Menu Expand
Data Integrity and Data Governance

Data Integrity and Data Governance

R D McDowall

(2018)

Abstract

Data integrity is the hottest topic in the pharmaceutical industry. Global regulatory agencies have issued guidance, after guidance after guidance in the past few years, most of which does not offer practical advice on how to implement policies, procedures and processes to ensure integrity. These guidances state what but not how. Additionally, key stages of analysis that impact data integrity are omitted entirely.
The aim of this book is to provide practical and detailed help on how to implement data integrity and data governance for regulated analytical laboratories working in or for the pharmaceutical industry. It provides clarification of the regulatory issues and trends, and gives practical methods for meeting regulatory requirements and guidance. Using a data integrity model as a basis, the principles of data integrity and data governance are expanded into practical steps for regulated laboratories to implement. The author uses case study examples to illustrate his points and provides instructions for applying the principles of data integrity and data governance to individual laboratory needs. This book is a useful reference for analytical chemists and scientists, management and senior management working in regulated laboratories requiring either an understanding about data integrity or help in implementing practical solutions. Consultants will also benefit from the practical guidance provided.

Table of Contents

Section Title Page Action Price
Cover Cover
Data Integrity and Data Governance: Practical Implementation in Regulated Laboratories i
Preface v
Acknowledgements vii
Glossary, Abbreviations and Data Integrity Terms ix
Contents xxxiii
Chapter 1 - How to Use This Book and an Introduction to Data Integrity 1
1.1 Aims and Objectives 1
1.2 Structure of This Book 2
1.2.1 Chapter Structure 2
1.2.2 You Do Not Read the Regulations! 2
1.2.3 The Regulatory Environment 4
1.2.4 Data Governance 4
1.2.5 Data Integrity 6
1.2.6 Quality Oversight for Data Integrity 8
1.3 Mapping This Book to the Data Integrity Model 9
1.4 Pharmaceutical Quality System and Data Integrity 9
1.4.1 Integration Within the Pharmaceutical Quality System 9
1.4.2 No Chapter on Risk Management 11
1.4.3 Back to the Future 1: Understanding Current in cGMP 11
1.4.4 The European Equivalent of cGMP 11
1.5 What Is Data Integrity 12
1.5.1 How Many Definitions Would You Like 12
1.5.2 What Do These Definitions Mean 12
1.5.3 ALCOA+ Criteria for Integrity of Laboratory Data 13
1.6 Data Quality and Data Integrity 15
1.6.1 From Sample to Reportable Result 15
1.6.2 Contextual Metadata and a Reportable Result 16
1.6.3 Data Integrity – Can I Trust the Data 18
1.6.4 Data Quality – Can I Use the Data 20
1.6.5 The Proposed FDA GLP Quality System 20
1.6.6 Continual Versus Continuous Improvement 21
1.7 Static Versus Dynamic Data 21
1.8 Important Data Integrity Concepts 22
1.8.1 Data Integrity Is More than Just Numbers 22
1.8.2 Quality Does Not Own Quality Anymore 23
1.8.3 Data Integrity Is Not Just 21 CFR 11 or Annex 11 Compliance 23
1.8.4 Data Integrity Is an IT Problem 24
1.8.5 Data Integrity Is a Laboratory Problem 24
1.8.6 We Are Research – Data Integrity Does Not Impact Us 24
1.9 It’s Déjà vu all Over Again! 24
References 25
Chapter 2 - How Did We Get Here 28
2.1 Barr Laboratories 1993: You Cannot Test into Compliance 28
2.1.1 Background to the Court Case 29
2.1.2 Key Laboratory Findings from the Judgement 29
2.1.3 Regulatory Response 30
2.2 Able Laboratories 2005: You Cannot Falsify into Compliance 30
2.2.1 Background to the Inspection 30
2.2.2 483 Observations 30
2.2.3 Regulatory Response 31
2.3 Ranbaxy Warning Letters and Consent Decrees 32
2.3.1 Background to the Regulatory Action 32
2.3.2 Details of the 2012 Consent Decree 32
2.4 Court Case for GLP Data Falsification 33
2.5 Semler Research Data Falsification 34
2.6 The Cost of Data Integrity Non-compliance 34
2.6.1 Relative Costs of Compliance Versus Non-compliance 35
2.6.2 Is It Worth It 36
2.7 A Parcel of Rogues: FDA Laboratory Data Integrity Citations 36
2.7.1 Why Use Only FDA Warning Letters and 483 Observations 36
2.7.2 Quality Management System Failures 37
2.7.3 Instrument Citations 39
2.7.4 Citations for Lack of Laboratory Controls 41
2.7.5 Failure to Have Complete Laboratory Records 41
2.7.6 Too Much Data – Duplicate Record Sets 43
2.7.7 Industrial Scale Shredding and Discarding of GMP Documents 43
2.7.8 Responses by the Regulatory Authorities 44
References 44
Chapter 3 - The Regulators' Responses 47
3.1 What Do the Regulators Want 47
3.1.1 EU Good Manufacturing Practice Chapter 1 47
3.1.2 EU GMP Chapter 4 on Documentation 48
3.1.3 21 CFR 211 cGMP Regulations for Finished Pharmaceutical Goods 48
3.1.4 EU GMP Annex 11 on Computerised Systems 50
3.1.5 Regulatory Requirements Summary 50
3.2 The Proposed FDA GLP Quality System 50
3.2.1 Background to the Proposed Regulation 50
3.2.2 New Data Quality and Integrity Requirements 51
3.2.3 A New Data Integrity Role for the Study Director 52
3.2.4 The GLP Study Report 52
3.2.5 No Hiding Place for GLP Data Integrity Issues 52
3.3 Overview of Regulatory Guidance Documents for Data Integrity 53
3.4 Food and Drug Administration Guidance Documents 55
3.4.1 FDA Guide to Inspection of Pharmaceutical Quality Control Laboratories 55
3.4.2 FDA Compliance Program Guide 7346.832 on Pre Approval Inspections 56
3.4.3 FDA Level 2 Guidance 57
3.4.4 Delaying, Denying, Limiting or Refusing an FDA Inspection 58
3.4.5 FDA Guidance on Data Integrity and Compliance with cGMP 58
