You are on page 1of 384

Seth Kwabena Amponsah

Yashwant V. Pathak Editors

Recent Advances in
Therapeutic Drug
Monitoring and
Clinical Toxicology
Recent Advances in Therapeutic Drug
Monitoring and Clinical Toxicology
Seth Kwabena Amponsah
Yashwant V. Pathak
Editors

Recent Advances in
Therapeutic Drug
Monitoring and Clinical
Toxicology
Editors
Seth Kwabena Amponsah Yashwant V. Pathak
Department of Medical Pharmacology USF Health Taneja College of Pharmacy
University of Ghana Medical School University of South Florida
Accra, Ghana Tampa, FL, USA

Editorial Contact
Carolyn Spence

ISBN 978-3-031-12397-9    ISBN 978-3-031-12398-6 (eBook)


https://doi.org/10.1007/978-3-031-12398-6

© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature
Switzerland AG 2022
This work is subject to copyright. All rights are solely and exclusively licensed by the Publisher,
whether the whole or part of the material is concerned, specifically the rights of translation,
reprinting, reuse of illustrations, recitation, broadcasting, reproduction on microfilms or in any
other physical way, and transmission or information storage and retrieval, electronic adaptation,
computer software, or by similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are
exempt from the relevant protective laws and regulations and therefore free for general use.
The publisher, the authors, and the editors are safe to assume that the advice and information in
this book are believed to be true and accurate at the date of publication. Neither the publisher nor
the authors or the editors give a warranty, expressed or implied, with respect to the material
contained herein or for any errors or omissions that may have been made. The publisher remains
neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This Springer imprint is published by the registered company Springer Nature Switzerland AG
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
I dedicate this book to my mother, Comfort Aboagye-Adu, who
through her tireless efforts saw me through my education. Also
to my wife, Twumwaa, and children, Ethan and Elsye, who
continuously encourage me. I also dedicate this book to my
sister, Adwoa, for all her support.

I cannot forget, Prof. George Obeng Adjei, Prof. Jorgen


Kurtzhals, and Prof. Kwasi Agyei Bugyei, who have been my
academic mentors.
—Seth Kwabena Amponsah

To the loving memories of my parents and Dr. Keshav Baliram


Hedgewar, who gave proper direction to my life, to my beloved
wife Seema who gave positive meaning, and my son
Sarvadaman who gave a golden lining to my life.

I would like to dedicate this book to the loving memories of Ma


Chamanlaljee, Ma Lakshmanraojee Bhide, and Ma Madhujee
Limaye, who mentored me selflessly and helped me to become a
good and socially useful human being.
—Yashwant V. Pathak
Foreword

Over the years, there has been growing interest in the fields of therapeutic
drug monitoring and clinical toxicology. However, there appears to be few
books that address recent trends. The current book, which I am very glad to
write a foreword, has an excellent compilation of information on clinical
toxicology and therapeutic drug monitoring.
Due to the fact that medicine is a fast-evolving field, it is important that
information on current trends is documented. This book offers thorough, but
succinct, details on drug monitoring and clinical toxicology. The book
includes dedicated chapters for key topics, including analytical techniques in
therapeutic drug monitoring and clinical toxicology, the role of artificial
intelligence in therapeutic drug monitoring and clinical toxicity, and analys-
ing data from therapeutic drug monitoring, pharmacokinetics and clinical
toxicological studies. I am very confident that this book will be very benefi-
cial to researchers, toxicologists, clinicians and students in the field of bio-
medicine and clinical sciences. The book contains 21 chapters with rich
content, presenting fundamental facts, as well as practical and clinically
related data, and ends with challenges and future directions of therapeutic
drug monitoring and clinical toxicology. Each chapter is well written, clear,
precise and easy to understand. The text integration is a bonus, making it
appropriate for all in the sciences.
As a biomedical scientist, I strongly recommend this book to all research-
ers and students. I consider this book as essential for any reference library.
My heartiest congratulations to Seth Kwabena Amponsah and Yashwant
V. Pathak for such a laudable initiative. I would definitely have this book on
my desk.

Gordon A. Awandare
Pro-Vice Chancellor (Academic and Student Affairs), University of Ghana
Accra, Ghana

vii
Preface

The correlation between drug concentration in body fluids and outcome is


stronger than between drug dose and outcome. Hence, measuring systemic
drug concentration is an essential part of therapeutic drug monitoring (TDM).
Aside its role in therapy, TDM can also prevent unnecessary therapeutic
interventions and subsequently reduce healthcare costs. There is no doubt
that recent advances in TDM will shape clinical practice.
Toxicology is multidisciplinary, hence, contributions by diverse scientists.
In the modern era, toxicologists share scientific knowledge to obtain accurate
data about unwanted effects of different agents. Over the years, advanced
tools used in toxicological and epidemiological research have been
discovered.
This book gives an overview of TDM and its clinical application (analyti-
cal techniques, pharmacokinetic models, etc.). The book also highlights
recent advances in toxicological studies.
Furthermore, this book focuses on major aspects of emerging and recent
advances in TDM and clinical toxicology. The highlights include

(i) Analytical techniques in TDM and clinical toxicology


(ii) TDM and pharmacokinetic studies
(iii) TDM of drugs with narrow therapeutic indices
(iv) Artificial intelligence in TDM and clinical toxicology
(v) Future directions and challenges

The editors hope that this book will provide current information on TDM
and clinical toxicology to healthcare professionals and academicians who
work in the field of pharmacokinetics, toxicology, and pharmaceutical chem-
istry. Additionally, this book will affordably provide information on TDM
and clinical toxicology to those interested in drug safety and the need for
individualized therapy. The editors envisage that this book will be more of a
reference and resource for all stakeholders in the health sciences.
The editors thank all contributing authors, who continue to play critical
roles in the field of TDM and clinical toxicology. The editors also thank
Springer Nature, for accepting to get this book published.

Accra, Ghana Seth Kwabena Amponsah


Tampa, FL, USA Yashwant V. Pathak

ix
Contents

1 Therapeutic and Toxic Concentrations of Drugs


in Biological Matrices��������������������������������������������������������������������    1
Seth Kwabena Amponsah and Yashwant V. Pathak
2 Analytical Techniques for Therapeutic Drug
Monitoring and Clinical Toxicology ����������������������������������������������   9
Samuel O. Bekoe, Samuel Asare-Nkansah, and Kwabena F.
M. Opuni
3 Plasma Therapeutic Drug Monitoring
and Clinical Toxicology������������������������������������������������������������������   21
Gregory Fishberger, Nicole Natarelli, Dao Le, Deborah Liaw,
Afrin Naz, Caroline Ward, Michael Young, and Charles Preuss
4 Dried Blood Spots in Therapeutic Drug Monitoring
and Toxicology��������������������������������������������������������������������������������   43
Raphael N. Alolga, Qun Liu, and Qi Lian-Wen
5 The Role of Artificial Intelligence in Therapeutic
Drug Monitoring and Clinical Toxicity����������������������������������������   67
Surovi Saikia, Jinga B. Prajapati, Bhupendra G. Prajapati,
Vijaya V. Padma, and Yashwant V. Pathak
6 Therapeutic Drug Monitoring and Optimal Pharmacotherapy
with Medicines of Narrow Therapeutic Index����������������������������   87
Anthony Kwaw, Arnold Forkuo Donkor, and Kwame Ohene
Buabeng
7 Therapeutic Drug Monitoring (TDM) and Toxicological
Studies in Alternative Biological Matrices����������������������������������   95
Biswajit Basu, Bhupendra G. Prajapati, Swarupananda
Mukherjee, Tapas Kumar Roy, Arnab Roy, Chowdhury
Mobaswar Hossain, Jigna B. Prajapati, and Jayvadan Patel
8 
Analyzing Data from Therapeutic Drug Monitoring,
Pharmacokinetics, and Clinical Toxicology Studies ������������������ 117
Abdul Malik Sulley
9 Reducing Toxicity in Critically Ill Patients by Using
Therapeutic Drug Monitoring������������������������������������������������������ 143
Zalak Panchal, Khushboo Faldu, and Jigna Shah

xi
xii Contents

10 Quality
 Assurance of Samples for Therapeutic
Drug Monitoring and Clinical Toxicology ���������������������������������� 161
Samuel O. Bekoe, Samuel Asare-Nkansah, and Kwabena F.
M. Opuni
11 Therapeutic
 Drug Monitoring and Toxicology of
Anticancer Drugs �������������������������������������������������������������������������� 165
Seema Kohli and Lavakesh Kumar Omray
12 Therapeutic
 Drug Monitoring and Toxicology of
Immunosuppressant���������������������������������������������������������������������� 181
Anshul Shakya, Rajdeep Sarma, Neha Ghimire, Surajit Kumar
Ghosh, Hans Raj Bhat, and Obaidur Rahman
13 Therapeutic
 Drug Monitoring and Toxicology: Relevance of
Measuring Metabolites������������������������������������������������������������������ 197
James Akingbasote, Sandra Szlapinski, Elora Hilmas, Patrik
Miller, and Natalie Rine
14 Recent
 Advances in Nanosensors for Therapeutic
Drug Monitoring (TDM) �������������������������������������������������������������� 233
Percy Selasi Agogo-Mawuli and David P. Siderovski
15 Organ
 Toxicity by Immunosuppressive Drugs in
Solid Organ Transplantation�������������������������������������������������������� 255
George J. Dugbartey and Alp Sener
16 A
 rtificial Intelligence-Based Techniques to Assess
Drug Toxicity in Drug-Induced Liver
Injury (DILI) Disease�������������������������������������������������������������������� 273
Munish Puri
17 Drug
 Dose and Therapy Individualization���������������������������������� 285
Ashley Mason, Gavin Lockard, Vance Cantrell, Snow Pinxue
Li, Kirtan Patel, Sierra Klein, Andre Elder, Melissa Sur, and
Charles Preuss
18 Models
 for Drug Individualization: Patient to
Population Level���������������������������������������������������������������������������� 303
Sierra Klein, Ashley Mason, Gavin Lockard, Vance Cantrell,
Snow Pinxue Li, Kirtan Patel, Andre Elder, Melissa Sur, and
Charles Preuss
19 Toxicity
 Evaluation of Nanomedicine������������������������������������������ 323
Archna Panghal and Swaran Jeet Singh Flora
20 Biochemical
 Indices of Drug Toxicity������������������������������������������ 347
Emmanuel Kwaku Ofori
21 Therapeutic
 Drug Monitoring and Clinical Toxicology:
Challenges and Future Directions������������������������������������������������ 369
Seth Kwabena Amponsah and Yashwant V. Pathak

Index�������������������������������������������������������������������������������������������������������� 379
About the Editors

Seth Kwabena Amponsah, PhD is a senior lecturer and head of the


Department of Medical Pharmacology, University of Ghana Medical School.
He has an MPhil and PhD in pharmacology. He has over 10 years’ experience
in teaching and research. 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 therapeutic drug monitoring, population
pharmacokinetic modeling, non-compartment pharmacokinetic and
pharmacodynamic estimation, and pharmacokinetic evaluation of new drug
formulations. He has published over 40 research articles, 3 book chapters,
and several conference abstracts.

Yashwant V. Pathak has over 13 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). He now holds the position
of associate dean for faculty affairs and tenured professor of pharmaceutical
sciences at USF Health Taneja College of Pharmacy, University of South
Florida and an adjunct professor at Faculty of Pharmacy, Airlangga University,
Surabaya, Indonesia. Dr. Pathak is an internationally recognized scholar,
researcher, and educator in the areas of healthcare education, nanotechnology,
drug delivery systems, and nutraceuticals. He has published extensively with
over 50 edited volumes in the area of nanotechnology, drug delivery systems,
artificial neural networks, conflict management, and cultural studies. Dr.
Pathak has over 300 research papers, reviews, and chapters in books and has
presented in many national and international conferences. 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, and the International Commission for
Human Rights and Religious Freedom.

xiii
Contributors

Percy Selasi Agogo-Mawuli Department of Pharmacology & Neuro-


science, University of North Texas Health Science Center at Fort Worth, Fort
Worth, TX, USA
James Akingbasote Regulatory Toxicologist, London, ON, Canada
Raphael N. Alolga State Key Laboratory of Natural Medicines, School of
Traditional Chinese Pharmacy, Department of Pharmacognosy, China
Pharmaceutical University, Nanjing, China
Seth Kwabena Amponsah Department of Medical Pharmacology,
University of Ghana Medical School, Accra, Ghana
Samuel Asare-Nkansah Department of Pharmaceutical Chemistry, Faculty
of Pharmacy and Pharmaceutical Sciences, Kwame Nkrumah University of
Science and Technology, Kumasi, Ghana
Biswajit Basu Bengal School of Technology (A College of Pharmacy),
Sugandha, Hooghly, West Bengal, India
Samuel O. Bekoe Department of Pharmaceutical Chemistry, Faculty of
Pharmacy and Pharmaceutical Sciences, Kwame Nkrumah University of
Science and Technology, Kumasi, Ghana
Hans Raj Bhat Department of Pharmaceutical Sciences, Faculty of Science
and Engineering, Dibrugarh University, Dibrugarh, Assam, India
Kwame Ohene Buabeng Department of Pharmacy Practice, Faculty of
Pharmacy and Pharmaceutical Sciences, College of Health Sciences, Kwame
Nkrumah University of Science and Technology, Kumasi, Ghana
Vance Cantrell University of South Florida Morsani College of Medicine,
MD Program, Tampa, FL, USA
George J. Dugbartey Department of Surgery, Division of Urology, London
Health Sciences Center, Western University, London, ON, Canada
Department of Pharmacology and Toxicology, School of Pharmacy, College
of Health Sciences, University of Ghana, Accra, Ghana

xv
xvi Contributors

Andre Elder University of South Florida Morsani College of Medicine, MD


Program, Tampa, FL, USA
Khushboo Faldu Department of Pharmacology, Institute of Pharmacy,
Nirma University, Ahmedabad, Gujarat, India
Gregory Fishberger University of South Florida Morsani College of
Medicine, MD Program, Tampa, FL, USA
Swaran Jeet Singh Flora Department of Pharmacology and Toxicology,
National Institute of Pharmaceutical Education and Research, SAS Nagar,
Mohali, India
Arnold Forkuo Donkor Department of Pharmacology, Faculty of Pharmacy
and Pharmaceutical Sciences, College of Health Sciences, Kwame Nkrumah
University of Science and Technology, Kumasi, Ghana
Neha Ghimire Department of Pharmaceutical Sciences, Faculty of Science
and Engineering, Dibrugarh University, Dibrugarh, Assam, India
Surajit Kumar Ghosh Department of Pharmaceutical Sciences, Faculty of
Science and Engineering, Dibrugarh University, Dibrugarh, Assam, India
Elora Hilmas Nationwide Children’s Hospital, Columbus, OH, USA
Chowdhury Mobaswar Hossain Department of Pharmaceutical Science &
Technology, Maulana Abul Kalam Azad University of Technology,
Haringhata, Nadia, West Bengal, India
Sierra Klein University of South Florida Morsani College of Medicine, MD
Program, Tampa, FL, USA
Seema Kohli Department of Pharmaceutical Sciences, Kalaniketan
Polytechnic College, Jabalpur, Madhya Pradesh, India
Anthony Kwaw Department of Pharmacology, Faculty of Pharmacy and
Pharmaceutical Sciences, College of Health Sciences, Kwame Nkrumah
University of Science and Technology, Kumasi, Ghana
Dao Le University of South Florida Morsani College of Medicine, MD
Program, Tampa, FL, USA
Snow Pinxue Li University of South Florida Morsani College of Medicine,
MD Program, Tampa, FL, USA
Qi Lian-Wen Clinical Metabolomics Center, Department of Pharmacognosy,
China Pharmaceutical University, Nanjing, China
Qun Liu State Key Laboratory of Natural Medicines, School of Traditional
Chinese Pharmacy, Department of Pharmacognosy, China Pharmaceutical
University, Nanjing, China
Deborah Liaw University of South Florida Morsani College of Medicine,
MD Program, Tampa, FL, USA
Contributors xvii

Gavin Lockard University of South Florida Morsani College of Medicine,


MD Program, Tampa, FL, USA
Ashley Mason University of South Florida Morsani College of Medicine,
MD Program, Tampa, FL, USA
Patrik Miller Nationwide Children’s Hospital, Columbus, OH, USA
Swarupananda Mukherjee NSHM Knowledge Campus, Kolkata – Group
of Institutions, Kolkata, India
Nicole Natarelli University of South Florida Morsani College of Medicine,
MD Program, Tampa, FL, USA
Afrin Naz University of South Florida Morsani College of Medicine, MD
Program, Tampa, FL, USA
Emmanuel Kwaku Ofori Department of Chemical Pathology, University
of Ghana Medical School, Accra, Ghana
Lavakesh Kumar Omray Radharaman Institute of Pharmaceutical
Sciences, Bhopal, Madhya Pradesh, India
Kwabena F. M. Opuni Department of Pharmaceutical Chemistry, School of
Pharmacy, University of Ghana, Legon, Ghana
Vijaya V. Padma Translational Research Laboratory, Department of
Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
Zalak Panchal Department of Pharmacology, Institute of Pharmacy, Nirma
University, Ahmedabad, Gujarat, India
Archna Panghal Department of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali,
India
Jayvadan Patel Nootan Pharmacy College, Faculty of Pharmacy,
Sankalchand Patel University, Visnagar, Gujarat, India
Kirtan Patel University of South Florida Morsani College of Medicine, MD
Program, Tampa, FL, USA
Yashwant V. Pathak USF Health Taneja College of Pharmacy, University of
South Florida, Tampa, FL, USA
Bhupendra G. Prajapati Shree S K Patel College of Pharmaceutical
Education and Research, Ganpat University, Mahesana, Gujarat, India
Jinga B. Prajapati Acharya Motibhai Patel Institute of Computer Studies,
Ganpat University, Kherva, Mahesana, Gujarat, India
Charles Preuss University of South Florida Morsani College of Medicine,
Department of Molecular Pharmacology & Physiology, Tampa, FL, USA
Munish Puri Artificial Intelligence, Immunology, AbbVie Pharmaceutical,
Worcester, MA, USA
xviii Contributors

Obaidur Rahman Department of Pharmaceutical Sciences, Faculty of


Science and Engineering, Dibrugarh University, Dibrugarh, Assam, India
Natalie Rine Central Ohio Poison Center, Nationwide Children’s Hospital,
Columbus, OH, USA
Arnab Roy Department of Pharmacology and Toxicology, NIPER,
Hyderabad, India
Tapas Kumar Roy Ocular Pharmacology and Pharmacy Division, Dr. R.P
Centre, AIIMS, New Delhi, India
Surovi Saikia Translational Research Laboratory, Department of
Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
Rajdeep Sarma Department of Pharmaceutical Sciences, Faculty of Science
and Engineering, Dibrugarh University, Dibrugarh, Assam, India
Alp Sener Department of Microbiology & Immunology, Schulich School of
Medicine & Dentistry, University of Western Ontario, London, ON, Canada
Department of Surgery, Division of Urology, London Health Sciences Center,
Western University, London, ON, Canada
Jigna Shah Department of Pharmacology, Institute of Pharmacy, Nirma
University, Ahmedabad, Gujarat, India
Anshul Shakya Department of Pharmaceutical Sciences, Faculty of Science
and Engineering, Dibrugarh University, Dibrugarh, Assam, India
David P. Siderovski Department of Pharmacology & Neuroscience,
University of North Texas Health Science Center at Fort Worth, Fort Worth,
TX, USA
Abdul Malik Sulley IQVIA RDS Inc., Kirkland, QC, Canada
Melissa Sur University of South Florida Morsani College of Medicine, MD
Program, Tampa, FL, USA
Sandra Szlapinski Regulatory Toxicologist, London, ON, Canada
Caroline Ward University of South Florida Morsani College of Medicine,
MD Program, Tampa, FL, USA
Michael Young University of South Florida Morsani College of Medicine,
MD Program, Tampa, FL, USA
Therapeutic and Toxic
Concentrations of Drugs
1
in Biological Matrices

