Basics and Clinical Applications of Drug Disposition in Special Populations -  - E-Book

Basics and Clinical Applications of Drug Disposition in Special Populations E-Book

0,0
194,99 €

-100%
Sammeln Sie Punkte in unserem Gutscheinprogramm und kaufen Sie E-Books und Hörbücher mit bis zu 100% Rabatt.
Mehr erfahren.
Beschreibung

An up-to-date exploration of techniques for effectively treating patients from special populations

In Basics and Clinical Applications of Drug Disposition in Special Populations, a team of distinguished researchers delivers a timely and authoritative discussion of how to predict drug disposition in special populations, including people with obesity, pediatric patients, geriatric patients, and patients with renal and hepatic impairment. The authors use pharmacokinetic models to account for variabilities between populations and to better predict drug disposition.

The book offers a collection of 15 chapters written by recognized experts in their respective fields. They cover topics ranging from the optimization of drug dosing regimens in specialized populations to model-based approaches in drug treatment among pediatrics.

Readers will also find:

  • A thorough introduction to considerations and regulatory affairs for clinical research in special populations
  • Comprehensive explorations of drug disposition in geriatrics, patients with hepatic insufficiency, and patients with renal insufficiency
  • Practical discussions of model-based pharmacokinetic approaches
  • Complete treatments of artificial intelligence in drug development

Perfect for practicing pharmacologists, pharmacists, and clinical chemists, Basics and Clinical Applications of Drug Disposition in Special Populations will also benefit medical professionals who provide medical and pharmaceutical care to special populations.

Sie lesen das E-Book in den Legimi-Apps auf:

Android
iOS
von Legimi
zertifizierten E-Readern

Seitenzahl: 847

Veröffentlichungsjahr: 2025

Bewertungen
0,0
0
0
0
0
0
Mehr Informationen
Mehr Informationen
Legimi prüft nicht, ob Rezensionen von Nutzern stammen, die den betreffenden Titel tatsächlich gekauft oder gelesen/gehört haben. Wir entfernen aber gefälschte Rezensionen.