3.4.6 Key Points from the FDA Data Integrity Guidance 60
3.5 MHRA Data Integrity Guidance Documents 60
3.5.1 Initial Request to Industry December 2013 60
3.5.2 MHRA GMP Data Integrity Guidance for Industry 61
3.5.3 MHRA GXP Data Integrity Guidance for Industry 61
3.5.4 MHRA Definition of Raw Data 62
3.6 PIC/S Guidance Documents 63
3.6.1 PIC/S PI-041 Good Practices for Data Management and Integrity in Regulated GMP/GDP Environments 64
3.7 WHO Guidance on Good Data and Records Management Practices 64
3.8 GAMP Guide for Records and Data Integrity 65
3.9 PDA Technical Report 80 68
3.9.1 Regulatory Trends for Data Integrity Issues 70
3.9.2 Data Integrity in Microbiology Laboratories 70
3.9.3 Data Integrity in Analytical QC Laboratories 71
3.9.4 How to Remediate Breaches in Data Integrity 72
3.10 Understanding the Meaning of Raw Data and Complete Data 72
3.10.1 Are Raw Data First-capture or Original Observations 72
3.10.2 In the Beginning … 72
3.10.3 Later, Much Later in Europe … 74
3.10.4 The GLP Quality System – The Proposed 21 CFR 58 Update 74
3.10.5 Extracting Principles for Laboratory GXP Raw Data 75
3.10.6 Visualising What Raw Data Mean 76
3.10.7 Summary: Raw Data Is the Same as Complete Data 78
3.11 Regulations and Data Integrity Guidance Summary 78
References 79
Chapter 4 - What Is Data Governance 82
4.1 What Do the Regulators Want 82
4.1.1 EU GMP Chapter 1 Pharmaceutical Quality System 82
4.1.2 FDA Proposed GLP Quality System Update 83
4.1.3 MHRA GXP Data Integrity Guidance 84
4.1.4 WHO Guidance on Good Records and Data Management Practices 85
4.1.5 PIC/S PI-041 – Good Practices for Data Management and Integrity in Regulated GMP/GDP Environments 87
4.1.6 EMA Questions and Answers on Good Manufacturing Practice – Data Integrity 88
4.1.7 Summary of Regulatory Guidance 88
4.2 The Rationale for Data Governance: Regulatory Boot or Business Imperative 88
4.3 Perspectives of Data Governance Outside the Pharmaceutical Industry 89
4.4 Key Data Governance Elements 90
4.4.1 Summary of Regulatory Guidance for Data Governance 90
4.4.2 Main Data Governance Areas 92
4.4.3 Further Data Governance Chapters in this Book 93
References 94
Chapter 5 - A Data Integrity Model 96
5.1 A Data Integrity Model 96
5.1.1 A Logical Organisation of Data Integrity Elements 97
5.1.2 Descriptions of the Four Levels in the Model 97
5.1.3 An Analogy of Building a House 99
5.1.4 Focus on the Laboratory Levels of the Data Integrity Model 100
5.2 Foundation Level: The Right Corporate Culture for Data Integrity 101
5.2.1 Role of Senior Management 101
5.2.2 Data Governance Functions in the Foundation Level 101
5.3 Level 1: The Right Analytical Instrument and Computer System for the Job 104
5.3.1 Analytical Instrument Qualification and Computerised System Validation (AIQ and CSV) 104
5.3.2 Data Governance Functions in Level 1 105
5.4 Level 2: The Right Analytical Procedure for the Job 105
5.4.1 Validation of Analytical Procedures 105
5.4.2 Verification of Pharmacopoeial Methods 106
5.4.3 Bioanalytical Method Validation Guidance 106
5.4.4 Manual Analytical Procedures Must Be Designed for Data Integrity 106
5.5 Level 3: Right Analysis for the Right Reportable Result 107
5.6 Quality Oversight for Data Integrity 107
5.6.1 Quality Oversight of Laboratory Procedures and Work 107
5.6.2 Data Integrity Audits 108
5.6.3 Data Integrity Investigations 108
5.7 Linking the Data Integrity Model to the Analytical Process 108
5.7.1 The Data Integrity Model in Practice 108
5.7.2 Quality Does Not Own Quality Anymore 110
5.8 Mapping the WHO Guidance to the Data Integrity Model 110
5.9 Assessment of Data Integrity Maturity 112
5.9.1 Data Management Maturity Model 112
5.9.2 Data Integrity Maturity Model 115
References 117
Chapter 6 - Roles and Responsibilities in a Data Governance Programme 119
6.1 What Do the Regulators Want 119
6.1.1 ICH Q10 Pharmaceutical Quality Systems 119
6.1.2 EU GMP Chapter 1 120
6.1.3 PIC/S-041 Good Practices for Data Management and Integrity in Regulated GMP/GDP Environments 121
6.1.4 WHO Guidance on Good Data and Record Management Practices 122
6.1.5 Update of the US GLP Regulations 123
6.1.6 GAMP Guide Records and Data Integrity 124
6.1.7 A Summary of Regulatory and Industry Guidance Documents 124
6.2 Data Governance Roles and Responsibilities – Corporate Level 125
6.3 Data Integrity Policy 129
6.4 Management, Monitoring and Metrics 129
6.5 Data Integrity and Data Governance Roles and Responsibilities – Process and System Level 131
6.5.1 From Data Governance to Data Ownership 131
6.5.2 Process Owner and System Owner 132
6.5.3 Can a Process Owner Be a Data Owner 132
6.5.4 Other Data Governance Roles at the System Level 133
6.5.5 Data Owner 135
6.5.6 Data Steward 136
6.5.7 Is a Lab Administrator a Data Steward 136
6.5.8 Is a Technology Steward a System Owner 137
6.5.9 Segregation of Roles and Duties 137
6.6 The Short Straw ...… 137
6.6.1 Where Are We Now 137
6.6.2 The Hybrid System Nightmare 138
6.7 Cascade of Roles and Responsibilities: from Boardroom to Bench 140
References 140
Chapter 7 - Data Integrity Policies, Procedures and Training 142
7.1 What Do the Regulators Want 142
7.1.1 EU GMP Chapter 4 on Documentation 142
7.1.2 WHO Guidance on Good Data and Record Management Practices 143
7.1.3 PIC/S PI-041 Good Practices for Data Management and Integrity in Regulated GMP/GDP Environments 144
7.1.4 Regulatory Requirements Summary 144
7.2 Environmental Analysis and an Approach to Data Integrity 145
7.2.1 Background to EPA and Data Integrity 145
7.2.2 NELAC and Laboratory Accreditation 146
7.2.3 NELAC Quality System 146
