Design of Experiments for Analytical Method Development and Validation



Similar documents
Assay Development and Method Validation Essentials

QbD Approach to Assay Development and Method Validation

Risk assessment is used as a

Step-by-Step Analytical Methods Validation and Protocol in the Quality System Compliance Industry

VALIDATION OF ANALYTICAL PROCEDURES: TEXT AND METHODOLOGY Q2(R1)

Guidance for Industry

ICH Topic Q 2 (R1) Validation of Analytical Procedures: Text and Methodology. Step 5

ANALYTICAL METHODS INTERNATIONAL QUALITY SYSTEMS

Validation and Calibration. Definitions and Terminology

Analytical Methods: A Statistical Perspective on the ICH Q2A and Q2B Guidelines for Validation of Analytical Methods

GUIDELINES FOR THE VALIDATION OF ANALYTICAL METHODS FOR ACTIVE CONSTITUENT, AGRICULTURAL AND VETERINARY CHEMICAL PRODUCTS.

Applying Statistics Recommended by Regulatory Documents

Content Sheet 7-1: Overview of Quality Control for Quantitative Tests

Workshop B Control Strategy

Workshop A Design Space (DS)

Analytical Test Method Validation Report Template

QbD Considerations for Analytical Methods - FDA Perspective

Assay Qualification Template for Host Cell Protein ELISA

Validating Methods using Waters Empower TM 2 Method. Validation. Manager

Lifecycle Management of Analytical Procedures: Method Development, Procedure Performance Qualification, and Procedure Performance Verification a

To avoid potential inspection

Implementing New USP Chapters for Analytical Method Validation

International GMP Requirements for Quality Control Laboratories and Recomendations for Implementation

USING CLSI GUIDELINES TO PERFORM METHOD EVALUATION STUDIES IN YOUR LABORATORY

PROCEDURES FOR HANDLING OOS RESULTS

USE OF REFERENCE MATERIALS IN THE LABORATORY

Journal of Chemical and Pharmaceutical Research

Lifecycle Management of Analytical Procedures; What is it all about? Jane Weitzel Independent Consultant

Develop a Quantitative Analytical Method for low (» 1 ppm) levels of Sulfate

American Association for Laboratory Accreditation

Multiple Optimization Using the JMP Statistical Software Kodak Research Conference May 9, 2005

Intelligent use of Relative Response Factors in Gas Chromatography-Flame Ionisation Detection

ICH Topic Q 1 A Stability Testing Guidelines: Stability Testing of New Drug Substances and Products

EXTRACTABLES AND LEACHABLES FOR MEDICAL DEVICES: MEETING THE 510 (k) REQUIREMENTS

Qualification Study CHO 360-HCP ELISA (Type A to D)

Fusion Pro QbD-aligned DOE Software

Reversed Phase High Presssure Liquid Chromatograhphic Technique for Determination of Sodium Alginate from Oral Suspension

FOOD FOR THOUGHT Topical Insights from our Subject Matter Experts UNDERSTANDING WHAT IS NEEDED TO PRODUCE QUALITY DATA

Chemistry 111 Laboratory Experiment 7: Determination of Reaction Stoichiometry and Chemical Equilibrium

SPECIFICATIONS AND CONTROL TESTS ON THE FINISHED PRODUCT

Quality Risk Management The Pharmaceutical Experience Ann O Mahony Quality Assurance Specialist Pfizer Biotech Grange Castle

Process Validation: Practical Aspects of the New FDA Guidance

Real-time PCR: Understanding C t

IN VITRO BINDING BIOEQUIVALENCE STUDY SUMMARY TABLES AND SAS TRANSPORT FORMATTED TABLES FOR DATASET SUBMISSION

Chapter 4 and 5 solutions

STABILITY TESTING OF ACTIVE SUBSTANCES AND PHARMACEUTICAL PRODUCTS

ASSURING THE QUALITY OF TEST RESULTS

Product Quality Management

What to control? CQAs and CPPs

4.2 Bias, Standards and Standardization

Annex 2 Stability testing of active pharmaceutical ingredients and finished pharmaceutical products