Ähnliche


Table of Contents

Cover

Table of Contents

Title Page

Copyright Page

Dedication Page

About the Editors

List of Contributors

Foreword

Preface

1 Pharmacokinetic Principles and Concepts: An Overview

1.1 Introduction

1.2 Pharmacokinetic Parameters

1.3 Pharmacokinetic Models

1.4 Applications

1.5 Conclusion

References

2 Model‐Based Pharmacokinetic Approaches

2.1 Introduction

2.2 Basics of Pharmacokinetics

2.3 Pharmacokinetic (PK) Models

2.4 Model Development and Validation

2.5 Applications of Model‐Based Approaches

2.6 Modeling in Special Populations

2.7 Software and Tools for PK Modeling

2.8 Regulatory Perspectives of PK Modeling

2.9 Future Directions of PK Modeling

2.10 Conclusion

Abbreviations

References

3 Physiologically Based Pharmacokinetic Modeling

3.1 Introduction

3.2 Significance of PBPK Modeling

3.3 Principles for the Development of PBPK for Special Populations

3.4 Data Integration for Special Populations

3.5 Applications of PBPK Modeling

3.6 Regulatory Applications/Pre–Post Market Utilization

3.7 Case Studies

3.8 Lessons Learned

3.9 Conclusion

References

4 Therapeutic Drug Monitoring in Special Populations

4.1 Introduction

4.2 Pediatrics

4.3 TDM Practices in Pediatrics

4.4 Conclusion

4.5 Pregnancy

4.6 The Elderly

4.7 Conclusion

4.8 Hepatic and Renal Impairments

4.9 Conclusion

4.10 Overall Conclusion and Future Direction

Acknowledgment

References

5 Optimization of Drug Dosing Regimen

5.1 Introduction

5.2 Dosing Regimen Optimization Approaches and Strategies

5.3 Dosing Regimen in Special Populations

5.4 Conclusion

References

6 Artificial Intelligence in Drug Development

6.1 Introduction

6.2 Application of AI in Drug Design

6.3 AI Use in Drug Formulation

6.4 Drug Release Characterization Using AI

6.5 AI‐Based Dose/Dosing Regimen

6.6 Dissolution Rate Predictions with AI

6.7 Clinical End‐Point Evaluation with AI

6.8 AI in Prediction of Fate of Drugs Administered Via Mucosal, Transdermal, and Parenteral Routes

6.9 AI‐Integrated Mechanistic Modeling Platform for Drug Delivery and Monitoring

6.10 AI‐Based Tools for Metabolism and Clearance Prediction

6.11 Limitations of Existing Tools

6.12 Conclusions

6.13 Conflict of Interest

Acknowledgments

References

7 Drug Disposition in Neonates and Infants

7.1 Introduction

7.2 Drug Absorption in Neonates and Infants

7.3 Drug Distribution in Neonates and Infants

7.4 Hepatic Metabolism of Drugs in Neonates and Infants

7.5 Drug Excretion in Neonates and Infants

7.6 Pharmacodynamics in Neonates and Infants

7.7 Age‐Related Dosing Regimen in Neonates and Infants

7.8 Conclusion

References

8 Drug Disposition in Adolescents

8.1 Introduction

8.2 Physiological Considerations in Adolescents

8.3 Medication Adherence Challenges in Adolescents

8.4 Psychological Development on Drug Disposition

8.5 Risk‐Taking behaviors and Their Implications on Medication Use

8.6 Drug Use Among Adolescents

8.7 Pharmacokinetic Variability in Adolescents Drug Examples

8.8 Legal and Ethical Considerations

8.9 Conclusion

References

9 Drug Disposition in Pregnancy

9.1 Introduction

9.2 Physiological Changes in Pregnancy

9.3 Placental Drug Disposition

9.4 Drug Classification in Pregnancy

9.5 Pharmacokinetic (PK) Modeling

9.6 Physiologically Based Pharmacokinetic (PBPK) Modeling

9.7 Limitations in PK and PBPK Models

9.8 PBPK Model Variables

9.9 Determining Treatment During Pregnancy

9.10 Fetal Blood Flow and Drug Processing

9.11 Teratogens

9.12 Conclusion

Abbreviations

References

10 Drug Disposition in Obesity

10.1 Introduction

10.2 Index of Obesity

10.3 Pathogenesis of Obesity/Overweight

10.4 Drug Disposition in Obesity

10.5 Drug Dose Calculations in Obese Patients

10.6 Disposition of Drugs in Obesity

10.7 Conclusion

References

11 Drug Disposition in Critical Care Patients

11.1 Introduction

11.2 Pharmacokinetic Considerations in Critical Care Patients

11.3 Dosing Algorithms for Commonly Administered Drugs in Critical Care Patients

11.4 Conclusion

References

12 Drug Disposition in Renal Insufficiency

12.1 Renal Physiology

12.2 Glomerular Filtration Rate

12.3 Acute Kidney Injury

12.4 Chronic Kidney Disease

12.5 Medication Dosing Modifications

12.6 Epidemiology and Outcomes of Patients with CKD

References

13 Drug Disposition in Hepatic Insufficiency

13.1 Introduction

13.2 The Spectrum of Liver Diseases

13.3 Liver Function and Drug Metabolism

13.4 Dosing Algorithms in Clinical Practice

13.5 Drug Disposition and Factors That Influence Drug Disposition

13.6 Major Classes of Drugs and Hepatic Insufficiency

13.7 Cases Demonstrating Application of Dosing Algorithms

13.8 Limitations of Current Dosing Strategies

13.9 Conclusion and Future Perspectives

References

14 Drug Disposition in Geriatrics

14.1 Introduction

14.2 Absorption

14.3 Distribution

14.4 Metabolism

14.5 Excretion

14.6 Hepatic

14.7 Renal

14.8 Cardiac

14.9 Sex Differences

14.10 Psychoactive Drugs

14.11 Anesthesiology Drugs

14.12 Drug Interactions

14.13 Drug Side Effects

14.14 Conclusion

Abbreviations

References

15 Considerations and Regulatory Affairs for Clinical Research in Special Populations

15.1 Introduction

15.2 Regulatory Frameworks for Clinical Research in Special Populations

15.3 Key Considerations for Clinical Trials in Special Population Groups

15.4 Pregnant Population Groups

15.5 Geriatric Populations

15.6 Critical Care

15.7 Summary Points

15.8 Conclusion

References

Index

End User License Agreement

List of Tables

Chapter 2

Table 2.1 Overview of model‐based approaches [2,4–7].

Table 2.2 Differences between traditional and model‐based pharmacokinetic a...

Table 2.3 Comparison of IV bolus, IV infusion, and oral administration (the...

Table 2.4 Model parameters and their unit [40].

Table 2.5 Clinical utility of model‐based approaches in PK studies. Adapted...

Table 2.6 Software used for PK modeling. Adapted from [69].

Chapter 3

Table 3.1 Physicochemical properties of compounds selected for case study (...

Chapter 4

Table 4.1 Application of precision dosing pediatrics

Table 4.2 Examples of drugs for which TDM could be beneficial during pregna...

Chapter 5

Table 5.1 Dosing adjustment for antimicrobials in continuous renal replacem...

Table 5.2 Advantages and disadvantages of antimicrobial stewardship strateg...

Chapter 7

Table 7.1 Factors that affect drug absorption in neonates and infants.

Table 7.2 Factors affecting drug distribution in neonates and infants.

Table 7.3 Ontogeny of phase II drug metabolizing enzymes.

Chapter 9

Table 9.1 Former FDA Pregnancy Categories (1979) [Adapted from the United S...

Table 9.2 Notable Drug Metabolizing CYPs.

Table 9.3 Selected, common pharmacological agents with teratogenic effects.

Chapter 10

Table 10.1 Classification of BMI.

Table 10.2 Impact of obesity on hepatic drug metabolism.

Table 10.3 Impact of obesity on renal drug excretion.

Table 10.4 Dose size descriptors that can be used in dose calculation.

Table 10.5 Examples of drugs and the body size descriptors that are used in...

Chapter 11

Table 11.1 Dosing algorithms of narcotic analgesics in adult ICU patients....

Table 11.2 Dosing algorithms for selected sedatives employed in ICUs.

Table 11.3 Dosing regimen for selected neuromuscular blockers.

Chapter 12

Table 12.1 Equations to assess GFR.

Table 12.2 The Rifle classification.

Table 12.3 AKIN classification.

Table 12.4 CKD stages.

Table 12.5 UACR risk stratification.

Table 12.6 Equations used to calculate GFR.

Table 12.7 ACE inhibitor drug dosage management in patients with CKD.

Table 12.8 Beta‐blocker drug dosage management in patients with CKD.

Table 12.9 Diuretic drug dosage management in patients with CKD.

Table 12.10 Hypoglycemic drug dosage management in patients with CKD.

Table 12.11 Antimicrobial drug dosage management in patients with CKD.

Table 12.12 Statin drug dosage management in patients with CKD.

Chapter 14

Table 14.1 Changes in absorption, distribution, metabolism, and elimination...

Table 14.2 Physiological changes in geriatric patients.

Table 14.3 Change in the pharmacodynamic effect of common drugs in geriatri...

Table 14.4 Formulas to estimate the change in renal function with age.

Table 14.5 Pre‐, intra‐, and postoperative anesthesia drug considerations....

List of Illustrations

Chapter 1

Figure 1.1 Overview of the basic pharmacokinetic processes.

Chapter 2

Figure 2.1 Types of models.

Figure 2.2 Catenary model.

Figure 2.3 Mamillary model.

Figure 2.4 Cyclic model.

Figure 2.5 One‐compartment closed model.

Figure 2.6 One‐compartment open model.

Figure 2.7 Representation of one‐compartment open model showing absorption (...

Chapter 3

Figure 3.1 “Learn, confirm and apply” development cycle used to build and op...

Figure 3.2 Application of PBPK modeling in a pediatric population [5] / with...

Figure 3.3 PBPK modeling strategy employed to predict exposure in neonates a...

Chapter 4

Figure 4.1 The clinical condition and disease state of patients have signifi...

Chapter 5

Figure 5.1 Overview of dose selection during clinical trials.

Figure 5.2 Relationship between drug characteristics, PK parameters and crit...

Figure 5.3 Dynamic interaction between infection, preferred antimicrobial ag...

Figure 5.4 Tools available to optimize antimicrobials in ICU patients.

Figure 5.5 Factors to be considered during antimicrobial dose adjustment dur...

Figure 5.6 Application of MIDD in paediatric dose selection.

Chapter 8

Figure 8.1 An overview of drug profile in adolescents and challenges.

Chapter 9

Figure 9.1 Physiological changes in pregnancy

Figure 9.2 Placenta

Figure 9.3 Teratogenicity by organ system.

Chapter 10

Figure 10.1 Pathophysiological changes in obesity that can affect drug dispo...

Chapter 13

Figure 13.1 A summary of drug disposition in hepatic insufficiency.

Chapter 14

Figure 14.1 Decline in liver volume by age.