7.2.4 NELAC Data Integrity Training 147
7.3 Corporate Data Integrity Policy Coupled with Effective Training 149
7.3.1 Contents of a Corporate Data Integrity Policy 151
7.3.2 Training in the Data Integrity Policy 152
7.3.3 Agreeing to Comply with the Policy 155
7.4 Suggested Data Integrity Procedures 155
7.5 Principles of Good Documentation Practice 155
7.5.1 Say What You Do 156
7.5.2 Do What You Say 157
7.5.3 Document It 157
7.5.4 Automating Procedure Execution 157
7.6 Training to Collect and Manage Raw Data and Complete Data 158
7.6.1 Principles for GXP Laboratory Raw Data and Complete Data 158
7.6.2 Approach to Training for Complete and Raw Data in the Laboratory 159
7.6.3 Example 1 – Paper Records from a Manual Test 159
7.6.4 Example 2 – Spectroscopic Analysis Using a Hybrid System 161
7.6.5 Example 3 – Chromatographic Analysis with a CDS Interfaced with a LIMS 163
7.6.6 Additional Raw Data 165
7.7 Good Documentation Practice for Paper Records 165
7.7.1 Recording Observations and Results 166
7.7.2 Examples of Good and Poor Documentation Practice for Handwritten Records 167
7.7.3 Fat Finger, Falsification and Fraud – Take 1 168
7.7.4 Original Records and True Copies 169
7.8 Good Documentation Practice for Hybrid Records 169
7.8.1 Record Signature Linking for Hybrid Systems – Spreadsheet Example 171
7.9 Good Documentation Practice for Electronic Records 172
7.9.1 Good Documentation Practice for Electronic Records 173
7.10 Good Documentation Practice Training 173
7.11 Role of the Instrument Log Book 173
7.11.1 EU GMP Chapter 4 on Documentation 175
7.11.2 FDA Good Laboratory Practice 21 CFR 58 177
7.11.3 FDA 21 CFR 211 cGMP for Finished Pharmaceutical Products 177
7.11.4 FDA Inspection of Pharmaceutical QC Laboratories 177
7.11.5 Instrument Lag Books in Practice 178
7.12 Training for Generating, Interpreting and Reviewing Laboratory Data 178
7.12.1 Data Integrity Training for a Chromatography Data System: Operational SOPs 178
7.12.2 Training Is of Little Value without an Open Culture 179
References 179
Chapter 8 - Establishing and Maintaining an Open Culture for Data Integrity 181
8.1 What Do the Regulators Want 181
8.1.1 WHO Guidance on Good Data and Record Management Practices 181
8.1.2 PIC/S PI-041 Good Practices for Data Management and Integrity in Regulated GMP/GDP Environments 182
8.1.3 MHRA “GXP” Data Integrity Guidance and Definitions 183
8.1.4 Regulatory Guidance Summary 183
8.2 Bad Culture: Cressey's Fraud Triangle and Organisational Pressure 184
8.2.1 Cressey's Fraud Triangle 184
8.2.2 Breaking the Fraud Triangle 185
8.2.3 Managerial and Peer Pressures Can Influence Analytical Results 186
8.3 ISPE Cultural Excellence Report 187
8.4 Management Leadership 188
8.4.1 Generate and Communicate the Data Integrity Vision 188
8.4.2 Talk the Talk and Walk the Walk 188
8.4.3 Reinforcing an Open Culture for Data Integrity 189
8.4.4 FDA Expectations for Analysts 189
8.5 Mind Set and Attitudes 189
8.5.1 Quality Does Not Own Quality Anymore 190
8.5.2 The Iceberg of Ignorance 190
8.5.3 How Do I Raise Problems to Management 190
8.6 Gemba Walks 192
8.6.1 Where Does a Gemba Walk Fit in a QMS 192
8.6.2 What Gemba Walks Are and Are Not 193
8.6.3 Why Bother with a Gemba Walk 194
8.6.4 Activation Energy for a Gemba Walk 194
8.6.5 Performing the Gemba Walk 195
8.6.6 Keep the Focus on the Process 196
8.6.7 Generic Questions for a Gemba Walk 196
8.6.8 Let Management See Analytical Instruments First Hand 197
8.7 Fat Finger, Falsification and Fraud – Take 2 197
8.7.1 To Err Is Human 197
8.7.2 Verification of Data Entry 198
8.7.3 What Is the Fat Finger Rate in a Laboratory 198
8.7.4 Learning from Health Service Studies 199
8.8 Maintaining the Open Culture 200
References 200
Chapter 9 - An Analytical Data Life Cycle 202
9.1 What Do the Regulators Want 202
9.1.1 MHRA GXP Data Integrity Guidance 202
9.1.2 WHO Guidance on Good Data and Record Management Practices 203
9.1.3 PIC/S PI-041 Good Practices for Data Management and Integrity in Regulated GMP/GDP Environments 203
9.1.4 Regulatory Requirements Summary 204
9.2 Published Data Life Cycles 205
9.2.1 GAMP Guide on Records and Data Integrity 205
9.2.2 Validation of Chromatography Data Systems 206
9.2.3 Critique of the Two Life Cycle Models 207
9.3 An Analytical Data Life Cycle 208
9.3.1 Overview of an Analytical Data Life Cycle 208
9.3.2 Controlling the Analytical Data Life Cycle 209
9.3.3 Phases of the Analytical Data Life Cycle 210
9.3.4 Generic Data Life Cycles Do Not Work in the Laboratory 212
9.3.5 The Requirement for Flexibility to Adapt to Different Analytical Procedures 212
9.4 Establishing Data Criticality and Inherent Integrity Risk 215
9.4.1 Spectrum of Analytical Instruments and Laboratory Computerised Systems 215
9.5 Risks to Data Over the Data Life Cycle 218
9.5.1 PIC/S PI-041 Good Practices for Data Management and Integrity in Regulated GMP/GDP Environments 218
9.5.2 Initial Assessment of Risk of the Analytical Data Life Cycle Phases 219
9.5.3 Phases of the Data Life Cycle are Equal but Some are More Equal than Others 220
9.5.4 Summary Risks in the Analytical Data Life Cycle 221
References 221
Chapter 10 - Assessment and Remediation of Laboratory Processes and Systems 223
10.1 What Do the Regulators Want 224
10.1.1 WHO Guidance on Good Data and Record Management Practices 224
10.1.2 PIC/S PI-041 Good Practices for Data Management and Integrity in Regulated GMP/GDP Environments 224
10.1.3 MHRA GXP Data Integrity Guidance and Definitions 224
10.1.4 Regulatory Guidance Summary 225
10.2 Business Rationale for Assessment and Remediation 225