2.500 Threshold e Threshold. Exponential phase. Cycle Number

KEYS TO SUCCESSFUL DESIGNED EXPERIMENTS

How to Verify Performance Specifications

TEST REPORT: SIEVERS M-SERIES PERFORMANCE SPECIFICATIONS

ICH guideline Q8, Q9 and Q10 - questions and answers volume 4

Contents. Abstract...i. Committee Membership... iii. Foreword... vii. 1 Scope...1

A commitment to quality and continuous improvement

ICH Topic Q 1 E Evaluation of Stability Data. Step 5 NOTE FOR GUIDANCE ON EVALUATION OF STABILITY DATA (CPMP/ICH/420/02)

IMPURITIES IN NEW DRUG PRODUCTS

Analytical Methods for Cleaning Validation

Lean Six Sigma Analyze Phase Introduction. TECH QUALITY and PRODUCTIVITY in INDUSTRY and TECHNOLOGY

Lean Six Sigma Black Belt Body of Knowledge

Control Charts and Trend Analysis for ISO Speakers: New York State Food Laboratory s Quality Assurance Team

STABILITY TESTING OF NEW DRUG SUBSTANCES AND PRODUCTS

Fairfield Public Schools

Method Validation/Verification. CAP/CLIA regulated methods at Texas Department of State Health Services Laboratory

Evaluating Current Practices in Shelf Life Estimation

Quality by Design Approaches to Analytical Methods -- FDA Perspective. Yubing Tang, Ph.D. FDA/CDER/ONDQA AAPS, Washington DC October 25, 2011

COMMITTEE FOR PROPRIETARY MEDICINAL PRODUCTS (CPMP)

ProteinPilot Report for ProteinPilot Software

Assessment of Accuracy and Precision

Commercial Manufacturing - Qualification & Validation-related GMP Deficiencies and Other Lifecycle Considerations

Measurement Information Model

Hazard Analysis and Critical Control Points (HACCP) 1 Overview

Guidance for Industry

Cleaning Validation in Active pharmaceutical Ingredient manufacturing plants

Guideline on Process Validation

Analytical Chemistry Lab Reports

Working with ICH Quality Guidelines - the Canadian Perspective

Metrologically-related out-of-specification test results

The Scheduled MRM Algorithm Enables Intelligent Use of Retention Time During Multiple Reaction Monitoring

White paper: FDA Guidance for Industry Update Process Validation

ICH Topic Q 1 A (R2) Stability Testing of new Drug Substances and Products. Step 5

Control Strategy Case Studies

Expectations for Data to Support Clinical Trial Drugs

Draft agreed by Pharmacokinetics Working Party January Adoption by CHMP for release for consultation 17 February 2011

A Stability Program for the Distribution of Drug Products

3.4 TEST FOR BACTERIAL ENDOTOXINS. Final text for revision of The International Pharmacopoeia

11.I In-process control Authors: Dr. Christian Gausepohl / Paolomi Mukherji / Update 07

Guidance for Industry Q1A(R2) Stability Testing of New Drug Substances and Products

ALLEN Mouse Brain Atlas

Conducting Effective OOS or OOT Investigations for Unexpected Results from the BET Assay

Environmental Water Testing: Surface Water, Groundwater, Hard Water, Wastewater, & Seawater

GOOD DOCUMENTATION AND QUALITY MANAGEMENT PRINCIPLES. Vimal Sachdeva Technical Officer (Inspector), WHO Prequalification of Medicines Programme

Measurement Systems Correlation MSC for Suppliers

Graphite Furnace AA, Page 1 DETERMINATION OF METALS IN FOOD SAMPLES BY GRAPHITE FURNACE ATOMIC ABSORPTION SPECTROSCOPY (VERSION 1.