Figure 14.2 Decline in the number of functional nephrons by age.

Figure 14.3 Flow chart for prescribing medications in geriatric patients.

Guide

Cover Page

Table of Contents

Title Page

Copyright Page

Dedication

About the Editors

List of Contributors

Foreword

Preface

Begin Reading

Index

WILEY END USER LICENSE AGREEMENT

Pages

iii

iv

v

xxi

xxii

xxiii

xxiv

xxv

xxvi

xxvii

xxviii

xxix

xxx

xxxi

xxxii

1

2

3

4

5

6

7

8

9

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

51

53

54

55

56

57

58

59

60

61

62

63

64

65

66

67

68

69

70

71

72

73

74

75

76

77

78

79

80

81

82

83

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

100

101

102

103

104

105

106

107

108

109

110

111

112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

127

128

129

130

131

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

153

154

155

156

157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

175

176

177

179

180

181

182

183

184

185

186

187

188

189

190

191

192

193

194

195

196

197

198

199

200

201

203

204

205

206

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

223

224

225

226

227

229

230

231

232

233

234

235

236

237

238

239

240

241

242

243

244

245

246

247

248

249

250

251

252

253

254

255

256

257

258

259

260

261

262

263

265

266

267

268

269

270

271

272

273

274

275

276

277

278

279

280

281

282

283

284

285

286

287

288

289

290

291

292

293

294

295

296

297

298

299

300

301

302

303

304

305

306

307

308

309

310

311

312

313

314

315

316

317

318

319

320

321

322

323

324

325

326

327

328

329

330

331

332

333

334

335

336

337

338

339

340

341

342

343

344

345

346

347

348

349

350

351

352

353

354

355

356

357

358

359

360

361

362

363

364

365

366

367

368

369

370

371

372

373

374

375

376

377

378

379

380

381

382

383

384

385

386

387

388

389

390

391

392

393

394

395

396

397

398

399

400

401

402

403

404

405

406

407

408

409

410

411

412

413

414

415

416

417

418

419

420

421

422

423

424

425

426

427

428

429

430

431

432

433

435

436

437

438

439

440

441

442

443

444

445

446

447

448

449

Basics and Clinical Applications of Drug Disposition in Special Populations

Edited by

Seth Kwabena Amponsah

University of Ghana Medical SchoolAccra, Ghana

Yashwant V. Pathak

USF Health Taneja College of PharmacyTampa, Florida, USA

Copyright © 2025 by John Wiley & Sons, Inc. All rights reserved, including rights for text and data mining and training of artificial technologies or similar technologies.

Published by JohnWiley & Sons, Inc., Hoboken, New Jersey.Published simultaneously in Canada.

No part of this publication may be reproduced, stored in a retrieval system, or transmitted in any form or by any means, electronic, mechanical, photocopying, recording, scanning, or otherwise, except as permitted under Section 107 or 108 of the 1976 United States Copyright Act, without either the prior written permission of the Publisher, or authorization through payment of the appropriate per‐copy fee to the Copyright Clearance Center, Inc., 222 Rosewood Drive, Danvers, MA 01923, (978) 750‐8400, fax (978) 750‐4470, or on the web at www.copyright.com. Requests to the Publisher for permission should be addressed to the Permissions Department, John Wiley & Sons, Inc., 111 River Street, Hoboken, NJ 07030, (201) 748‐6011, fax (201) 748‐6008, or online at http://www.wiley.com/go/permission.

The manufacturer’s authorized representative according to the EU General Product Safety Regulation is Wiley‐VCH GmbH, Boschstr. 12, 69469 Weinheim, Germany, e‐mail: [email protected].

Trademarks: Wiley and the Wiley logo are trademarks or registered trademarks of John Wiley & Sons, Inc. and/or its affiliates in the United States and other countries and may not be used without written permission. All other trademarks are the property of their respective owners. John Wiley & Sons, Inc. is not associated with any product or vendor mentioned in this book.

Limit of Liability/Disclaimer of Warranty: While the publisher and author have used their best efforts in preparing this book, they make no representations or warranties with respect to the accuracy or completeness of the contents of this book and specifically disclaim any implied warranties of merchantability or fitness for a particular purpose. No warranty may be created or extended by sales representatives or written sales materials. The advice and strategies contained herein may not be suitable for your situation. You should consult with a professional where appropriate. Further, readers should be aware that websites listed in this work may have changed or disappeared between when this work was written and when it is read. Neither the publisher nor authors shall be liable for any loss of profit or any other commercial damages, including but not limited to special, incidental, consequential, or other damages.

For general information on our other products and services or for technical support, please contact our Customer Care Department within the United States at (800) 762‐2974, outside the United States at (317) 572‐3993 or fax (317) 572‐4002.

Wiley also publishes its books in a variety of electronic formats. Some content that appears in print may not be available in electronic formats. For more information aboutWiley products, visit our web site at www.wiley.com.

Library of Congress Cataloging‐in‐Publication Data Applied for:

Hardback ISBN: 9781394251285

Cover Design: WileyCover Image: Courtesy of Seth Kwabena Amponsah and Yashwant V. Pathak

Dedication

I dedicate this book to my academic mentors, Professor Kwasi Agyei Bugyei and Professor George Obeng Adjei. I also dedicate this book to a very good friend, Dr. Enoch Narh Kudjordjie.

—Seth Kwabena Amponsah

This book is dedicated to the elders and people of ancient traditions and cultures who believe in working for humanity.

—Yashwant V. Pathak

About the Editors

Seth Kwabena Amponsah is an associate professor and former head of Department of Medical Pharmacology, University of Ghana Medical School. He has an MPhil and PhD in pharmacology. He has had postdoctoral fellowships under BANGA‐Africa Project and BSU III (DANIDA—Denmark). He has over 13 years’ experience in teaching and research. He teaches students in the medical school, school of pharmacy, school of nursing and midwifery, and school of biomedical and allied health sciences. His research focus includes clinical pharmacology (infectious disease and antimicrobial stewardship): prudent use of antimicrobials, antimicrobial level monitoring, and efficacy of antimicrobials in patients. He also has experience in population pharmacokinetic modeling, non‐compartment pharmacokinetic estimation, and pharmacokinetic evaluation of new drug formulations. He has supervised several undergraduate and postgraduate students. He has published over 60 research articles, 3 books, 20 book chapters and several conference abstracts. He is an academic editor for PLOS One and an associate editor for Pan African Medical Journal.

