Analytical Methods Validation

Similar documents
ASEAN GUIDELINES FOR VALIDATION OF ANALYTICAL PROCEDURES

serve the goal of analytical lmethod Its data reveals the quality, reliability and consistency of

Chapter 4: Verification of compendial methods

International Journal of Pharmacy and Pharmaceutical Sciences ISSN Vol 2, Suppl 3, 2010

The Theory of HPLC. Quantitative and Qualitative HPLC

Analytical Performance & Method. Validation

REVIEW. Comparison of various international guidelines for analytical method validation

Analytical Method Validation: An Updated Review

INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE

VALIDATION OF ANALYTICAL METHODS. Presented by Dr. A. Suneetha Dept. of Pharm. Analysis Hindu College of Pharmacy

Regulatory and Alternative Analytical Procedures are defined as follows 2 :

Instrumental Chemical Analysis

Introduction to Pharmaceutical Chemical Analysis

A Laboratory Guide to Method Validation, (Eurachem).

Validation of an Analytical Method

DETERMINATION OF DRUG RELEASE DURING DISSOLUTION OF NICORANDIL IN TABLET DOSAGE FORM BY USING REVERSE PHASE HIGH PERFORMANCE LIQUID CHROMATOGRAPHY

Critical Pairs in Column Chromatography: A Primer for Pharmaceutical Method Validation

VALIDATION OF ANALYTICAL METHODS FOR PHARMACEUTICAL ANALYSIS

Work plan & Methodology: HPLC Method Development

The Role of and the Place of Method Validation in Drug Analysis Using Electroanalytical Techniques

Modern Analytical Techniques in Pharmaceuticals

Method Validation Essentials, Limit of Blank, Limit of Detection and Limit of Quantitation

Simultaneous Estimation of Residual Solvents (Isopropyl Alcohol and Dichloromethane) in Dosage Form by GC-HS-FID

The Analysis of Residual Solvents in Pharmaceutical Products Using GC-VUV and Static Headspace

Streamlining the Analysis of Oral Contraceptives Using the ACQUITY UPLC H-Class System

VALIDATION OF A UPLC METHOD FOR A BENZOCAINE, BUTAMBEN, AND TETRACAINE HYDROCHLORIDE TOPICAL SOLUTION

PART II VIII. CONCERNING CHEMICAL. PHARMACEUTICAL AND BIOLOGICAL DOCUMENTATION FOR RADIOPHARMACEUTICAL PRODUCTS.

LABORATORY EXERCISE: USING SPECTROPHOTOMETRY FOR QUALITY CONTROL: NIACIN

Int. J. Pharm. Sci. Rev. Res., 30(2), January February 2015; Article No. 09, Pages: 63-68

Traceability, validation and measurement uncertainty 3 pillars for quality of measurement results. David MILDE

Rosemary extract liquid

A Simple, Novel Validated Stability Indicating RP-HPLC method for estimation of Duloxetine HCl in Capsule Pharmaceutical Formulation

Development And Validation Of Rp-Hplc Method For Determination Of Velpatasvir In Bulk

BRIEFING. (EXC: K. Moore.) RTS C Propylparaben C 10 H 12 O Benzoic acid, 4 hydroxy, propyl ester; Propyl p hydroxybenzoate [ ].

Ministry of Health MS Brazilian Health Surveillance Agency - ANVISA

International Journal of PharmTech Research CODEN (USA): IJPRIF, ISSN: , ISSN(Online): Vol.9, No.7, pp , 2016

Copyright ENCO Laboratories, Inc. II. Quality Control. A. Introduction

COMMITTEE FOR HUMAN MEDICINAL PRODUCTS (CHMP) GUIDELINE ON RADIOPHARMACEUTICALS

To control the consistency and quality

Since 1988 High Force Research has worked with organizations operating in practically all end use sectors requiring chemical synthesis input, and

Development of Validated Analytical Method of Mefenamic Acid in an Emulgel (Topical Formulation)

Agilent 1200 Infinity Series HDR DAD Impurity Analyzer System for the Quantification of Trace Level of Genotoxic Impurity

2.1 2,3 Dichloro Benzoyl Cyanide (2,3 DCBC) and survey of. manufactured commonly for the bulk drug industry, few references were

