speed and parallel processing | Directly affects samples per hour metric |
The integration of these tools and materials into the method development workflow enables researchers to systematically assess and optimize the practical aspects of chromatographic methods for impurity profiling. The BAGI software applications, in particular, provide the formal framework for converting method parameters into standardized practicality scores, facilitating objective comparison between different methodological approaches [66] [67].
The implementation of the Blue Applicability Grade Index (BAGI) represents a significant advancement in the comprehensive assessment of chromatographic methods for impurity profiling. By providing a standardized framework for evaluating practical methodology aspects, BAGI complements established green chemistry metrics to support the principles of White Analytical Chemistry. The integration of BAGI into method development workflows ensures that new chromatographic approaches for impurity analysis are not only environmentally sustainable but also practically viable for routine implementation in pharmaceutical quality control laboratories.
As the pharmaceutical industry continues to emphasize both sustainability and operational efficiency, the adoption of BAGI is poised to grow significantly. Future developments may include the integration of BAGI with laboratory information management systems for automated data collection and assessment, as well as the establishment of BAGI benchmarking databases for specific analytical applications. Through its systematic approach to practicality evaluation, BAGI enables researchers and drug development professionals to make informed decisions about method selection and optimization, ultimately contributing to more sustainable, practical, and effective impurity profiling in pharmaceutical development.
Within the framework of a thesis on green chromatographic methods for impurity profiling, the evaluation of sample preparation is paramount. Sample preparation is often the most polluting step in an analytical method, characterized by high consumption of organic solvents and reagent waste generation [70]. The principles of Green Analytical Chemistry (GAC) aim to minimize this environmental impact by promoting safer solvents, reducing waste, and improving energy efficiency [4] [11]. While advanced detection techniques like LC-MS-MS can sometimes reduce the need for extensive sample preparation, most complex pharmaceutical matrices still require efficient and clean extraction steps to accurately profile impurities without matrix interference [70].
To systematically evaluate and improve the environmental footprint of these sample preparation procedures, specialized metrics are required. The AGREEprep metric has emerged as a dedicated tool for assessing the greenness of sample preparation methods [44]. It provides a comprehensive, quantitative evaluation based on ten key assessment criteria that align with the broader objectives of GAC, enabling researchers to benchmark, optimize, and justify their sample preparation strategies within a sustainability context [44] [11]. This application note details the implementation of AGREEprep for the specialized evaluation of sample preparation steps in pharmaceutical impurity profiling.
The AGREEprep tool (Analytical GREEnness Metric for Sample Preparation) is a recently developed open-access software that delivers a standardized, holistic assessment of sample preparation procedures [44]. Its design focuses specifically on the sample preparation step, which is critical for reducing the overall environmental impact of analytical methods in pharmaceutical quality control.
AGREEprep evaluates sample preparation procedures against ten carefully selected principles, each contributing to the overall greenness score [44]. The criteria, along with their objectives and typical data requirements for assessment, are summarized in Table 1.
Table 1: The Ten Assessment Criteria of the AGREEprep Metric
| Criterion Number | Primary Objective | Typical Data for Assessment |
|---|---|---|
| 1 | Minimize or eliminate sample collection | Need for sample collection, microextraction vs. exhaustive |
| 2 | Perform in-place analysis | On-site vs. lab-based analysis |
| 3 | Minimize sample size | Sample volume/mass required |
| 4 | Use safer solvents/reagents | Solvent toxicity, volume, and hazard profile |
| 5 | Minimize waste generation | Total waste produced per sample |
| 6 | Maximize operator safety | Operator exposure, automation, hazard controls |
| 7 | Minimize energy consumption | Energy demand of equipment (e.g., heating, cooling) |
| 8 | Maximize analysis throughput | Number of samples processed per unit time |
| 9 | Minimize or eliminate derivatization | Need for chemical derivatization |
| 10 | Use available resources | Integration with other steps, method simplicity |
The AGREEprep software algorithm calculates a total score on a scale from 0 to 1, where a higher score indicates a greener sample preparation procedure [44]. This score is presented within an intuitive, circular pictogram, similar to the original AGREE metric. The pictogram is divided into ten sections, each corresponding to one of the assessment criteria. Each section is color-coded, providing an immediate visual indication of performance across all principles. This visual output allows scientists to quickly identify the specific strengths and weaknesses of their method, guiding targeted improvements for a more sustainable workflow [44] [11].
