+353-1-416-8900REST OF WORLD
+44-20-3973-8888REST OF WORLD
1-917-300-0470EAST COAST U.S
1-800-526-8630U.S. (TOLL FREE)

Patient Centric Blood Sampling and Quantitative Analysis. Edition No. 1. Wiley Series on Pharmaceutical Science and Biotechnology: Practices, Applications and Methods

  • Book

  • 400 Pages
  • October 2023
  • John Wiley and Sons Ltd
  • ID: 5863964
PATIENT CENTRIC BLOOD SAMPLING AND QUANTITATIVE ANALYSIS

Authoritative resource providing a complete overview of patient centric blood sampling, as well as its benefits and challenges

Patient Centric Blood Sampling and Quantitative Analysis focuses on the growing interest in alternative means to standard phlebotomy and analytical workflows for the collection and analysis of high-quality human biological samples for the quantitative determination of circulating drugs, their metabolites, and endogenous substances for clinical trials, routine healthcare and neonatal screening. The book clearly explains the benefits and constraints of having patients collect small volumes of blood in locations outside of a clinic (e.g at home), including: patient convenience; less invasive procedures; increased frequency of sampling; applicability to collecting samples from the young, elderly, and those in remote locations; greater frequency; and lower cost per sample. Readers will learn about approaches for successfully implementing patient centric sampling workflows in a number of scenarios, including the clinical setting and in the analytical laboratory.

Edited by four recognized experts in this field, with additional specialists in the discipline enlisted to write the component chapters, enabling greater depth and detail to be added and further raising the scientific standing of the publication, Patient Centric Blood Sampling and Quantitative Analysis includes information on: - Basics of patient centric blood sampling and techniques and approaches that are available and in development for the collection and analysis of the samples - Science behind patient centric blood sampling and its implications regarding human healthcare and wellbeing - Application areas of patient centric sampling, including drug development, clinical chemistry/pathology, therapeutic drug monitoring, and more - Practical approaches to successful implementation for existing and developing purposes and workflows, and case studies to support implementation within an organization

Giving the reader a broad understanding of what patient centric sampling is and where it might be applied for existing and potential future areas, Patient Centric Blood Sampling and Quantitative Analysis is an essential resource on the subject for many different types of laboratories, areas of clinical research and healthcare, including those in pharmaceutical, clinical, and research functions.

Table of Contents

List of Contributors xiii

Foreword xvii

Preface xix

1 Patient Centric Healthcare - What’s Stopping Us? 1
Jenny Royle and Rachel Jones

1.1 The Evolution of Future Health Systems 1

1.2 Exploring the Barriers to Home Sampling 3

1.2.1 Barrier One - The Discord Between Innovation and Practice 4

1.2.2 Barrier Two - Ethical and Operational Considerations 5

1.2.3 Barrier Three - Where Does the Liability Sit? 7

1.2.4 Barrier Four - Addressing the Technology Challenge 8

1.2.5 Barrier Five - The Human Touch 9

1.2.6 Barrier Six - Trust in Data Security 10

1.2.7 Barrier 7 - Adherence to Service Change 11

1.3 Conclusion: The Changing Role of Home Sampling 12

References 13

2 Tips for Successful Quantitative Assay Development Using Mitra Blood Sampling with Volumetric Absorptive Microsampling 17
James Rudge