Center for Drug Evaluation and Research (CDER) Reviewer Guidance. Validation of Chromatographic Methods. November 1994 CMC 3

DEVELOPMENT AND VALIDATION OF GC-FID METHOD FOR THE DETERMINATION OF ETHANOL RESIDUE IN MARJORAM OINTMENT

High Performance Liquid Chromatography

Validated HPLC Methods

Chromatography & instrumentation in Organic Chemistry

Stability-indicating HPLC determination of tolterodine tartrate in pharmaceutical dosage form

Tomorrow s quantitation with the TSQ Fortis mass spectrometer: quantitation of phenylephrine hydrochloride for QA/QC laboratories

Impact factor: 3.958/ICV: 4.10 ISSN:

THERAPEUTIC GOODS SAMPLING Application to Tablet Manufacture and Ingredients. David Edmonds CMC Regulatory

STABILITY INDICATING METHOD OF RELATED IMPURITIES IN VENLAFAXINE HYDROCHLORIDE SUSTAINED RELEASE TABLETS

Development and validation a RP-HPLC method: Application for the quantitative determination of quetiapine fumarate from marketed bulk tablets

Implementation of Methods Translation between Liquid Chromatography Instrumentation

Development and Validation of a HPLC Method for Chlorphenamine Maleate Related Substances in Multicomponents Syrups and Tablets

Part IVB Quality Assurance/Validation of Analytical Methods

Method Development and Validation Of Prasugrel Tablets By RP- HPLC

Substance Characterisation for REACH. Dr Emma Miller Senior Chemist

Quality by Design and Analytical Methods

CHAPTER INTRODUCTION OF DOSAGE FORM AND LITERATURE REVIEW

COLA Mass Spec Criteria

Mashhour Ghanem 1 and Saleh Abu-Lafi 2 * ABSTRACT ARTICLE INFO

QUALITY CONTROL CRITERIA FOR CHEMISTRY EXCEPT RADIOCHEMISTRY.

PART III B METHODS OF ANALYSIS/ANALYTICAL SCHEME FOR IDENTIFICATION OF DRUGS OR CHEMICALS

7. Stability indicating analytical method development and validation of Ramipril and Amlodipine in capsule dosage form by HPLC.

Validation and Standardization of (Bio)Analytical Methods

3.1.1 The method can detect, identify, and potentially measure the amount of (quantify) an analyte(s):

Validation of HPLC Instrumentation

The Claviature of Gas Analysis

SIMULTANEOUS RP HPLC DETERMINATION OF CAMYLOFIN DIHYDROCHLORIDE AND PARACETAMOL IN PHARMACEUTICAL PREPARATIONS.

Guideline/SOP: Handling of Laboratory Gross Errors/Data History

INTERNATIONAL RESEARCH JOURNAL OF PHARMACY

International Journal of Pharmaceutical Research & Analysis

Method Validation. Role of Validation. Two levels. Flow of method validation. Method selection

Introduction to E&Ls 1

Limit of Detection and Its Establishment in Analytical Chemistry

RP-HPLC Method Development and Validation of Dapagliflozin in Bulk and Tablet formulation

Routine MS Detection for USP Chromatographic Methods

Starting Materials For Active Substances

PROCEDURES. Pharmacopeial Forum 2 Vol. 36(1) [Jan. Feb. 2009]

GUIDELINES ON PERFORMANCE CRITERIA FOR METHODS OF ANALYSIS FOR THE DETERMINATION OF PESTICIDE RESIDUES IN FOOD AND FEED CXG Adopted in 2017.

Quality control analytical methods- Switch from HPLC to UPLC

MEDROXYPROGESTERONE INJECTION

A RP-HPLC METHOD DEVELOPMENT AND VALIDATION OF PARA- PHENYLENEDIAMINE IN PURE FORM AND IN MARKETED PRODUCTS

Method Validation Characteristics through Statistical Analysis Approaches. Jane Weitzel

CHAPTER - IV. Acharya Nagarjuna University, Guntur 105

Pharmacopeial Forum Vol. 36(1) [Jan. Feb. 2010] 1

Automated and accurate component detection using reference mass spectra

DETERMINATION OF ORGANIC VOLATILE IMPURITIES IN NEPAFENAC BY GC METHOD

Overview of HACCP Principles. OFFICE OF THE TEXAS STATE CHEMIST Texas Feed and Fertilizer Control Service Agriculture Analytical Service

Validation of ACQUITY UPLC Methods

Application of mathematical, statistical, graphical or symbolic methods to maximize chemical information.