Seth Kwabena Amponsah and Yashwant V. Pathak

Abstract Keywords

Therapeutic drug monitoring (TDM) describes Bioanalysis · Drug levels · Matrices ·


the measurement of chemical parameters of Pharmacokinetics · Toxic concentration
drugs during clinical laboratory testing. TDM
aids estimation of the efficacy and safety of
drugs, often a determinant of future dosing 1.1 Introduction
pattern. It combines knowledge of pharma-
ceutics, pharmacokinetics, and pharmacody- For several decades, drug levels in biological sam-
namics of drugs. TDM typically involves ples have been estimated. Estimation of levels of
measuring of drug concentration in various drugs and other toxic substances in biological
biological fluids (matrices). Drug levels can matrices has proven essential in the field of medi-
be assayed in blood, urine, hair, tears, etc. The cine, toxicology, pharmacology, forensic science,
concentration of drugs measured in these and environmental research. Bioanalysis of drugs
matrices helps to estimate whether a drug is and toxic substances aids decision making in phar-
within its therapeutic range. Usually, when macotherapy and, under certain circumstances,
drug levels in these matrices attain toxic con- legal decisions [1]. Furthermore, assay of drugs in
centrations, it will lead to potential adverse biological matrices has seen tremendous techno-
effects, thus the need for documented data on logical advancement over the years [2, 3], and this
therapeutic and toxic concentrations of drugs has improved the practice of this science.
in the various biological matrices. Detection of drugs and metabolites in tissues
can aid clinical decision in pharmacotherapy,
detection of illicit drugs and foreign substances
(in toxicological and forensic contexts), estima-
tion of trace elements and toxicants in biological
matrices, and estimation of foreign compounds in
S. K. Amponsah (*) biological materials for signs of poison and
Department of Medical Pharmacology, University of sometimes post-mortem [4]. The equilibrium
Ghana Medical School, Accra, Ghana
e-mail: skamponsah@ug.edu.gh
between body fluids means that a drug present in
the blood will also be present in oral fluid (saliva),
Y. V. Pathak
USF Health Taneja College of Pharmacy, University
but this concentration may be very low, some-
of South Florida, Tampa, FL, USA times below the analytical detection limit. In
e-mail: ypathak1@usf.edu other instances, low levels of drugs may be
1
© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022
S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_1
2 S. K. Amponsah and Y. V. Pathak

deposited in growing hair. It is noteworthy that drug 1.3.1 Blood


in biological matrices may be affected by several
factors, including sample collection time, sample Due to advancements in sample preparation,
preparation, and stability of the drug [4–7]. chromatography, and detection techniques, whole
blood may be used as a screening matrix for
drugs [10]. In one matrix, both identification and
1.2 Therapeutic Drug quantification can be done. Blood is a very uni-
Monitoring form matrix because physiological factors vary
within restricted boundaries. Serum, plasma, and
Many decades ago, it became clear that the whole blood are among the most important matri-
administered dose of a drug alone does not pre- ces in TDM [11]. Measurement of plasma or
dict drug exposure. This prompted determination blood concentration may be a useful surrogate or
of systemic concentrations of drugs and linking indicator of the body’s exposure to drugs [8].
this to efficacy or adverse effects. This monitor- Sampling of blood for TDM should be done
ing of drug in biological matrices has made it steady state, which generally occurs at least five
possible to tailor drug treatment in individual half-lives into dosage regimen [12]. If a loading
patients [5]. dose is administered, steady state could be
Therapeutic drug monitoring (TDM) describes reached earlier. Before steady state is reached,
the measurement of a chemical parameter of a however, patients with hepatic or renal impair-
drug during clinical laboratory testing. This ment should be monitored to ensure that they do
parameter can, when paired with the right medi- not experience drug toxicity [8].
cal interpretation, have a direct impact on future
drug dosing [6]. TDM combines knowledge of
pharmaceutics, pharmacokinetics, and pharma- 1.3.2 Urine
codynamics. It aids in the individualization of
drug dosing by keeping concentrations of drug in Urine sample is more commonly used than blood
plasma or blood within a therapeutic range or to test for drugs of abuse [13]. The collection of
window [7]. Clinical pharmacists and pharma- urine and analysis of drug are relatively easy to
cologists use pharmacokinetic principles to inter- undertake. A urine sample will test positive for a
pret TDM data. TDM can be used to assess drug over a longer period than blood. Drug
compliance to drug regimen and drug-drug inter- metabolites or the parent moiety can be found in
actions. TDM may also be necessary when there urine for several days after a single dose [14].
is suspicion of toxicity, when there is subthera- There are countless applications of urinary drug
peutic effect, and when the manifestations of tox- determination in literature, even if a significant
icity and disease state are similar [8]. part of them may only be of toxicological impor-
tance. However, assaying drugs in urine can be
used in several contexts [15–17].
1.3 Biological Matrices Two of the main drawbacks of TDM using
urine are inconvenience of collecting samples
The most common biological samples used to and the possibility of a loss of integrity. Unless
estimate drug levels are serum, plasma, and urine urine voidance is observed, the authenticity of the
[9]. Drug level assays in whole blood, saliva, and sample can be questioned. It has been widely
cerebrospinal fluid can also be done, albeit less documented that urine adulterated with chemi-
frequently. Drug concentrations in different bio- cals or diluted can lead to misleading results [18].
logical matrices will not be the same since the Witnessing the collection of samples is essential
drug is not uniformly distributed within the body to prevent adulteration. However, this can be an
[3]. Characteristics of the various biological extremely time-consuming and impractical
matrices are summarized in Table 1.1. sometimes [19].
1 Therapeutic and Toxic Concentrations of Drugs in Biological Matrices 3

Table 1.1 Characteristics of different biological matrices


Characteristic Blood Urine Saliva Hair
Maximum drug detection 1–2 days 2–4 days 1–2 days 3–6 months
time Yes Yes No Yes
Intrusive sampling None High Low Medium
Potential for adulteration High Medium Low High
Refusal rate

1.3.3 Saliva 1.3.4 Cerebrospinal Fluid (CSF)

Water constitutes almost 99% of saliva, a viscous Using alternative matrices to conduct TDM can
oral fluid. Additionally, saliva contains salts, reduce pain, stress, and the general invasiveness
enzymes, peptides, hormones, lipids, sugars, epi- of sampling. However, it is sometimes necessary
thelial cells, food fragments, and microorganisms to conduct highly invasive sampling to assess
[20, 21]. In addition to maintaining the mucosa, drug disposition over time [29]. The CSF is one of
saliva also aids in chewing, mineralization of the most important sites for drug delivery because
teeth, regulating microorganisms, enhancing the blood-brain barrier (BBB) can vary the ratio
taste, and digestion [22]. of CSF to plasma concentration for many drugs
Oral fluids are now being considered as viable [23]. The study of drug levels in the CSF and
candidates for TDM, despite their limited use in directly in the brain has helped characterize com-
the past due to numerous restrictions [23]. Direct partmental pharmacology of drugs used for dis-
expectoration (spitting) is a good way to collect eases of the central nervous system [30].
large amounts of saliva (more than 1 ml). Drugs injected at clinically safe doses must be
Alternatively, saliva collectors in the form of able to penetrate the brain and CSF for effective
absorbing pads, wipes, or sponges may also be treatment of brain or meningeal diseases.
used [22]. Knowing the concentration of a drug at the site of
In pharmacokinetic studies, the use of saliva is action (on-target or off-target) can assist in guid-
advantageous because saliva contains fewer pro- ing pharmacokinetic and pharmacodynamic eval-
teins than plasma [24]. Thus, a drug is less likely uations, which in turn guides dosing decisions.
to bind to proteins in saliva. It is, therefore, pos- The brain and CSF are not readily or repeatedly
sible to quantify the biologically active forms of accessible compartments, and given that plasma
unbound drugs (or their metabolites) [25]. In and CSF clear drugs differently, the dynamics of
addition to providing noninvasive sampling and a drug concentrations in CSF cannot be accurately
great number of samples, saliva can be recovered predicted or extrapolated from plasma concentra-
from different types of patients (sometimes criti- tion data [31, 32].
cally ill ones) [26]. In some cases, saliva is con-
sidered a substitute for urine samples in
toxicology, since there is less chance that the 1.3.5 Hair
patient will deliberately adulterate the sample
[27]. Nonetheless, saliva samples are smaller Human hair consists of hair shaft and hair folli-
than blood and urine samples; hence, drug con- cle. Unlike the hair shaft, which consists of dead
centrations can be substantially low [22]. The keratinized epithelial cells, the hair follicle con-
analytical method used in assaying saliva sam- tains live epithelial cells. An intricate network of
ples must be able to identify and quantify several blood capillaries surrounds each hair follicle,
analytes from a small sample volume at low con- supplying it with nutrients. Each hair follicle is
centrations, which places some constraints on the directly connected to the sebaceous gland (oil
sample [28]. gland). There are three axial layers in every hair
4 S. K. Amponsah and Y. V. Pathak

shaft: the medulla (inner layer), cortex (middle single therapeutic range for all indications, regard-
layer), and cuticle (outer layer). There are less of age, co-medication, or comorbidity.
65–95% proteins in the hair matrix, mainly kera-
tins, water, lipids, and minerals [33].
Hair analysis may provide evidence of drug use 1.5 Toxic Concentration of Drugs
over an extended period. Blood and urine concen-
trations can only reflect use of drugs over hours Drug toxicity occurs when a drug’s therapeutic
and days, respectively [34]. Although the specific effect is exceeded; nevertheless, toxic and thera-
process for drug integration into hair is unknown, peutic responses can occur at the same time [42].
it is thought that drugs enter through blood during The manifestation of drug toxicity could be
hair development, sebum and sweat, and the exter- behavioral and physiological. Drug toxicity can
nal environment [35]. Blood sampling is more manifest itself behaviorally in a variety of ways,
intrusive than hair sampling. Hair with a width of including decreased locomotor activity, loss of
around a pencil and a weight of about 200 mg is motor coordination, and cognitive impairment.
typically taken from the back of the head [36]. The Tissue damage, neuronal death, and hormone
sample should be wrapped in aluminum foil and cycle disruptions are examples of physiological
kept dry at room temperature. It is important to manifestations of toxicity [43]. Safety is one of
thoroughly decontaminate it by washing it with the most significant challenges in drug develop-
various solvents before drug testing. ment. Clinical trials are affected by unexpected
toxicities, and post-market safety concerns can
lead to the withdrawal of new drugs from the
1.4 Therapeutic Concentration market [44]. For most drugs, there are therapeutic
of Drugs and toxic concentration ranges (Table 1.2).
Another principle that explains the toxic con-
The main goal of clinical pharmacokinetics is to centration of drugs is therapeutic index (TI),
improve efficacy of drugs, as well reduce toxic- which compares the dose of a drug that causes
ity. The discovery of robust correlations between therapeutic effect to the dose that causes death (in
systemic drug concentration and pharmacologi- animals) or toxicity (in humans) [45]. TI can be
cal effect has allowed clinicians to apply pharma- computed in animal research by dividing the
cokinetic principles to real-life patient settings lethal dose of a drug for 50% of the population
[37]. Drug concentration at the receptor site (and (LD50) by the minimal effective dose for 50% of
other tissues) can be affected by changes in the the population (ED50), i.e., TI = LD50/ED50.
plasma drug concentration. Increasing the con- Depending on the drug, the difference between
centration of the drug in plasma will often lead to the ED50 and the TD50 can be significant. The
a corresponding increase in the concentration of safer the drug, the larger the TI. A drug with a
the drug in most tissues [38]. narrow TI, on the other hand, has a steep
The therapeutic range of a drug is the range of concentration-­response relationship for efficacy,
doses or plasma (serum) concentrations that typi- toxicity, or both, resulting in a relatively low risk-­
cally leads to the desired therapeutic effect. While benefit range [42].
a patient may achieve benefit when drug concen- In clinical practice, TI can be the range of doses
trations are below the minimum threshold, he or at which drugs are considered effective in clinical
she may also experience adverse effects when trials for a median of participants without causing
drug concentrations at that level continue for pro- unacceptable adverse reactions [47]. This range is
longed periods [39]. It is important to consider the sufficient for most drugs, so when the recom-
benefit-to-risk ratio when determining the lower mended doses of a drug are prescribed, the maxi-
and upper limits of a treatment regimen. In the mum plasma concentration and the area under the
1960s and 1970s, pharmacokinetic studies and concentration-time curve are sufficiently above
expert opinions were used to assign therapeutic the minimum therapeutic concentration and below
ranges to drugs [40, 41]. In general, drugs have a the toxic concentration [46]. It can therefore be
1 Therapeutic and Toxic Concentrations of Drugs in Biological Matrices 5

Table 1.2 Therapeutic and toxic concentrations of some drugs in blood


Therapeutic blood concentration Toxic blood concentration
Drug (mg/L) (mg/L) References
Acetylsalicylic acid 20–200 300–350 [48]
(aspirin)
Alfuzosin 0.003–0.06 0.12 [49, 50]
Alprazolam 0.005–0.05 (−0.08) 0.1–0.4 [51]
Baclofen 0.08–0.4 1.1–3.8 [52]
Bisoprolol 0.01–0.1 0.2 [53]
Bromocriptine 0.1–0.3a 8a [51]
Cabergoline 0.058–0.144a 0.39a [51]
Candesartan 0.08–0.18 (−0.4) 0.54 [54]
Cetirizine Appr. 0.1–0.6 2–5 [54]
Dapsone 0.5–2 10 [55]
Dexamethasone Appr. 0.05–0.27 0.8 [56]
Ergotamine 0.36–0.42a 0.82a [54]
Furosemide (Frusemide) 2–5 (−10) 25–30 [54]
Gentamicin (2–) 4–10 12 [57]
Ibuprofen 15–30 (−50) 200 [54]
Levodopa (L-dopa) 0.3–2 5–20 [51]
Metformin 0.1–2 5–10 [49]
Naproxen (20–) 50–100 200 [54]
Omeprazole 0.05–4 8 [50]
Paracetamol (5–) 10–25 100–150 [58]
Quinine 1–7 10 [59]
Rabeprazole 0.2–1.8 3.6 [49]
Sulfasalazine 5–30 50 [49]
Tetracycline 1–5 (5–10) 30 [49]
Vancomycin 10–20 30–40 [57]
Warfarin 1–3 10–12 [60]
Zidovudine 0.1–0.3 2–3 [54]
Units are in ng/mL
a

assumed that at recommended doses, drugs are


clinically effective and are relatively safe. References
1. Amponsah SK, Boadu JA, Dwamena DK, Opuni
KFM. Bioanalysis of aminoglycosides using high-­
1.6 Conclusion performance liquid chromatography. ADMET
DMPK. 2022;10(1):27–62.
2. Reid E, Wilson ID, editors. Drug determination in
The concentration of drugs measured in bodily
therapeutic and forensic contexts. Elsevier B.V; 1986.
fluids or tissues is important since it helps to esti- p. 495.
mate whether a drug is within therapeutic range. 3. Peng GW, Chiou WL. Analysis of drugs and other
The most common biological samples used to toxic substances in biological samples for pharma-
cokinetic studies. J Chromatogr B Biomed Sci Appl.
determine drug levels are urine, serum, and
1990;531:3–50.
plasma. When a decision has been made to moni- 4. Wong SHY, et al. Microbore liquid chromatogra-
tor a drug’s concentration, it is critical to obtain a phy for therapeutic drug monitoring and toxicology:
biological sample that is clinically meaningful. clinical analyses of theophylline, caffeine, procain-
amide, and N-acetyl procainamide. J Liq Chromatogr.
Indeed, the relevance of literature on therapeutic
1987;10(2–3):491–506.
and toxic concentration of drugs in biological 5. Eliasson E, et al. Therapeutic drug monitoring for
matrices during drug monitoring cannot be tomorrow. Eur J Clin Pharmacol. 2013;69(Suppl 1):
overemphasized. 25–32.
6 S. K. Amponsah and Y. V. Pathak

6. Amponsah SK, et al. A pharmacokinetic evalua- 22. Drummer OH. Drug testing in oral fluid. Clin
tion of a pectin based oral multiparticulate matrix Biochem Rev. 2006;27(3):147–59.
carrier of carbamazepine. Adv Pharm Pharm Sci. 23. Avataneo V, et al. LC-MS application for therapeu-
2021;5527452:7. tic drug monitoring in alternative matrices. J Pharm
7. Opuni KFM, Boadu JA, Amponsah SK, Okai Biomed Anal. 2019;166:40–51.
CA. High performance liquid chromatography: a ver- 24. Nunes LAS, Brenzikofer R, Macedo DV. Reference
satile tool for assaying antiepileptic drugs in biologi- intervals for saliva analytes collected by a standard-
cal matrices. J Chromatogr B. 2021;1179:122750. ized method in a physically active population. Clin
8. Gross AS. Best practice in therapeutic drug monitor- Biochem. 2011;44(17):1440–4.
ing. Br J Clin Pharmacol. 2001;52(S1):5–9. 25. Burckhardt BB, Tins J, Laeer S. Liquid chromatog-
9. Hadland SE, Levy S. Objective testing: urine and raphy–tandem mass spectrometry method for deter-
other drug tests. Child Adolesc Psychiatr Clin N Am. mination of aliskiren in saliva and its application to a
2016;25(3):549–65. clinical trial with healthy volunteers. J Pharm Biomed
10. Dankyi BO, et al. Chitosan coated hydroxypropyl- Anal. 2014;96:118–26.
methyl cellulose microparticles of levodopa (and car- 26. Ghareeb M, Akhlaghi F. Alternative matrices for ther-
bidopa): in vitro and rat model kinetic characteristics. apeutic drug monitoring of immunosuppressive agents
Curr Ther Res. 2020;93:100612. using LC-MS/MS. Bioanalysis. 2015;7(8):1037–58.
11. Moeller MR, Steinmeyer S, Kraemer T. Determination 27. Petrides AK, et al. Monitoring opioid and benzodiaz-
of drugs of abuse in blood. J Chromatogr B Biomed epine use and abuse: is oral fluid or urine the preferred
Sci Appl. 1998;713(1):91–109. specimen type? Clin Chim Acta. 2018;481:75–82.
12. Gross AS. Best practice in therapeutic drug monitor- 28. Marchei E, et al. New synthetic opioids in biological
ing. Br J Clin Pharmacol. 1998;46(2):95–9. and non-biological matrices: a review of current ana-
13. Qian H-Z, et al. Current drug use and lack of HIV lytical methods. TrAC, Trends Anal Chem (Regular
virologic suppression: point-of-care urine drug screen ed). 2018;102:1–15.
versus self-report. BMC Infect Dis. 2014;14(1):508. 29. Cory TJ, et al. Overcoming pharmacologic sanctuar-
14. Johansson E, Halldin MM. Urinary excretion half-­ ies. Curr Opin HIV AIDS. 2013;8(3):190–5.
life of delta 1-tetrahydrocannabinol-7-oic acid in 30. Rizk ML, et al. Importance of drug pharmaco-
heavy marijuana users after smoking. J Anal Toxicol. kinetics at the site of action. Clin Transl Sci.
1989;13(4):218–23. 2017;10(3):133–42.
15. Michely JA, Meyer MR, Maurer HH. Power of 31. Best BM, et al. Low atazanavir concentrations in cere-
Orbitrap-based LC-high resolution-MS/MS for brospinal fluid. AIDS (London). 2009;23(1):83–7.
comprehensive drug testing in urine with or with- 32. Smith PB, et al. Population pharmacokinetics of
out conjugate cleavage or using dried urine spots Meropenem in plasma and cerebrospinal fluid of
after on-spot cleavage in comparison to established infants with suspected or complicated intra-abdominal
LC–MSn or GC–MS procedures. Drug Test Anal. infections. Pediatr Infect Dis J. 2011;30(10):844–9.
2018;10(1):158–63. 33. Boumba V, Ziavrou K, Vougiouklakis T. Hair as a bio-
16. Michely JA, Meyer MR, Maurer HH. Paper spray logical indicator of drug use, drug abuse or chronic
ionization coupled to high resolution tandem mass exposure to environmental toxicants. Int J Toxicol.
spectrometry for comprehensive urine drug test- 2006;25(3):143–63.
ing in comparison to liquid chromatography-­ 34. Mali, N., M. Karpe, and V.J. Kadam, A review on
coupled techniques after urine precipitation or biological matrices and analytical methods used for
dried urine spot workup. Anal Chem (Washington). determination of drug of abuse. 2011.
2017;89(21):11779–86. 35. Kintz P, Villain M, Cirimele V. Hair analysis for drug
17. Lee HH, et al. Simultaneous drug identification in detection. Ther Drug Monit. 2006;28(3):442–6.
urine of sexual assault victims by using liquid chro- 36. Gjerde H, et al. Detection of illicit drugs in oral fluid
matography tandem mass spectrometry. Forensic Sci from drivers as biomarker for drugs in blood. Forensic
Int. 2018;282:35–40. Sci Int. 2015;256:42–5.
18. Fu S. Adulterants in Urine Drug Testing. Adv Clin 37. Amponsah SK, et al. Effect of Cellgevity® supple-
Chem. 2016;76:123–63. ment on selected rat liver Cytochrome P450 enzyme
19. Wolff K, et al. File name: Expert Panel Review of activity and pharmacokinetic parameters of carba-
alternative biological matrices for use as an eviden- mazepine. Evid Based Complement Alternat Med.
tial sample for drug driving Expert Panel Review of 2020;7956493:8.
alternative biological matrices for use as an evidential 38. Amponsah SK, Opuni KFM, Antwi KA, Kunkpeh
sample for drug driving. Department for Transport VP. Effect of aminophylline on the pharmacokinet-
Expert Panel; 2017. ics of amikacin in Sprague-Dawley rats. J Infect Dev
20. Dawes C, et al. The functions of human saliva: a Countries. 2019;13(3):251–4.
review sponsored by the World Workshop on Oral 39. Amponsah SK, Opuni KFM, Donkor AA. Animal
Medicine VI. Arch Oral Biol. 2015;60(6):863–74. model investigation suggests betamethasone alters
21. Zhang C-Z, et al. Saliva in the diagnosis of diseases. the pharmacokinetics of amikacin. ADMET &
Int J Oral Sci. 2016;8(3):133–7. DMPK. 2018;6(4):279–83.
1 Therapeutic and Toxic Concentrations of Drugs in Biological Matrices 7

40. Cereghino JJ, et al. Carbamazepine for epilepsy. cation blood concentrations for prescription and
A controlled prospective evaluation. Neurology. over-the-counter medications. JAMA Netw Open.
1974;24(5):401. 2018;1(7):e184196.
41. Smith TW, Haber E. Digoxin intoxication: the rela- 51. Hiemke C, et al. Consensus guidelines for therapeu-
tionship of clinical presentation to serum digoxin tic drug monitoring in Neuropsychopharmacology:
concentration. J Clin Invest. 1970;49(12):2377–86. update 2017. Pharmacopsychiatry. 2018;51(1/02):
42. Amponsah SK, et al. Population pharmacokinetic 9–62.
characteristics of amikacin in suspected cases of neona- 52. Meyer FP. Indicative therapeutic and toxic drug
tal sepsis in a low-resource African setting: a prospec- concentrations in plasma : a tabulation. Int J Clin
tive non-randomized single-site study. Curr Ther Res. Pharmacol Ther. 1994;32(2):71–81.
2017;84:e1–e6. 53. Schulz M, et al. Beta receptor blockers. Principles
43. AL R, K S. Drug toxicity. In: Stolerman IP, editor. for drug selection for rational therapy. Medizinische
Encyclopedia of psychopharmacology. Berlin, Monatsschrift fur Pharmazeuten. 1989;12(8):237.
Heidelberg: Springer; 2010. 54. Drummer OH, Baselt RC. Disposition of toxic drugs
44. Basile AO, Yahi A, Tatonetti NP. Artificial intelligence and chemicals in man, Biomedical Publications, Seal
for drug toxicity and safety. Trends Pharmacol Sci Beach, CA. Elsevier Ireland Ltd; 2015. p. 12.
(Regular ed). 2019;40(9):624–35. 55. Piscitelli SC, et al. Therapeutic monitoring and
45. Katzung BG. Basic & clinical pharmacology. 14th pharmacist intervention in a Hansen's disease clinic.
ed. McGraw-Hill's AccessMedicine. New York: Ann Pharmacother. 1993;27(12):1526–31.
McGraw-Hill Education LLC; 2018. 56. Richter O, et al. Pharmacokinetics of dexametha-
46. Reiffel JA. Formulation substitution and other phar- sone in children. Pediatr Pharmacol (New York).
macokinetic variability: underappreciated variables 1983;3(3–4):329.
affecting antiarrhythmic efficacy and safety in clinical 57. Butler DR, Kuhn RJ, Chandler MHH. Pharmacokinetics
practice. Am J Cardiol. 2000;85(10):46–52. of anti-infective agents in paediatric patients. Clin
47. Reiffel JA. Issues in the use of generic antiarrhythmic Pharmacokinet. 1994;26(5):374–95.
drugs. Curr Opin Cardiol. 2001;16(1):23–9. 58. Skeith KJ, Brocks DR. Pharmacokinetic optimisation
48. Johnson MW, et al. Time course of pharmacokinetic and of the treatment of osteoarthritis. Clin Pharmacokinet.
hormonal effects of inhaled high-dose salvinorin A in 1994;26(3):233–42.
humans. J Psychopharm (Oxford). 2016;30(4):323–9. 59. Jurgens G, Graudal NA, Kampmann JP. Therapeutic
49. Regenthal R, et al. Drug levels: therapeutic and toxic drug monitoring of antiarrhythmic drugs. Clin
serum/plasma concentrations of common drugs. J Pharmacokinet. 2003;42(7):647–63.
Clin Monit Comput. 1999;15(7):529–44. 60. White RH, et al. Changes in plasma warfarin levels
50. Sutherland JJ, et al. Assessment of patient medication and variations in steady-state prothrombin times. Clin
adherence, medical record accuracy, and medi- Pharmacol Ther. 1995;58(5):588–93.
Analytical Techniques
for Therapeutic Drug Monitoring
2
and Clinical Toxicology

Samuel O. Bekoe, Samuel Asare-Nkansah,


and Kwabena F. M. Opuni

Abstract also possess seamless workflow. This chapter,


thus, discusses the various high-throughput
Therapeutic drug monitoring (TDM) and clin- analytical techniques employed in TDM and
ical toxicology (CT) studies play significant CT, as well as the challenges associated with
roles in understanding and controlling the their respective applications as reported in the
observed variabilities in therapeutic response literature. It must be emphasized that consid-
of administered drug products, as well as prof- eration for a suitable analytical method for
fering measures to improve the safety and effi- TDM and CT comes with careful planning
cacy of treatments that patients receive. and decision making. Factors to be considered
However, the optimization of patient care include but are not limited to the availability
through TDM continues to remain a challenge of expertise (clinical and laboratory), equip-
in many health jurisdictions despite the ment/instrument, the physicochemical nature
numerous advancements and progress in ana- of the target analyte (drug, metabolite, toxi-
lytical techniques and technology. The prac- cant, or toxin), and the clinical situation pre-
tice of TDM and CT in the optimization of senting the need for TDM. Other important
patient care is still evolving and requires a factors such as sample preparation and stor-
myriad of technical and material resources to age, analytical method development and vali-
achieve the needed optimal health outcomes. dation, and interferences associated with
One of the critical elements in this endeavour matrix effect also require careful consider-
is the availability of analytical techniques that ation in order to assure the reliability and
are sensitive, cost effective, and high perform- quality of TDM or CT data needed for
ing in terms of accuracy and precision and informed clinical decision.