10.2.1 Improve Business Processes 225
10.2.2 Ensure Regulatory Compliance 225
10.2.3 Release Product Earlier 226
10.2.4 The Problem is Management 226
10.3 Current Approaches to System Assessment and Remediation 226
10.3.1 The Rationale for Current Approaches 226
10.3.2 Assessment of Validated Computerised Systems 227
10.4 Data Process Mapping 229
10.4.1 The Problem with Checklists 229
10.4.2 What is Data Process Mapping 229
10.4.3 Instrument Data System with Spreadsheet Calculations 232
10.4.4 Spreadsheets Used for GMP Calculations Are High Risk 233
10.4.5 Critical Activities in a Process 234
10.4.6 Fix and Forget versus Delivering Business Benefits 235
10.4.7 Short Term Remediation Leading to Long Term Solution 236
10.5 Data Integrity Issues with Analysis by Observation 238
10.5.1 Potential Problems with Analysis by Observation 238
10.5.2 A Risk Based Approach to Analysis by Observation 238
10.5.3 Melting Point Determination 239
10.6 Data Integrity Issues with Paper Records 239
10.6.1 Blank Forms Must be Controlled with Accountability 240
References 241
Chapter 11 - Data Integrity and Paper Records: Blank Forms and Instrument Log Books 242
11.1 What Do the Regulators Want – Blank Forms 242
11.1.1 Focus on the Key Data Integrity Issues with Paper Records 242
11.1.2 FDA Guide to Inspection of Quality Control Laboratories 243
11.1.3 MHRA GMP Data Integrity Guidance 243
11.1.4 MHRA Draft GXP Data Integrity Guidance 243
11.1.5 MHRA GXP Data Integrity Guidance and Definitions 244
11.1.6 WHO Guidance on Good Data and Record Management Practices 244
11.1.7 FDA Data Integrity and Compliance with cGMP 244
11.1.8 PIC/S PI-041 Good Practices for Data Management and Integrity in Regulated GMP/GDP Environments 245
11.1.9 EMA GMP Questions and Answers on Data Integrity 246
11.1.10 Regulatory Guidance Summary 246
11.2 Control of Master Templates and Blank Forms 247
11.2.1 Understanding Master Templates and Blank Forms 247
11.2.2 Requirements for the Design, Approval and Storage of Master Templates 248
11.2.3 Process for Generation, Review and Approval of a Master Template 248
11.2.4 Requirements for the Issue and Reconciliation of Blank Forms 251
11.2.5 Process for Issue and Reconciliation of Blank Forms 253
11.2.6 Process for Issue and Reconciliation of Blank Forms 254
11.2.7 Completing Blank Forms and Creating GXP Records 255
11.3 What Do the Regulators Want – Instrument Log Books 255
11.3.1 EU GMP Chapter 4 on Documentation 255
11.3.2 FDA GMP 21 CFR 211 255
11.3.3 FDA Good Laboratory Practice 21 CFR 58 257
11.3.4 OECD GLP Regulations 257
11.3.5 Summary of Regulatory Requirements for an Instrument Log Book 257
11.4 The Role of an Instrument Log Book for Ensuring Data Integrity 258
11.4.1 Why is an Instrument Log Book Important 258
11.4.2 What Needs to be Entered in the Log Book 259
11.4.3 Inspectors Know the Importance of an Instrument Log 260
11.4.4 FDA Citations for Laboratory Log Books 261
11.4.5 Instrument Log Books in Practice 261
11.5 Role of the Instrument Log Book in the Second Person Review 262
11.5.1 Is an Instrument Performing OK 262
11.6 Automating Blank Forms and Instrument Log Books 263
11.6.1 Automating Master Templates and Blank Forms 263
11.6.2 Instrument Log Book 264
Acknowledgements 265
References 265
Chapter 12 - The Hybrid System Problem 267
12.1 What Do the Regulators Want 267
12.1.1 Electronic Records and Electronic Signatures Regulations (21 CFR 11) 267
12.1.2 WHO Guidance on Good Data and Record Management Practices 268
12.1.3 PIC/S PI-041 Good Practices for Data Management and Integrity in Regulated GMP/GDP Environments 269
12.1.4 EU GMP Chapter 4 on Documentation 269
12.1.5 FDA Guidance for Industry Data Integrity and cGMP Compliance 269
12.1.6 FDA Level 2 Guidance for Records and Reports 271
12.1.7 Regulatory Summary 272
12.2 What Is a Hybrid System 272
12.2.1 WHO Definition of a Hybrid System 272
12.2.2 Key Features of a Hybrid System 273
12.3 The Core Problems of Hybrid Systems 273
12.3.1 A Typical Hybrid System Configuration 273
12.3.2 File Organisation and Printing Results 275
12.3.3 Synchronising Paper Printouts and Electronic Records 276
12.3.4 A Simple Way to Reduce Paper with Hybrid Systems 278
12.4 Eliminate Hybrid Systems 278
References 280
Chapter 13 - Get Rid of Paper: Why Electronic Processes are Better for Data Integrity 281
13.1 What Do the Regulators Want 281
13.1.1 WHO Guidance on Good Data and Record Management Practices 281
13.1.2 PIC/S PI-041 Good Practices for Data Management and Integrity in Regulated GMP/GDP Environments 282
13.1.3 EU GMP Annex 11 Computerised Systems 283
13.1.4 Regulatory Summary 283
13.2 Why Bother with Paper 284
13.2.1 Tradition – Why Change Our Approach 284
13.2.2 Back to the Future 2: Understanding the Current in cGMP 284
13.2.3 The Pharmaceutical Industry is a Two Sigma Industry 285
13.2.4 Are the Regulations Part of the Data Integrity Problem 286
13.2.5 Is Paper a Realistic Record Medium Now 287
13.3 Design Principles for Electronic Working 287
13.4 Designing Data Workflows 1 – Analytical Balances 289
13.4.1 Weighing a Reference Standard or Sample 290
13.4.2 Recording a Weight by Observation 290
13.4.3 Recording Balance Weights with a Printer 291
13.4.4 Connecting the Balance to an Instrument Data System 292
13.5 Designing Data Workflows 2 – Chromatography Data Systems and LIMS 295
13.5.1 Options for Interfacing 295
13.5.2 Manual Data Transfer Between CDS and LIMS 297
13.5.3 Unidirectional Interfacing from CDS to LIMS 297
13.5.4 Bidirectional Interfacing Between CDS and LIMS 299
13.6 Impact on Data Integrity and Second Person Review 301