+ H 2 O Equation 1. + NaOH CO 2 Na

INTERNATIONAL CHEMISTRY TESTING. 258 Main Street- Suite 311- Milford, MA Tel: Fax:

Validation of analytical methods. Adrian Covaci Toxicological Center, University of Antwerp

CDER Risk Assessment to Evaluate Potential Risks from the Use of Nanomaterials in Drug Products

First Quarter OSHA Hazardous Chemical Labeling Requirements

Introduction. Chapter 1. Learning Objectives

Reverse Phase High Performance Liquid Chromatography method for determination of Lercanidipine hydrochloride in bulk and tablet dosage form

Transcription:

Analytical Methods Validation In-Process Control Methods for the Manufacture of Active Pharmaceutical Ingredients Jose Zayas*, Victor Sanchez, and Michelle Talley The authors propose a strategy for classifying and validating inprocess testing COMSTOCK Jose Zayas, PhD, is a technical director at Zaycor Industries Corp., 1602 Bori Street, Urb. Caribe, San Juan, PR 00927, tel. 787.763.3016, fax 787.763.5652, jzayas@zaycor.com. Victor Sanchez is an analytical support supervisor at Schering- Plough, SA, Madrid, Spain. Michelle Talley is a consultant at the Mintanciyan Consulting Group, New York, NY. *To whom all correspondence should be addressed. In-process methods are key components of quality control in a manufacturing plant. These methods ensure that a production step conducted by trained operators within the entire validated process will produce a quality entity in the expected yields. The presence of impurities and related compounds (derived from the or secondary s) is a critical parameter that determines a synthetic material s quality. Chemical processing differs from product manufacturing. For example, the manufacture of a finished product typically involves a molecular entity that is stable under normal conditions and can be stored for prolonged periods without losing its physical and characteristics. Most s, however, require very tight controls and close monitoring of their progress because any of several potential result paths may be followed if conditions are not monitored closely. Other factors such as temperature and pressure are critical parameters for the successful completion of the conversion process. Each is unique. Consider the combination of reactants and the resulting end products, for example. One must examine conditions such as temperature, light, heat, environment, and the vessel s surface. In addition, whether the is or biological is an important factor. Therefore, each process must be analyzed separately and classified according to the International Conference on Harmonization (ICH) Q7A guidance. This task is important because a step may generate an impurity that may be carried over to the active pharmaceutical ingredient (), regardless of how far apart that process may be from the. ICH s Q7A guidance briefly mentions analytical methods validation and does not discuss the in-process control methods for each. The guidance does indicate, however, that as the process gets closer to the manufacture of the key intermediate and the, the current good manufacturing practices (CGMP) requirements become more demanding. This article outlines a plan for classifying and validating in-process testing methods and is intended as a foundation for assessing the parameters and acceptance criteria needed for validation. Matrix and interferences To characterize a by means of an analytical method, it may be necessary to prepare a matrix, which functions similarly 154 Pharmaceutical Technology APRIL 2005