S. O. Bekoe · S. Asare-Nkansah (*) Keywords


Department of Pharmaceutical Chemistry, Faculty of
Pharmacy and Pharmaceutical Sciences, Kwame Analytical techniques · Bioanalysis · Clinical
Nkrumah University of Science and Technology,
Kumasi, Ghana toxicology · Pharmacokinetics ·
e-mail: sankansah.pharm@knust.edu.gh Pharmacodynamics · Therapeutic drug
K. F. M. Opuni (*) monitoring
Department of Pharmaceutical Chemistry, School of
Pharmacy, University of Ghana, Legon, Ghana
e-mail: kfopuni@ug.edu.gh

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 9


S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_2
10 S. O. Bekoe et al.

2.1 Introduction (i) Suboptimal concentration of drug molecules


in the plasma and at the receptor site or site
Therapeutic drug monitoring (TDM) involves the of action in the patient because of potentially
optimization of therapy through adjustment of poor physicochemical quality of the drugs
dose at the individual level by monitoring con- (ii) An interruption in drug-receptor interactions
centrations of drug or drug metabolite in body and disruption of signal transduction path-
fluids (e.g. blood, plasma, or serum) or a suitable ways or processes [2, 6, 7, 9]
physiological matrix [1, 2]. The scientific basis
and practice of TDM date to 1946 with the estab- On the other hand, CT has been defined as a
lishment of a correlation between the pharmaco- ‘medical subspecialty focusing on the diagnosis,
logical activity of a drug and its corresponding management, and prevention of poisoning and
plasma concentration [3]. This scientific estab- other adverse health effects due to medications,
lishment resulted in the practice of dose adjust- occupational and environmental toxins and bio-
ment for individuals with peculiar demographic logical agents’ [10]. The strategy for CT includes
and clinical characteristics, among others, to among others the confirmation of the toxicant
obtain optimized therapy. Such decisions, how- and/or poison using the appropriate analytical
ever, are premised on the fact that the pharmaco- methods [11]. Since the dose of a drug makes it a
logical responses being observed by clinicians poison, almost all drugs are studied under CT
can be measured either clinically (e.g. in the use [12].
of analgesic or sedative drug) or with the use of As earlier mentioned, the relevance of appro-
an appropriate laboratory marker (e.g. in the use priate and sensitive analytical techniques in
of lipid-lowering drug) [1, 2, 4, 5]. The situation, TDM and CT cannot be overemphasized. We
however, becomes a bit more complex and unpre- propose in this book chapter that if sensitive
dictable when drug response cannot be easily high-­throughput analytical techniques and
evaluated clinically or when toxic side effects of detection technologies were more widely avail-
the drug cannot be easily monitored or detected able, user friendly, and affordable, there would
until irreparable damage has been caused. Thus, be better treatment outcomes, patient adher-
TDM can involve both the pharmacokinetic and ence, and reduced side effects for patients on
pharmacodynamic aspects of drug action in certain drugs including antibiotics, anticancer
applied pharmacotherapy. Therefore, analytical agents, and immunosuppressants (cyclosporin),
methods for the purposes of TDM should have among others. In order to make available such
the power to address relevant pharmacokinetic simple, cost-­effective, and efficient analytical
and pharmacodynamic parameters [2, 6–9]. methods, rigorous efforts that involve an invest-
To wit, different doses of a drug may be ment of resources into analytical technology
required by different patients/individuals in order development and transfer and, more impor-
to observe similar optimal therapeutic effects tantly, availability of resources to train and build
because of an individual patient’s pharmacoki- capacity for TDM and CT implementation will
netic and pharmacodynamic variabilities. be required in the global health delivery space
Different individuals absorb, distribute, metabo- [2, 6, 9].
lize, and excrete drugs after administration dif- Analytical techniques that have found suc-
ferently and as well are influenced by the action cessful use in TDM and CT can be very diverse,
of a drug based on the unique characteristics of depending on the depth of the investigation
the patient [6, 7]. Beyond the pharmacokinetic being undertaken (i.e. the physicochemical prop-
and pharmacodynamic factors that influence drug erties of the drug, pKa values, ionized or union-
action, other variables include: ized status at physiological pH, etc.), parameters
2 Analytical Techniques for Therapeutic Drug Monitoring and Clinical Toxicology 11

being monitored, the biological matrix (saliva, 2.2 Immunoassays


urine, plasma, etc.), and others. For instance, to
establish a scientifically sound correlation These analytical techniques which involve the
between plasma drug concentration and pharma- development and application of antibodies have
cological response, an analytical method must be found significant use in various disciplines
developed and validated. Analytical techniques including TDM and CT. The key aspect of the
earlier employed for TDM studies included colo- development and application of immunoassays
rimetry, electrochemical techniques, spectropho- borders mainly on antibodies. Antibodies, usu-
tometry, and spectrofluorimetry [13–18]. Some of ally generated by beta-lymphocytes following
these methods currently find very little or no use exposure to substances such as foreign cells or
at all in TDM studies due to their lack of specific- proteins, required to trigger the immune system
ity and inability to distinguish clear effects due to of mammals, are proteins. The main purpose of
matrix from the drug of interest [13, 17]. Although antibodies is to help fight infection in mammals.
such limitations exist for the majority of drugs, Various subtypes including IgA, IgD, IgE, IgG,
the application of colorimetry as well as flame and IgM have been reported and each has specific
atomic emission spectroscopy is still relevant for roles or functions within various compartments
TDM of lithium [19]. Furthermore, bioassay tech- of the human body acting as sentinel sites in
niques were previously used for TDM analysis of cases of re-infection or attack of new infections.
many antibiotics. However, this technique was Although this technique comes with its own
found as laborious with poor specificity for poly- unique limitations (including the probability of
pharmacy patients and very slow in obtaining data obtaining false results, the effect of matrix on
required to make an informed clinical decision. antibodies, poor specificity resulting in cross-­
The technique has also been reported as imprecise reactivity with compounds that share similar
for present-day applications [2]. physicochemical properties amongst others),
Thin-layer chromatography has also experi- immunoassays, with its cost effective, rapid
enced a significant shift in its use for TDM anal- detection, and robust nature, continue to play
ysis currently, as its limitations for total highly significant roles in the detection and quan-
quantitative estimates are a challenge for such tification of minute amounts of target analytes in
purposes [2]. Current high-throughput analyti- various complex matrices such as hair, plasma,
cal techniques, which mostly find wide applica- and others. Immunoassays are usually designed
tions for routine monitoring and measurement for specific analytical purposes. For example, it
of plasma drug concentrations, are electropho- could be designed and developed for solely quali-
resis, immunoassays, gas chromatography tative purposes as well as for both qualitative and
(GC), conventional high-performance liquid quantitative uses. Furthermore, it finds applica-
chromatography (HPLC), and recently, ultra- tion for both low and large molecular weight
performance liquid chromatography (UPLC). molecules (compounds with molecular weights
The availability and compatibility of hyphen- ≥250 daltons). The major common elements of
ated modes such as LC-UV, LC-MS/MS, interest in the design and development of an
LC-MS, and GC-MS have positively impacted immunoassay technique include (i) target analyte
on sensitivities of such techniques in the suc- (antibodies), (ii) a drug derivative needed to link
cessful quantitation of drug molecules in vari- to a reporter (hapten), (iii) the target for assay
ous biological matrices [2, 6, 20, 21]. (analyte), (iv) required buffers or conditions such
This chapter focuses on a description of mod- as pH of the sample, (v) a location to immobilize
ern analytical techniques employed in TDM and antibody (solid phase), (vi) the sample being ana-
CT, appraisal of the strengths and limitations of lysed (matrix), and (vii) a recorder that amplifies
the various techniques described, as well as pro- the result (usually in the form of enzymes, fluo-
vision of some examples of relevant studies rescence, chemiluminescence, and radioimmu-
reported in the literature. noassay). Although various forms exist for
12 S. O. Bekoe et al.

various purposes, the basic elements enumerated Table 2.1 Analytical methods for TDM and/or CT of
drugs
above remain basically the same for all immuno-
assay methods. Analytical
method Analyte Reference
The development of such analytical tech-
Immunoassay Immunosuppressants [28, 29]
niques first involves the creation of the required Antiepileptic drugs [30–32]
antibody, followed by its production, and finally Antibiotics [30, 33–36]
the immunoassay design. The evaluation of an Bronchodilators [30]
antibody available for the design of the assay is a Antimalarial agents [37]
critical stage that informs the assay format that Psychotropic drugs [38, 39]
will be employed for the assay. Assay formats are Cardiovascular agents [40]
Anticancer agents [35, 41, 42]
generally grouped into two categories namely, (i)
Monoclonal antibodies [43–45]
heterogenous and (ii) homogenous immunoas- Electrophoresis Antiepileptic drugs [31, 32, 47,
says. Heterogenous immunoassays require a sep- 48]
aration step to remove materials that did not bind Inflammatory bowel [49]
immunologically (separately bound from free disease drugs
materials). As an analytical method, certain key Nonsteroidal anti-­ [50, 51]
inflammatory drugs
parameters required to evaluate the performance Cardiovascular drugs [47]
of the analytical technique under consideration Psychotropic agents [47, 52]
include specificity/cross-reactivity, precision, Diuretics [47]
limit of detection, interferences/adulteration, and Vasodilators [47]
stability. The optimization of such parameters Antibiotics [53]
assures the quality of data obtained from such Anthelmintics [54]
Anaesthetic agents [55]
determinations [2, 22–27].
Biosensors Anticancer agents [56–59]
Examples of compounds that have undergone Anticoagulants [60]
TDM and reported in the literature employing Monoclonal antibodies [61–63]
various immunoassay techniques include immu- Antibiotics [64–72]
nosuppressants [28, 29], antiepileptic drugs [30– Bronchodilators [73]
32], antibiotics [30, 33–36], bronchodilators Anticonvulsants [74]
[30], antimalarial agents [37], psychotropic drugs Substance of abuse [75]
HPLC/UPLC Anti-infective agents [76]
[38, 39], cardiovascular agents [40], anticancer
Immunosuppressants [28, 77–83]
agents, [35, 41, 42], and monoclonal antibodies Antifungal agents [81, 83, 84]
[43–45] (Table 2.1). Also, immunoassays have Anti-arrhythmic drugs [85]
been used in CT studies of diverse drugs [46]. Monoclonal antibodies [86]
Antibiotics [21, 83,
87–89]
2.3 Electrophoresis Antiepileptic drugs [31, 32, 83,
90, 91]
Anticancer drugs [42, 83, 92,
Electrophoresis is a separation technique involv- 93]
ing the use of a high-voltage electric field Antiviral agents [83, 94]
(electro-­driven) to migrate and separate charged Cardiovascular drugs [83]
particles in a capillary-shaped separation com- Psychotropic agents [38, 83,
95–105]
partment. Efficient separations resulting from
Anticoagulants [83]
capillary electrophoresis (CE) are usually Antidiabetic agents [83]
employed in the analysis of charged compounds Substance of abuse [97]
or drug molecules under the influence of an elec-
tric field generated uniformly across the separa-
tion compartment. The application of this certain unique features that CE provides, and
technique especially in TDM is influenced by these include robustness of the instrument with
2 Analytical Techniques for Therapeutic Drug Monitoring and Clinical Toxicology 13

its high separation efficiency and sensitivity and 2.4 Biosensors


minimal application of samples (sample size) and
solvents, coupled with the versatile nature of its Biosensor-based techniques use antibodies,
applications. The above-mentioned applications enzymes, membranes, molecularly imprinted
together with highly improved resolution, polymers, and aptamers for the recognition of
decreased separation time, and automation of the analytes of interest based on binding affinity
instrument provide the required real-time detec- [110]. Biosensors may be classified as either
tion needed for such sensitive tasks. The princi- electrochemical, optical, piezoelectric, or nano-
ple of electrophoresis involves the high-voltage mechanical [110]. This technique is advanta-
influenced migration of charged species between geous due to low sample consumption, nearly
oppositely charged electrodes, dependent on non-invasive sample collection procedure,
electrostatic and electroosmotic forces. Three reduced reagent consumption, reduced analysis
key parameters (i.e. size, charge, and shape of the time, multiple analyte detection, and portability
analyte of interest) significantly influence the [111]. These advantages notwithstanding, there
efficiency of separation. The commonly encoun- are challenges associated with sensitivity, quali-
tered CE modes are capillary gel electrophoresis tative, or semi-quantitative results obtained with
(CGE), capillary isoelectric focusing (CIEF), the use of biosensors for TDM [112].
capillary electrochromatography (CEC), capil- Biosensors have been used for the TDM of
lary zone electrophoresis (CZE), and micellar anticancer agents [56–59], anticoagulants [60],
electrokinetic chromatography (MEKC). Quite a monoclonal antibodies [61–63], antibiotics [64–
significant number of drug molecules have had 72], bronchodilators [73], anticonvulsants [74],
their levels in biological matrices measured using and opioids [75] (Table 2.1). Biosensors have
these named modes or techniques in TDM stud- also been applied in CT studies [113].
ies. Though CZE finds extensive use and applica-
tion in TDM, all the other modes of separation
are also utilized as appropriate for some drug 2.5 Conventional HPLC
molecules. It is also worth mentioning that and Emerging UHPLC
hyphenation of CZE mode with mass spectrom- Techniques
etry detectors is made possible through efficient
compatibility and this further enhances structural For decades, HPLC and recently (from the first
elucidation for metabolite profiling [2, 106–108]. decade of the twenty-first century) UHPLC have
It must also be noted that of all the advantages seen significant applications in various disci-
listed for CE in TDM, some limitations do exist plines for the efficient separation and quantifica-
for its application, and these include the tendency tion of various analytes in various matrices. The
of target analytes to undergo adsorption (which ability of this unique and versatile technique to
can be reversible or irreversible) onto the nega- separate and analyse complex samples, both
tively charged surface of silica-based capillaries. small and large molecules, continues to gain
It is as well difficult in handling the very small prominence in almost all basic and applied sci-
sample sizes and volumes with precision. ences of which TDM is no exception [114–119].
Electrophoretic methods have been used for These liquid chromatography techniques have
the TDM of antiepileptic drugs [31, 32, 47, 48], found versatile application and use in diverse set-
inflammatory bowel disease drugs [49], nonste- tings as a result of its ability to separate a wide
roidal anti-inflammatory drugs [50, 51], cardio- range of sample types, exceptional resolution
vascular drugs [47], psychotropic agents [47, 52], power, speed of separation, and compatibility
diuretics [47], vasodilators [47], antibiotics [53], with a wide scope of highly sensitive detectors
anthelmintics [54], and anaesthetic agents [55] including mass spectrometric detectors. Such
(Table 2.1). CT studies have been reported for dif- advantages that conventional HPLC as well as
ferent drugs using electrophoretic methods [109]. emerging UHPLC techniques provide have influ-
14 S. O. Bekoe et al.

enced their high use and applications in forensic, stant composition for an entire analysis (isocratic
biological, and pharmaceutical research includ- mode) or being able to change/modify the com-
ing TDM and CT [2]. position of the solvent system for analysis with
Both conventional HPLC and UHPLC have time (gradient mode) provides the required elu-
similar mechanisms of separation and resolution. tion modes for efficient separation of compounds
However, when analysts and scientists in various in even highly complex matrices.
fields wish to have faster separation without com- However, there are several other factors that
promising data quality, then UHPLC becomes may influence the reliability of HPLC/UHPLC
the obvious choice for such tasks. UHPLC also data for TDM and CT. These include the nature
provides the requisite technological improve- of the matrix being studied (urine, blood, liver,
ments in stationary support material (column) saliva, etc.), probable interference from endoge-
chemistry, detectors, and overall hardware nous substances/compounds from the matrix, the
required for ultra-fast separation without com- levels of the analyte available for detection and
promising the quality of data obtained. quantification (sensitivity of the detector being
The efficient separation of components of dif- employed), and nature of sample preparation pro-
ferent samples could be due to the availability of cedures used prior to pre-concentration of sample
diverse modes of separation including adsorp- and analysis [2, 115].
tion, partition, ion exchange, and size exclusion. Examples of drug compounds for which TDM
The selection of the mode of separation usually has been performed and reported in literature
depends on the type of task to be performed and with the use of the liquid chromatography tech-
the physicochemical properties (polar or non-­ nique include anti-infective agents [76], immu-
polar nature, etc.) of the analyte. Thus, a wide nosuppressants [28, 77–83], antifungal agents
range of options with respect to the mode of [81, 83, 84], anti-arrhythmic drugs [85], mono-
separation such as normal phase, reversed phase, clonal antibodies [86], antibiotics [21, 83, 87–
ion exchange, or size exclusion chromatography 89], antiepileptic drugs [31, 32, 83, 90, 91],
are available for well-defined tasks. The sensitiv- anticancer agents [42, 83, 92, 93], antiviral agents
ity of such analytical methods would depend on [83, 94], cardiovascular drugs [83], psychotropic
the type of detectors employed to monitor the agents [38, 83, 95–105], anticoagulants [83],
column eluates, and these could be optical detec- antidiabetic agents [83], and substance of abuse
tors such as ultraviolet (UV) absorption, fluores- [97] (Table 2.1). CT studies of drugs using liquid
cence, diode array, and photodiode array chromatography have also been reported in the
detectors. It must be noted that the diode array literature [11, 120–122].
(DAD) and photodiode array (PDA) detectors
are advanced forms of UV detectors. Other
equally well-known detectors include refractive 2.6 Gas Chromatography (GC)
index and electrochemical detectors (two types,
namely, the coulometric detector and ampero- This analytical technique, which also borders
metric detector). on partition chromatography, has similarities to
The development and introduction of hyphen- the HPLC technique already described.
ated techniques especially with mass spectromet- However, there are some significant differences
ric detectors some decades ago have also seen with respect to the instrumentation, stationary
significant improvement in the ability of conven- phase support material (column), the mobile
tional HPLC and UHPLC to separate and iden- phase, and the nature of analytes. In GC, com-
tify samples in highly complex matrices such as pounds suitable for analysis must be volatile in
observed in TDM, and these include HPLC-MS, nature or if not volatile can be derivatized to
LC-MS/MS, and UPLC-MS/MS [2, 117]. provide samples that can be volatilized during
Further to all of the above advancements, the analysis without thermal decomposition
availability of using a solvent system with con- because of the high temperatures utilized in
2 Analytical Techniques for Therapeutic Drug Monitoring and Clinical Toxicology 15

such analysis. Unlike HPLC, the GC separation make room for appropriate decisions on the opti-
is ensured and performed on a stationary phase mal selection of techniques and instrumentation
which is usually a steel capillary tube that is to generate the required data for clinical interpre-
supplied with a continuous flow of inert gases tation and application. TDM should have com-
or supercritical fluid (SCF) as a mobile phase in prehensive sample collection and handling
a temperature-regulated oven (usually around protocols in addition to overarching quality con-
400 °C). The mode of separation could be gas- trol measures to govern sample analysis, data
solid or gas-liquid in nature, depending on acquisition, data interpretation, and data manage-
whether the column contains either a solid ment. The continuous introduction of novel and
(polymers) or liquid (polysiloxanes) stationary more efficacious drugs for the treatment of dis-
phase [2, 117]. eases, which can also be potentially toxic, further
The detection of compounds is successfully strengthens the need to have clinical and analyti-
achieved using any of the following detectors: cal mechanisms to improve drug therapy. For this
purpose, advances in the various analytical tech-
(i) Nitrogen-phosphorus detector niques and related instrumentations for TDM and
(ii) Alkali flame ionization detector CT have led to the availability of many analytical
(iii) Electron-capture detector tools with diverse costs and complexity as already
(iv) Atomic emission detector described. Among the several analytical tech-
(v) Flame ionization detector niques, the immunoassay technique appears to be
the most commonly used, with a lot of immuno-
Hyphenated modes with MS detectors are also assay kits commercially available. However, this
available for GC in TDM analysis. Limitations technique might not be suitable in multicompo-
such as lack of regular preventive maintenance nent analyses, especially in paediatrics where
and the presence of even trace levels of contami- small sample volumes are used to monitor mul-
nants in the mobile phase (carrier gas), among tiple analytes. Under such circumstances, the
others, could result in significant variabilities in HPLC/UHPLC technique and its hyphenated
data acquisition. Due to this challenge, few drug forms are considered more appropriate because
compounds have had their TDM studies reported of the capacity to analyse and detect multicom-
in literature with the use of GC due to the huge ponent analytes in a sample with a highly accept-
tasks associated with the monitoring and control able level of accuracy and precision, even when
of variabilities in data acquisition. certain analytes co-elute (when mass spectromet-
Examples of drug compounds for which TDM ric detection is applied). As a result of this, the
data has been determined and reported with the HPLC/UHPLC technique has been shown to be
use of GC include antiepileptic drugs [31, 32], suitable for a wide scope of drugs and matrices.
psychotropic drugs [123–125], antihistamines Though the initial capital investment into the liq-
[126], and narcotic analgesics [127] (Table 2.1). uid chromatographs is high, they are known to
GC has also been applied in CT studies [128]. have low running costs, making them a prudent
choice for resource constraint environments that
may need TDM to enhance therapeutic and clini-
2.7 Conclusion and Outlook cal outcomes. Despite its limited role in TDM as
a result of potential variabilities in data acquisi-
In actualizing the cost benefits of TDM in clinical tion, GC has been very useful in CT screening,
settings, it is imperative to re-emphasize the ver- especially in its hyphenated modes (GC-mass
satility of TDM in the establishment of bench- spectrometric detection).
marks for individualization of therapy, dose With the assessment of the advantages and
optimization, screening for drug interactions, and disadvantages of the other analytical techniques
prevention of drug toxicity. The objectives of any for TDM and CT studies, an appropriate decision
TDM project should be clearly defined in order to can be taken on optimal technique and instru-
16 S. O. Bekoe et al.