Statistical estimation using confidence intervals

Measuring Line Edge Roughness: Fluctuations in Uncertainty

Draft Agreed by Pharmacokinetics Working Party January End of consultation (deadline for comments) 31 May 2011

Transcription:

Design of Experiments for Analytical Method Development and Validation Thomas A. Little Ph.D. 2/12/2014 President Thomas A. Little Consulting 12401 N Wildflower Lane Highland, UT 84003 1-925-285-1847 drlittle@dr-tom.com Design of experiments (DOE) is a well proven characterization approach within product and process development and a key aspect of Quality by Design. Recently more attention has been

placed on applying DOE to analytical methods. DOE for analytical methods has three major applications: 1) method development for new methods or those that need improvement, 2) method validation and 3) quantitation of the influence of analytical methods on product and process acceptance and out-of-specification rates. Method development seeks to understand where critical process parameters are in the analytical method and to minimize their influence on accuracy and precision. DOE for method validation seeks to validate the analytical method for a range of concentrations so that changes in formulation or concentration will not require additional validation as they are changes within a characterized design space. Once methods have been developed, qualified and validated the impact they have on out-of-specification rates and process capability needs to be quantified and evaluated to determine their fitness for use. A systematic approach for using DOE for analytical method development and validation is discussed in this paper and was written in line with the International Conference of Harmonization (ICH) Q2(R1), Q8(R2) and Q9 guidelines. A quantitative understanding of the factors that influence resolution, linearity, precision and accuracy, is integral to applying DOE to method development. Text book approaches to DOE generally suggest a sequential approach to DOE: 1) screening studies, 2) characterization studies and 3) optimization of the method or process. This approach applied to analytical methods is often not practical as 10-20 methods are often used for drug substance and drug product evaluation and the amount of time and materials needed to follow the three step, screen, characterize and optimize would consume unreasonable amounts of resources. The sequence generally recommended by the author for method development is 1) understanding the purpose of the study, 2) perform risk assessments to screen out factors that may or may not have an influence on the analytical method (screening variables by logic and an examination of their scientific potential for influence) and 3) characterization studies to quantify and minimize their influence on precision, accuracy and linearity. Assays and measurement systems must be viewed as a process. The measurement process is made up of methods, standards, software, materials, chemistry, reagents, analysts, sample preparation methods, environmental conditions and instrumentation/equipment. Quality risk management and statistical data analysis techniques should be used to examine the process of measurement and identify factors that may influence precision, accuracy, linearity, signal to noise, limits of detection and quantification and/or any other assay attributes to achieve optimal assay results. DOE for Method Development Figure 1. Measurement Process Elements Design of experiments can be applied to many aspects of method development; however, the following will provide the typical steps for designing and analyzing experiments for analytical methods. 1. Define the purpose (repeatability, intermediate precision, accuracy, LOD/LOQ linearity, resolution, etc.)

2. Define the range of concentrations the method will be used to measure and the solution matrix it will be measured in. 3. Develop/define the reference standards for bias and accuracy studies. 4. Define the steps in the method and any associated documentation. 5. Determine the responses that are aligned to the purpose of the study. 6. Complete a risk assessment of all materials, equipment, analysts and method components aligned to the purpose of the study and the key responses that will be quantified. 7. Design the experimental matrix and sampling plan. 8. Identify the error control plan and run the study. 9. Analyze the study and determine settings and processing conditions that improve method precision and minimize bias errors. Document the design space of the method and associated limits of key factors. 10. Run confirmation tests to confirm settings improve precision, linearity and bias. Evaluate the impact of the method on product acceptance rates and process capability. 1. Identify the Purpose of the Method Experiment Make sure the purpose of the analytical method experiment is clear (repeatability, intermediate precision, linearity, resolution, etc.) The structure of the study, the sampling plan, and ranges used in the study all depend on the purpose of the study. Designing a study for accuracy determination is very different from a study that is designed to explore and improve precision. Accuracy, for example does not require sample replicates to estimate the mean change in the response, precision; however, requires replicates and duplicates to evaluation variation in the sample preparation and in other aspects of the method. The purpose of the study should drive the study design. 2. Define the Range of Concentrations to be Evaluated Define the range of concentrations the used to measure and the solution matrix. Ranges of the concentration will generate the characterized design space so they should be selected carefully as it will put restrictions on how the method may be used in the future. Normally five concentrations should be evaluated per ICH Q2R1.