13.6.1 Ensuring Data Integrity with Electronic Working 301
13.6.2 Impact on Second Person Review 302
13.6.3 Summary of an Approach for Electronic Working that Ensures Data Integrity 302
References 302
Chapter 14 - Data Integrity Centric Analytical Instrument Qualification and Computerised System Validation 305
14.1 What the Regulators Want 306
14.1.1 21 CFR 211 Current GMP for Finished Pharmaceutical Products 306
14.1.2 21 CFR 58 GLP for Non-clinical Studies 306
14.1.3 United States Pharmacopoeia ၘ on Analytical Instrument Qualification 306
14.1.4 EU GMP Annex 11 307
14.1.5 ICH Q7 and EU GMP Part 2: Good Manufacturing Practice Guide for Active Pharmaceutical Ingredients 307
14.1.6 WHO Guidance on Good Data and Record Management Practices 307
14.1.7 PIC/S PI-041 Good Practices for Data Management and Integrity in Regulated GMP/GDP Environments 308
14.1.8 Regulatory Summary 308
14.2 GMP Regulations and the Pharmacopoeias 309
14.2.1 Relationship Between GMP and the Pharmacopoeias 309
14.2.2 Importance of USP ၘ 310
14.2.3 Use the USP ၘ Principles for GLP Instruments and Systems 311
14.3 Why Is Instrument Qualification Important 311
14.3.1 Data Quality Triangle 311
14.3.2 Data Integrity Model 312
14.4 Why a New Revision of USP ၘ 312
14.4.1 Problems with the 2008 Version 312
14.4.2 Revision Path of USP ၘ 313
14.5 What Has Changed with USP ၘ 314
14.5.1 Differences Between the Old and New Versions of USP ၘ 314
14.5.2 Omitted Sections in the New Version 314
14.5.3 Additions and Changes to USP ၘ 315
14.5.4 Roles and Responsibilities 315
14.5.5 An Updated 4Qs Model 315
14.5.6 Harmonisation of Qualification Approaches 319
14.6 Importance of the Laboratory URS for Analytical Instruments 320
14.6.1 Role of the URS 320
14.6.2 Understand Your Intended Use 321
14.6.3 A Role of the Supplier: Write Meaningful Specifications 321
14.6.4 How Minimal Is Minimal 322
14.6.5 Do Not Forget the Software! 323
14.6.6 Purchasing a Second Instrument 323
14.6.7 It's all About Investment Protection 323
14.7 Software Validation Changes to USP ၘ 324
14.7.1 Improving the Analytical Process 325
14.7.2 A Validated System with Vulnerable Records Means Data Integrity Problems 326
14.7.3 Change the Validation Approach to Ensure Data Integrity 328
14.7.4 Brave New CSV World 328
14.7.5 Turning Principles into Practice 329
14.7.6 Qualified, Validated and Released for Operational Use 331
14.8 Performance Qualification 331
14.8.1 Changes to USP ၘ and the Impact on Understanding of PQ 332
14.8.2 Linking the URS, OQ, and PQ 333
14.8.3 PQ for Group A Instruments 334
14.8.4 PQ for Group B Instruments 334
14.8.5 PQ for Group C Instruments 335
14.8.6 System Suitability Tests as Part of a PQ 337
14.8.7 Keep It as Simple as Possible – But No Simpler 338
14.8.8 Holistic HPLC PQ Test 338
Acknowledgement 339
References 339
Chapter 15 - Validating Analytical Procedures 342
15.1 What the Regulators Want 343
15.1.1 US GMP 21 CFR 211 343
15.1.2 EU GMP Chapter 6 on Quality Control 343
15.1.3 EU GMP Annex 15: Qualification and Validation 343
15.1.4 Bioanalytical Method Validation Guidances 343
15.1.5 Regulatory Requirements Summary 345
15.1.6 Outsource Analytical Work with Care 345
15.2 Current Method Validation Guidance 346
15.2.1 Terminology: Analytical Method or Analytical Procedure 346
15.2.2 Business Rationale for Procedure Validation/Verification 346
15.2.3 ICH Q2(R1) Validation of Analytical Procedures: Text and Methodology 348
15.2.4 FDA Guidance for Industry on Analytical Procedure Validation 348
15.2.5 Update of ICH Q2(R1) to a Life Cycle Approach 348
15.2.6 Pharmacopoeial Methods Do Not Work as Written 349
15.3 Role of Analytical Procedure Validation in Data Integrity 350
15.3.1 Method Validation in the Data Integrity Model 350
15.3.2 Equating the Data Integrity Model with the USP ၘ Data Quality Triangle 351
15.4 Current Approaches to Validation and Verification of Procedures 351
15.4.1 Good Manufacturing Practice 351
15.4.2 Bioanalytical Method Validation 351
15.4.3 Validation Documentation for Analytical Procedures 353
15.4.4 Validation Parameters 354
15.5 Overview of the Life Cycle of Analytical Procedures 354
15.5.1 USP ሠ and Stimuli to the Revision Process 354
15.5.2 Life Cycle of Analytical Procedures 355
15.6 Defining the Analytical Target Profile (ATP) 356
15.6.1 Specification for an Analytical Procedure 356
15.6.2 Advantages and Limitations of an Analytical Target Profile 356
15.7 Stage 1: Procedure Design and Development 357
15.7.1 Overview 357
15.7.2 Information Gathering and Initial Procedure Design 357
15.7.3 Iterative Method Development and Method Optimisation 358
15.7.4 Risk Assessment and Management 360
15.7.5 Analytical Control Strategy: Identifying and Controlling Risk Parameters 361
15.7.6 Procedure Development Report 361
15.8 Stage 2: Procedure Performance Qualification 361
15.8.1 Planning the Validation 361
15.8.2 Validation Report 362
15.8.3 Analytical Procedure Transfer 363
15.9 Stage 3: Procedure Performance Verification 364
15.9.1 Routine Monitoring of Analytical Performance 364
15.9.2 Changes to an Analytical Procedure 364
15.9.3 Validated Analytical Procedure 364
References 365
Chapter 16 - Performing an Analysis 367
16.1 What the Regulators Want 368
16.1.1 EU GMP Chapter 1 Pharmaceutical Quality System 368
16.1.2 US GMP 21 CFR 211 GMP for Finished Pharmaceutical Products 368
16.1.3 FDA Guide for Inspection of Pharmaceutical Quality Control Laboratories 369
16.2 The Analytical Process 370
16.2.1 Linking the Data Integrity Model to the Analytical Process 370
16.2.2 Process Overview 372
16.2.3 Analytical Instruments Are Qualified and/or Validated 372