Definitions Although the International Conference on Harmonization established several definitions in its Q7A and Q2B guidances, the following terms are not defined in those documents: Intermediate-release method:a method developed and used for the release of an isolated intermediate material produced during the manufacture of an active pharmaceutical ingredient () before the material is used,stored,or shipped.it may contain more than one specification for product release. In-process control method: a method developed and used for monitoring the progress of a (e.g., disappearance or formation of intermediates) or a critical attribute of a such as moisture or ph; Marker: a stable material, characterized by nuclear magnetic resonance, mass spectrometry, or another scientifically recognized characterization method, that is related to the process being monitored. Its identity must be reconfirmed periodically; Step (or stage):the synthesis progression for converting one molecular entity to another in an manufacturing process.this step may have several conditions.a step is complete when the converted intermediate either is isolated or is stable enough that it can be converted in situ to another molecule. to a placebo for a finished product. A matrix is the combination of the reactants without the main component or precursor being converted. Because of the nature of some s, the combination of reagents may not be possible. On the other hand, adducts or complexes can be formed, which would not otherwise be formed in the presence of a component being converted into a product. The preparation of the matrix must be judged by the scientists working with the process. Classification of methods For monitoring purposes, analytical methods can be classified according to the manufacturing step in which they are applied. The document indicates that the GMP requirements become more stringent as the synthesis steps approach the. Beginning with the introduction of the starting material into the process, manufacturing processes can be divided into three classes, which reflect the practices established in the ICH Q7A guidance. intermediates production (e.g., alkylation, hydrogenation) isolation and purification (e.g., washing, crystallization) physical processing and packaging (e.g., micronization). For the purposes of this article, intermediates production is subdivided into intermediates and key intermediates production. The classification of the methods (e.g., in-process controls and intermediate-release methods) is determined by how far the stage or step is removed from the. Figure 1 represents the application of the ICH guidance to these classes. The ICH Q2B guidance enables chromatographic resolution to be used as an indicator of specificity for critical separations, which means that peak purity is not necessary. Furthermore, peak purity should not be a consideration because samples are submitted only to confirm the disappearance of the starting material and the formation of the desired adduct. No peak will be as pure as required when the analysis is a crude mixture. Solution and standard stability should be included as part of some of the studies. The length of the stability study is defined by the process. The classification is exclusive to methods used for in-process control (and monitoring) of intermediate steps during an manufacturing process. The classification pertains to s that are at least two steps from the processing of the key intermediate. Because the formation or source of impurities should be known and each impurity identified, it is possible that in some instances, the classification becomes Class 2 several steps before the key intermediate production. Class 2.Class 2 is exclusive to methods used for in-process control (and monitoring) of intermediate steps during an manufacturing process. The classification pertains to those s that precede the formation of the key intermediate.. This classification includes methods used as intermediate-release methods when the product formed is an intermediate that will be used further after isolation or supplied as a starting material for another synthesis. This classification pertains to the key intermediate or isolated entity that eventually will be converted to an. An example of how processes would be classified is shown in Figure 2. Substance D is the key intermediate, one step before the formation. Substance E represents the final molecule before purification. Substance B, for example, could be subject to intermediate-release method testing if the material is isolated and the starting material is used in a parallel synthesis. In this case, the purification step does not involve any conversion and the is structurally identical to Substance E. The purification step can be recrystallization, micronization, or any other physical manipulation of the active that does not involve a conversion or change in structure. In addition, a sequence may involve the isolation of an intermediate that is several steps away from the formation of the key intermediate. The intermediate would be classified as an intermediate-release method or. Validation of The suitability of all methods used as inprocess control methods and as intermediate-release methods should be verified and documented under actual conditions of use. Each category has a recommended suitability procedure defined. The degree of analytical validation performed must reflect the purpose and stage of the production process. All analytical equipment must be qualified before it is used for method validation. Complete records must be maintained for any and all equipment modifications made to validate analytical The validation process may require that intermediates be characterized, isolated, and used as reference markers for establishing the relative retention times. The preparation of a matrix or mixture without the active may help establish unknown peaks and potential interferences. The matrix must be treated according to the procedures established for in-process control monitoring If an intermediate is not isolated, but reacted in situ to a later step, isolation may not be necessary for its characterization if it is an unstable entity. Should it be a stable molecule, however, its isolation and characterization may be necessary. Chromatographic methods Chromatographic methods are validated according to their classification, as discussed previously. The method validation pro- 156 Pharmaceutical Technology APRIL 2005