Yashwant V. Pathak has over 16 years of versatile administrative experience in an Institution of Higher education as dean (and over 30 years as faculty and as a researcher in higher education after his PhD). Presently holds the position for associate dean for faculty affairs and tenured professor of pharmaceutical sciences. He is an internationally recognized scholar, researcher, and educator in the areas of health care education, nanotechnology, drug delivery systems, and nutraceuticals. He has received many international and national awards including four Fulbright Fellowships, Endeavour Executive Fellowship by Australian Government, four outstanding faculty awards, and he was selected as fellow of American Association for Advancement of Science (AAAS) in 2021. He has published over 350 research publications, reviews, and chapters in various books. He has edited over 60 books in various fields including nanotechnology, nutraceuticals, conflict management, and cultural studies. He is also actively involved many nonprofit organizations, to mention a few, Hindu Swayamsevak Sangh, USA, Sewa International USA, International accreditation council for Dharma Schools and Colleges, International commission for Human rights and religious freedom, and Uberoi Foundation for religious studies, among others.

List of Contributors

Fried A. AbilbaDepartment of Paediatric and ChildHealth PharmacyTamale Teaching HospitalTamaleGhana

James A. AkingbasoteRegulatory ToxicologistLondon, OntarioCanada

Raphael N. AlolgaState Key Laboratory of NaturalMedicines, Department ofPharmacognosyChina Pharmaceutical UniversityNanjingChinaandClinical Metabolomics CenterDepartment of PharmacognosyChina Pharmaceutical UniversityNanjingChina

Emmanuel B. AmoafoDepartment of PharmaceuticalSciencesNorth Dakota State UniversityFargo, NDUSA

Seth K. AmponsahDepartment of Medical PharmacologyUniversity of Ghana Medical SchoolAccraGhana

Aparna AnandanTranslational Research LaboratoryDepartment of BiotechnologyBharathiar UniversityCoimbatore, Tamil NaduIndia

Unais AnnenkottilTranslational Research LaboratoryDepartment of BiotechnologyBharathiar UniversityCoimbatoreTamil Nadu, India

Vishnu P. AthilingamTranslational Research LaboratoryDepartment of BiotechnologyBharathiar UniversityCoimbatore, Tamil NaduIndia

Michael M. AttahDivision of Clinical PharmacologyIndiana University School ofMedicineIndianapolis, INUSAandDepartment of Pharmacy PracticeCollege of PharmacyPurdue UniversityWest Lafayette, INUSA

Jacob A. AyembillaDepartment of Science LaboratoryTechnologyAccra Technical UniversityAccraGhana

Stefanos BelavilasUniversity of South Florida MorsaniCollege of MedicineTampa, FLUSA

Gabriella BlancoUniversity of South Florida MorsaniCollege of MedicineTampa, FLUSA

Alexandra BurtonNationwide Children's HospitalColumbus, OHUSA

Nishanth ChalasaniUniversity of South Florida MorsaniCollege of MedicineTampa, FLUSA

Samuel CockeyUniversity of South Florida MorsaniCollege of MedicineTampa, FLUSA

Justin ColeUniversity of South Florida MorsaniCollege of MedicineTampa, FLUSA

Dominique CookUniversity of South Florida MorsaniCollege of MedicineTampa, FLUSA

Aparoop DasDepartment of PharmaceuticalSciencesDibrugarh UniversityDibrugarh, AssamIndia

Dibyajyoti DasDepartment of PharmaceuticalSciencesDibrugarh UniversityDibrugarh, AssamIndiaandPratiksha Institute of PharmaceuticalScienceGuwahati, AssamIndia

Mansa Fredua‐AgyemanDepartment of Pharmaceutics andMicrobiology, School of PharmacyUniversity of GhanaAccraGhana

Anuradha K. GajjarDepartment of PharmaceuticalChemistryL. M. College of PharmacyAhmedabad, GujaratIndia

Urvashee GogoiDepartment of PharmaceuticalSciencesDibrugarh UniversityDibrugarh, AssamIndia

David GyamfiDepartment of PharmaceuticalSciencesNorth Dakota State UniversityFargo, NDUSA

Arindam HalderSun Pharmaceutical Industries Ltd.Vadodara, GujaratIndia

Rana HannaUniversity of South Florida MorsaniCollege of MedicineTampa, FLUSA

Kartik HariharanSun Pharmaceutical Industries Ltd.Vadodara, GujaratIndia

Elora HilmasNationwide Children's HospitalColumbus, OHUSA

Md Ariful IslamDepartment of PharmaceuticalSciencesDibrugarh UniversityDibrugarh, AssamIndia

Partha P. KalitaFaculty of ScienceAssam down town UniversityGuwahati, AssamIndia

Ali KarimiUniversity of South Florida MorsaniCollege of MedicineTampa, FLUSA

Ajay J. KhopadeSun Pharmaceutical Industries Ltd.Vadodara, GujaratIndia

Seema KohliPharmacy DepartmentK N Polytechnic CollegeJabalpur, MPIndia

Awo A. KwapongDepartment of Pharmaceuticsand Microbiology, School ofPharmacyUniversity of GhanaAccraGhana

Stephanie LeighDepartment of Pharmacy andPharmacology, Faculty of HealthSciencesUniversity of the WitwatersrandJohannesburgSouth Africa

Millena LevinUniversity of South Florida MorsaniCollege of MedicineTampa, FLUSA

Deborah LiawUniversity of South Florida MorsaniCollege of MedicineTampa, FLUSA

Mahesh P. MoreNovel Formulation LaboratorySekkei Bio Pvt Ltd.Bangalore, KAIndia

Chinenye E. MuolokwuDepartment of PharmaceuticalSciences, School of PharmacyCollege of Health and HumanSciencesNorth Dakota State UniversityFargo, NDUSA

Sarah NestlerUniversity of South Florida MorsaniCollege of MedicineTampa, FLUSA

Viswanadha V. PadmaTranslational ResearchLaboratory, Department ofBiotechnologyBharathiar UniversityCoimbatore, Tamil NaduIndia

Amitkumar K. PatelSaffron Health LLCEast Brunswick, NJUSA

Jayvadan K. PatelVie Saine Pharma LLCSheridan, WYUSA

Kashyap M. PatelDepartment of PharmaceuticsL. M. College of PharmacyAhmedabad, GujaratIndia

Manish P. PatelDepartment of PharmaceuticsL. M. College of PharmacyAhmedabad, GujaratIndia

Vivek PatelApex professional UniversityPasighat, Arunachal PradeshIndiaandSun Pharmaceutical Industries Ltd.Vadodara, GujaratIndia

Kalyani PathakDepartment of PharmaceuticalSciencesDibrugarh UniversityDibrugarh, AssamIndia