16.2.4 System Suitability Tests and Point of Use Checks 372
16.3 The Scope of Analytical Procedures 374
16.4 Sampling and Sample Management 375
16.4.1 What the Regulators Want 375
16.4.2 Sampling Is Critical 376
16.4.3 GMP Sample Plan and Sampling 377
16.4.4 GLP Protocol and Sampling 377
16.4.5 Ensure Correct Sample Labelling 378
16.4.6 Transport to the Laboratory 379
16.4.7 Sample Receipt and Storage 380
16.4.8 Sample Collection Best Practice 381
16.5 Reference Standards and Reagents 382
16.5.1 What the Regulators Want 382
16.5.2 Preparation of Reference Standards and Solutions 383
16.5.3 Sweep Under the Carpet or Own Up to a Mistake 384
16.5.4 What Is the FDA's View of Analyst Mistakes 385
16.6 Sample Preparation 386
16.6.1 What the Regulators Want 386
16.6.2 Sample Preparation Current Practices 386
16.6.3 Automate Where Technically Feasible 387
16.7 Recording Data by Observation 388
16.7.1 Typical Tests Recording Results by Observation 388
16.7.2 Instruments with No Printer or Data Transfer Capability 388
16.7.3 Pharmacopoeial Indicator Tests 389
16.8 Sample Preparation Followed by Instrumental Analysis Methods 389
16.8.1 An Illustrative Analytical Procedure 389
16.8.2 Ensuring Data Integrity 390
16.8.3 Consider Alternate Analytical Approaches 390
16.9 Methods Involving Instrumental Analysis and Data Interpretation 391
16.9.1 What the Regulators Want 391
16.9.2 Near Infra-red (NIR) Identity Testing 392
16.9.3 Building a Spectral Library 392
16.9.4 Performing the Analysis 393
16.10 Chromatographic Analysis and CDS Data Interpretation 393
16.10.1 What the Regulators Want 394
16.10.2 Setting Up the Chromatograph and Acquiring Data 394
16.10.3 Entering Factors, Weights, and Other Assay Values into the Sequence File 394
16.10.4 An Alternate Approach to Weights and Factors 396
16.10.5 System Evaluation Injections 397
16.10.6 System Suitability Tests – What the Regulators Want 398
16.10.7 Integrating Chromatograms 399
16.10.8 General Principles for Ensuring Good Chromatographic Integration 400
16.10.9 SOP for Integration of Chromatograms 401
16.10.10 Bioanalytical Guidance for Integration of Chromatograms 404
16.10.11 Incomplete (Aborted) Runs 405
16.10.12 Other Unplanned Injections 406
16.10.13 Data Storage Locations 406
16.10.14 Chromatography Falsification Practices 1: Peak Shaving and Enhancing 406
16.10.15 Chromatography Falsification Practices 2: Inhibiting Integration 407
16.10.16 Chromatography Falsification Practices 3: Integrating Samples First 408
16.11 Calculation of Reportable Results 408
16.11.1 What the Regulators Want 409
16.11.2 General Considerations for Calculations 410
16.11.3 Avoid Using Spreadsheets for Analytical Calculations Whenever Possible 411
16.11.4 Calculation of Reportable Results and Out of Specification Results 411
16.11.5 Completion of Testing 412
Acknowledgement 412
References 412
Chapter 17 - Second Person Review 415
17.1 What Do the Regulators Want 416
17.1.1 cGMP for Finished Pharmaceutical Products (21 CFR 211) 416
17.1.2 EU GMP Chapter 6 Quality Control 416
17.1.3 EU GMP Annex 11 416
17.1.4 MHRA GXP Data Integrity Guidance and Definitions 417
17.1.5 FDA Guidance on Data Integrity and cGMP Compliance 417
17.1.6 WHO Guidance on Good Data and Record Management Practices 419
17.1.7 Regulatory Compliance Summary 420
17.2 What the Regulators Want: Out of Specification (OOS) Results 420
17.2.1 21 CFR 211 420
17.2.2 EU GMP Chapter 6 Quality Control 421
17.2.3 FDA Guidance for Industry on Investigating OOS Test Results 421
17.2.4 FDA Guidance on Quality Metrics 422
17.2.5 OOS Definitions 422
17.2.6 OOS Regulatory Summary 422
17.3 Procedures for the Second Person Review 423
17.3.1 Who Should Conduct a Second Person Review 423
17.3.2 The Scope of the Procedure 423
17.3.3 The Troika of Record Review 424
17.3.4 Timeliness of the Second Person Review 425
17.3.5 Documenting the Audit Trail Review 425
17.3.6 Training for Second Person Review 425
17.3.7 Out of Specification (OOS) Procedure 426
17.4 Second Person Review of Analytical Procedures Involving Observation 426
17.4.1 What Is an Analytical Procedure Involving Observation 426
17.4.2 Improving Manual Analytical Procedures 426
17.4.3 Witness Testing or Second Person Review 427
17.5 Sample Preparation and Instrumental Analysis 428
17.5.1 Loss on Drying Analysis 428
17.5.2 Review of the Second Person Review of the Analytical Records 429
17.6 Second Person Review of a Hybrid System Records 431
17.6.1 Increased Scope of Record and Data Review 431
17.6.2 Technical Versus Procedural Controls for Second Person Review 431
17.6.3 The Scope of an Analytical Procedure Involving a Hybrid System 432
17.6.4 Technical Controls to Aid a Second Person Review 433
17.6.5 Paper and Electronic Records to be Reviewed 434
17.6.6 Recording the Work Performed and the Review 434
17.6.7 Original Record or True Copy 435
17.6.8 Have Critical Data Been Entered into the Instrument Data System 436
17.6.9 Review of Electronic Records, Metadata and Audit Trail 436
17.6.10 Second Person Review to Ensure Data Have Not Been Falsified 437
17.6.11 Do You Really Want to Work This Way 437
17.7 Risk Based Audit Trail Review 438
17.7.1 MHRA GXP Data Integrity Guidance and Definitions 438
17.7.2 Which Audit Trail Should Be Reviewed 439
17.7.3 How Regular Is a Regular Review of Audit Trail Entries 439
17.8 Second Person Review of Electronic Systems and Data 442
17.8.1 LIMS Interfaced with a CDS 442
17.8.2 A Second Person Review Is Process Not System Centric 444
17.9 Recording and Investigating Out of Specification Results 447