mentation for any clearly defined TDM or CT 16. Donbrow M. Instrumental methods in analyti-
cal chemistry: their principles and practice-optical
programmes. It is still unclear when TDM will methods. Pitman and Sons; 1967.
become routine in the health systems of emerging 17. Fernandez Torres R, Callejon Mochon M,
economies. Jimenez Sanchez JC, Bello Lopez MA, Guiraum
PA. Electrochemical behaviour and determination
of acrivastine in pharmaceuticals and human urine. J
Pharm Biomed Anal. 2002;30(4):1215–22.
References 18. Gorham JD, Walton KG, McClellan AC, Scott
MG. Evaluation of a new colorimetric assay for serum
1. Buclin T, Thoma Y, Widmer N, et al. The steps to lithium. Ther Drug Monit. 1994;16(3):277–80.
therapeutic drug monitoring: a structured approach 19. Popov P, Otruba V, Sommer L. Determination of lith-
illustrated with imatinib. Front Pharmacol. ium in blood serum--a comparison between molecu-
2020;11:177. lar absorption spectrophotometry and emission flame
2. Moffat AC, Osselton MD, Widdop B, Watts spectrometry. Clin Chim Acta. 1986;154(3):223–5.
J. Clarke’s analysis of drugs and poisons. London: 20. Jang SH, Yan Z, Lazor JA. Therapeutic drug moni-
Pharmaceutical Press; 2011. toring: a patient management tool for precision
3. Marshall E, Dearborn EH. The relation of the plasma medicine. Clin Pharmacol Ther. 2016;99(2):148–50.
concentration of quinacrine to its antimalarial activ- 21. Xu L, Cheng X, Zhu G, Hu J, Li Q, Fan G. Therapeutic
ity. J Pharmacol Exp Ther. 1946;88(2):142–53. drug monitoring of amikacin: quantification in
4. Abrantes JA, Jonsson S, Karlsson MO, Nielsen plasma by liquid chromatography-tandem mass
EI. Handling interoccasion variability in model-­ spectrometry and work experience of clinical phar-
based dose individualization using therapeu- macists. Eur J Hosp Pharm. 2022;29(e1):e77–82.
tic drug monitoring data. Br J Clin Pharmacol. 22. Dandliker WB, Kelly RJ, Dandliker J, Farquahar J,
2019;85(6):1326–36. Levin J. Fluorescence polarization immunoassay.
5. Bhattaram VA, Booth BP, Ramchandani RP, et al. Theory and experimental method. Immunochemistry.
Impact of pharmacometrics on drug approval and 1973;10(4):219–27.
labeling decisions: a survey of 42 new drug applica- 23. Henderson DR, Friedman SB, Harris JD,
tions. AAPS J. 2005;7(3):E503–12. Manning WB, Zoccoli MA. CEDIA, a new
6. Clarke W. Overview of therapeutic drug monitoring. homogeneous immunoassay system. Clin Chem.
In: Clinical challenges in therapeutic drug monitor- 1986;32(9):1637–41.
ing. Elsevier; 2016. p. 1–15. 24. Khanna P. Homogeneous enzyme immunoassay. In:
7. Collins FS, Varmus H. A new initiative on precision Principles and practice of immunoassay. Springer;
medicine. N Engl J Med. 2015;372(9):793–5. 1991. p. 326–64.
8. Cremers S, Guha N, Shine B. Therapeutic drug mon- 25. Ullman E. Homogeneous immunoassays. The
itoring in the era of precision medicine: opportuni- immunoassay handbook. London: Nature Publishing
ties! Br J Clin Pharmacol. 2016;82(4):900–2. Group; 2001.
9. Csajka C, Verotta D. Pharmacokinetic-­ 26. Ullman EF, Maggio ET. Principles of homogeneous
pharmacodynamic modelling: history and per- enzyme-immunoassay. In: Enzyme immunoassay.
spectives. J Pharmacokinet Pharmacodyn. CRC Press; 1980. p. 105–34.
2006;33(3):227–79. 27. Yalow RS, Berson SA. Assay of plasma insulin in
10. Vale A, Bradberry S. Clinical toxicology. In: human subjects by immunological methods. Nature.
General, applied and systems toxicology. Wiley; 1959;184(Suppl 21):1648–9.
2011. 28. Khoschsorur G, Fruehwirth F, Zelzer S, Stettin
11. Hallbach J, Degel F, Desel H, Felgenhauer M, Halwachs-Baumann G. Comparison of flu-
N. Analytical role in clinical toxicology: impact on orescent polarization immunoassay (FPIA)
the diagnosis and treatment of poisoned patients. J versus HPLC to measure everolimus blood concen-
Lab Med. 2009;33(2):1–15. trations in clinical transplantation. Clin Chim Acta.
12. Grandjean P. Paracelsus revisited: the dose concept 2007;380(1–2):217–21.
in a complex world. Basic Clin Pharmacol Toxicol. 29. Vicente FB, Smith FA, Peng Y, Wang S. Evaluation
2016;119(2):126–32. of an immunoassay of whole blood sirolimus in
13. Abdine H, Belal F. Polarographic behaviour and pediatric transplant patients in comparison with
determination of acrivastine in capsules and human high-performance liquid chromatography/tan-
urine. Talanta. 2002;56(1):97–104. dem mass spectrometry. Clin Chem Lab Med.
14. Bertholf RL, Savory MG, Winborne KH, Hundley 2006;44(4):497–9.
JC, Plummer GM, Savory J. Lithium determined in 30. Jolley ME. Fluorescence polarization immunoassay
serum with an ion-selective electrode. Clin Chem. for the determination of therapeutic drug levels in
1988;34(7):1500–2. human plasma. J Anal Toxicol. 1981;5(5):236–40.
15. Christian GD. Analytical strategies for the mea- 31. Kang J, Park YS, Kim SH, Kim SH, Jun MY. Modern
surement of lithium in biological samples. J Pharm methods for analysis of antiepileptic drugs in the
Biomed Anal. 1996;14(8–10):899–908. biological fluids for pharmacokinetics, bioequiva-
2 Analytical Techniques for Therapeutic Drug Monitoring and Clinical Toxicology 17

lence and therapeutic drug monitoring. Korean J 45. Tikhonova IA, Yang H, Bello S, et al. Enzyme-linked
Physiol Pharmacol. 2011;15(2):67–81. immunosorbent assays for monitoring TNF-alpha
32. Krasowski MD. Therapeutic drug monitoring of the inhibitors and antibody levels in people with rheu-
newer anti-epilepsy medications. Pharmaceuticals matoid arthritis: a systematic review and economic
(Basel). 2010;3(6):1909–35. evaluation. Health Technol Assess (Winchester,
33. Amponsah SK, Opuni KFM, Antwi KA, Kunkpeh England). 2021;25(8):1.
VP. Effect of aminophylline on the pharmacokinet- 46. Clarke W. Chapter 5 – Immunoassays for therapeutic
ics of amikacin in Sprague-Dawley rats. J Infect Dev drug monitoring and clinical toxicology. In: Hempel
Ctries. 2019;13(3):251–4. G, editor. Handbook of analytical separations.
34. Amponsah SK, Opuni KFM, Donkor AA. Animal Elsevier Science B.V.; 2020. p. 97–114.
model investigation suggests betamethasone alters 47. Evenson MA, Wiktorowicz JE. Automated capillary
the pharmacokinetics of amikacin. ADMET DMPK. electrophoresis applied to therapeutic drug monitor-
2018;6(4):279–83. ing. Clin Chem. 1992;38(9):1847–52.
35. Mendu DR, Chou PP, Soldin SJ. An improved 48. Thormann W, Theurillat R, Wind M, Kuldvee
application for the enzyme multipled immunoassay R. Therapeutic drug monitoring of antiepileptics by
technique for caffeine, amikacin, and methotrex- capillary electrophoresis: characterization of assays
ate assays on the Dade-Behring Dimension RxL via analysis of quality control sera containing 14
Max clinical chemistry system. Ther Drug Monit. analytes. J Chromatogr A. 2001;924(1):429–37.
2007;29(5):632–7. 49. Marakova K, Piestansky J, Zelinkova Z, Mikus
36. Wu XJ, Zhang J, Yu JC, et al. Establishment of nor- P. Capillary electrophoresis hyphenated with mass
vancomycin fluorescence polarization immunoassay spectrometry for determination of inflammatory
for therapeutic drug monitoring. J Antibiot (Tokyo). bowel disease drugs in clinical urine samples.
2012;65(1):35–9. Molecules. 2017;22(11):1973.
37. Khalil IF, Alifrangis M, Recke C, et al. Development 50. Makino K, Itoh Y, Teshima D, Oishi R. Determination
of ELISA-based methods to measure the anti-­ of nonsteroidal anti-inflammatory drugs in
malarial drug chloroquine in plasma and in pharma- human specimens by capillary zone electropho-
ceutical formulations. Malar J. 2011;10(1):249. resis and micellar electrokinetic chromatography.
38. Hackett LP, Dusci LJ, Ilett KF. A comparison of Electrophoresis. 2004;25(10–11):1488–95.
high-performance liquid chromatography and fluo- 51. Zinellu A, Carru C, Sotgia S, Porqueddu E, Enrico
rescence polarization immunoassay for therapeutic P, Deiana L. Separation of aceclofenac and diclof-
drug monitoring of tricyclic antidepressants. Ther enac in human plasma by free zone capillary elec-
Drug Monit. 1998;20(1):30–4. trophoresis using N-methyl-D-glucamine as an
39. Midha K, Loo J, Charette C, Rowe M, Hubbard J, effective electrolyte additive. Eur J Pharm Sci.
McGilveray I. Monitoring of therapeutic concentra- 2005;24(4):375–80.
tions of psychotropic drugs in plasma by radioim- 52. Musenga A, Saracino MA, Spinelli D, et al. Analysis
munoassays. J Anal Toxicol. 1978;2(5):185–92. of the recent antipsychotic aripiprazole in human
40. Kagawa Y, Iwamoto T, Matsuda H, Mukohara R, plasma by capillary electrophoresis and high-­
Sawada J, Kojima M. Comparative evaluation of performance liquid chromatography with diode array
digoxin concentrations determined by three assay detection. Anal Chim Acta. 2008;612(2):204–11.
systems: TDx, IMx and OPUS. Biopharm Drug 53. Oguri S, Miki Y. Determination of amikacin in human
Dispos. 2004;25(1):21–6. plasma by high-performance capillary electropho-
41. Broto M, McCabe R, Galve R, Marco MP. A resis with fluorescence detection. J Chromatogr B
high throughput immunoassay for the thera- Biomed Sci Appl. 1996;686(2):205–10.
peutic drug monitoring of tegafur. Analyst. 54. Prochazkova A, Chouki M, Theurillat R, Thormann
2017;142(13):2404–10. W. Therapeutic drug monitoring of albendazole:
42. Descoeur J, Dupuy AM, Bargnoux AS, Cristol JP, determination of albendazole, albendazole sulfoxide,
Mathieu O. Comparison of four immunoassays to and albendazole sulfone in human plasma using non-
an HPLC method for the therapeutic drug monitor- aqueous capillary electrophoresis. Electrophoresis.
ing of methotrexate: influence of the hydroxylated 2000;21(4):729–36.
metabolite levels and impact on clinical threshold. J 55. Chik Z, Johnston A, Tucker AT, Burn RT, Perrett
Oncol Pharm Pract. 2022;28(1):55–63. D. Validation and application of capillary electropho-
43. Afonso J, Lopes S, Goncalves R, et al. Proactive resis for the analysis of lidocaine in a skin tape strip-
therapeutic drug monitoring of infliximab: a com- ping study. Biomed Chromatogr. 2007;21(8):775–9.
parative study of a new point-of-care quantitative 56. Yang J, Tan X, Shih WC, Cheng MM. A sandwich
test with two established ELISA assays. Aliment substrate for ultrasensitive and label-free SERS
Pharmacol Ther. 2016;44(7):684–92. spectroscopic detection of folic acid/methotrexate.
44. Perez I, Fernandez L, Sanchez-Ramon S, et al. Biomed Microdevices. 2014;16(5):673–9.
Reliability evaluation of four different assays for ther- 57. Yockell-Lelièvre H, Bukar N, Toulouse JL, Pelletier
apeutic drug monitoring of infliximab levels. Ther JN, Masson JF. Naked-eye nanobiosensor for thera-
Adv Gastroenterol. 2018;11:1756284818783613.
18 S. O. Bekoe et al.

peutic drug monitoring of methotrexate. Analyst. 71. Tomassetti M, Conta G, Campanella L, et al. A flow
2016;141(2):697–703. SPR immunosensor based on a sandwich direct
58. Zhao SS, Bichelberger MA, Colin DY, Robitaille R, method. Biosensors (Basel). 2016;6(2):22.
Pelletier JN, Masson J-F. Monitoring methotrexate 72. Zhu Y, Qu C, Kuang H, et al. Simple, rapid and
in clinical samples from cancer patients during che- sensitive detection of antibiotics based on the side-­
motherapy with a LSPR-based competitive sensor. by-­side assembly of gold nanorod probes. Biosens
Analyst. 2012;137(20):4742–50. Bioelectron. 2011;26(11):4387–92.
59. Zhao SS, Bukar N, Toulouse JL, et al. Miniature 73. Hanbury CM, Miller WG, Harris RB. Antibody
multi-channel SPR instrument for methotrexate characteristics for a continuous response fiber
monitoring in clinical samples. Biosens Bioelectron. optic immunosensor for theophylline. Biosens
2015;64:664–70. Bioelectron. 1996;11(11):1129–38.
60. Rusnati M, Bugatti A. Surface plasmon resonance 74. Astles JR, Miller WG. Measurement of free phenyt-
analysis of heparin-binding angiogenic growth fac- oin in blood with a self-contained fiber-optic immu-
tors. Methods Mol Biol. 2016;1464:73–84. nosensor. Anal Chem. 1994;66(10):1675–82.
61. Beeg M, Nobili A, Orsini B, et al. A Surface Plasmon 75. Dillon PP, Daly SJ, Manning BM, O’Kennedy
Resonance-based assay to measure serum concentra- R. Immunoassay for the determination of
tions of therapeutic antibodies and anti-drug anti- morphine-­ 3-glucuronide using a surface plasmon
bodies. Sci Rep. 2019;9(1):2064. resonance-based biosensor. Biosens Bioelectron.
62. Lu J, Spasic D, Delport F, et al. Immunoassay for 2003;18(2–3):217–27.
detection of infliximab in whole blood using a fiber-­ 76. Veringa A, Sturkenboom MGG, Dekkers BGJ,
optic surface plasmon resonance biosensor. Anal et al. LC-MS/MS for therapeutic drug monitoring
Chem. 2017;89(6):3664–71. of anti-infective drugs. TrAC Trends Anal Chem.
63. Van Stappen T, Bollen L, Vande Casteele N, 2016;84:34–40.
et al. Rapid test for infliximab drug concentra- 77. Baldelli S, Zenoni S, Merlini S, Perico N, Cattaneo
tion allows immediate dose adaptation. Clin Transl D. Simultaneous determination of everolimus and
Gastroenterol. 2016;7(12):e206. cyclosporine concentrations by HPLC with ultravio-
64. Cappi G, Spiga FM, Moncada Y, et al. Label-free let detection. Clin Chim Acta. 2006;364(1–2):354–8.
detection of tobramycin in serum by transmission-­ 78. Khoschsorur G. Simultaneous measurement of siro-
localized surface plasmon resonance. Anal Chem. limus and everolimus in whole blood by HPLC with
2015;87(10):5278–85. ultraviolet detection. Clin Chem. 2005;51(9):1721–4.
65. Cooper MA, Fiorini MT, Abell C, Williams 79. Korecka M, Shaw LM. Review of the newest HPLC
DH. Binding of vancomycin group antibiot- methods with mass spectrometry detection for deter-
ics to D-alanine and D-lactate presenting self-­ mination of immunosuppressive drugs in clinical
assembled monolayers. Bioorg Med Chem. practice. Ann Transplant. 2009;14(2):61–72.
2000;8(11):2609–16. 80. Seger C, Tentschert K, Stoggl W, Griesmacher A,
66. Losoya-Leal A, Estevez MC, Martinez-Chapa SO, Ramsay SL. A rapid HPLC-MS/MS method for
Lechuga LM. Design of a surface plasmon reso- the simultaneous quantification of cyclosporine
nance immunoassay for therapeutic drug monitoring A, tacrolimus, sirolimus and everolimus in human
of amikacin. Talanta. 2015;141:253–8. blood samples. Nat Protoc. 2009;4(4):526–34.
67. Luo Q, Yu N, Shi C, Wang X, Wu J. Surface plas- 81. Takasaki S, Hirasawa T, Sato Y, et al. Simultaneous
mon resonance sensor for antibiotics detection analysis of drugs administered to lung-transplanted
based on photo-initiated polymerization molecularly patients using liquid chromatography-tandem mass
imprinted array. Talanta. 2016;161:797–803. spectrometry for therapeutic drug monitoring.
68. Rao J, Yan L, Xu B, Whitesides GM. Using sur- Biomed Chromatogr. 2021;35(6):e5067.
face plasmon resonance to study the binding of 82. Taylor PJ. Therapeutic drug monitoring of immu-
vancomycin and its dimer to self-assembled mono- nosuppressant drugs by high-performance liquid
layers presenting d-Ala-d-Ala. J Am Chem Soc. chromatography-­ mass spectrometry. Ther Drug
1999;121(11):2629–30. Monit. 2004;26(2):215–9.
69. Spiga FM, Maietta P, Guiducci C. More DNA-­ 83. Tuzimski T, Petruczynik A. Review of chromato-
aptamers for small drugs: a capture-SELEX coupled graphic methods coupled with modern detection
with surface plasmon resonance and high-­throughput techniques applied in the therapeutic drugs monitor-
sequencing. ACS Comb Sci. 2015;17(5):326–33. ing (TDM). Molecules. 2020;25(17):4026.
70. Tenaglia E, Ferretti A, Decosterd LA, et al. 84. Yousefian S, Dastan F, Marjani M, et al.
Comparison against current standards of a DNA Determination of voriconazole plasma concentration
aptamer for the label-free quantification of by HPLC technique and evaluating its association
tobramycin in human sera employed for thera- with clinical outcome and adverse effects in patients
peutic drug monitoring. J Pharm Biomed Anal. with invasive aspergillosis. Can J Infect Dis Med
2018;159:341–7. Microbiol. 2021;2021:5497427.
85. Gui Y, Lu Y, Li S, et al. Direct analysis in real
time-mass spectrometry for rapid quantification of
2 Analytical Techniques for Therapeutic Drug Monitoring and Clinical Toxicology 19