Manash P. PathakFaculty of Pharmaceutical SciencesAssam Down Town UniversityGuwahati, AssamIndia

Yahwant V. PathakUSF Health Taneja College ofPharmacyUniversity of South FloridaTampa, FLUSA

Goonaseelan C. PillaiDivision of Clinical PharmacologyUniversity of Cape TownRondeboschSouth AfricaandCP+ Associates GmbHBaselSwitzerland

Amruta PotdarUniversity of South Florida MorsaniCollege of MedicineTampa, FLUSA

Pallab PramanikDepartment of PharmaceuticalSciencesDibrugarh UniversityDibrugarh, AssamIndia

Charles PreussDepartment of MolecularPharmacology & PhysiologyUniversity of South Florida MorsaniCollege of MedicineTampa, FLUSA

Jon J. SahariahDepartment of PharmaceuticalSciencesDibrugarh UniversityDibrugarh, AssamIndia

Riya SaikiaDepartment of PharmaceuticalSciencesDibrugarh UniversityDibrugarh, AssamIndia

Surovi SaikiaTranslational Research LaboratoryDepartment of BiotechnologyBharathiar UniversityCoimbatore, Tamil NaduIndia

Dhruv ShahSun Pharmaceutical Industries Ltd.Vadodara, GujaratIndia

Ellen SiUniversity of South Florida MorsaniCollege of MedicineTampa, FLUSA

Ankita A. SinghPharmacy DepartmentK N Polytechnic CollegeJabalpur, MPIndia

Sandra K. SzlapinskiRegulatory ToxicologistLondon, OntarioCanada

Rahul S. TadeH R Patel Institute of PharmaceuticalEducation and ResearchDhule, MSIndia

Benjamin TagoeDepartment of PharmaceuticalSciences, School of PharmacyCollege of Health and HumanSciencesNorth Dakota State UniversityFargo, NDUSA

Teresa TravnicekUniversity of South Florida MorsaniCollege of MedicineTampa, FLUSA

Jacob TreanorUniversity of South Florida MorsaniCollege of MedicineTampa, FLUSA

Maxine TurnerDepartment of Pharmacy andPharmacology, Faculty of HealthSciencesUniversity of the WitwatersrandJohannesburgSouth Africa

Shakil Z. VhoraDepartment of PharmaceuticsL. M. College of PharmacyAhmedabad, GujaratIndia

Kyle WestonRegulatory ToxicologistToronto, OntarioCanada

Yelena WuNationwide Children's HospitalColumbus, OHUSA

Foreword

The fundamentals of pharmacokinetics are essential in understanding the fate of administered drugs in patients. Pharmacokinetic knowledge is also relevant in drug‐dose adjustment in patients undergoing treatment or management in medical practice. Pharmacokinetic considerations are even more important in determining optimal drug dosing in special patient populations, such as pediatrics, critically ill patients, and geriatrics. These populations often exhibit variations in drug disposition of administered drugs compared to healthy adults, from whom pharmacokinetic data of drugs are usually derived. As such, understanding these differences is essential in providing effective and safe pharmacotherapy in these vulnerable populations.

In pediatric patients for instance, age, weight, and maturation of organs influence drug disposition. Additionally, renal function can impact on drug clearance in this cohort of the population.

Geriatrics also experience age‐related changes in disposition of drugs. Decrease in renal and liver functions and a decline in total body water can alter drug distribution, metabolism, and excretion. In geriatrics, these changes can lead to increased drug exposure and susceptibility to adverse drug reactions.

Likewise, hemodynamic, metabolic, and biochemical derangements in critically ill patients can affect drug disposition. Critical illness can alter drug protein binding, volume of distribution, hepatic metabolism, and renal clearance. Indeed, understanding the unique pharmacokinetic profiles of these populations is essential for healthcare providers to make informed decisions regarding drug dosing and monitoring.

Unfortunately, few books are readily available on the market that focus on drug disposition in special populations. This edited book has 15 chapters that discusses relevant topics such as model‐based pharmacokinetic approaches; physiologically based pharmacokinetic modeling; optimization of drug‐dosing regimen; drug disposition in neonates, infants, adolescents, geriatrics, and critically ill patients. Regulatory affairs for clinical research in special populations are also extensively discussed.

Distinguished scientists and researchers have made contributions to this book; with each chapter well written and easy to understand.

I believe that this book will be a great resource for clinicians, biomedical scientists, and students of the medical fraternity. I am, therefore, very happy to write the foreword for this book, Basics and Clinical Applications of Drug Disposition in Special Populations, edited by Seth K. Amponsah and Yaswant V. Pathak.

I congratulate the editors and contributing authors and look forward to seeing this book on the market.

Professor Kwasi Agyei Bugyei

Pharmacokineticist and Former Head, Department of Pharmacology, University of Ghana Medical School

Preface

Over the years, there has been growing interest in the field of pharmacokinetics/pharmacodynamics and its applications. Currently, pharmacokinetic and pharmacodynamic models aid better understanding of drug disposition and effect. Indeed, models can range in complexity from models with a single compartment to models containing multiple compartments. Each type of model from the simplest, one‐compartment model to more complex models has its own applications and can be used to gain valuable information from preclinical and clinical data.

It is well documented that clinical response to drugs vary widely between individuals and that most of this variability is at the pharmacokinetic level. In general, variability arises because of interindividual differences in rates of drug absorption, distribution, and elimination (metabolism and excretion). The pharmacokinetics of a drug can vary from person to person, and it is affected by age, gender, diet, environment, body weight, pathophysiology, genetics, and drug–drug or drug–food interactions. Indeed, the disposition (pharmacokinetics) of a drug can vary in special populations such as paediatrics, geriatrics, obese individuals, and patients with renal and hepatic impairment. To better explain the time course of drugs, pharmacokinetic models that take into account these possible variabilities have been used to predict drug disposition.

After carefully studying literature, we found that there are few books available on the market that compile the various aspects of the pharmacokinetics of drugs in special populations; hence, this book will be a great resource for clinicians, scientists, and researchers. This book covers topics such as optimization of drug‐dosing regimen, model‐based approaches in drug treatment, therapeutic drug monitoring, and pharmacotherapeutic considerations in special populations (neonates, infants, adolescents, geriatrics, among others). The book also contains 15 chapters with rich content, presenting fundamental facts, as well as practical and clinically related data. Renowned scientists/researchers in the field of pharmacokinetics, pharmacodynamics, mathematical, and computational science have contributed to this book. The current book will be a good resource for pharmacometricians, health care professionals (pediatricians, geriatricians, and endocrinologists), and academic institutions.