17.9.1 Phase 1: Initial OOS Laboratory Investigation 448
17.9.2 Phase 2A Production 450
17.9.3 Phase 2B Additional Laboratory Testing 450
17.9.4 OOS Investigations: Prevention Is Better than the Cure 451
References 451
Chapter 18 - Record Retention 453
18.1 What Do the Regulators Want 453
18.1.1 WHO Guidance on Good Data and Record Management Practices 453
18.1.2 EU GMP Annex 11 454
18.1.3 GLP Regulations: 21 CFR 58 454
18.1.4 US GMP Regulations: 21 CFR 211 455
18.1.5 21 CFR 11 Requirements 455
18.1.6 MHRA GXP Data Integrity Guidance and Definitions 456
18.1.7 FDA Guidance on Data Integrity and cGMP Compliance 456
18.1.8 EU GMP Chapter 4 Documentation 457
18.1.9 FDA Guidance for Industry Part 11 – Scope and Application Guidance 457
18.1.10 FDA Inspection of Pharmaceutical Quality Control Laboratories 458
18.1.11 OECD GLP Regulations 458
18.1.12 OECD GLP Guidance on Application of GLP to Computerised Systems 459
18.1.13 Regulatory Requirements Summary 459
18.2 Laboratory Data File Formats and Standards 460
18.2.1 JCAMP-DX Data Format for Spectroscopy 460
18.2.2 Current CDS Data Standards 461
18.2.3 Progress Towards Universal Data File Formats 461
18.3 Options for Electronic Records Retention and Archive 462
18.3.1 Backup Is Not Archive (Unless You Are the FDA) 462
18.3.2 Organising Electronic Records to Retain 463
18.3.3 Options for Electronic Archive 464
18.3.4 Can I Read the Records 465
18.3.5 Impact of a Changed Data System File Format 466
18.3.6 Selection of Off-line Archive Media 466
18.3.7 Changing the Instrument Data System – What Are the Archive Options 467
18.3.8 Overview of Some Options 467
18.3.9 Assessment of Option Feasibility 467
18.4 OECD Guidance for Developing an Electronic Archive 468
18.4.1 Definitions 468
18.4.2 Roles and Responsibilities 469
18.4.3 Archive Facilities 469
18.4.4 Archiving Electronic Records 470
References 472
Chapter 19 - Quality Metrics for Data Integrity 474
19.1 What Do the Regulators Want 474
19.1.1 EU GMP Chapter 6 Quality Control 474
19.1.2 FDA Quality Metrics Guidance for Industry 475
19.1.3 WHO Guidance on Good Data and Record Management Practices 475
19.1.4 PIC/S PI-041 Good Practices for Data Management and Integrity in Regulated GMP/GDP Environments 476
19.1.5 MHRA GXP Data Integrity Guidance and Definitions 477
19.1.6 Regulatory Guidance Summary 477
19.2 KPIs and Metrics for the Laboratory 477
19.2.1 Understanding Laboratory Metrics 478
19.2.2 Metrics Must Be Generated Automatically 478
19.2.3 Why Metrics for Data Integrity 479
19.2.4 Do Quality Metrics Lead Behaviour 479
19.2.5 Are Incidents Hidden Metrics 481
19.3 Data Integrity Metrics in an Organisation 481
19.3.1 Overview: Start Small and Expand 481
19.3.2 Scope of the Organisation 482
19.3.3 Some Suggested Data Integrity Metrics 482
19.4 DI Policies, Assessment and Remediation of Processes and Systems 482
19.4.1 Data Integrity Policy and Associated Procedures 482
19.4.2 Assessment of Processes and Systems 483
19.4.3 Executed Remediation Plans 484
19.5 Laboratory Data Integrity Metrics 486
19.5.1 Some Preliminary Considerations for Laboratory Data Integrity Metrics 486
19.5.2 Outsourced Laboratory Testing 487
19.6 Quality Assurance DI Metrics 487
19.7 Management Review of DI Metrics 488
19.7.1 Management Are Responsible for Data Integrity and the PQS 488
19.7.2 How Regular Is Regular Review 489
Acknowledgement 489
References 489
Chapter 20 - Raising Data Integrity Concerns 491
20.1 What Do the Regulators Want 491
20.1.1 WHO Guidance on Good Data and Record Management Practices 491
20.1.2 PIC/S PI-041 Good Practices for Data Management and Integrity in Regulated GMP/GDP Environments 492
20.1.3 NELAC Quality Standard 493
20.1.4 Regulatory Guidance Summary 493
20.2 Data Integrity Problem or Concern 493
20.3 What Is Needed to Raise a Data Integrity Concern 494
20.3.1 A Section in the Corporate Data Integrity Policy 494
20.3.2 Communicate and Train How to Raise Data Integrity Concerns 494
20.3.3 Raising a Concern or Airing a Grievance 495
20.3.4 What Should Be Reported 495
20.3.5 Protecting the Whistleblower 495
20.3.6 Confidentiality 495
20.3.7 Raising Concerns Anonymously 496
20.4 Raising a Concern 496
20.4.1 Who Should You Raise Your Concern with 496
20.4.2 How to Raise a Concern 496
20.4.3 Raise an Issue via Management or Quality Assurance 497
20.4.4 What the Organisation Must Do 497
20.4.5 What If the Company Is the Problem 498
References 498
Chapter 21 - Quality Assurance Oversight for Data Integrity 499
21.1 What Do the Regulators Want 499
21.1.1 EU GMP Chapter 9 Self-inspections 499
21.1.2 US GMP 21 CFR 211 Current Good Manufacturing Practice for Finished Pharmaceutical Products 500
21.1.3 FDA Compliance Program Guide 7346.832 for Pre-approval Inspections 500
21.1.4 21 CFR 58 Good Laboratory Practice for Non-clinical Laboratory Studies 501
21.1.5 MHRA GXP Data Integrity Guidance and Definitions 501
21.1.6 WHO Guidance on Good Data and Record Management Practices 502
21.1.7 PIC/S-PI-041 Good Practices for Data Management and Integrity in Regulated GMP/GDP Environments 503
21.1.8 Regulatory Compliance Summary 504
21.1.9 Role of the Laboratory in Ensuring Data Integrity 505
21.2 Data Integrity Audits: Planning and Execution 505
21.2.1 Rationale for Data Integrity Audits 505
21.2.2 What Are the Objectives of a Laboratory Data Integrity Audit 505
21.2.3 What Will We Audit The Data Integrity Inventory and Data Criticality 506
21.2.4 What Is the Order and Frequency of Audit 506
21.2.5 Who Will Conduct the Audit 508
21.2.6 Data Integrity Audits and Periodic Reviews of Computerised Systems 508
21.2.7 Procedure and Checklist for a Data Integrity Audit 509