Figure 2. Concentration Ranges and Other Constituents 3. Define all Reference Standards Used in the Study Develop/define the reference standards for bias and accuracy studies. Without a well characterized reference standard bias/accuracy cannot be determined for the method. Care should be made in selecting, storing and using reference materials. Stability of the reference is a key consideration and accounting for degradation when replacing standards is critical. 4. Identify all Steps in the Analytical Method Layout the flow or sequence used in the analytical method. Define the steps in the method (SOPs, procedures or work instructions), all chemistries, reagents, plates and materials used in the method and all instruments/sensors and equipment. Identify any steps in the process, materials, analyst techniques or equipment that may influence bias or precision. 5. Determine the Reponses Determine the responses that are aligned to the purpose of the study. Raw data and statistical measures such as bias, intermediate precision, signal to noise ratio and CV are all responses and should be considered as independent results from the method. Make sure the data table is set up to collect both the raw data and then the statistics can be easily generated from the raw data and there is a direct link from the statistics to the data. 6. Perform a Risk Assessment A risk assessment of the analytical method is used to identify areas/steps in the method that may influence precision, accuracy, linearity, selectivity, signal to noise etc.

Specifically the risk question is Where do we need characterization and development for this assay? Complete a risk assessment of all materials, equipment, analysts and method components aligned to the purpose of the study and the key responses. The outcome of the risk assessment a small set (3-8) of risk ranked factors that may influence the reportable result of the assay. There are many kinds of factors, so factor identification and how to treat the factor in the analysis is critical to designing valid experiments. There are controllable factors: continuous, discrete numeric, categorical, and mixture. There are uncontrollable factors: covariate and uncontrolled. In addition there are factors used in error control: blocking and constants. CQAs ICH Parameter Safety Identify Purity/impurity Potency Stability Yield CQA/assay name (release) USL Target LSL Assay/Test name for Characterization Only Analytical Method Process Step and or Process Changes Risk Analysis Unit Operation Number Unit Operation Name Baseline (optional) Change (optional) Difference (optional) Potential Risk, Influence or Failure Mode Severity and/or Influence (1,3,5,7,10) Probability and/or Uncertainty (1,3,5,7,10) Detectability (optional) (1,3,5,7,10) Risk Score (RPN) Severity x Probability Only Risk Score (RPN) Severity x Prob x Detect Description Unit Operation Delta ( ) 1 5 3 5 0 75 2 0 0 2 0 0 2 0 0 3 0 0 3 0 0 3 0 0 4 0 0 Figure 3. Analytical Method Risk Assessment Example 7. Design the Experimental Matrix and Sampling Plan For small studies using two or three factors a full factorial type design may be appropriate. When the number of factors rises above three a D-optimal type custom DOE design should be used to more efficiently explore the design space and determine factors that impact the method. There are many good software programs today that help the user define statistically valid experiments and can be customized to meet the user s needs. The experimental matrix is one consideration and the sampling plan is another. Replicates and duplicates are essential to quantification of factor influence on precision. Replicates are complete repeats of the method including repeats of the sample preparation, duplicates are single sample preparations but with multiple measurements or injections using the final chemistry and instrumentation. Replicates provide total method variation and duplicates provide instrument, plate and chemistry precision independent of sample preparation errors. If the experiment is designed properly many of the requirements for method validation (figure 4, Method Validation) can be directly met from the outcomes of the method DOE.