The editors appreciate the effort of each contributor who shared knowledge through writing chapters in this book. The editors are also grateful to John Wiley & Sons Inc. for facilitating all processes involved in getting this book published.

Seth Kwabena Amponsah and Yashwant V. Pathak

1Pharmacokinetic Principles and Concepts: An Overview

Seth K. Amponsah1 and Yahwant V. Pathak2

1 Department of Medical Pharmacology, University of Ghana Medical School, Accra, Ghana

2 USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA

1.1 Introduction

Pharmacokinetics, a subset of pharmacology, describes the disposition of drugs in the body. The word “pharmacokinetics” is derived from two root words: “kinetics,” which relates to the study of how things change with time, and “pharmaco,” which pertains to pharmaceutical agents or drugs [1]. Pharmacokinetics, which describes the absorption, distribution, metabolism, and excretion of drugs (Figure 1.1), is not just theoretical but a practical tool in ensuring the efficacy and safety of drugs in a patient. Pharmacokinetics is an important arm of the drug development process, helping to achieve the correct dosage, frequency, and delivery method [2].

Indeed, pharmacokinetics provides insights into how the body handles drugs, paving the way for the development of personalized medicine. Pharmacokinetics also assesses the temporal effects of toxic agents in the body [3]. A good understanding of pharmacokinetic principles is essential for healthcare professionals and researchers in the pharmaceutical industry who are at the forefront of developing personalized medicine. Furthermore, comprehension of the basic principles of pharmacokinetics is essential in optimizing drug therapy and ensuring patients receive the best care.

Figure 1.1 Overview of the basic pharmacokinetic processes.

1.2 Pharmacokinetic Parameters

There are four main pharmacokinetic parameters: absorption, distribution, metabolism, and excretion. Although distinct, these parameters are interrelated and usually occur after a drug is administered to the body [4]. Importantly, some drugs access circulation without going through the process of absorption: drugs given intravenously [5].

1.2.1 Absorption

The rate and extent to which a drug is absorbed are dependent on the route of administration (oral, subcutaneous, intrathecal, intravenous, etc.), the formulation, the physicochemical properties, and the physiological factors that affect the absorption site [6]. After an oral (p.o.) administration of a drug, it is absorbed across the intestinal lumen into the portal vein and then to the liver. Subsequently, it will often undergo first‐pass metabolism before entering systemic circulation, reducing drug bioavailability [4]. Bioavailability, therefore, can be defined as the proportion of an administered drug that reaches systemic circulation in its active form. It comprises both the rate and the extent of drug absorption, and they are both influenced by several factors, such as the physicochemical properties of the drug, formulation, and the physiological conditions of the gastrointestinal tract (GIT). Following oral administration, drugs must undergo dissolution in GIT fluids before traversing the walls of the gut for absorption. Dissolution is heavily dependent on the solubility of the drug, as well as gastric pH and GIT motility.

When a drug is administered intravenously, there is no absorption because the drug goes directly from the administration site into circulation—almost near 100% bioavailability. Other routes may reduce the bioavailability of drugs because of incomplete absorption [6]. Generally, lipid‐soluble drugs at physiological pH are effectively absorbed via passive diffusion. Additionally, drug absorption can occur through different mechanisms, such as active and facilitated diffusion (carrier‐mediated membrane transport) and other nonspecific transporter systems, such as the P‐glycoprotein transporter [7]. In addition, the presence of food in the stomach can either enhance or hinder the absorption of certain drugs, which is also dependent on the characteristics of the drugs. For example, high‐fat meals may enhance the absorption of lipophilic drugs [8].

Various strategies are employed to enhance drug bioavailability, some of which include formulation modifications (such as nanoparticles and liposomes) and techniques such as micronization. These methods enhance dissolution and absorption rates [5]. Indeed, an in‐depth understanding of drug absorption principles is essential in drug development and in ensuring maximal therapeutic outcomes. As such, researchers and health professionals need to consider the aforementioned factors that affect drug absorption [2].

1.2.2 Distribution

After a drug is absorbed into the systemic circulation, it undergoes equilibration between the vascular compartment and several body compartments, including interstitial and intracellular spaces. It is worth noting that the molecular structure of a drug determines its degree of distribution in different tissues: adipocytes, muscle, and brain. Notably, the brain and testes possess barriers that confer unique characteristics, rendering drugs less susceptible to distribution within these organs [9]. Following entry into the bloodstream, drugs can bind to plasma proteins, such as albumin, or remain unbound. The fraction of the bound drug acts as a reservoir, facilitating the gradual release of the drug. At the same time, the unbound portion remains pharmacologically active and is available for distribution to several tissues [10]. The degree of protein binding significantly influences drug distribution, duration of action, and elimination [11].

Many factors influence how drugs are distributed in the body. Some of these factors include the degree of blood flow to tissues, the permeability of tissues, and the physicochemical properties of the drug. For example, organs with high blood flow, such as the brain, kidney, and liver, receive drugs much quicker than tissues with low blood flow, such as fat and muscle tissues [12]. The ability of a drug to cross cell membranes is often determined by its solubility profile and molecular size. Lipophilic drugs can cross membranes more easily and accumulate in fatty tissues, while hydrophilic drugs are more likely to remain in the extracellular fluid [13].

Often, the extent to which drugs are distributed can be represented as the volume of distribution (Vd). Vd can be defined as the theoretical volume in which the total amount of drug must be homogenously distributed to produce the observable blood concentration. A drug with a large Vd signifies that it is extensively distributed into tissues, while a small Vd suggests limited distribution, chiefly within the vascular compartment [14]. For instance, digoxin shows a large Vd due to its extensive tissue binding, especially in skeletal and cardiac muscles [15].

Understanding drug distribution is essential, as this will help optimize therapeutic effects and minimize adverse events. Variability in drug distribution can be a result of age, body composition, and pathological conditions.

1.2.3 Metabolism

After drugs are absorbed into the systemic circulation, a number of them have to undergo metabolism in the liver. Drugs that undergo metabolism are usually lipophilic. The process of metabolism converts lipophilic drugs into more water‐soluble metabolites to aid their excretion. In the liver, drugs undergo chemical modifications through enzymatic reactions [16]. Metabolism does not only facilitate the excretion of drugs but also plays a key role in regulating the pharmacological activity of drugs. Metabolism can potentially convert inactive prodrugs into active compounds [17]. Also, parent drugs that are active can be metabolized into less active or inactive forms.