Table I: Summary of the requirements, per classification, for chromatographic Performance parameter Class 2 System suitability Accuracy Precision Quantitation limit Detection limit Specificity Linearity Robustness Standard and solution stability tocol should include a discussion of the method s classification and the justification for the classification. The validation described for each classification is for quantitative chromatographic Chromatographic identification and semiquantitative techniques such as thin-layer methods must be validated (described later in this article). These methods require the determination of accuracy in their semiquantitative level. Table I summarizes the requirements of each classification..description. methods are limit tests and must be validated accordingly. This validation should include a demonstration of the method s detection limit specificity and determination because these steps are far removed from the formation of the key intermediate and. Therefore, one must be able to identify the peak of interest, properly resolved from the starting materials (reactants). The method should confirm the disappearance of the reactants or the formation of the adduct. Requirements.The testing needed to monitor the intermediate steps during manufacture falls into. According to ICH Q7A, this classification suits steps far removed from the formation of the and for which CGMP requirements are not as demanding. As is typical for every method, system suitability is a requirement and usually includes injecting a marker for methods using response normalization for quantitation. Precision is not necessarily a requirement at this stage. Knowledge of the relative retention times of the reactants being monitored is essential, however. The method s sensitivity should be established once the target entity monitored is quantified and the limit is established. The limits can be established by following ICH Q2B recommendations, which suggest a linearity experiment. At this stage, running a three-point linearity experiment around the target value can be used to estimate the detection limit. For example, consider a in which the transformed or consumed reactant will be monitored until the area of the peak of its signal is 0.5% of the product being formed. The linearity experiment can be executed to include 0.25, 0.50, and 0.75% of the reactant. Linear regression results would provide the intercept s slope and standard error, and thus, the estimation of the detection limit for this classification. Alternatively, estimation solutions can be prepared. When a signal-to-noise (3.3:1) ratio is 158 Pharmaceutical Technology APRIL 2005 obtained for the solution, the figure becomes the detection limit for the method. Class 2. Description.Class 2 method validation includes the limittest requirements described previously (specificity, detection limit), the determination of the method quantitation limit, and the demonstration of the method linearity covering the entire range of the method (i.e., reactants and adduct being formed to determine the specified limit). Thus, the detection limit and quantitation limit values should be calculated from the linearity data generated during the execution of the validation protocol. The testing required for monitoring the formation of the key intermediate falls under Class 2. As with, a marker solution can be injected to establish the system-suitability criteria such as resolution and tailing factors. Again, precision may not be an issue because the criticality of this monitoring is the appearance or disappearance of a reactant or an adduct relative to each other. The determination of the quantitation limit would be part of the limit-test requirement. In this case, because we are further into the ICH 7A-suggested criteria for CGMPs, it is recommended that the linearity experiment be conducted over its full range. This range would be from the quantitation limit to at least 125% of the concentration range monitored. If the target value lies far from the quantitation limit (e.g., the quantitation limit is 0.01% and the target concentration is 5%), then a three-point calibration should be run close to the quantitation limit. This step would be followed by a normal five-point linear regression from 50 to 125% of the target. It is useful to compare the precision between the curves and to compare the response factors obtained from both curves. Solution and standard stability are essential components at this stage of the synthesis. The CGMP requirements mandate that these solutions be stable for the time of use. Any degradation could generate false data and miscalculations/misidentifications of unknowns. The length of stability will be defined by the process.description. In essence, methods are finished-product (release) methods that control the final product of the manufacturing process. As such, the extent of validation for this type of method approximates the extent of validation for the finished product methods described in the US Pharmacopeia (USP). Class 3 methods validation includes linearity, accuracy, precision, specificity, and intermediate precision. The ranges applied to each parameter are subject to internal procedure The rationale for the ranges used must be clearly explained in the methods validation protocol. The impurity method parameters will be applied when the reactant is the quantified chromatographic component. Requirements.The testing required for the monitoring s for the formation of the final molecule falls into. The following are the specific requirements for validating such methods: The standard must be fully characterized by scientifically recognized USP standards should be used if they are available. Semiquantitative thin-layer chromatography methods are in this category. This technique is a two-dimensional chromato-