five anti-arrhythmic drugs in human serum: appli- performance liquid chromatography method coupled
cation to therapeutic drug monitoring. Sci Rep. with tandem mass spectrometry for determination of
2020;10(1):15550. forty-two therapeutic drugs and drugs of abuse in
86. Becher F, Ciccolini J, Imbs DC, et al. A simple and human urine. Anal Chem. 2010;82(13):5636–45.
rapid LC-MS/MS method for therapeutic drug moni- 98. Gutteck U, Rentsch KM. Therapeutic drug monitor-
toring of cetuximab: a GPCO-UNICANCER proof ing of 13 antidepressant and five neuroleptic drugs
of concept study in head-and-neck cancer patients. in serum with liquid chromatography-electrospray
Sci Rep. 2017;7(1):2714. ionization mass spectrometry. Clin Chem Lab Med.
87. Feferbaum R, Kobol Machado JK, de Albuquerque 2003;41(12):1571–9.
Diniz EM, et al. Vancomycin monitoring in term 99. Kirschbaum KM, Muller MJ, Zernig G, et al.
newborns: comparison of peak and trough serum Therapeutic monitoring of aripiprazole by HPLC
concentrations determined by high performance liq- with column-switching and spectrophotometric
uid chromatography and fluorescence polarization detection. Clin Chem. 2005;51(9):1718–21.
immunoassay. Rev Hosp Clin Fac Med Sao Paulo. 100. Kollroser M, Schober C. Simultaneous determi-
2001;56(5):149–52. nation of seven tricyclic antidepressant drugs in
88. Feliu C, Konecki C, Candau T, et al. Quantification human plasma by direct-injection HPLC-APCI-­
of 15 antibiotics widely used in the critical care MS-MS with an ion trap detector. Ther Drug Monit.
unit with a LC-MS/MS system: an easy method 2002;24(4):537–44.
to perform a daily therapeutic drug monitoring. 101. Lancelin F, Djebrani K, Tabaouti K, et al.
Pharmaceuticals (Basel). 2021;14(12):1214. Development and validation of a high-performance
89. Verhoven SM, Groszek JJ, Fissell WH, et al. liquid chromatography method using diode array
Therapeutic drug monitoring of piperacillin and detection for the simultaneous quantification of
tazobactam by RP-HPLC of residual blood speci- aripiprazole and dehydro-aripiprazole in human
mens. Clin Chim Acta. 2018;482:60–4. plasma. J Chromatogr B Analyt Technol Biomed
90. Baldelli S, Cattaneo D, Giodini L, et al. Development Life Sci. 2008;867(1):15–9.
and validation of a HPLC-UV method for the quanti- 102. Luo M, Qin Y-P, Yu Q, et al. Determination of acam-
fication of antiepileptic drugs in dried plasma spots. prosate calcium in human plasma by RP-HPLC-MS/
Clin Chem Lab Med. 2015;53(3):435–44. MS. Chin J New Drugs. 2007;16:163–6.
91. Martinavarro-Dominguez A, Capella-Peiro ME, 103. Qi Y, Liu G. Ultra-performance liquid
Gil-Agusti M, Marcos-Tomas JV, Esteve-Romero chromatography-­ tandem mass spectrometry for
J. Therapeutic drug monitoring of anticonvulsant simultaneous determination of antipsychotic drugs in
drugs by micellar HPLC with direct injection of human plasma and its application in therapeutic drug
serum samples. Clin Chem. 2002;48(10):1696–702. monitoring. Drug Des Devel Ther. 2021;15:463–79.
92. Farkouh A, Ettlinger D, Schueller J, Georgopoulos 104. Theurillat R, Thormann W. Monitoring of tricy-
A, Scheithauer W, Czejka M. A rapid and simple clic antidepressants in human serum and plasma
HPLC assay for quantification of capecitabine by HPLC: characterization of a simple, laboratory
for drug monitoring purposes. Anticancer Res. developed method via external quality assessment. J
2010;30(12):5207–11. Pharm Biomed Anal. 1998;18(4–5):751–60.
93. Liang J, Zhang Z, Zhao H, et al. Simple and rapid mon- 105. Wen Y-G, Shi Y-X, Wang G-F. Determination of
itoring of doxorubicin using streptavidin-­modified aripiprazole in plasma by RP-HPLC with UV detec-
microparticle-based time-resolved fluorescence tion. Chin J Hosp Pharm. 2006;26(1):50.
immunoassay. RSC Adv. 2018;8(28):15621–31. 106. Haselberg R, de Jong GJ, Somsen GW. Capillary
94. Lamorde M, Fillekes Q, Sigaloff K, et al. Therapeutic electrophoresis-mass spectrometry for the
drug monitoring of nevirapine in saliva in Uganda analysis of intact proteins. J Chromatogr A.
using high performance liquid chromatography and 2007;1159(1–2):81–109.
a low cost thin-layer chromatography technique. 107. Staub A, Guillarme D, Schappler J, Veuthey
BMC Infect Dis. 2014;14:473. JL, Rudaz S. Intact protein analysis in the bio-
95. Aymard G, Livi P, Pham YT, Diquet B. Sensitive and pharmaceutical field. J Pharm Biomed Anal.
rapid method for the simultaneous quantification of 2011;55(4):810–22.
five antidepressants with their respective metabolites 108. Staub A, Schappler J, Rudaz S, Veuthey
in plasma using high-performance liquid chromatog- JL. CE-TOF/MS: fundamental concepts, instrumen-
raphy with diode-array detection. J Chromatogr B tal considerations and applications. Electrophoresis.
Biomed Sci Appl. 1997;700(1–2):183–9. 2009;30(10):1610–23.
96. Bose D, Durgbanshi A, Martinavarro-Dominguez 109. Thormann W, Aebi Y, Lanz M, Caslavska J. Capillary
A, et al. Amitriptyline and nortriptyline serum electrophoresis in clinical toxicology. Forensic Sci
determination by micellar liquid chromatography. J Int. 1998;92(2–3):157–83.
Pharmacol Toxicol Methods. 2005;52(3):323–9. 110. Ates HC, Roberts JA, Lipman J, Cass AEG, Urban
97. Chiuminatto U, Gosetti F, Dossetto P, et al. GA, Dincer C. On-site therapeutic drug monitoring.
Automated online solid phase extraction ultra high Trends Biotechnol. 2020;38(11):1262–77.
20 S. O. Bekoe et al.

111. Meneghello A, Tartaggia S, Alvau MD, Polo F, 121. Pragst F, Herzler M, Erxleben BT. Systematic
Toffoli G. Biosensing technologies for thera- toxicological analysis by high-performance liquid
peutic drug monitoring. Curr Med Chem. chromatography with diode array detection (HPLC-­
2018;25(34):4354–77. DAD). Clin Chem Lab Med. 2004;42(11):1325–40.
112. Sin ML, Mach KE, Wong PK, Liao JC. Advances 122. Viette V, Hochstrasser D, Fathi M. LC-MS (/MS)
and challenges in biosensor-based diagnosis in clinical toxicology screening methods. Chimia
of infectious diseases. Expert Rev Mol Diagn. (Aarau). 2012;66(5):339–42.
2014;14(2):225–44. 123. Girault J, Gobin P, Fourtillan JB. Determination of
113. Wang XH, Wang S. Sensors and biosensors for the calcium acetylhomotaurinate in human plasma and
determination of small molecule biological toxins. urine by combined gas chromatography-­ negative-­
Sensors (Basel). 2008;8(9):6045–54. ion chemical ionization mass spectrometry. J
114. Amponsah SK, Boadu JA, Dwamena DK, Opuni Chromatogr B Biomed Sci Appl. 1990;530:295–305.
KFM. Bioanalysis of aminoglycosides using high-­ 124. Volin P. Therapeutic monitoring of tricyclic antide-
performance liquid chromatography. ADMET pressant drugs in plasma or serum by gas chroma-
DMPK. 2022;10(1):27–62. tography. Clin Chem. 1981;27(10):1785–7.
115. Halász I, Endele R, Asshauer J. Ultimate limits in 125. Wilson JM, Williamson LJ, Raisys VA. Simultaneous
high-pressure liquid chromatography. J Chromatogr measurement of secondary and tertiary tricyclic anti-
A. 1975;112:37–60. depressants by GC/MS chemical ionization mass
116. Hansen SH. Column liquid chromatography on fragmentography. Clin Chem. 1977;23(6):1012–7.
dynamically modified silica. I. J Chromatogr A. 126. Chang SY, Nelson FR, Findlay JWA, Taylor
1981;209(2):203–10. LCE. Quantitative gas chromatographic-mass spec-
117. Miller JM. Chromatography: concepts and contrasts. trometric analysis of acrivastine and a metabolite in
John Wiley & Sons; 2005. human plasma. J Chromatogr B Biomed Sci Appl.
118. Opuni KFM, Boadu JA, Amponsah SK, Okai 1989;497:288–95.
CA. High performance liquid chromatography: a 127. Kintz P, Tracqui A, Lugnier AJ, Mangin P, Chaumont
versatile tool for assaying antiepileptic drugs in bio- AA. Simultaneous screening and quantification of
logical matrices. J Chromatogr B Analyt Technol several nonopiate narcotic analgesics and phency-
Biomed Life Sci. 2021;1179:122750. clidine in human plasma using capillary gas chro-
119. Snyder LR. Principles of adsorption chromatogra- matography. Methods Find Exp Clin Pharmacol.
phy; the separation of nonionic organic compounds. 1990;12(3):193–6.
Marcel Dekker, Inc; 1968. 128. Maurer HH. Role of gas chromatography-mass
120. Bekoe SO, Orman E, Asare-Nkansah S, et al. spectrometry with negative ion chemical ioniza-
Detection and quantification of antibiotic residues in tion in clinical and forensic toxicology, doping
urine samples of healthy individuals from rural and control, and biomonitoring. Ther Drug Monit.
urban communities in Ghana using a validated SPE-­ 2002;24(2):247–54.
LC-­MS/MS method. SN Appl Sci. 2020;2(11):1903.
Plasma Therapeutic Drug
Monitoring and Clinical Toxicology
3
Gregory Fishberger, Nicole Natarelli, Dao Le,
Deborah Liaw, Afrin Naz, Caroline Ward,
Michael Young, and Charles Preuss

Abstract as well as age-related changes in drug metabo-


lism, further complicates medication dosing.
Therapeutic drug monitoring (TDM) is a Therapeutic drug monitoring in the context of
branch of clinical pharmacology that seeks to each patient serves to address these factors
optimize and maintain the concentration of and requires collaboration between pharmacy,
medications within the bloodstream. The abil- nursing, and medical teams.
ity to predict a drug concentration within the
serum or plasma following a predetermined Keywords
dose is essential in treating disease; however,
a given dose of a drug may not produce identi- Therapeutic monitoring · Pharmacokinetic ·
cal serum concentrations between patients due Pharmacodynamic · Drug metabolism ·
to variations in drug formulation, drug interac- Optimal dosing
tions, environmental factors, genetic variation,
and renal and hepatic function. Therein lies
the risk of undertreatment or overtreatment, 3.1 Overview of Therapeutic
particularly in drugs with narrow therapeutics Drug Monitoring (TDM)
indices. Identifying drug concentrations at
steady state using proper measurement tech- Therapeutic drug monitoring (TDM) is a branch
niques specific to the drug of interest is both of clinical pharmacology in which serum or
practical and maximizes safety and efficacy. plasma medication concentrations are regularly
Of note, pharmacogenetic variation in cyto- measured to maintain a constant concentration in
chrome P-450 enzymes between individuals, the patient’s bloodstream. The practice of thera-
peutic drug monitoring stems from the observa-
tion that a given dose of a drug does not
G. Fishberger (*) · N. Natarelli · D. Le · D. Liaw necessarily produce the same plasma or serum
A. Naz · C. Ward · M. Young concentrations in every patient; instead, a multi-
University of South Florida Morsani College of
Medicine, MD Program, Tampa, FL, USA tude of patient factors and behaviors can influ-
e-mail: gfishberger@usf.edu ence drug absorption, distribution, metabolism,
C. Preuss and elimination, ultimately impacting concentra-
University of South Florida Morsani College of tions in the bloodstream [1]. By measuring drug
Medicine, Department of Molecular Pharmacology & concentrations in the serum or plasma at desig-
Physiology, Tampa, FL, USA nated intervals, physicians can tailor treatment to
e-mail: cpreuss@usf.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 21


S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_3
22 G. Fishberger et al.

maximize therapeutic benefit and minimize the metabolites for proper interpretation of results,
risk of toxicity [2]. which is usually not possible in routine monitor-
ing. In addition, there is reduced utility of TDM
for drugs with a large therapeutic range such that
3.1.1 Assumptions high doses can be prescribed with little risk for
toxicity, such as penicillin [5]. Instead, drugs
The concept of therapeutic drug monitoring to with narrow therapeutic ranges and thus higher
maintain efficacious and safe plasma or serum risk of toxicity or undertreatment, such as lith-
concentrations relies on two primary assump- ium, cyclosporine, and aminoglycoside antibiot-
tions. First, TDM requires a definable relation- ics, are often indicated for TDM to ensure patient
ship between dose and plasma or serum drug serum concentrations are maintained within a
concentration [3]. There may be individual predetermined window and to inform changes to
patient factors or behaviors that influence this dose regimens if necessary. Although TDM
relationship, including drug formulation, drug assumes a clear dose-concentration and
interactions, environmental factors, genetic vari- concentration-­ effect relationship, specific drug
ation, and renal and hepatic function [2]; how- characteristics further impact its usefulness.
ever, the dose must somewhat predict plasma
concentration. If a medication does not adhere to
this assumption, then physicians would be less 3.1.2 Process of Therapeutic Drug
equipped to predict how a change in a dosing Monitoring
regimen would affect patient plasma concentra-
tions, undermining the process of therapeutic Therapeutic drug monitoring can begin immedi-
drug monitoring. ately following diagnosis and selection of a drug
In addition to a defined dose-blood concentra- or when an alteration in drug regimen is required
tion relationship, the concept of therapeutic drug at a later stage of treatment. First, the physician
monitoring relies on the assumption that a serum must determine if the drug is an appropriate can-
or plasma concentrations can predict therapeutic didate for TDM. Next, the normal range for the
or toxic effects [3]. In other words, a predeter- drug’s concentration must be determined. Buclin
mined therapeutic range, describing the blood et al. (2020) describe two types of percentiles that
plasma or serum concentration that is expected to can inform the appropriate range: population per-
achieve a therapeutic response while minimizing centiles, describing the range of concentrations
toxicity [4], is only valuable if therapeutic or expected for a given dose across the whole target
toxic effects can be somewhat predicted based on population, and a priori percentiles, which take
concentrations. It remains true that individual into consideration a given set of individual
patient factors may influence the concentration-­ covariate values that better match the characteris-
effect relationship, such as drug interactions, tics of the patient [6]. Given this information, the
electrolyte balance, acid-base balance, age, bac- medical team then designs a dosing regimen to
terial resistance, and protein binding [2]; how- reach the predetermined window of plasma con-
ever, a defined relationship must exist to ensure centrations deemed appropriate for the specific
therapeutic ranges are not arbitrary and to maxi- patient. After administration of the drug, plasma
mize the utility of therapeutic drug monitoring. or serum concentrations are measured and clini-
Although these assumptions apply to several cal assessments are performed to inform whether
drugs, Aronson and Hardman (1992) describe adjustments are necessary [3]. Clinical judgment
specific criteria that decrease the utility of thera- and proper interpretation are necessary to deter-
peutic drug monitoring: drug metabolism to mine how to adjust the dose regimen to bring
active metabolites and a low toxic to therapeutic concentrations closer to the target.
ratio [2]. Drugs that produce active metabolites
require plasma measurement of both the drug and
3 Plasma Therapeutic Drug Monitoring and Clinical Toxicology 23

3.2 Indications for Measuring Poor adherence may be indicated by a plasma


Plasma Concentrations concentration that is unlikely to be associated
with the prescribed dose [3]. In addition, physi-
Although the primary purpose of TDM is to max- cians can use previous measurements to guide the
imize efficacy and safety, other indications interpretation of plasma concentrations. If poor
include individualizing therapy, diagnosing tox- patient adherence is suspected, as opposed to
icity or undertreatment, monitoring adherence undertreatment, then physicians must implement
and drug interactions, prophylaxis, and guiding strategies to improve medication adherence, such
withdrawal of therapy [7]. as educating and empowering patients, reducing
the barriers to obtaining and taking medication,
improving communication during office visits,
3.2.1 Avoiding or Diagnosing and understanding the underlying causes of non-
Toxicity adherence [8].

Therapeutic drug monitoring is not only utilized


to prevent toxicity, but to aid the interpretation of 3.2.3 Prophylaxis
instances in which toxic effects are difficult to
distinguish from the effect of the disease itself. Whereas some drugs, such as antimicrobials, are
For example, aminoglycoside nephrotoxicity prescribed for the therapeutic resolution of dis-
clinically mimics that of generalized infection. In ease or illness, prophylactic drugs are adminis-
such cases, therapeutic drug monitoring of serum tered to prevent the onset of disease or illness.
or plasma concentrations may better inform the For example, lithium is often prescribed to
cause of clinical symptoms, whether it be toxicity patients with affective disorders to prevent
or infection [3]. This insight is necessary to manic-depressive attacks [9]. Furthermore,
appropriately determine how to proceed with cyclosporine can be prescribed to transplant
treatment. patients to suppress the immune system and pre-
vent transplant rejection. As these specific indica-
tions of lithium and cyclosporine use are
3.2.2 Diagnosing Undertreatment prophylactic, physicians are unable to monitor
and Monitoring Patient clinical response. Instead, patients can undergo
Adherence therapeutic drug monitoring to ensure plasma or
serum concentrations indicate therapeutic benefit
Just as high plasma drug concentrations are with minimized risk for toxicity. However, in
avoided to minimize the risk of toxicity, low such cases, physicians must continue to “treat the
plasma drug concentrations must be corrected as patient, not the plasma drug concentration” [7];
the patient would otherwise experience subthera- whereas the plasma drug concentration can sug-
peutic effects. Low plasma concentrations can be gest therapeutic benefits or risk for toxicity, indi-
indicative of undertreatment or low patient adher- vidual patient factors must continue to be
ence, as the therapeutic benefit of many medica- implemented into prophylactic and treatment
tions is strongly dependent on patient adherence strategies.
to dosage regimens [7]. As such, it is incredibly
important that physicians use clinical judgment
to determine how to correct for low plasma or 3.2.4 Drug Interactions
serum concentration. If the dosage is increased and Termination of Treatment
without any consideration of patient adherence,
the patient could potentially be at risk for future One aspect of individualizing therapy is the man-
toxicity. agement of drug interactions. When an interact-
ing drug is prescribed, such as for an unrelated
24 G. Fishberger et al.

condition or by a different provider, measure- surements regarding a patient’s plasma drug con-
ment of plasma concentrations can inform drug centration. Following the onset of treatment,
regimen alterations to avoid unanticipated toxic- plasma concentration values should be measured
ity or undertreatment. This is especially relevant once the drug has reached a steady state concen-
for interacting drugs that impact hepatic or renal tration [11]. Steady state occurs once equilibrium
metabolism. For example, rifampin, an enzyme has been established between administration and
inducer, increases the clearance of cyclosporine, elimination of the drug, which takes place after at
which may put patients receiving cyclosporine least five half-lives, when the drug is eliminated
and rifampin at risk for undertreatment [10]. In by 94–97% [12]. This means that when patients
this case, therapeutic drug monitoring can better are placed on a continuous dosing schedule, the
ensure that the patient is receiving an adequate steady state occurs after approximately five
dosage of cyclosporine for therapeutic benefit, respective cycles of elimination and administra-
despite increased clearance. Lastly, TDM can be tion. Errors associated with improper timing of
used to guide the withdrawal of treatment, spe- sample collection can negatively affect the inter-
cifically when the plasma concentration is below pretation of plasma drug concentrations. For
therapeutic range in a patient with an acceptable example, when blood samples are collected pre-
clinical condition and when the plasma concen- maturely, plasma concentrations may be higher
tration is high with little therapeutic benefit [7]. than expected due to the reduced time available
In the latter case, the increased risk for toxicity at for tissue absorption. Most drug concentrations
high concentrations would likely outweigh the peak after 1 to 2 hours; however, in cases of
marginal therapeutic benefit, suggesting treat- delayed absorption or other distribution impair-
ment should be terminated and other options ments, the time at which the drug levels peak
explored. These two indications for withdrawal may be prolonged [3]. In most cases of therapeu-
of treatment depict the risk of analyzing plasma tic drug monitoring, samples are collected prior
or serum drug concentrations in isolation; to to the administration of the following dose, oth-
appropriately perform therapeutic drug monitor- erwise known as the trough period, when plasma
ing, one must utilize both plasma concentrations drug concentrations are at their lowest [11].
and clinical assessment to inform adjustments in Samples acquired during the trough period are
treatment. reflective of the drug’s elimination phase and
more accurately assess the steady state concen-
tration. Table 3.1 highlights the timing necessary
3.3 Proper Use of Measurements for measuring steady state concentrations for
both individual drugs and drug classes [7].
The timing of blood samples in therapeutic drug
monitoring (TDM) is critical, as inaccurate read-
ings can dramatically affect the interpretation of 3.3.2 Type of Sample Used
results and potentially diminish the value of the
measured plasma concentrations. Four factors are In addition to the proper timing of sample collec-
needed to ensure the accuracy and success of tion, the type of sample used has an important
TDM: timing of blood samples, types of samples effect on the validity and reliability of plasma
used, measurement technique, and individualiza- drug concentrations. For most drugs, the sample
tion of results [7]. should be allowed to clot in its tube, a process
called heparinization [3]. Prior to measurement,
there are no important storage restrictions for
3.3.1 Timing of Blood Samples most drug classes. Lithium and aminoglycosides
differ from most drugs, as they require heparin-
The adequate timing of blood sample collection ization followed by separation within 1 hour of
is important in providing clinically useful mea- collection [3]. Cyclosporine also differs as it
3 Plasma Therapeutic Drug Monitoring and Clinical Toxicology 25

Table 3.1 Variability in timing of serum drug measurement by drug class and route of administration
Drug/Drug Class Route of Administration Timing of Blood Sample
Aminoglycosides Intravascular (IV) Peak – 15 mins after infusion.
Trough – before next dose.
Intramuscular (IM) Peak – 1 hour after injection.
Trough –before next dose.
Cyclosporine PO, IV Before next dose, measure at same time of day.
Digoxin PO Approximately 6 hours after the last dose.
Lithium PO 12 hours after the last dose.
Phenytoin PO, IV Timing not applicable.
Theophylline During an infusion 4–6 hours after infusion starts; stop infusion for 15 min before sample.
Oral Before next dose, measure at the same time of day.

requires the consultation of local laboratory facil- falsely low values of digoxin upon immunoassay
ities for the necessary sampling and post-dosage [13]. This demonstrates the importance of ensur-
collection techniques [3]. Ensuring that the ing adequate immunoassay techniques, which
proper sample types and collection techniques includes the evaluation of possible interferences
are utilized in TDM is critical, as errors in these and mitigation of any associated adverse effects.
stages may adversely affect the laboratory assay
results and greatly reduce the value of plasma
drug concentrations when providing patient care. 3.3.4 Individualization of Results