Drug metabolism occurs through different chemical reactions, which are classified as phase I (functionalization) and phase II (conjugation) [18]. In phase I reactions, lipophilic drugs are biotransformed through processes such as oxidation, reduction, and hydrolysis. These reactions are capable of converting inactive prodrugs into active forms. With oxidation reactions, metabolites often retain some of their pharmacological activity. For instance, diazepam undergoes phase I reaction to become desmethyldiazepam, which is then further metabolized into oxazepam. The two metabolites exhibit pharmacological effects similar to those of diazepam. The cytochrome P450 enzyme (CYP) system, also known as microsomal mixed function oxidase, is the notable catalyst for most phase I reactions [19, 20].

During phase II metabolism, drugs are conjugated with polar endogenous substrates like glucuronic acid, sulfate, and glutathione. Conjugation reactions usually make the drug pharmacologically inactive and water soluble. Phase II reactions occur mainly in the liver but can also occur in the kidneys, lungs, and intestines [21]. Due to genetic differences (single nucleotide polymorphism), the activity of CYP enzymes can vary significantly among individuals. These variations could influence how drugs are metabolized, potentially impacting drug effectiveness and safety. For instance, polymorphisms in CYP2D6 can lead to different phenotypes of enzymes, and this can affect the metabolism of a drug like codeine, which is converted into its active form, morphine [22].

In addition to genetic factors, metabolism can be affected by age, liver disease, diet, and concomitant drug use. For example, neonates and geriatrics frequently exhibit reduced enzyme activity, and this has to be adequately catered for with dosage adjustments [23].

Furthermore, liver diseases such as cirrhosis have been shown to reduce drug metabolism, possibly leading to the accumulation of the drug and ultimately an increased risk of adverse effects. Also, specific diets may induce or inhibit CYP enzymes, thereby influencing the metabolism of some drugs. For example, grapefruit juice inhibits CYP3A4 and can significantly raise the plasma concentration of drugs metabolized by this enzyme [24].

1.2.4 Excretion

Excretion is the process of physically removing a drug from the body, either as the parent compound or a metabolite. Two of the most important channels for drug excretion are urine and bile. While polar compounds are effectively excreted without metabolism in the liver, lipophilic compounds usually require metabolism [25]. Drugs move to the kidneys through circulation and are filtered from the blood in the glomerulus. Also, drugs can undergo active tubular secretion in the proximal tubules, usually involving transporters that pump drugs into the renal tubules. Parent drugs and metabolites may be reabsorbed into circulation from the kidneys. Tubular reabsorption can be affected by the ionization state of the drug and the pH of urine [26]. For instance, weak acidic drugs are more likely to be reabsorbed in acidic urine, while weak basic drugs are reabsorbed in alkaline urine [27]. Indeed, if a drug is lipophilic, it is most likely to be reabsorbed.

Aside from renal excretion, drugs can be secreted into bile (biliary excretion); usually large molecular weight drugs and conjugate drug metabolites. In biliary excretion, drugs are eliminated in feces or undergo enterohepatic circulation. Enterohepatic circulation can prolong the duration of action and/or half‐life of drugs [28].

1.3 Pharmacokinetic Models

Pharmacokinetic models involve fitting mathematical equations to experimental observations of drug concentration in plasma. They are useful in predicting drug concentration‐time profiles. Common examples include compartmental, non‐compartmental, and physiologically based pharmacokinetic (PBPK) models. Compartmental models limit the human body to one or more compartments, each proposed to distribute drugs equally [29]. The one‐compartment model portrays the body as a single homogenous compartment and assumes that the drug quickly reaches equilibrium. This model is best suited for drugs that enter the body rapidly and uniformly, such as intravenous drugs. Conversely, the two‐compartment model divides the body into core compartments associated with the blood and well‐perfused organs and a peripheral compartment associated with less perfused tissues. For drugs that exhibit a dispersion phase followed by an elimination phase, this model is best suited [30].

Pharmacokinetic data can be assessed through non‐compartmental analysis (NCA), which deviates from the compartmental organization presuppositions. The NCA, which was founded on statistical moment theory, can calculate the Vd, clearance (CL), and area under the curve (AUC) directly from the concentration–time data. This method is appropriate when choosing a model‐independent analysis or when the complexity of compartmental models is deemed unnecessary [31].

Using physiologically based pharmacokinetic (PBPK) models becomes essential when a more comprehensive and mechanical approach, integrating physiological, biochemical, and anatomical data about the human body, is required. Based on actual organ and tissue volume, rate of blood flow, and specific characteristics of drugs, this model partitions the body into distinct compartments [32].

1.4 Applications

Applying pharmacokinetic principles in clinical settings is relevant for optimizing drug therapy. Pharmacokinetic principles find great use in individualized dosing, which involves adjusting drug doses based on specific patient factors such as age, weight, renal and hepatic function, and genetic makeup. Furthermore, therapeutic drug monitoring (TDM) is often used for drugs that have narrow therapeutic indices. This is to ensure accurate dosing and also to prevent subtherapeutic or toxic effects. For instance, the dosing of phenytoin, an antiepileptic drug, is tailored through TDM to maintain plasma drug concentrations within the therapeutic range [33]. Another notable application of pharmacokinetic principles in the clinical setting is the management of drug–drug interactions. A robust understanding of the pharmacokinetics of drugs enables healthcare workers to anticipate and effectively address potential interactions. For example, CYP enzyme inhibitors, such as ketoconazole, can increase the plasma concentrations of drugs metabolized by these enzymes, eventually increasing the risk of toxicity. On the other hand, enzyme inducers, such as rifampin, can decrease drug concentrations and subsequently decrease drug efficacy [34].

1.5 Conclusion

Pharmacokinetics processes serve as a keystone in clinical pharmacology and advance the safe and effective use of drugs. The application of these critical principles helps healthcare workers customize drug dosing according to patient‐specific requirements, manage drug interactions, and optimize therapeutic outcomes. Aside from enhancing patient care, pharmacokinetic models play a crucial role in the drug development continuum, from preclinical studies to market authorization. Pharmacokinetic research holds great potential for further enhancing drug therapy and fostering its precision.

References

1

Byers, J.P. and Sarver, J.G. (2009). Pharmacokinetic modeling. In:

Pharmacology

(ed. M. Hacker, W. Messer, and K. Bachmann), 201–277. Academic Press.

2

Shargel, L., Wu‐Pong, S., and Yu, A.B.C. (2012).

Applied Biopharmaceutics & Pharmacokinetics

, 6e. McGraw‐Hill's AccessPharmacy. New York, N.Y: McGraw‐Hill Education LLC.

3

Benet, L.Z. and Zia‐Amirhosseini, P. (1995). Basic principles of pharmacokinetics.