Maximum CGMP Strong validation Class 2 Medium CGMP Mild validation Minimum CGMP Weak validation Figure 1: The classes to which an manufacturing process is divided, according to the International Conference on Harmonization Q7A guidance document. graphic procedure. As such, the system must be tested and the procedure must be clear. The accuracy and linearity of the technique being qualified as a semiquantitative method must be verified. The reference standards must be prepared at concentrations ranging from a quantitation limit to at least 125% of the concentration targeted in the analysis. This information is a visual calibration for the method. On the other hand, specifications must be set for the response factor (R f ) of the spots of reference material and any other reference substances that are critical for the test. This process will establish the conditions for system suitability. Furthermore, the sensitivity of the method must be determined to enable visual calibration to be established and a reliable limit to be set for the test. Along these lines, the composition of the mobile phase and the length of the plate run are critical to this test. The concentration established for the test must be such that the separation yields good resolution (R) between the closest eluting components of the mixture. The process also involves changes in chromatographic Table II: Summary of the requirements for nonchromatographic Performance parameter Titration Atomic absorption UV -vis Calibration Accuracy Precision * * Quantitation limit Detection limit Specificity Linearity NA NA Standard and solution stability *Intermediate precision. NA indicates not applicable plates and analyst-to-analyst comparison. Therefore, robustness also is required. A comparison of a unique R f for the main substances monitored or quantified with the R f of other major components of the would yield the test s specificity. Once again, standard and solution stability are critical as controls for the methodology and for ensuring reliable results. Nonchromatographic instrument methods Among nonchromatographic methods, titration can be used for monitoring sequences. Titration methods will require validation beyond equipment qualification. Linearity and accuracy for the technique are important. Linearity is established by preparing three-point calibration curves. Regarding accuracy, the titration would require the determination of matrix contributions, if any, to the end point of the procedure. The preparation of spiked solutions, as well as the titration of blanks containing matrix elements, is required for validating the reliability of the technique. Thus, precision can be assessed from the experiments conducted under linearity and accuracy. The and its stoichiometric relationship can be presented to assess the specificity of the method. If a method is not specific, an explanation or the rationale for using a nonspecific method must be provided in the method validation protocol.

Class 2 A B C D E F G Starting material Intermediate Production of intermediates (ICH-Q7A) Key intermediate Figure 2: An example of how processes can be classified. Unpurified physical Isolation or purification Purified physical Physical processing Micronized In-process spectrophotometric methods With the exception of chromatographic methods, in-process spectrophotometric methods are those for which the end detection technique is spectrophotometric. When these methods are used for identification purposes, only sensitivity, standard, and solution stability data are required. The calibration of each technique is required before analytical work can begin. The validation of atomic absorption techniques should include linearity, accuracy, precision, and specificity. Although specificity is inherent to the technique, the protocol and its report should indicate the analysis specificity of the particular metal. Furthermore, the sensitivity (detection limit) and standard and solution stability also should be included. For system suitability, the %RSD of a standard s replicate sampling measurements usually must be reported. At minimum, most systems include a calibration curve and a standard confirmation reading. Systems that do not report %RSD values and/or include a calibration curve must be addressed in the methods validation protocol and report. Table II summarizes the validation requirements for nonchromatographic UV-vis spectrophotometry should include linearity, accuracy, precision, sensitivity (detection limit), and standard and solution stability. This technique is nonspecific. A calibration curve is an essential component for analyses such as the American Pharmaceutical Association s standard for absorption of light (color) and requires the preparation of primary standards. If a calibration curve must cover a range of time (e.g., 7 days), the statement s validity must be established and documented. Conclusion Monitoring processes for the formation of an is the first step to ensuring quality in pharmaceutical manufacturing. Having reliable and reproducible methods will enable the production plant to guarantee the consistency of drugs batch after batch. Furthermore, it may simplify the characterization of such processes and their profile. Through the years, vast publications and general information have been presented to pharmaceutical industry specialists about the validation of analytical Federal and international regulatory groups have published various guidelines to shed light on analytical method validation. No such emphasis has been given, or guidances described, however, the validation of in-process control This article intends to establish a starting point for discussions about the validation of in-process References 1. International Conference on Harmonization (ICH), ICH Quality Guidelines: Good Manufacturing Practice Guidance for Active Pharmaceutical Ingredients Q7A (ICH, Geneva, Switzerland, 2001). 2. US Food and Drug Administration, Guidance Document for Industry, Analytical Procedures and Methods Validation, (FDA, Rockville, MD, 2000). 3. ICH, ICH Quality Guidelines Validation on Analytical Procedures: Methodology Q2B (ICH, Geneva, Switzerland, 1996). 4. General Chapter 1225, Validation of Compendial Methods, USP 27 NF 22 (US Pharmacopeial Convention, Rockville, MD, 2003). PT Please rate this article. On the Reader Service Card, circle a number: 348 Very useful and informative 349 Somewhat useful and informative 350 Not useful or informative Your feedback is important to us. 162 Pharmaceutical Technology APRIL 2005