Following sample collection and laboratory anal-


3.3.3 Measurement Technique ysis, the results of the immunoassays must be
interpreted in a way that is tailored to the patient’s
Following sample collection and processing, the status. Phenytoin is an example of a drug that
laboratory performs a specific drug assay to exhibits variation between individuals, especially
determine the plasma drug concentration. The those with comorbid conditions, and requires
current standard practice involves the use of close monitoring of plasma concentrations as a
immunobinding assay procedures, most com- result. The recommended dosage of phenytoin is
monly fluorescence polarization immunoassay 4–9 μmol/L, which is calculated based on the
(FPIA), enzyme immunoassay (EMIT), and ideal plasma concentration of 40–80 μmol/L and
enzyme-linked immunosorbent assay (ELISA) the high affinity for binding albumin, as only
[3]. These assays should be performed within 10% is unbound in the blood [14]. However,
24 hours, especially when determining dosage comorbid conditions that increase drug metabo-
adjustments or possible toxicity, as changes need lism, alter albumin binding, or decrease albumin
to be made quickly in both scenarios to avoid concentrations can lead to larger unbound con-
complications [7]. The specificity of these assay centrations of phenytoin, which alters the ideal
methods contributes to their high accuracy; how- plasma concentration range. One patient with
ever, there may be potential interferences associ- chronic renal failure was treated with 300 mg of
ated with cross-reactivity between the assay and phenytoin per day for tonic-clonic seizures and
the drug’s metabolites, or the assay and structur- was found to be asymptomatic 6 weeks later with
ally similar compounds [3]. These cross reactions a plasma concentration of 30 μmol/L, which is
may falsely increase or decrease the drug’s below the target range for phenytoin [14]. The
plasma concentration, ultimately affecting the clinician increased the dosage to 400 mg per day
reliability of the immunoassay’s results. For to fall within the recommended therapeutic
example, digoxin toxicity has been discovered range; however, 2 weeks later the patient exhib-
when patients were co-administered spironolac- ited mental dullness and difficulty walking, both
tone or canrenone, as these two drugs led to being adverse effects associated with high
26 G. Fishberger et al.

p­henytoin plasma concentrations [14]. The properties of a given drug affect individuals by
patient’s plasma concentration was found to be treating disease states, by producing adverse side
60 μmol/L, and the clinician subsequently effects, and by producing tolerance [15]. On a
reduced the dose back to 300 mg per day, after broader scale, pharmacogenomics, defined as the
which the patient returned to an asymptomatic effect of an individual’s genetic composition on
state [14]. This patient displayed drug toxicity at response to medications, has become increas-
a plasma concentration within the reference ingly recognized as a critical part of drug dosing
range, ultimately due to the concurrent renal fail- [16]. Essentially, it is the pharmacodynamic,
ure altering the characteristics of phenytoin’s pharmacokinetic, and pharmacogenomic aspects
absorption and metabolism. The patient’s posi- of drug metabolism that impact drug therapeutic
tive response at a lower therapeutic index indi- range and efficacy.
cates the need for repeated plasma monitoring,
especially when altering the dosage regimen in
the face of comorbid conditions. 3.4.1 Formulation and Diet
Although phenytoin is just one example of a
drug with varying effects between patients on the Drug absorption is described is the process by
same dosage regimen, this case demonstrates the which drugs enter the systemic circulation, which
importance of combining the immunoassay requires the passage through cell membranes.
results with the patient’s full clinical picture to Passage through cell membranes is affected by
provide a more precise therapeutic range, regard- molecular shape and size, as well as the solubility
less of the drug prescribed. In addition, plasma of the drug in water defined by the lipid-water
drug concentrations should not be used in isola- partition coefficient [17]. Highly lipid-soluble
tion, as other therapeutic indices, such as protein drugs rapidly cross cell membranes without the
binding ability in this scenario, are equally need for molecular transport proteins, whereas
important when determining the dosage regimen drugs with a low lipid-water partition coefficient
necessary to achieve a desired effect. require a protein carrier [18]. Thiopental is an
Overall, therapeutic drug monitoring is a use- example of highly lipid-soluble barbiturate used
ful tool when evaluating a patient’s response to a for rapid intravenous general anesthesia. The
drug and determining plasma concentrations in drug rapidly crosses the blood-brain barrier due
comparison to a desired therapeutic range. to its high lipid solubility [19]. When a drug
Through the proper collection techniques, sam- administered orally is absorbed through the gas-
ple types, laboratory evaluation, and individual- trointestinal tract through the hepatic portal sys-
ization of results, TDM conveys a considerable tem and undergoes biotransformation in the liver,
benefit for clinicians in providing patient care, the drug is converted to a more water-soluble
especially when used among other therapeutic metabolite [20]. For drugs, such as thiopental, for
monitoring techniques. which their high lipid solubility is critical in dis-
tribution and mechanism of action, parenteral
administration would be preferred.
3.4 Individualizing Therapy With lipophilic drugs, a patient’s body mass
index (BMI) and obesity status is highly relevant.
It’s well known that for a given dose of a drug, Individuals with a greater degree of adipose tis-
the physiologic impact varies greatly between sue tend to retain lipid-soluble drugs, thereby
individuals of both the same and different ethnic reducing drug elimination. Halothane is an
and geographical backgrounds. Variation exists inhaled general anesthetic that deposits in adi-
in terms of the pharmacokinetics of a given drug, pose tissue and undergoes reduced hepatic
in that an individual may absorb the drug more metabolism in obese individuals and increases
readily, distribute it more rapidly, or eliminate it the risk of hepatitis [21]. Halothane additionally
more quickly [3]. Further, the pharmacodynamic has a high blood-gas partition coefficient, which
3 Plasma Therapeutic Drug Monitoring and Clinical Toxicology 27

indicates that it binds to plasma proteins (such as is of high clinical relevance as we begin to dis-
albumin) in blood quite readily [22]. cuss cytochrome P450 enzymes and variations in
functionality of these enzymes between
individuals.
3.4.2 Pharmacokinetics: Ion
Trapping
3.4.4 Cytochrome P450 Enzymes
The unionized form of a drug readily crosses cell
membranes; however, the ionized form becomes Cytochrome P450 (CYP) oxidases consist of a
trapped and is unable to cross membranes. For family of more than 50 heme-dependent enzymes
example, a weakly acidic drug in the plasma will that play a key role in hormone synthesis but
become trapped on the interstitial side if that sidemore importantly in xenobiotic and drug metabo-
has a higher pH due to deprotonation of the drug lism [29]. CYP enzymes are found in the smooth
[23]. Acetazolamide is a weak diuretic that is endoplasmic reticulum of cells and are most
used to treat glaucoma, epilepsy, altitude sick- prominent in the liver, emphasizing the essential
ness, and fluid retention [24]. The drug also role of the liver in drug metabolism and clear-
decreases the plasma pH producing a mild meta- ance. CYP enzymes metabolize approximately
bolic acidosis. Doxorubicin is a chemotherapeu- 90% of drugs. Many drugs exist as prodrugs,
tic drug that is a weak base and is prone to ion such that the oral form of a given drug must be
trapping in the presence of low pH environments. bioactivated by CYP enzyme(s) into its active
Consequently, concomitant use of acetazolamide functional form. For example, prasugrel is an
and doxorubicin would lead to elevated concen- antiplatelet drug that targets the P2Y12 receptor
trations of doxorubicin and increase the risk of on the platelet and is useful in the treatment and
doxorubicin toxicity [25]. management of acute coronary syndrome.
Prasugrel is converted by esterases to an inactive
form that is subsequently primarily converted by
3.4.3 Pathologic Disease States CYP3A enzymes and CYP2B6 to its final active
form [30].
Drugs typically undergo renal and/or hepatic Individuals possess pharmacogenetic varia-
clearance. Therefore, the physiologic status of tion in CYP enzymes alleles such that for a given
these organs may have a direct impact on a drug’s CYP enzyme, an individual may metabolize the
systemic concentration and biological effects. drug at a more rapid or a slow rate. Clopidogrel
Metformin is frequently prescribed for type 2 has the same function as prasugrel but is metabo-
diabetes mellitus and is a hydrophilic drug that is lized differently. Clopidogrel is bioactivated
excreted unchanged by the kidneys [26]. At high mainly by CYP2C19 from a prodrug form into a
doses of metformin, there is a risk of life-­ primary metabolite, which is subsequently acted
threatening lactic acidosis, which may result upon again by CYP2C19 to generate a second
from drug accumulation in instances of severe metabolite that is the true active form of the drug
renal impairment [27]. Therefore, in individuals [31]. CYP2C19 has two key polymorphisms that
with chronic kidney disease (CKD), metformin influence drug serum concentrations and clopido-
dosing must be carefully monitored and is con- grel activity. CYP2C19*2 produces a loss-of-­
traindicated in late-stage CKD when the esti- function polymorphism such that the less of the
mated glomerular filtration rate is <30 mL/ active form of clopidogrel is produced. In a com-
min/1.73m2 [28]. In the context of an individual plete homozygous case of CYP2C19*2, individ-
with hepatic cirrhosis when liver function is uals should not be given clopidogrel; however, in
severely reduced, the dosage of drugs undergoing the heterozygous case, a higher dosage would
hepatic modification or clearance may require need to be administered to achieve the desired
adjustment. Hepatic biotransformation of drugs drug effect. This polymorphism is present in up
28 G. Fishberger et al.

to 70% of Asian individuals. Conversely, that worsen a syndrome or disease, drugs with
CYP2C19*17 results in increased transcription cautioned used, significant drug-drug interac-
of CYP2C19, thus producing more active clopi- tions to be avoided in older adults, drugs with
dogrel and elevated drug potency for a given strong anticholinergic effects, and drugs that are
dose. Administration of clopidogrel to individu- contraindicated or require reduced dosage for
als with the CYP2C19*17 polymorphism would patients with reduced kidney function.
require reduced amounts of the drug to achieve
the desired drug effect.
Certain drugs can induce or inhibit the activity 3.5 Serum Versus Plasma
of CYP enzymes that metabolize a given drug Monitoring
resulting in reduced or elevated serum concentra-
tions of the latter drug, respectively. Warfarin is a Pioneer scientist and clinicians of drug monitor-
popular anticoagulant used in the prophylaxis ing demonstrated that incidence of toxicity could
and treatment of deep vein thrombosis, pulmo- be reduced to drugs such as digoxin, phenytoin,
nary embolism, and atrial fibrillation [32]. The lithium, and theophylline when patients are mon-
drug is inactivated by CYP2C9. Therefore, inhi- itored within a therapeutic range [37]. This is a
bition of CYP2C9 increases serum concentra- practice mainly utilized for drugs that have nar-
tions of warfarin and consequently increases the row therapeutic indexes, variable pharmacoki-
risk of severe bleeding [29]. Given that the anti- netic properties, and adverse side effects [3].
microbial metronidazole has been shown to Drug monitoring consists of extracting various
inhibit CYP2C9, it would be necessary to reduce blood components to measure the drug concen-
the dosage of warfarin and carefully monitor the trations within them and then comparing the con-
patient for both external and internal bleeding if centration to clinical parameters [3]. Several key
concomitant use of the drugs is necessary. factors are important to pay attention to such as
the timing of the blood sample and the type of
blood sample used.
3.4.5 Age Components within blood are distinct as well
as the processing after the blood draw. Whole
The guidelines, contraindications, and dosage for blood should be used if the compounds are con-
medications vary depending on an individual’s centrated in the erythrocytes such as lead, cya-
age. For infants and young children, drugs may nide, mercury, carbon monoxide, and
be more potent than desired due to incomplete chlorthalidone [41] or if the binding protein is on
development of the physiological systems that the erythrocyte such as with many immunosup-
metabolize medications [33]. However, in older pressants (e.g., cyclosporin, tacrolimus, azathio-
children, other drugs may be less active for a prine) [38, 39]. Whole blood that has been
drug dosed by weight because the liver and CYP centrifuged becomes plasma and mainly consists
enzymes develop more quickly than the relative of fibrinogen, clotting factors, plasma proteins
increase in body mass [34]. In older and elderly (e.g., albumin), electrolytes (e.g., sodium, potas-
patients, the activity of drug-metabolizing sium, bicarbonate, chloride, calcium), and immu-
enzymes as well as kidney function declines with noglobulins. The chief difference in processing
age [35]. Beers Criteria for potentially inappro- between plasma and serum is that anticoagulants
priate medication use in older adults was first are added to plasma, whereas coagulation
developed in 1991 to provide recommendations enhancers are added to serum [40]. These coagu-
on prescription drug practices for adults 65 years lation enhancers facilitate clotting and separation
and older. The goal of the guidelines is to reduce of erythrocytes via centrifuge. Therefore, serum
the risk to benefit ratio and reduce drug interac- contains no fibrinogen or clotting factors.
tions that result from polypharmacy in older Frequently, serum and plasma are used
adults [36]. Drugs to be avoided include those
3 Plasma Therapeutic Drug Monitoring and Clinical Toxicology 29

i­nterchangeably, and reasons are not stated as to toms despite therapeutic doses of ACE inhibitors
why one is used over the other. and diuretics. Digoxin can be used chronically as
One of the advantages to using plasma is that a palliative medication or until a higher interven-
plasma has a larger volume. Because the yield for tional step can be implemented [44]. However,
plasma is higher with about 50% of the original utility of the drug is associated with adverse
sample versus 30–50% for serum, a greater drug effects and a narrow therapeutic window. In addi-
amount may generate a more accurate drug anal- tion, there are variable associations between clin-
ysis. Another advantage is that there is no delay ical symptoms, plasma concentration, and
from the long duration of the clotting process. therapeutic/toxic responses that make digoxin
Clotting can occur even after centrifugation if unpredictable and therefore further necessitate
enough time is not allotted. There will also be a plasma monitoring. There is a higher correlation
decreased risk of hemolysis. When erythrocytes between clinical signs and therapeutic/toxic
lyse, they increase the plasma concentration. The effects of digoxin in treating atrial fibrillation
effect will be more pronounced in serum [41]. with a fast ventricular rate. Typically, the slowing
Some of the disadvantages to using plasma are of the ventricular rate is a good indicator that
related to the effect of anticoagulants on the sam- digoxin is having corrective effects. On the other
ple. Anticoagulants can have a variable effect on hand, it’s more difficult to measure the therapeu-
the free concentration of the drug. For example, tic response for patients with congestive heart
anticoagulants can increase lipolytic activity, failure due to overlapping symptoms between
resulting in an increase in non-esterified fatty congestive heart failure and digoxin toxicity such
acid that often displaces phenytoin. Phenytoin as anorexia, nausea and vomiting, confusion, and
will then concentrate in erythrocytes and decrease cardiac arrhythmias. While evidence shows
the plasma concentration of phenytoin compared increasing the dosage of digoxin within the thera-
to the original. On the other hand, ibuprofen con- peutic window produces an increasing therapeu-
centration can increase in the plasma with hepa- tic effect when treating atrial fibrillation, there is
rin usage because anticoagulants can disrupt not an established one for treating heart failure.
protein binding. Some other potential disadvan- The therapeutic effect is generally seen between
tages are associated with the additives or impuri- 1 and 3.8 nmol/l with the risk of digoxin toxicity
ties found within the anticoagulants such as zinc, starting to increase above 2.6 nmol/l [45]. The
lead, aluminum, copper, and fluoride. They can recommended range for plasma digoxin concen-
falsely elevate the concentrations of these ele- tration has decreased over time due to evidence
ments in the plasma [41]. In conclusion, while of better outcomes with lower dosages [46, 47].
there are some advantages and disadvantages to Demographics of patients who are more prone to
utilizing plasma and serum, the concentrations digoxin toxicity are the elderly, especially
overall are similar. The differences in whether patients who have poor kidney perfusion [42].
one is used over the other may depend more on
the hospital system.
3.5.2 Drugs Requiring Monitoring:
Phenytoin
3.5.1 Drugs Requiring Monitoring:
Digoxin Phenytoin is an anticonvulsant medication used
in the management of epilepsy, generalized tonic-­
Digoxin is one of the oldest cardiovascular medi- clonic seizures, complex partial seizures, and sta-
cations, having been used for over 200 years to tus epilepticus [48]. Phenytoin is another drug
treat atrial fibrillation with fast ventricular rate that has a narrow therapeutic window between 40
and congestive heart failure with sinus rhythm and 80 μmol/L with mainly neurological toxici-
[42, 43]. They are used in patients with later stage ties occurring above 100 μmol/L. Binding sites
systolic heart failure that have recurrent symp- become saturated at relatively low doses and
30 G. Fishberger et al.

therefore any increase in dosage beyond the satu- 3.6 Renal Clearance
ration point rapidly increases the plasma concen- and Metabolism
tration [14]. Furthermore, there is also a large in Therapeutic Drug
variation in the dosage taken by patients and the Monitoring
resulting plasma concentration. Individual differ-
ences in hepatic enzyme system cytochrome Another consideration when it comes to thera-
P450 (CYP), predominantly CYP2C9 and peutic drug monitoring is renal clearance and
CYP2C19 along with other drug interferences metabolism. Many disease states, medications,
with this system, can contribute to the discrep- and individual variations contribute to differ-
ancy between the dosage given to patients and the ences in renal clearance among patients. It is
resulting plasma concentration. For example, imperative to take these variations into account
drugs that inhibit CYP are cimetidine, amioda- when monitoring drug concentrations for differ-
rone, allopurinol, azapropazone, chlorpromazine, ent patient populations to ensure proper thera-
imipramine, isoniazid, metronidazole, flucon- peutic effects and avoid toxicity. This section will
azole, omeprazole, sulfonamides, thioridazine discuss therapeutic drug monitoring regarding
[48], and valproic acid [49], leading to an increase renally eliminated drugs in multiple patient pop-
in plasma phenytoin. Conversely, medications ulations with altered renal clearance including
that induce CYP are alcohol, rifampin, carbam- critically ill individuals, the geriatric population,
azepine, barbiturates, theophylline, etc., leading the pediatric population, and individuals with
to an increase in phenytoin breakdown and there- acute or chronic renal failure.
fore a decrease in its plasma concentration [50].
Medical conditions that cause hypoalbuminemia,
such as chronic liver disease, nephrotic syn- 3.6.1 Critically Ill Patients
drome, and pregnancy, will additionally increase
plasma concentration due to a decrease in albu- Critically ill patients may experience a phenome-
min binding sites. Approximately 90% of the non called augmented renal clearance in which they
time, phenytoin is bound to albumin and is inac- have elevated creatinine clearance and thus
tive as a result [51]. Therefore, monitoring the increased renal elimination of renally cleared drugs.
plasma concentration is important to ensure the This phenomenon is especially prevalent in indi-
availability of phenytoin stays within the thera- viduals with neurologic injury, sepsis, trauma, and
peutic range for sufficient anti-seizure effects and burns [52]. While the normal range of creatinine
avoidance of neurological toxicities with higher clearance is 110 to 150 mL/min in males and 100 to
concentrations. 130 mL/min in females, creatinine clearance in
Phenytoin therapeutic effects can be well patients with these conditions averages from
demarcated by clinical evidence for patients with 170 mL/min to more than 300 mL/min [53].
more severe seizure conditions but may not be as Specifically, neurologic injury, sepsis, trauma,
straightforward with patients that have milder burns, and interventions such as vasopressor use
symptoms or are taking phenytoin prophylacti- and fluid resuscitation in critically ill individuals
cally for surgery. On the other hand, phenytoin’s increase renal blood flow, resulting in elevated renal
toxic effects are also insidious and difficult to clearance [54]. Augmented renal clearance may
distinguish from symptoms of other neurological contribute to subtherapeutic drug concentrations in
conditions. In many of these cases, the measure- critically ill patients, especially regarding most anti-
ment of plasma concentration within the thera- microbials, e.g., beta-lactams, vancomycin, and
peutic range can serve as an adequate marker for carbapenems due to their renal mechanism of clear-
its efficacy [2]. ance and standard use in septic patients [52].
3 Plasma Therapeutic Drug Monitoring and Clinical Toxicology 31

Though augmented renal clearance is a well-­


documented phenomenon in many critically ill
patients, it is also important to remember that
many critically ill patients may also suffer from
acute kidney injury (AKI) which may reduce
GFR and subsequently renal drug clearance.
Therapeutic drug monitoring in patients with AKI
will be further discussed later in this section.
In terms of therapeutic drug monitoring, it has
been deemed to be most useful in monitoring
renally cleared antimicrobials, namely, amino-
glycosides and beta-lactams, in critically ill
patients as opposed to other types of medications
in this population [55]. The bactericidal effects of
aminoglycosides, specifically, are dependent on
its concentration in the plasma. It has been rec-
ommended that therapeutic drug monitoring be
performed in critically ill patients on aminogly- Fig. 3.1 Concentration-time profile commonly used in
cosides utilizing a concentration-time profile therapeutic drug monitoring
(Fig. 3.1) and the area under the concentration-­
time curve (AUC) in (mmol/L) x min divided by
the minimum inhibitory concentration (MIC) in 3.6.2 Geriatric Patients
mmol [56]. The maximum concentration of ami-
noglycoside in the plasma and the renal elimina- Though glomerular filtration rate (GFR) and thus
tion half-life can be used to approximate the AUC renal function and clearance are generally
with mathematical modeling [57]. For critically believed to decline with age, studies have shown
ill patients, the target AUC/MIC was determined that about one-third of elderly individuals do not
to be approximately 80–100 as opposed to 30–50 experience a decline in renal function. Two-thirds
for non-critically ill patients [58]. Multiple days of elderly individuals do experience an age-­
of prolonged elevated AUC values have corre- related decline in renal function and clearance,
lated with increased ototoxicity and nephrotoxic- though these changes seem to correlate with
ity in critically ill patients though this population other comorbidities such as cardiovascular dis-
may require a higher initial single dose as well as ease [61]. Individuals affected by an age-related
an extended interval of administration due to decline in renal function and clearance may be
augmented renal clearance [55]. For beta-­ more susceptible to supratherapeutic drug con-
lactams, therapeutic drug monitoring for criti- centrations and adverse drug events due to
cally ill patients is also recommended to optimize decreased renal elimination. Despite potential
dosing and reduce the chances of toxicity [59]. differences in renal clearance in comparison to
Currently, the minimum concentration at steady the general population, therapeutic drug monitor-
state in a dosing interval, or trough sample, is the ing is not routinely used by default in this popula-
main way in which beta-lactam concentrations tion unless there is a proven deficit in renal
are monitored in the intensive care unit, but more function [62].
accurate methods and models are currently
explored to improve the monitoring of beta-­
lactams. The goal trough concentration for beta-­ 3.6.3 Pediatric Patients
lactams is equivalent to each antimicrobial’s
respective minimum inhibitory concentration It is difficult to pinpoint renal function and clear-
[60]. ance trends in the pediatric population due to the
32 G. Fishberger et al.