This protocol provides a step-by-step guide for utilizing the AGREEprep tool to evaluate a sample preparation method used in the impurity profiling of a new drug substance.
https://mostwiedzy.pl/AGREEprep).Table 2: Data Requirements for AGREEprep Assessment of a Solid-Phase Extraction (SPE) Method
| Criterion | Example Data from a Conventional SPE Method | Example Data from a Miniaturized/Green Method |
|---|---|---|
| Sample size | 1 mL plasma [71] | 100 µL plasma [71] |
| Solvent consumption | 6 mL acetonitrile (for protein precipitation) [71] | 300 µL acetonitrile (for phospholipid removal) [71] |
| Waste generated | ~6.1 mL per sample (including solvents and consumables) | ~0.4 mL per sample |
| Operator hazards | Handling of mL volumes of toxic solvent (acetonitrile) | Handling of µL volumes; potential for automation [72] |
| Energy demand | ~0.05 kWh/sample (for centrifugation and evaporation) | ~0.02 kWh/sample (vortex mixing only) |
| Throughput | 12 samples in 2 hours | 96 samples in 2 hours via parallel processing [44] |
| Derivatization | Not required | Not required |
The following workflow diagram illustrates the logical process of conducting an AGREEprep assessment and utilizing its results for method improvement.
Background: A bioanalytical laboratory aims to evaluate and improve the greenness of its sample preparation method for analyzing drug impurities and metabolites in plasma using LC-MS/MS. The current method is conventional protein precipitation [71].
Objective: To compare the greenness and analytical performance of a composite PLR technology against conventional protein precipitation.
Materials:
Procedure:
Data from the two protocols were used for the AGREEprep assessment. The results demonstrate the clear environmental advantages of the miniaturized PLR approach.
Table 3: AGREEprep Input Data and Resulting Scores for Case Study
| Assessment Criterion | Conventional Protein Precipitation | Miniaturized PLR Method |
|---|---|---|
| Sample Size | 100 µL | 100 µL |
| Solvent Volume | 3000 µL | 300 µL |
| Estimated Waste | High (~3.1 mL) | Low (~0.4 mL) |
| Operator Hazards | Higher (mL solvent handling) | Lower (reduced solvent, plate-based) |
| Energy Demand | Medium (centrifugation) | Low (vortex mixing, positive pressure) |
| Throughput | Medium (12 samples/run) | High (96-well parallel processing) |
| Calculated AGREEprep Score | ~0.45 | ~0.81 |
The case study shows that the miniaturized PLR method scores significantly higher on the AGREEprep scale. This is largely due to a drastic reduction in solvent consumption and waste generation, improved operator safety, and higher potential for automation and parallel processing [44] [71]. Analytically, the PLR method also outperforms protein precipitation by effectively removing phospholipids, thereby reducing ion suppression in the mass spectrometer, minimizing source contamination, and extending column lifetime [71].
The implementation of green sample preparation strategies often relies on specialized consumables and reagents. The following table lists key solutions used in the development of sustainable sample preparation workflows for impurity profiling.
Table 4: Key Research Reagent Solutions for Green Sample Preparation
| Item Name | Function/Description | Application in Green Sample Prep |
|---|---|---|
| Microlute PLR Plates | Composite technology plates for selective phospholipid removal from biological samples [71]. | Reduces solvent use vs. traditional PPT; minimizes matrix effects and instrument maintenance. |
| Solid-Phase Microextraction (SPME) Fibers | Solventless extraction technique where a coated fiber absorbs analytes directly from sample headspace or liquid [70]. | Eliminates solvent use; enables on-site sampling and automation. |
| Ionic Liquids / Deep Eutectic Solvents | Safer, biodegradable solvents with low volatility used as alternatives to traditional organic solvents [4]. | Replace hazardous solvents (e.g., acetonitrile, chlorinated solvents) in liquid-liquid extraction. |
| Molecularly Imprinted Polymers (MIPs) | Synthetic polymers with tailor-made recognition sites for specific target analytes [4]. | Provide high selectivity, reducing need for extensive clean-up and solvent consumption. |
| Automated SPE Systems | Instrument systems that perform solid-phase extraction steps (conditioning, loading, washing, elution) with minimal user intervention [72]. | Improves reproducibility, increases throughput, and reduces operator exposure to hazards. |
The AGREEprep metric provides a critical, specialized tool for quantifying the environmental performance of sample preparation methods in pharmaceutical impurity profiling. By offering a standardized, ten-criteria assessment, it moves the field beyond subjective claims and enables data-driven decisions for achieving sustainability goals. As demonstrated in the case study, AGREEprep can effectively guide scientists away from traditional, wasteful practices like large-scale protein precipitation and towards modern, miniaturized, and automated alternatives. Integrating AGREEprep into routine method development and validation is a vital step for the pharmaceutical industry to align with the principles of Green Analytical Chemistry, ensuring that the crucial first step in analysisâsample preparationâcontributes to a more sustainable laboratory practice without compromising analytical integrity.
The integration of green chromatographic methods into pharmaceutical impurity profiling is no longer a niche concept but a necessary evolution for sustainable and economically viable quality control. This synthesis of foundational principles, advanced techniques, troubleshooting strategies, and robust validation metrics demonstrates that environmental responsibility can be achieved without compromising analytical performance. Future progress hinges on interdisciplinary collaboration, supportive regulatory frameworks that incentivize green practices, and the adoption of disruptive innovations like machine learning for method development. Embracing this holistic approach will be crucial for the pharmaceutical industry to meet its ethical and environmental responsibilities while ensuring drug safety and efficacy.