21.3 Conducting a Laboratory Data Integrity Audit 510
21.3.1 Relationship Between the Data Integrity Model and a Data Integrity Audit 510
21.3.2 Overview of the Analytical Process for a Laboratory Data Integrity Audit 511
21.3.3 Expectations for Laboratory Records 513
21.3.4 Auditing Records and Data from Sampling to Report 513
21.3.5 Checking the Configuration Settings of Computerised Systems 515
21.3.6 Identification and Investigation of Laboratory Out of Specification Results 516
21.3.7 Photographs to Support Audit Observations and Findings 516
21.3.8 Reporting the Audit 517
21.4 What Is a Forensic Approach to Data Checking 517
21.4.1 Forensic Data Analysis 517
21.4.2 Recovery of Deleted Files 518
21.4.3 Forensic Data Analysis Techniques 519
21.5 Triggers for a Data Integrity Investigation 519
References 520
Chapter 22 - How to Conduct a Data Integrity Investigation 521
22.1 What the Regulators Require 521
22.1.1 WHO Guidance on Good Data and Record Management Practices 522
22.1.2 FDA Guidance on Data Integrity and Compliance with CGMP 523
22.1.3 FDA Application Integrity Policy 523
22.1.4 PIC/S PI-041 Good Practices for Data Management and Integrity in Regulated GMP/GDP Environments 524
22.1.5 Summary of Data Investigation Regulations and Guidance 525
22.2 Case Study 1: Software Error Investigation 527
22.2.1 Case Study 1 Background 527
22.2.2 Sequester a Copy of the System and the Data 527
22.2.3 Temporary Resolution of the Problem 528
22.2.4 Systems Approach to the Issue 528
22.2.5 Time Frame of the Potential Data Integrity Vulnerability 528
22.2.6 Investigating the Impacted Database 529
22.2.7 Informing Regulatory Authorities 529
22.3 Case Study 2: Data Falsification Investigation 530
22.3.1 Case Study Background 530
22.3.2 Meeting the Intent of the Application Integrity Policy 531
22.3.3 Scope of the Data Integrity Investigation 533
22.3.4 Approaches to the Investigation of Laboratory Data Integrity Issues 533
22.3.5 Do Not Just Focus on Data Integrity Violations – Look Also for Poor Practices 534
22.3.6 Investigation of Tests Using Observation 534
22.3.7 Investigation of Simple Analytical Testing 535
22.3.8 Investigation of Analytical Testing by Chromatography 535
22.3.9 Staff Interviews 536
22.3.10 Findings and Their Classification 537
22.3.11 Root Cause of Data Integrity and Poor Data Management Practices 540
22.3.12 Assessment of Material Impact 543
22.3.13 CAPA Plans: Short-term Remediation and Long-term Solutions 544
22.4 Summary 545
References 545
Chapter 23 - Data Integrity and Outsourcing 547
23.1 What the Regulators Want 547
23.1.1 WHO Guidance on Good Data and Record Management Practices 547
23.1.2 PIC/S PI-041 Good Practices for Data Management and Integrity in Regulated GMP/GDP Environments 549
23.1.3 Regulatory Guidance Summary 551
23.2 Cetero Research Laboratories Data Falsification Case 552
23.3 Include Data Integrity in Supplier Assessment/Audit 553
23.3.1 Current Approaches to Laboratory Audit 553
23.3.2 Extending Assessment to Include Data Integrity 554
23.4 Initial Data Integrity Assessment of a Facility 554
23.4.1 Initial Selection of the Contract Laboratory 555
23.4.2 Do Not Forget the Scientific and Technical Competence of the Supplier 556
23.4.3 Request for Pre-audit Information 556
23.4.4 Planning the Audit 557
23.4.5 Data Governance and Data Integrity in the Context of a PQS 557
23.4.6 Investigate Electronic Record Controls 558
23.4.7 Conclusion of the Audit 559
23.5 Agreements and Contracts for Data Integrity 560
23.5.1 Main Data Integrity Contractual Responsibilities 560
23.5.2 Using the Same Chromatography Data System 561
23.5.3 Storage of the Records Generated 561
23.6 On-going Monitoring of Work and Audits 561
23.6.1 Risk Based Approaches to Monitoring 562
23.6.2 Monitoring the Results 562
23.6.3 Remote Assessment of Work Packages 563
23.6.4 On-site Audits 563
23.6.5 Contract Analytical Work with Your Eyes Open 564
References 564
Chapter 24 - Data Integrity Audit Aide Memoire 565
24.1 What the Regulators Want 565
24.1.1 EU GMP Chapter 9 Self-inspections 565
24.1.2 Data Integrity Guidances for Audits 566
24.1.3 Regulatory Requirements Summary 566
24.2 Audit Aide Memoire for the Foundation Layer: Data Governance 566
24.2.1 Management Leadership for Data Integrity 567
24.2.2 Corporate Data Integrity and Ethics Policy 568
24.2.3 Data Integrity Training 568
24.2.4 Data Ownership for Computerised Systems 570
24.2.5 Data Ownership for Manual Processes 570
24.2.6 Establishment and Maintenance of an Open Culture 570
24.3 Audit Aide Memoire for Level 1: AIQ and CSV 571
24.3.1 Overview 571
24.3.2 Analytical Instrument Qualification 571
24.3.3 Computerised System Validation 571
24.3.4 Validating Interfaces Between Computerised Systems 571
24.4 Audit Aide Memoire for Level 2: Analytical Procedure Validation Life Cycle 571
24.4.1 Procedure Design (Method Development) 574
24.4.2 Analytical Procedure Performance Qualification (Method Validation) 576
24.4.3 Method Application: Control and Monitoring 576
24.5 Level 3: Study and Batch Analysis Data Integrity Aide Memoire 576
24.5.1 Routine Analysis Data Integrity Aide Memoire 577
24.5.2 Audit of Paper Analytical Records 579
24.5.3 Audit of Hybrid Laboratory Computerised Systems 579
24.5.4 Validation and Use of a Spreadsheet 579
24.5.5 Chromatography Data System Aide Memoire 579
24.6 Quality Assurance Oversight Aide Memoire 579
24.6.1 Routine Checks of Study or Batch Records 582
24.6.2 Data Integrity Audits 582
24.6.3 Data Integrity Investigations 584
Acknowledgements 584
References 584
Subject Index 586