numerous metabolic changes in infancy and early venient method of monitoring mycophenolate
childhood. In general, metabolic clearance is mofetil concentrations in children may be neces-
very low in the newborn but increases rapidly to sary. In addition, data shows that total immuno-
levels greater than metabolic clearance in adults suppressant monitoring in addition to
in early childhood [63]. Later childhood and ado- mycophenolate mofetil monitoring may be more
lescent stages display similar renal function pat- useful in promoting therapeutic effects in pediat-
terns to that of average adults [62]. ric transplant and nephrotic syndrome patients
Therapeutic drug monitoring is mainly uti- than sole mycophenolate mofetil monitoring
lized in the pediatric population for two specific [66].
types of medications: antiepileptic drugs and
immunosuppressants. The main renally cleared
antiepileptic drug utilized in pediatric popula- 3.6.4 Acute or Chronic Renal Failure
tions is levetiracetam [63]. Renal clearance is Patients
very high in children particularly from the ages
of 6 months to 6 years, requiring a dose per kilo- Regardless of an acute or chronic etiology,
gram per body weight of levetiracetam about patients with renal failure have severely decreased
30% greater at these ages relative to later child- renal function and clearance. This makes them
hood, adolescence, and adulthood [64]. Because more susceptible to supratherapeutic drug con-
of this variation in required dosage due to rapidly centrations, putting them at higher risk for toxic-
changing renal function during childhood, thera- ity. Many of these patients are on dialysis,
peutic drug monitoring may be useful in discov- however, and this further changes their pharma-
ering the best therapeutic dose of levetiracetam cokinetic profile. Drug clearance in dialysis
for individual children [62]. Therapeutic drug patients is affected by the membrane, dialysate
monitoring for levetiracetam utilizes trough sam- and its flow rate, duration of dialysis, and the
ples instantly before the next dose due to its short individual properties of the drug. High molecular
half-life of 6–8 hours, which may result in great weight, high protein affinity, and large volume of
variability in serum drug concentration if sam- distribution are all properties that decrease the
ples are taken at other time points [64]. The goal likelihood of clearance by renal replacement
trough concentration for levetiracetam is cur- therapy [68].
rently 12–46 mg/L as established by the Therapeutic drug monitoring plays an exten-
International League Against Epilepsy [65]. In sive role in the medical treatment of renal failure
terms of immunosuppressants in transplant and dialysis patients due to their respective
patients, mycophenolate mofetil is a commonly increased risks for toxicity and sometimes unpre-
used renally cleared immunosuppressant in the dictable drug reactions with dialysis [68]. The
pediatric population. Therapeutic drug monitor- antibiotic vancomycin can best illustrate this
ing of this drug has the potential to decrease the concept in these patient populations. Patients
likelihood of underexposure, which can improve with renal insufficiency have increased rates of
outcomes for transplant and nephrotic syndrome nephrotoxicity with vancomycin, and therapeutic
patients. The current mechanism of monitoring drug monitoring utilizing trough concentrations
for mycophenolate mofetil involves measuring along with mathematical models forecasting the
AUC, which was discussed previously regarding trough concentration based on dosing has been
aminoglycosides [66]. Simulations have shown shown to decrease the incidence of vancomycin-­
an optimal AUC of 30 to 60 (mmol/L) x min for associated nephrotoxicity in patients with poor
pediatric transplant patients to decrease the risk renal function [69]. In the case of dialysis
of acute rejection [67]. The AUC method, how- patients, vancomycin is typically administered in
ever, requires excessive sampling within a the last hour of dialysis because though low-flux
12-hour period, which may not be feasible for dialyzers have no pronounced effect on vanco-
children; thus, an equally reliable but more con- mycin clearance, high-flux dialyzers may elimi-
3 Plasma Therapeutic Drug Monitoring and Clinical Toxicology 33

nate up to 40% of the vancomycin starting dose new evidence suggesting it is a prominent source
during dialysis [70]. Because of this, therapeutic of liver regeneration in settings with or without
drug monitoring with trough concentrations is liver injury likely due to hepcidin antimicrobial
considered a necessity in dialysis patients to peptide 2 (Hamp2) gene expression [75].
ensure proper drug concentrations are reached to The accepted model on liver clearance is the
treat the patient’s infection [71]. Traditionally, physiologically based pharmacokinetic (PBPK)
the goal trough concentration for vancomycin model by Rowland et al. The PBPK model is a
has been 15–20 mg/L, though there has been compartmental model but deviates from its clas-
increased nephrotoxicity associated with these sical predecessors in that the compartments are
concentrations with no proven improvement in equivalent to the actual organs and their volumes,
therapeutic outcome. A goal trough concentra- and it considers the sequential processing in the
tion of 10–15 mg/L for renal insufficiency and primary organ of metabolism, accounts for drug
dialysis patients may result in decreased inci- clearance in organs where secondary metabolites
dence of vancomycin-associated nephrotoxicity form, considers the differing kinetics between the
[72]. various primary and downstream metabolites,
and differentiates between the permeability and
transport of the different metabolites [76].
3.7 Role of Cytochrome P450 Current PBPK models account for the role of
Systems in Liver Clearance futile cycling in phase I and II reactions which
and Metabolism involve reduction/oxidation and conjugation/
deconjugation reactions, respectively [77]. There
The liver is the site of drug modification and is a combined PBPK model which accounts for
clearance. To appreciate the properties of the first-pass metabolism occurring within the intes-
drug and its downstream effects, understanding tinal and hepatic systems [76]. The traditional
the mechanics of the liver is valuable. For drugs model focuses on flow and volume of the intesti-
that are taken orally, it is the site of first-pass nal compartment; meanwhile the segregated flow
metabolism which can result in secondary activa- (SFM) model differentiates between serosal and
tion of the drug or removal. The heterogeneity of absorptive or enterocyte layers of the intestine
the liver can be attributed to its unique portal [76]. Majority of the blood flow is shunted to the
venous system, the biliary system, the protein serosa rather than the enterocyte layer [76]. This
interactions occurring at the hepatocyte and sinu- fact accounts for the variable intestinal and
soidal surfaces, and the variety of transporters hepatic processing which occurs when drugs are
and enzymes located intracellularly [73]. administered orally or intravenously since there
There is functional diversity in the liver lobule is minimal secondary metabolite formation asso-
given the zonal location. For instance, zone 1 is ciated with the latter [76]. Although there is less
known as the periportal zone and is sensitive to biochemical or phase I and II reactions involving
changes in oxygen and nutrients. This region the intestines compared to the liver, the dose-­
plays a role in carbohydrate, fat, and urea metab- dependent exposure of metabolites and drugs to
olism. This region is particularly vulnerable to the liver varies largely when drugs are dosed IV
oxidative stress [74]. In zone 3, which is closest or orally [77]. The first-pass effect by the liver
to the central vein and furthest from the portal changes when the intestines are summed into the
triad, there is increased expression of cytochrome equation [76]. Even if the intestines play a minute
P450 enzymes which has a wide range of bio- role in the overall metabolic processing, the
chemical activities especially drug metabolism, organ is the gateway to downstream liver
in addition to bile acid synthesis. This region is ­processing [76]. The PBPK intestinal and liver
more susceptible to ischemia [74]. Although model is becoming the more commonly employed
there is not much evidence highlighting the syn- approach for drug development and understand-
thetic or metabolic properties of zone 2, there is ing drug processing.
34 G. Fishberger et al.

3.7.1 Acetaminophen When APAP is dosed, the sulfation pathway


followed by the glucuronidation pathways
Acetaminophen (APAP) is metabolized by the becomes saturated [79]. Normally these path-
cytochrome P450 (CYP) system which generates ways are activated prior to NAPQI formation
N-acetyl-p-benzoquinone imine (NAPQI), an [79]. Toxic doses of APAP can cause hepatic
electrophile, that reacts with glutathione (GSH) damage which limit sulfation and glucuronida-
and other proteins (particularly in the mitochon- tion [79]. Sulfotransferases (SULT) increase the
dria) [78]. The NAPQI-GSH conjugate is how propensity of APAP elimination by increasing its
APAP is detoxified from the liver [78]. Other hydrophilicity [79]. The primary sulfotransfer-
metabolic reactions include sulfonation and gluc- ases are mediated by SULT2A1 and SULT1A1
uronidation which are also hepatoprotective [78]. [79]. Glucuronidation is mediated by UDP-­
To a lesser degree, there is oxidation of APAP to glucuronosyltransferases (UGT), particularly by
3-OH-APAP [78]. The main CYP enzymes which UGT1A9 and UGT1A1, at normal or toxic doses,
metabolize APAP are CYP2E1, CYP1A2, and UGT1A6 at lower doses [79].
CYP3A4, and CYP2A6 [78]. CYP3A4 more N-Acetylcysteine (NAC) treatment of APAP
commonly bioactivates APAP and generates the overdose increases flux to the SULTs to limit pro-
toxic NAPQI metabolite [78]. In organisms duction of the NAPQI and promote elimination
where CYP3A4 completed the greatest bioactiva- of APAP and its conjugates [79].
tion, there were fewer sulfonate and glucuronide
conjugates [78]. CYP3A4-specific inhibitors like
troleandomycin did not show a reduction in 3.7.2 Rifampin
NAPQI production [78]. When adding GSH there
was some enhancement of CYP3A4 activity with Rifampin is an antituberculosis medication that is
certain substrates [78]. These specific drug-drug a notable cytochrome P450, i.e., CYP3A4,
interactions suggest that there is possible alloste- inducer. Given this property, it enhances warfarin
ric modulation involved in CYP3A4 activity. and benzodiazepine metabolism. In addition,
CYP2E1 was a significant activator at low doses there is induction of P-glycoprotein (P-gp) activ-
of APAP; meanwhile CYP1A2 does so at high ity which can also reduce bioavailability and
doses of APAP [78]. mitigate intestinal absorption and lower plasma
In addition to the cytochrome P450 system, concentrations [81]. Hence, drugs which are
there are other adjacent reactions occurring in the metabolized by the cytochrome P450 system
liver such as conjugation to proteins via the sulf- may be initially excreted by MDR1 proteins [81].
hydryl group and some GSH depletion [79]. The Without considering this relationship, the role of
original presumption was that APAP-CYS cytochrome P450 metabolism in clearance or
adducts would be detectable only when hepato- bioactivation of certain drugs may be inflated.
cyte necrosis and cell death were occurring [80]. To monitor CYP3A4 activity and its expres-
It is now suggested that these adducts can be sion is normally done by measuring the area
present without alanine aminotransferase (ALT) under midazolam plasma concentration-time
elevations, suggesting that hepatic necrosis does curve (AUC), but a more accurate measure would
not concomitantly occur when APAP is conjugat- be to see increased CYP3A4 expression in the
ing to proteins. Protein adducts can be generated gastrointestinal and hepatic systems [82]. The
without significant GSH depletion [80]. administration of rifampin caused reductions of
Originally, it was assumed that major reductions midazolam by 75% [82]. This is mediated by the
in GSH would permit for an environment where activation of the nuclear pregnane X receptor
protein adducts could be created. APAP-CYS (PXR). CYP3A4 enzymes were more commonly
adducts form regardless of whether low or high present in the intestines than in the liver, although
concentrations of APAP are administered [80]. it was found to be that the cytochrome P450 sys-
3 Plasma Therapeutic Drug Monitoring and Clinical Toxicology 35

tem in the liver was rate-limiting in terms of major differences in turnover which can likely be
CYP3A4 activation and deactivation [83]. due to minimal allelic differences [85]. It is dif-
It is important to consider the role of hepato- ficult to isolate the role of competitive inhibition
toxicity after oral rifampin administration given given that MBI is still at play. The role of MBI
there are elevations in alanine aminotransferase was so significant between certain CYP3A4 phe-
(ALT) as seen in mice studies [84]. In addition, notypes and variants that inhibitory potencies,
there were increases in bilirubin, LDL, and total K1, had upwards of six-fold difference [85].
cholesterol. Alkaline phosphatase (ALP) concen- With respect to CYP3A4 inhibition, studies
trations did not increase [84]. The elevations in were completed to investigate the role of erythro-
lipid concentrations are associated with increased mycin with biopterin administration. When
expression of peroxisome proliferator-activated erythromycin was administered, the AUC of
receptor-γ (PPARγ) and upregulation can also be biopterin was doubled [86]. The inhibitory role
induced by rifampin [84]. Overall, this suggests of CYP3A4 increases the bioavailability of sub-
that activation of the PPARγ and CYP3A4 activ- strates but is unclear if it does so with CYP inhib-
ity can both contribute to hepatotoxicity follow- itors such as erythromycin. Erythromycin is
ing rifampin administration [84]. converted to N-desmethylerythromycin by the
enzymes and was present in equal quantities
when administered with biopterin, suggesting
3.7.3 Erythromycin that biopterin has little to no role in CYP3A4
metabolism [86]. Erythromycin relies on P-gp for
Erythromycin is a macrolide antibiotic and a transport and biopterin is a known P-gp inhibitor
known cytochrome P450 inhibitor. To mediate its [86]. Erythromycin increased after P-gp adminis-
effects on the CYP system, it relies on both com- tration, suggesting that this mechanism is the
petitive inhibition and mechanism-based inacti- likely explanation for elevated plasma concentra-
vation (MBI) where intermediates bind with the tions and not due to CYP3A4 inhibition, although
enzyme and impair reactions [85]. Given that the both mechanisms are pathways that play a role in
latter method occurs through covalent binding, clearance [86].
overall drug clearance does not regenerate the
enzyme due to the powerful bond created by the
intermediate [85]. Hence, there is allosteric inhi- 3.8 Antiretrovirals and Glucose
bition when these reactive metabolites are in Therapeutic Drug
generated. Monitoring
It’s important to consider the CYP3A4 vari-
ants present in patients. In particular, the changes 3.8.1 The Role and Influence
in hydrophobicity in the helical structure of these of Glucose in Therapeutic
enzymes are heavily responsible for substrate Drug Monitoring
interaction and changes in the Km [85]. The vari-
ants also have changes in their allosteric sites, as Glucose monitoring in diabetic patients is one of
well [85]. Certain substrates are required prior to the most common, routine forms of serum moni-
activation of the enzyme at the allosteric site first toring. Multiple variations of a “glucometer”
[85]. These substrates can be rate-limiting factors exist, ranging from test strips to continuous glu-
in terms of accessibility for the main substrates cose monitoring with feedback, also known as
required at the activation site. “closed-loop” systems. Common, at-home blood
Given that CYP3A4 enzymes are heavily glucose tests require a whole capillary blood
affected by MBI, the rate of degradation and syn- sample in which glucose reacts with either the
thesis of these enzymes influence bioactivation glucose oxidase, glucose dehydrogenase, or
and downstream reactions normally mediated by hexokinase enzymes, leading to a chemical reac-
this system [85]. In vitro studies have not shown tion [87]. A particular challenge with this mecha-
36 G. Fishberger et al.

nism is that it must be done quickly, as glucose potential impact on drug clearance in the afore-
concentrations in a whole blood sample decline mentioned systems.
with time due to erythrocyte glycolysis and con-
sumption of glucose.
Hemoglobin A1C (HbA1C) has long been 3.8.2 Therapeutic Drug Monitoring
shown to be a surrogate for blood glucose levels of Antiretroviral Drugs
over a 3-month period and is routinely measured
in diabetic patients. The two main methods for Human immunodeficiency virus, commonly
determining HbA1C are in-office point of care known as HIV, is a retrovirus spread through
tests (POCT) and cation exchange chromatogra- bodily fluids, leading to significant immune sys-
phy [88]. Interestingly, studies have shown that tem impairment and eventual acquired immuno-
hemoglobinopathies such as sickle cell (HbS) deficiency syndrome (AIDS). As a result, many
can influence the accuracy of POCT HbA1C [88, pharmaceuticals have been developed to combat
89]. HIV, colloquially known as “antiretrovirals.”
Many drugs are cleared within hepatocytes via Many forms of antiretrovirals exist, ranging from
the cytochrome P450 enzymatic system. Varied integrase, entry, and protease inhibitors, as well
subtypes have been shown to be significantly as analogs including nucleoside and non-­
upregulated or downregulated in response to nucleoside reverse transcriptase inhibitors (NRTI
either hypo- or hyperglycemia, in vitro [90]. and NNRTI, respectively).
Further, prolonged hyperglycemia in cultured Although very effective, many of these anti-
hepatocytes was shown to significantly increase retroviral drugs have been associated with hepa-
accumulation of lipids as well as exhibit insulin totoxicity [92]. Further, many of these drugs are
resistance. Current data suggest hepatic lipid associated with resistance in HIV-infected indi-
accumulation, commonly known as hepatic ste- viduals [93]. Cases have highlighted the benefi-
atosis or nonalcoholic fatty liver disease, can cial effects of therapeutic drug monitoring
change the pharmacokinetics and pharmacody- (TDM) in HIV-infected individuals with comor-
namics of hepatic drug metabolism. bid hepatic and renal failure [94]. As a result,
Elevated serum glucose concentrations can TDM of these compounds can be beneficial to
have significant effects on the pharmacokinetic determine both minimum effective concentration
properties of drugs cleared by the kidney. and potential dose-dependent toxicity. Many of
Diabetes mellitus (DM) is associated with these compounds are bound to plasma proteins
increased glomerular filtration rate (GFR) and in vivo and only contribute therapeutic and toxic
subsequent renal “hyperfiltration,” with eventual effects when free and unbound [95]. Despite this,
progression to chronic kidney disease (CKD) many of these compounds have notable toxicity
[91]. Renal clearance of a drug is dependent on profiles and thus highlight the benefit of routine
the urine flow rate and inversely related to the TDM.
plasma concentration of the said drug. As a result, Protease inhibitors, including ritonavir, have
a patient with uncontrolled DM may develop sig- long been associated with toxic side effects
nificant changes in their GFR and thus varying including hepatotoxicity [96]. Cases of
concentrations of renally cleared substances in dolutegravir-­induced neurotoxicity following
their serum as the disease progresses. This war- supratherapeutic dosing of the drug as well as
rants potential changes to drug dose in patients severe thrombocytopenia have been noted [97,
with varying stages of CKD. 98]. Interestingly, many studies exist regarding
Glucose plays a very large and active role in TDM and ritonavir, specifically in the pregnant
drug metabolism via detrimental effects on the population. Drug toxicity in pregnant patients is
architecture of the liver and kidney. Maintaining important due to the potential consequences of
blood glucose within the normal range is ideal, placental transfer of toxic compounds.
especially in diabetic patients, as it will limit the Dolutegravir has been shown to cross the pla-
3 Plasma Therapeutic Drug Monitoring and Clinical Toxicology 37

centa in an ex vivo placental model with a fetal-­ transaminase (ALT), aspartate transaminase
to-­maternal concentration ratio of 0.6, which is (AST), gamma-glutamyl transferase (GGT), as
considered relatively high compared to other well as creatinine and complete blood count
antiretrovirals [99]. Further, mouse models of (CBC). Routine hematologic monitoring is rec-
dolutegravir have noted increased rates of neural ommended for zidovudine, hepatic monitoring
tube defects at therapeutic, but not suprathera- for nevirapine, and renal monitoring for tenofovir
peutic, concentrations [100]. The aforementioned [104]. Given that many antiretrovirals are admin-
studies highlight the benefits of TDM in HIV-­ istered together in a regimen known as highly
infected individuals. active antiretroviral therapy (HAART), using
Currently, the most common published meth- routine laboratory markers is ideal and more cost
ods for determining serum concentrations of anti- effective.
retrovirals are via high-performance liquid
chromatography (HPLC), mass spectrometry, and
ultrafiltration. Pharmacokinetics of common anti- 3.9 Conclusion
retroviral drugs can be seen in Table 3.2. Integrase
inhibitors such as raltegravir, dolutegravir, and The ability to accurately monitor drug concentra-
elvitegravir concentrations can be determined via tions remains a critical component of patient
HPLC as well as tandem mass spectrometry in care. Knowledge of the pharmacokinetic proper-
both plasma and cerebrospinal fluid (CSF) sam- ties of a given drug allows for members of the
ples [101]. Ultrafiltration has been noted to be a care team to determine the serum concentration
plausible technique for determining unbound, of the drug and predict the effect the drug will
active plasma concentrations of dolutegravir, have on the individual. Given that the absorption,
raltegravir, and darunavir [95]. These methods are biotransformation, and clearance of particular
ideal in resource-rich countries with widespread drugs may have interpatient variation, treating
access to complex laboratory techniques. In patients based on their known comorbidities,
resource-poor regions, the use of dried blood characteristics, and genetic profiles will maxi-
spots has been shown to be an effective method mize the therapeutic safety and efficacy of their
for TDM of antiretroviral drugs [102]. medical treatment. Furthermore, knowledge of
Despite the aforementioned routes of TDM, how to properly administer and follow the con-
the most common and routine method of TDM in centrations of higher risk drugs with narrow ther-
HIV-infected individuals is via surrogate labora- apeutic windows requires special consideration
tory markers. Patients may receive routine moni- and pharmacist involvement. Plasma provides
toring for hepatic, renal, or hematologic insight into the often cryptic manner by which
abnormalities. Common markers include alanine drugs are metabolized and function.

Table 3.2 Pharmacokinetic variations in antiretroviral drugs


Drug Bioavailability Hours to peak serum concentration Half-life (hours)
Abacavir 50% protein bound 0.7–1.7 1.5
Darunavir 95% protein bound 2.5–4 15
Dolutegravir >98% protein bound 2–3 14
Maraviroc 70% protein bound 0.5–4 14–18
Raltegravir 83% protein bound 3 9
Ritonavir 98–99% protein bound 2 3–5
Tenofovir <7% protein bound 1–2 17
Adapted from Brody’s Human Pharmacology: Mechanism Based Therapeutics [103]
38 G. Fishberger et al.