2.1 What is Volumetric Absorptive Microsampling? 17

2.2 Tip 1 - Ensure the Use of a Correct Sampling Procedure to Prevent Volume-Related Biases 18

2.3 Tip 2 - Working with Wet Whole Blood 19

2.3.1 Is Your Choice of Assay Biologically Relevant in Blood? 19

2.3.2 Working with Blood as a Matrix 20

2.3.3 Allowing Analytes to Equilibrate Ex Vivo 21

2.3.4 Bridging Between Venous Capillary Blood and the Role of Anticoagulants 22

2.4 Tip 3 - Working with Dried Whole Blood 24

2.4.1 Dried Blood Spot Cards 24

2.4.2 Volumetric Hematocrit Bias - Blood Viscosity 25

2.4.3 Dried Blood is a Complicated Matrix 26

2.4.4 Working with “Aged” Blood 26

2.4.5 Temporal Extraction Bias or Degradation? 27

2.5 Tip 4 - Optimizing Extraction Efficiencies from VAMS 29

2.5.1 Measuring Percentage Recovery from Mitra Samplers 30

2.5.2 Extraction Conditions - Where to Start 31

2.5.2.1 Consulting the Literature and Matching Physicochemical Properties 31

2.5.2.2 Adapting Published DBS Methods 32

2.5.2.3 Converting from a Wet (Whole Blood or Plasma) Method 32

2.5.2.4 Starting from a Blank Canvas - What to Consider? 33

2.5.2.5 Choice of Matrix 35

2.5.3 Aqueous Extraction Conditions 36

2.5.4 Organic Extraction Conditions 37

2.5.5 Generic Extraction Conditions 39

2.6 Conclusions 40

References 41

3 Preanalytical Considerations for Implementation of Microsampling Solutions 49
Bradley B. Collier, Peyton K. Miesse, and Russell P. Grant

3.1 Introduction 49

3.2 Sample Matrices 50

3.2.1 Venous Sample 51

3.2.2 Capillary Blood 54

3.2.3 Material Selection 62

3.2.4 Dried Samples 62

3.2.5 Conclusions 63

3.3 Alternate Sample Acceptance Criteria 63

3.4 Collection 65

3.4.1 Device and Kit Components 66

3.4.2 Training and Preparation 67

3.4.3 Wound Generation 68

3.4.4 Collecting Sample 70

3.4.5 Post Collection Processing 72

3.4.6 Conclusions 72

3.5 Transportation and Sample Stability 73

3.5.1 Specimen Matrix and Separation 73

3.5.2 Storage Condition 74

3.5.3 Measurement Technique 77

3.5.4 Hematocrit Effects 79

3.5.5 Conclusions 79

3.6 Preanalytical Processing 80

3.6.1 Separation of Plasma and Serum 80

3.6.2 Sample Dilution 81

3.6.3 Conclusions 82

3.7 Overall Conclusions 82

References 83

4 Collection and Bioanalysis of Quantitative Microsamples: Technological Innovations and Practical Implications 93
Regina V. Oliveira, Marc Yves Chalom, and Carlos Roberto V. Kiffer

4.1 Introduction 93

4.2 Practical Implications in Clinical Settings 94

4.2.1 Clinical Development 95

4.2.2 Clinical Analyses 96

4.3 Microsampling Devices - A Patient-Centered Approach 99

4.3.1 Collection Devices for Microsampling Analysis 99

4.3.1.1 Blood Sampling Techniques 102

4.3.1.2 Other Biological Matrices 124

4.4 New Development Areas 124

4.4.1 Automated Sample Collectors 124

4.4.2 Microfluidic Point-of-Care Devices 127

4.5 Summary of Currently Available Patient Centric Sampling Technologies 127

4.6 Microsampling Analysis by LC-MS - Analytical Considerations 127

4.6.1 Basic Principles of Liquid Chromatography (LC) and Mass Spectrometry (MS) for Bioanalysis of Microsamples 128

4.6.1.1 Microspray Ionization Sources 129

4.6.1.2 Microflow Liquid Chromatography 131

4.6.1.3 Microchip-Based LC 138

4.7 Conclusions 138

References 140

5 Automation in Microsampling: At Your Fingertips? 153
Sigrid Deprez, Liesl Heughebaert, Nick Verougstraete, Veronique Stove, Alain G. Verstraete, and Christophe P. Stove