Toxicologic Pathology

23 (2): 115–123.

4

Fan, J. and de Lannoy, I.A.M. (2014).

Pharmacokinetics

.

Biochemical Pharmacology

87 (1): 93–120.

5

Currie, G.M. (2018). Pharmacology part 2: introduction to pharmacokinetics.

Journal of Nuclear Medicine Technology

46 (3): 221–230.

6

Blaschke, T. (2012). Pharmacokinetics: absorption, distribution, metabolism, excretion overview. In:

Principles of Pharmacogenetics and Pharmacogenomics

(ed. R.B. Altman, D. Flockhart, and D.B. Goldstein), 21–26. Cambridge University Press.

7

Mayersohn, M. (1987). Drug absorption.

J Clin Pharmacol.

27 (9): 634–638.

8

Cheng, L. and Wong, H. (2020). Food effects on oral drug absorption: application of physiologically‐based pharmacokinetic modeling as a predictive tool.

Pharmaceutics

12 (7): 672.

9

Dong, X. (2018). Current strategies for brain drug delivery.

Theranostics

8 (6): 1481–1493.

10

Benet, L.Z. and Hoener, B.A. (2002).

Changes in plasma protein binding have little clinical relevance

.

Clinical Pharmacology and Therapeutics

71 (3): 115–121.

11

Kaur, G., Grewal, J., Jyoti, K. et al. (2018). Oral controlled and sustained drug delivery systems: Concepts, advances, preclinical, and clinical status. In:

Drug Targeting and Stimuli Sensitive Drug Delivery Systems

(ed. A.M. Grumezescu), 567–626. Elsevier.

12

Toutain, P.L. and Bousquet‐MÉLou, A. (2004). Plasma clearance.

Journal of Veterinary pharmacology and therapeutics

27 (6): 415–425.

13

Mittal, B. (2017). Pharmacokinetics and preformulation. In:

How to Develop Robust Solid Oral Dosage Forms From Conception to Post‐Approval

, 17–37. Elsevier.

14

Amponsah, S.K., Adjei, G.O., Enweronu‐Laryea, C. et al. (2017). Population pharmacokinetic characteristics of amikacin in suspected cases of neonatal sepsis in a low‐resource African setting: a prospective nonrandomized single‐site study.

Current Therapeutic Research, Clinical and Experimental

84: e1–e6.

15

Bhindi, A.K. (2010). Optimisation of digoxin maintenance dose in toxicity.

Journal of Pharmacy Practice and Research

40 (1): 50–53.

16

Open Resources for Nursing (Open RN) (2023).

Nursing Pharmacology [Internet]

(ed. K. Ernstmeyer and E. Christman), 19–47. Eau Claire (WI): Chippewa Valley Technical College.

17

Testa, B. and Krämer, S.D. (2006). The biochemistry of drug metabolism‐‐an introduction: part 1. Principles and overview.

Chemistry & Biodiversity

3 (10): 1053–1101.

18

Susa, S., Hussain, A., and Preuss, C. (2023).

Drug Metabolism

. Stat Pearls Publishing.

19

Azzam, A.A.H., McDonald, J., and Lambert, D.G. (2019). Hot topics in opioid pharmacology: mixed and biased opioids.

British Journal of Anaesthesia: BJA

122 (6): e136–e145.

20

Wilkinson, G.R. (2005). Drug metabolism and variability among patients in drug response.

The New England Journal of Medicine

352 (21): 2211–2221.

21

Fernandez, E., Perez, R., Hernandez, A. et al. (2011). Factors and mechanisms for pharmacokinetic differences between pediatric population and adults.

Pharmaceutics

3 (1): 53–72.

22

Ingelman‐Sundberg, M. (2005). Genetic polymorphisms of cytochrome P450 2D6 (CYP2D6): clinical consequences, evolutionary aspects and functional diversity.

The Pharmacogenomics Journal

5 (1): 6–13.

23

Hines, R.N. (2008). The ontogeny of drug metabolism enzymes and implications for adverse drug events.

Pharmacology & Therapeutics (Oxford)

118 (2): 250–267.

24

Bailey, D.G., Malcolm, J., Arnold, O., and Spence, J.D. (1998). Grapefruit juice–drug interactions.

British Journal of Clinical Pharmacology

46 (2): 101–110.

25

Talevi, A. and Bellera, C.L. (2024). Drug excretion. In:

ADME Processes in Pharmaceutical Sciences

(ed. A. Talevi and P.A. Quiroga), 81–96. Springer.

26

Ritter, J.M., Flower, R.J., Henderson, G., and Rang, H.P. (2011).

Dale's Pharmacology

, 7e. Elsevier/Churchill Livingstone.

27

Barreto, E., Larson, T., and Koubek, E. (2021).

Drug excretion

. In:

Reference Module in Biomedical Sciences

. Elsevier.

28

Roberts, M.S., Magnusson, B.M., Burczynski, F.J., and Weiss, M. (2002). Enterohepatic circulation: physiological, pharmacokinetic and clinical implications.

Clinical Pharmacokinetics

41 (10): 751–790.

29

Huang, X.H., Zheng, Q.S., Gabrielsson, J., and Weiner, D. (2010). Pharmacokinetic and pharmacodynamic data analysis: concepts and applications, fourth edition.

American Journal of Pharmaceutical Education

74 (3): 53b.

30

Sung, J., Srinivasan, B., Esch, M.B. et al. (2014). Using physiologically‐based pharmacokinetic‐guided “body‐on‐a‐chip” systems to predict mammalian response to drug and chemical exposure.

Exp Biol Med (Maywood)

239 (9): 1225–1239.

31

Bonate, P. (2018). Editorial to themed issue: recent advances in cardiovascular pharmacokinetic–pharmacodynamic modeling and simulation.

Journal of Pharmacokinetics and Pharmacodynamics

45 (3): 353–353.

32

Jones, H.M. et al. (2015). Physiologically based pharmacokinetic modeling in drug discovery and development: a pharmaceutical industry perspective.

Clinical Pharmacology and Therapeutics

97 (3): 247–262.

33

Touw, D.J., Neef, C., Thomson, A.H. et al. (2005). Cost‐effectiveness of therapeutic drug monitoring: a systematic review.

Therapeutic Drug Monitoring

27 (1): 10–17.

34

Hollenberg, P.F., Kent, U.M., and Bumpus, N.N. (2008). Mechanism‐based inactivation of human cytochromes P450s: experimental characterization, reactive intermediates, and clinical implications.

Chemical Research in Toxicology

21 (1): 189–205.