Standard: VALIDATION OF ANALYTICAL PROCEDURES Q2 R1, Nov 2005 Definition Assay Characterization Specificity Linearity Range Accuracy Understanding of the To provide an exact factors that influence the result which allows an mean and standard accurate statement on deviation/cv of the assay. the content or potency of the analyte in a sample. The linearity of an analytical procedure is its ability (within a given range) to obtain test results which are directly proportional to the concentration (amount) of analyte in the sample. The range of an analytical procedure is the interval between the upper and lower concentration (amounts) of analyte in the sample (including these concentrations) for which it has been demonstrated that the analytical procedure has a suitable level of precision, accuracy and linearity. The accuracy of an analytical procedure expresses the closeness of agreement between the value which is accepted either as a conventional true value or an accepted reference value and the value found. Typical Factors Excipients, Concentrations, Assay Methods (# Dilutions) Sample prep method, controlled impurities or sample matrix 3-5 concentrations are typical with 3 min. Concentration Well characterized standards with known potency etc. Recommended Data and Analysis Procedure For the establishment of linearity, a minimum of 5 concentrations is recommended. Other approaches should be justified. ICH Topic Q 2 (R1) Part II. Examination of residuals will indicate where the linear range has been established. Minimum of 9 determinations over a minimum of 3 concentration levels covering the specified range (e.g. 3 concentrations and 3 replicates each of the total analytical procedure). ICH Topic Q 2 (R1) Part II. 10 + determinations is even better for accuracy. Tip QRM, Process Mapping and FR Matrix to identify key factors in the analytical method Assay or analytical method designed to detect the specific drug attribute Linear fit, Ad Rsquare, equation (slope/intercept) and residuals plots Make sure concentrations exceed drug application ranges and refer to linearity study for range Measure mean shift from reference standard JMP Platform DOE, Full Factorial, Custom Designs Fit Model and or Fit Y by X Fit Y by X or Fit Model, Residuals Fit Y by X Fit Y by X, Distribution and Graph Builder Figure 4. Method Validation Quick Reference Guide 8. Identify the Error Control Plan Make sure to measure and record uncontrolled factors during the study, analyst name, equipment ID, out time, hold times, ambient temperature, temperature at the beginning and end of an operation, transfer times, ph, incubation time etc. may all hold valuable information on factors that impact the method. What factors will be restricted or held constant during the study? Do we need to block for batch, lot, sample prep or instruments that may have an influence on the reportable result. 9. Analyze the DOE and Determine the Settings and Design Space Use a good multiple regression/ancova software package that allows the DOE factors and any uncontrolled variables to be correctly evaluated. Analyze the study and determine settings and processing conditions that improve method precision and minimize bias errors. When using statistics from the method (CV, mean, standard deviation) rather than raw data make sure and weight the analysis by the number of replicates or duplicates in order to assure statistical tests and confidence intervals are meaningful. Determine the design space and allowable ranges for all key factors that influence the method.

Figure 5. DOE Design Space for Method Precision and Bias 10. Verify the Modal and Determine the Impact of the Method on Specifications and Capability Run confirmation tests to confirm settings improve precision, linearity and bias. Evaluate the impact of the method on product acceptance rates and process capability. Using an accuracy-to-precision (ATP) model it is possible to visualize the relationship of precision and accuracy on product acceptance rates. The ATP model shows how changes in precision and accuracy impact product acceptance rates and the assay error design space relative to product acceptance specification limits.

Summary Figure 6. Accuracy to Precision Modeling The attention paid to method development, validation and control will greatly improve the quality of drug development, patient safety and predictable, consistent outcomes. Design of experiment is a powerful and underutilized development tool for method characterization and method validation. Analytical professionals need to be comfortable using it to characterize and optimize the analytical method. If used properly and during development DOE will provide significant improvements in precision and a reduction in bias errors. It will further, help to avoid costly and time consuming validation studies as concentration are modified in formulations and dosing schemes are changed for drug product and drug substance. References: ICH Q2(R1) Validation of Analytical Procedures: Text and Methodology, 2005 ICH, Q8(R2) Pharmaceutical Development, 2009 ICH Q9 Quality Risk Management, 2006 Little, T.A. Engineering Statistics and Data Analysis-BPM, 2014