5.1 Introduction 153

5.1.1 Identifying the Current Bottlenecks for Routine Implementation of Microsampling in Clinical Practice 153

5.1.2 The Importance of Analytical Automation for Different Application Fields 157

5.2 Automation of Dried Blood Microsampling Analysis Coupled to (LC-) MS/MS: What’s Available? 159

5.2.1 Amenability of DBS Samples for Automation 159

5.2.2 Commercially Available Automated DBS Extraction Instruments 162

5.2.2.1 Automated Extraction of DBS: Workflow 162

5.2.2.2 Extraction Process 163

5.2.2.3 Extract Processing Strategy 164

5.2.2.4 Internal Standard Application 165

5.2.3 Points of Attention During Method Validation 166

5.2.3.1 Matrix Effects and Recovery 166

5.2.3.2 The Hct Effect 168

5.2.3.3 Calibration Curve and Dilution Integrity 169

5.2.4 Cross-Validation of Automated DBS Procedures 170

5.2.5 Current Applications of Automated Online DBS Extraction 173

5.2.6 Approaches for Automating Analysis with Other Microsampling Devices 181

5.2.7 Alternative Approaches for the Analysis of DBS Samples 183

5.2.7.1 Direct Analysis of DBS 183

5.2.7.2 Coupling DBS Analysis to Automated Immuno-Analyzers 184

5.3 Integration Into a Clinical Laboratory 185

5.3.1 Requirements, Challenges, and Advantages of Implementation 185

5.3.2 Cost-Effectiveness of Implementation of (Automated) DBS Analysis 187

5.4 Conclusions and Future Perspectives 192

Acknowledgments 192

References 193

6 Over 50 Years of Population-Based Dried Blood Spot Sampling of Newborns; Assuring Quality Testing and Lessons Learned 205
Amy M. Gaviglio, Kristina Mercer, Konstantinos Petritis, Carla D. Cuthbert, and Suzanne K. Cordovado

6.1 Overview of Population-Based Newborn Screening 205

6.2 Public Perceptions of NBS 208

6.3 Characteristics of the DBS Matrix and Its Utility in NBS 209

6.3.1 Recovery of Biochemical and Molecular Analytes from DBS for NBS 210

6.3.2 Evaluation of Lot-to-Lot Variability in NBS Collection Devices 211

6.3.3 Effect of Hematocrit on DBS Homogeneity, Data Analysis, and Results 212

6.3.4 DBS Specimen Collection Transport and Safe Handling 213

6.3.5 DBS Analyte Stability and Storage 216

6.3.6 Known Interferences with DBS use for NBS 221

6.3.7 History of NSQAP - 40 Years of Quality Assurance 222

6.4 Methods Used in NBS 232

6.4.1 Origins of NBS and Expansion of Biochemical Testing 232

6.4.2 Origins of Molecular DBS Testing and Expansion in NBS 238

6.4.3 How the Expansion of Genomics may Impact NBS 241

6.4.4 Expansion of DBS Utility, Including Direct Patient Use 244

6.5 Conclusion 245

Acknowledgments 246

Conflicts of Interest 246

References 246

7 Considerations for Implementation of Microsampling in Pediatric Clinical Research and Patient Care 263
Ganesh S. Moorthy, Christina Vedar, and Athena F. Zuppa

7.1 Introduction 263

7.2 Considerations for Implementation 264

7.2.1 Benefits 264

7.2.1.1 Clinical Research 266

7.2.1.2 Clinical Care 267

7.2.2 Challenges 268

7.2.2.1 Clinical Research 269

7.2.2.2 Clinical Care 270

7.2.3 Laboratory Challenges and Considerations 271

7.2.4 Survey Results on Feasibility 273

7.3 Conclusion 274

References 274

8 Simplification of Home Urine Sampling for Measurement of 2,8-Dihydroxyadenine in Patients with Adenine Phosphoribosyltransferase Deficiency 277
Unnur A. Thorsteinsdottir, Hrafnhildur L. Runolfsdottir, Vidar O. Edvardsson, Runolfur Palsson, and Margret Thorsteinsdottir

8.1 Introduction 277

8.1.1 Adenine Phosphoribosyltransferase Deficiency 278

8.1.2 Diagnosis of Adenine Phosphoribosyltransferase Deficiency 280

8.2 Methods 281

8.2.1 Sample Collection 281

8.2.2 Preparation of Urine Samples for Analysis 281

8.2.3 The UPLC-MS/MS Urinary 2,8-Dihydroxyadenine Assay 282

8.3 Results 283

8.3.1 Assay Development and Optimization 283

8.3.2 Comparison of First-Morning Void Urine Specimens and 24-hr Urine Collections for Assessment of 2,8-Dihydroxyadenine Excretion 288

8.4 Discussion 290

8.5 Conclusions and Future Directions 291

References 292

9 Utilization of Patient Centric Sampling in Clinical Blood Sample Collection and Protein Biomarker Analysis 297
Jinming Xing, Joseph Loureiro, Dmitri Mikhailov, and Arkady I. Gusev

9.1 Introduction 297

9.1.1 Challenges with the Current Clinical Trial Model 297

9.1.2 Clinical Trial Conduct Faced Unprecedented Challenges Brought by Covid-19 298

9.2 Current Patient Centric Sampling Landscape 299

9.3 Clinical Proteome Profiling Technologies for Testing Patient Centric Microsampling Devices 300

9.3.1 Biomarker and Profiling Can Be Used to Benchmark Patient Centric Sampling Technologies 300

9.3.2 Orthogonal Analysis Cultivates Confidence for Biomarker Test with Patient Centric Sampling 302

9.4 Clinical Sample Collection with Tap Device: A Clinical Case Study 303

9.4.1 Clinical Study with TAP Device 303

9.4.2 User Experience, TAP Device Performance, and Sample Hemolysis 306

9.4.3 SomaScan Blood Proteome Profiling Landscape 308

9.4.4 Orthogonal Confirmation Provided by Quantitative Immunoassay 313

9.4.5 Extending Protein Biomarkers Tested by Quantitative Immunoassay Beyond the Zone of Highest Concordance (Negative Controls) 314

9.5 Discussion 318

9.5.1 Utility of Patient Centric Sampling for Clinical Proteome Sample Collection 318

9.5.2 Considerations for Patient Centric Sampling Implementation in Clinical Trials 320

9.5.3 Future Outlook for Patient Centric Sampling 322

Acknowledgements 323

References 323

10 Enabling Patient Centric Sampling Through Partnership: A Case Study 327
Christopher Bailey, Cecilia Arfvidsson, Stephanie Cape, Paul Severin, Silvia Alonso Rodriguez, Robert Nelson, and Catherine E. Albrecht

10.1 Introduction 327

10.1.1 The Partnership 327

10.1.2 AstraZeneca’s Evolving PCS Approach 328

10.1.3 Why Change? 328

10.1.4 Patient Choice 329

10.1.5 The Challenges - Why Isn’t PCS Already the Norm? 329

10.2 Pre-Study Considerations 331

10.2.1 Early Engagement 331

10.2.2 Feasibility Assessment 332

10.3 The Case Study 332

10.3.1 Background 332

10.3.2 Scientific Considerations 335

10.3.3 Regulatory Agency Expectations Bridging 338

10.3.4 Study Operations Considerations 338

10.3.5 Route of Drug Administration and Potential for Sample Contamination 340

10.3.6 Sample Handling 341

10.3.7 Training and Patient Recruitment Challenges 342

10.3.8 Other Logistical, Data Protection, and Compliance Considerations 342

10.3.9 Study Participant Engagement 343

10.4 Summary 344

References 347

11 Perspectives on Adopting Patient Centric Sampling for Pediatric Trials 351
Enaksha Wickremsinhe

11.1 Overview and Why 351

11.1.1 Regulations and Legislation 351

11.1.2 Who are Pediatric Patients? 352

11.2 Challenges and Current Status 353

11.2.1 Conducting Pediatric Studies 353

11.2.2 Ethics, Consent, and Assent 353

11.2.3 Patient/Parent Burden 354

11.2.4 Blood Sampling 354

11.2.5 Blood Volume Limits 355

11.3 Solutions: How Do We Get This Done 356

11.3.1 Microsampling 356

11.3.2 Patient Centric Sampling 357

11.3.3 Pediatric PCS Devices/Techniques 358

11.3.4 COVID-19 Era 359

11.3.5 Training 359

11.4 Summary 359

References 360

Index 363

Authors

Neil Spooner Emily Ehrenfeld Joe Siple Mike S. Lee Milestone Development Services.