References Indian J Exp Biol. 2001;39(10):955–61. https://


www.ncbi.nlm.nih.gov/pubmed/11883519
17. Fan J, de Lannoy IA. Pharmacokinetics. Biochem
1. Raber S, Simonson W. How the body manages thera-
Pharmacol. 2014;87(1):93–120. https://doi.
peutic drugs: AN introduction to Pharmacokinetics.
org/10.1016/j.bcp.2013.09.007.
Lab Med. 1997;28(9):558–91.
18. Kaur G, Grewal J, Jyoti K, Jain UK, Chandra R,
2. Aronson JK, Hardman M. ABC of monitoring drug
Madan J. Chapter 15 - Oral controlled and sus-
therapy. measuring plasma drug concentrations.
tained drug delivery systems: Concepts, advances,
BMJ. 1992;305(6861):1078–80.
preclinical, and clinical status. In: Grumezescu
3. Kang JS, Lee MH. Overview of therapeutic drug
AM, editor. Drug targeting and stimuli sensitive
monitoring. Korean J Intern Med. 2009;24(1):
drug delivery systems. William Andrew Publishing;
1–10.
2018. p. 567–626. https://doi.org/10.1016/
4. Yip DW, Gerriets V. Penicillin. [Updated 2021 Sep
B978-­0-­12-­813689-­8.00015-­X.
30]. In: StatPearls [Internet]. StatPearls Publishing;
19. Russo H, Bres J, Duboin MP, Roquefeuil
2021 Jan.. https://www.ncbi.nlm.nih.gov/books/
B. Pharmacokinetics of thiopental after single and
NBK554560/
multiple intravenous doses in critical care patients.
5. Cooney L, Loke YK, Golder S, et al. Overview of
Eur J Clin Pharmacol. 1995;49(1-2):127–137.
systematic reviews of therapeutic ranges: methodol-
https://doi.org/10.1007/BF00192371.
ogies and recommendations for practice. BMC Med
20. Phang-Lyn S, Llerena VA. Biochemistry, biotrans-
Res Methodol. 2017;17(1):84.
formation. In: StatPearls; 2021. https://www.ncbi.
6. Buclin T, Thoma Y, Widmer N, et al. The steps to
nlm.nih.gov/pubmed/31335073.
therapeutic drug monitoring: a structured approach
21. De Baerdemaeker LEC, Mortier EP, Struys
illustrated with Imatinib. Front Pharmacol.
MMRF. Pharmacokinetics in obese patients. Contin
2020;11:177.
Educ Anaesth Crit Care Pain. 2004;4(5):152–5.
7. Reynolds DJ, Aronson JK. ABC of monitoring drug
https://doi.org/10.1093/bjaceaccp/mkh042.
therapy. Making the most of plasma drug concentra-
22. Bergadano A, Lauber R, Zbinden A, Schatzmann U,
tion measurements. BMJ. 1993;306(6869):48–51.
Moens Y. Blood/gas partition coefficients of halo-
8. Neiman AB, Ruppar T, Ho M, et al. CDC grand
thane, isoflurane and sevoflurane in horse blood. Br J
rounds: improving medication adherence for
Anaesth. 2003;91(2):276–8. https://doi.org/10.1093/
chronic disease management – innovations and
bja/aeg151.
opportunities. MMWR Morb Mortal Wkly Rep.
23. Mahoney BP, Raghunand N, Baggett B, Gillies
2017;66:1248–1251.
RJ. Tumor acidity, ion trapping and chemothera-
9. Prien RJ. Lithium in the prophylactic treat-
peutics. I. Acid pH affects the distribution of che-
ment of affective disorders. Arch Gen Psychiatry.
motherapeutic agents in vitro. Biochem Pharmacol.
1979;36(8):847–8.
2003;66(7):1207–18. https://doi.org/10.1016/
10. Vandevelde C, Chang A, Andrews D, Riggs W,
s0006-­2952(03)00467-­2.
Jewesson P. Rifampin and Ansamycin interac-
24. Farzam K, Abdullah M. Acetazolamide. In:
tions with Cyclosporine after renal transplantation.
StatPearls; 2021. https://www.ncbi.nlm.nih.gov/
Pharmacotherapy. 1991;11(1):88–9.
pubmed/30335315.
11. Gross AS. Best practice in therapeutic drug moni-
25. Gieling RG, Parker CA, De Costa LA, Robertson N,
toring. Br J Clin Pharmacol. 1998;46(2):95–9.
Harris AL, Stratford IJ, Williams KJ. Inhibition of
https://doi.org/10.1046/j.1365-­2125.1998.00770.x.
carbonic anhydrase activity modifies the toxicity of
PubMed PMID: 9723816; PubMed Central PMCID:
doxorubicin and melphalan in tumour cells in vitro.
PMC1873661
J Enzyme Inhib Med Chem. 2013;28(2):360–9.
12. Hallare J, Gerriets V. Half Life. Treasure Island:
https://doi.org/10.3109/14756366.2012.736979.
StatPearls; 2021.
26. Hsu WH, Hsiao PJ, Lin PC, Chen SC, Lee MY, Shin
13. Steimer W, Muller C, Eber B. Digoxin assays: fre-
SJ. Effect of metformin on kidney function in patients
quent, substantial, and potentially dangerous inter-
with type 2 diabetes mellitus and moderate chronic
ference by spironolactone, canrenone, and other
kidney disease. Oncotarget. 2018;9(4):5416–23.
steroids. Clin Chem. 2002;48(3):507–16.
https://doi.org/10.18632/oncotarget.23387
14. Aronson JK, Hardman M, Reynolds DJ. ABC
27. Graham GG, Punt J, Arora M, Day RO, Doogue
of monitoring drug therapy. Phenytoin. BMJ.
MP, Duong JK, Furlong TJ, Greenfield JR, Greenup
1992;305(6863):1215–8. https://doi.org/10.1136/
LC, Kirkpatrick CM, Ray JE, Timmins P, Williams
bmj.305.6863.1215. PubMed PMID: 1467727;
KM. Clinical pharmacokinetics of metformin. Clin
PubMed Central PMCID: PMC1883800
Pharmacokinet. 2011;50(2):81–98. https://doi.
15. Marino M, Jamal Z, Zito PM. Pharmacodynamics.
org/10.2165/11534750-­000000000-­00000.
In: StatPearls; 2021. https://www.ncbi.nlm.nih.gov/
28. Hur KY, Kim MK, Ko SH, Han M, Lee DW, Kwon
pubmed/29939568.
HS, Committee of Clinical Practice Guidelines,
16. Ganguly NK, Bano R, Seth SD. Human genome
K. D. A., & Committee of the Cooperative Studies,
project: pharmacogenomics and drug development.
K. S. o. N. Metformin treatment for patients with dia-
3 Plasma Therapeutic Drug Monitoring and Clinical Toxicology 39

betes and chronic kidney disease: A Korean Diabetes Weekbl. 1988;10:185–8. https://doi.org/10.1007/
Association and Korean Society of nephrology con- BF01956868.
sensus statement. Diabetes Metab J. 2020;44(1):3– 42. Eichorn EJ, Gheorghiade M. Digoxin. Prog
10. https://doi.org/10.4093/dmj.2020.0004. Cardiovasc Dis. 2002;44(4):251–66.
29. Lynch T, Price AL. The effect of cytochrome 43. Virgadamo S, Charnigo R, Darrat Y, Morales G,
P450 metabolism on drug response, interac- Elayi CS. Digoxin: A systematic review in atrial
tions, and adverse effects. Am Fam Physician. fibrillation, congestive heart failure and post myo-
2007;76(3):391–6. cardial infarction. World J Cardiol. 2015;7(11):808–
30. Dean L. Prasugrel therapy and CYP genotype. In: 16. https://doi.org/10.4330/wjc.v7.i11.808.
Pratt VM, Scott SA, Pirmohamed M, Esquivel 44. Tariq S, Aronow WS. Use of inotropic agents in
B, Kane MS, Kattman BL, Malheiro AJ, editors. treatment of systolic heart failure. Int J Mol Sci.
Medical genetics summaries; 2012b. https://www. 2015;16(12):29060–8. https://doi.org/10.3390/
ncbi.nlm.nih.gov/pubmed/28520385. ijms161226147.
31. Dean L. Clopidogrel therapy and CYP2C19 geno- 45. Aronson JK, Hardman M. ABC of monitoring
type. In: Pratt VM, Scott SA, Pirmohamed M, drug therapy. Digoxin. BMJ (Clinical research ed).
Esquivel B, Kane MS, Kattman BL, Malheiro AJ, 1992;305(6862):1149–52. https://doi.org/10.1136/
editors. Medical genetics summaries; 2012a. https:// bmj.305.6862.1149.
www.ncbi.nlm.nih.gov/pubmed/28520346. 46. Pincus M. Management of digoxin toxicity.
32. Herman D, Locatelli I, Grabnar I, Peternel P, Stegnar Aust Prescrib. 2016;39(1):18–20. https://doi.
M, Mrhar A, Breskvar K, Dolzan V. Influence of org/10.18773/austprescr.2016.006
CYP2C9 polymorphisms, demographic factors 47. Rathore SS, Curtis JP, Wang Y, Bristow MR,
and concomitant drug therapy on warfarin metabo- Krumholz HM. Association of serum digoxin con-
lism and maintenance dose. Pharmacogenomics centration and outcomes in patients with heart
J. 2005;5(3):193–202. https://doi.org/10.1038/ failure. JAMA. 2003;289(7):871–8. https://doi.
sj.tpj.6500308. org/10.1001/jama.289.7.871.
33. O'Hara K. Paediatric pharmacokinetics and drug 48. Gupta M, Tripp J. Phenytoin. [Updated 2021 Jul
doses. Aust Prescr. 2016;39(6):208–10. https://doi. 25]. In: StatPearls [Internet]. Treasure Island (FL):
org/10.18773/austprescr.2016.071 StatPearls Publishing; 2021 Jan-. Available from:
34. Wu A. Minimizing medication errors in pediatric https://www.ncbi.nlm.nih.gov/books/NBK551520/
patients. US Pharm. 2019;44(4):20–3. 49. Perucca E, Hebdige S, Frigo GM, Gatti G, Lecchini
35. Klotz U. Pharmacokinetics and drug metabolism S, Crema A. Interaction between phenytoin and val-
in the elderly. Drug Metab Rev. 2009;41(2):67–76. proic acid: plasma protein binding and metabolic
https://doi.org/10.1080/03602530902722679. effects. Clin Pharmacol Ther. 1980;28(6):779–89.
36. Fixen RD. American Geriatrics Society 2019 https://doi.org/10.1038/clpt.1980.235.
Updated AGS Beers Criteria(R) for potentially 50. Craig S. Phenytoin poisoning. Neurocrit Care.
inappropriate medication use in older adults. J 2005;3(2):161–70. https://doi.org/10.1385/
Am Geriatr Soc. 2019;67(4):674–94. https://doi. NCC:3:2:161.
org/10.1111/jgs.15767. 51. Iorga A, Horowitz BZ. Phenytoin toxicity. In:
37. Shenfield GM. Therapeutic drug monitor- StatPearls. StatPearls Publishing; 2021.
ing beyond 2000. Br J Clin Pharmacol. 52. Cook AM, Hatton-Kolpek J. Augmented renal clear-
2001;52 Suppl 1(Suppl 1):3S–4S. https://doi. ance. Pharmacotherapy. 2019;39(3):346–54.
org/10.1046/j.1365-­2125.2001.0520s1003.x. 53. Shahbaz H, Gupta M. Creatinine clearance.
38. Tsunoda SM, Aweeka FT. The use of therapeutic drug [Updated 2021 Jul 26]. In: StatPearls [Internet].
monitoring to optimise immunosuppressive therapy. Treasure Island: StatPearls Publishing; 2021 Jan-.
Clin Pharmacokinet. 1996;30(2):107–40. https://doi. Available from: https://www.ncbi.nlm.nih.gov/
org/10.2165/00003088-­199630020-­00003. books/NBK544228/
39. Akbas SH, Ozdem S, Caglar S, Tuncer M, Gurkan A, 54. Smith BS, Yogaratnam D, Levasseur-Franklin
Yucetin L, Senol Y, Demirbas A, Gultekin M, Ersoy KE, Forni A, Fong J. Introduction to drug phar-
FF, Akaydin M. Effects of some hematological macokinetics in the critically ill patient. Chest.
parameters on whole blood tacrolimus concentration 2012;141(5):1327–36.
measured by two immunoassay-based analytical 55. Abdul-Aziz MH, Alffenaar JC, Bassetti M, Bracht
methods. Clin Biochem. 2005;38(6):552–7. https:// H, Dimopoulos G, Marriott D, Neely MN, Paiva
doi.org/10.1016/j.clinbiochem.2005.02.011. JA, Pea F, Sjovall F, Timsit JF, Udy AA, Wicha SG,
40. Mathew J, Sankar P, Varacallo M. Physiology, blood Zeitlinger M, De Waele JJ, Roberts JA. Infection
plasma. [Updated 2021 Apr 28]. In: StatPearls Section of European Society of Intensive Care
[Internet]. Treasure Island: StatPearls Publishing; Medicine (ESICM); Pharmacokinetic/pharmaco-
2021 Jan. Available from: https://www.ncbi.nlm.nih. dynamic and Critically Ill Patient Study Groups
gov/books/NBK531504/ of European Society of Clinical Microbiology
41. Uges DRA. Plasma or Serum in therapeutic and Infectious Diseases (ESCMID); Infectious
drug monitoring and clinical toxicology. Pharm Diseases Group of International Association
40 G. Fishberger et al.

of Therapeutic Drug Monitoring and Clinical 67. Zeng L, Blair EY, Nath CE, Shaw PJ, Earl JW,
Toxicology (IATDMCT); Infections in the ICU and Stephen K, Montgomery K, Coakley JC, Hodson
Sepsis Working Group of International Society of E, Stormon M, McLachlan AJ. Population phar-
Antimicrobial Chemotherapy (ISAC). Antimicrobial macokinetics of mycophenolic acid in children and
therapeutic drug monitoring in critically ill adult young people undergoing blood or marrow and
patients: a Position Paper. Intensive Care Med. solid organ transplantation. Br J Clin Pharmacol.
2020;46(6):1127–53. 2010;70(4):567–79.
56. van Lent-Evers NA, Mathôt RA, Geus WP, van Hout 68. Smith BS, Yogaratnam D, Levasseur-Franklin KE,
BA, Vinks AA. Impact of goal-oriented and model-­ Forni A, Fong J. Introduction to drug pharmaco-
based clinical pharmacokinetic dosing of aminogly- kinetics in the critically ill patient. Chest. 2012
cosides on clinical outcome: a cost-effectiveness May;141(5):1327-1336.18. Rosansky SJ, Schell J,
analysis. Ther Drug Monit. 1999 Feb;21(1):63–73. Shega J, Scherer J, Jacobs L, Couchoud C, Crews
57. Lea-Henry TN, Carland JE, Stocker SL, Sevastos D, McNabney M. Treatment decisions for older
J, Roberts DM. Clinical Pharmacokinetics in kid- adults with advanced chronic kidney disease. BMC
ney disease: fundamental principles. Clin J Am Soc Nephrol. 2017 Jun 19;18(1):200. Nephrol Ther.
Nephrol 2018 ;13(7):1085-1095. 2017;18(1):200.
58. Bland CM, Pai MP, Lodise TP. Reappraisal of con- 69. Zhang Y, Wang T, Zhang D, You H, Dong Y, Liu Y,
temporary Pharmacokinetic and Pharmacodynamic Du Q, Sun D, Zhang T, Dong Y. Therapeutic drug
principles for informing aminoglycoside dosing. monitoring coupled with Bayesian forecasting could
Pharmacotherapy. 2018;38(12):1229–38. prevent vancomycin-associated nephrotoxicity in
59. Wong G, Briscoe S, McWhinney B, Ally M, Ungerer renal insufficiency patients: a prospective study
J, Lipman J, Roberts JA. Therapeutic drug monitor- and pharmacoeconomic analysis. Ther Drug Monit.
ing of β-lactam antibiotics in the critically ill: direct 2020;42(4):600–9.
measurement of unbound drug concentrations to 70. Taylor ME, Allon M. Practical vancomycin dosing
achieve appropriate drug exposures. J Antimicrob in hemodialysis patients in the era of emerging van-
Chemother. 2018;73(11):3087–94. comycin resistance: a single-center experience. Am
60. Heil EL, Nicolau DP, Farkas A, Roberts JA, Thom J Kidney Dis. 2010;55(6):1163–5.
KA. Pharmacodynamic target Attainment for 71. Crew P, Heintz SJ, Heintz BH. Vancomycin dosing
Cefepime, Meropenem, and Piperacillin-Tazobactam and monitoring for patients with end-stage renal
using a Pharmacokinetic/Pharmacodynamic-based disease receiving intermittent hemodialysis. Am J
dosing calculator in critically ill patients. Antimicrob Health Syst Pharm. 2015;72(21):1856–64.
Agents Chemother. 2018;62(9):e01008–18. 72. Oda K, Jono H, Nosaka K, Saito H. Reduced neph-
61. Shi S, Klotz U. Age-related changes in pharmacoki- rotoxicity with vancomycin therapeutic drug moni-
netics. Curr Drug Metab. 2011;12(7):601–10. toring guided by area under the concentration-time
62. Johannessen Landmark C, Johannessen SI, Patsalos curve against a trough 15-20 μg/mL concentration.
PN. Therapeutic drug monitoring of antiepileptic Int J Antimicrob Agents. 2020;56(4):106109.
drugs: current status and future prospects. Expert 73. Liu L, Pang SK. An integrated approach to
Opin Drug Metab Toxicol. 2020;16(3):227–38. model hepatic drug clearance. Eur J Pharm Sci.
63. Verrotti A, Iapadre G, Di Donato G, Di Francesco 2006;29:215–30.
L, Zagaroli L, Matricardi S, Belcastro V, Iezzi 74. Ahn J, Ahn J, Yoon S, Yoon SN, Son M, Oh J. Human
ML. Pharmacokinetic considerations for anti-­ three-dimensional in vitro model of hepatic zona-
epileptic drugs in children. Expert Opin Drug Metab tion to predict zonal hepatotoxicity. J Biol Eng.
Toxicol. 2019;15(3):199–211. 2019;13(22):1–15.
64. Sourbron J, Chan H, Wammes-van der Heijden EA, 75. Wei Y, Wang YG, Jia Y, Li L, Yoon J, Zhang S, Wang
Klarenbeek P, Wijnen BFM, de Haan GJ, van der Z, Zhang Y, Zhu M, Sharma T, Lin Y, Hsieh M,
Kuy H, Evers S, Majoie M. Review on the relevance Albrecht J, Le PT, Rosen CJ, Wang T, Zhu H. Liver
of therapeutic drug monitoring of levetiracetam. homeostasis is maintained by midlobular zone 2
Seizure. 2018;62:131–5. hepatocytes. Science. 2021;371(6532):1–10.
65. Patsalos PN, Berry DJ, Bourgeois BF, Cloyd JC, 76. Pang KS, Yang QJ, Noh K. Unequivocal evidence
Glauser TA, Johannessen SI, Leppik IE, Tomson supporting the segregated flow intestinal model
T, Perucca E. Antiepileptic drugs--best practice that discriminates intestine versus liver first-pass
guidelines for therapeutic drug monitoring: a posi- removal with PBPK modeling. Biopharm Drug
tion paper by the subcommission on therapeutic Dispos. 2017;38:231–50.
drug monitoring, ILAE Commission on Therapeutic 77. Pang KS, Durk MR. Physiologically-based pharma-
Strategies. Epilepsia. 2008;49(7):1239–76. cokinetic modeling for absorption, transport, metab-
66. Ehren R, Schijvens AM, Hackl A, Schreuder MF, olism and excretion. J Pharmacokinet Pharmacodyn.
Weber LT. Therapeutic drug monitoring of myco- 2010;37:591–615.
phenolate mofetil in pediatric patients: novel tech- 78. Laine JE, Auriola S, Pasanen M, Juvonen
niques and current opinion. Expert Opin Drug Metab RO. Acetaminophen bioactivation by human cyto-
Toxicol. 2021;17(2):201–13.
3 Plasma Therapeutic Drug Monitoring and Clinical Toxicology 41

chrome p450 enzymes and animal microsomes. 89. Little RR, Roberts WL. A review of variant hemo-
Xenobiotica. 2009;39(1):11–21. globins interfering with hemoglobin A1c measure-
79. Mazaleuskaya L, Sangkuhl K, Thorn CF, Fitzgerald ment. J Diabetes Sci Technol. 2009;3(3):446–51.
GA, Altman RB, Klein TE. PharmGKB summary: https://doi.org/10.1177/193229680900300307.
pathways of acetaminophen metabolism at the thera- PMID: 20144281; PMCID: PMC2769887
peutic versus toxic doses. Pharmacogenet Genomics. 90. Davidson M, Ballinger K, Khetani S. Long-term
2016;25(8):416–26. exposure to abnormal glucose levels alters drug
80. McGill MR, Lebofsky M, Norris HK, Slawson MH, metabolism pathways and insulin sensitivity in pri-
Bajt ML, Zie Y, Williams CD, Wilkins DG, Rollins mary human hepatocytes. Sci Rep. 2016;6:28178.
DE, Jaeschke H. Plasma and liver Acetaminophen-­ https://doi.org/10.1038/srep28178.
protein adduct levels in mice after Acetaminophen 91. Tonneijck L, Muskiet MH, Smits MM, et al.
treatment: dose-response, mechanisms, and Glomerular hyperfiltration in diabetes: mecha-
clinical implications. Toxicol Appl Pharmacol. nisms, clinical significance, and treatment. J Am
2013;269(3):240–9. Soc Nephrol. 2017;28(4):1023–39. https://doi.
81. Gorski JC, Vannaprsaht S, Hamman MA, Ambrosius org/10.1681/ASN.2016060666.
WT, Bruce MA, Haehner-Daniels B, Hall SD. The 92. Núñez M. Hepatotoxicity of antiretrovirals: inci-
effect of age, sex, and rifampin administration on dence, mechanisms and management. J Hepatol.
intestinal and hepatic cytochrome p450 3A activity. 2006;44(1 Suppl):S132–9. https://doi.org/10.1016/j.
Clin Pharm Ther. 2003;74:275–87. jhep.2005.11.027. Epub 2005 Nov 28
82. Kapetas AJ, Sorich MJ, Rodrigues AD, Rowland 93. Hughes A, Barber T, Nelson M. New treatment
A. Guidance for Rifampin and Midazolam dosing options for HIV salvage patients: an overview of
protocols to study intestinal and hepatic Cytochrome second generation PIs, NNRTIs, integrase inhibitors
P450 (CYP) 3A4 induction and de-induction. Am and CCR5 antagonists. J Infect. 2008;57(1):1–10.
Assoc Pharm Sci. 2019;21(78):1–11. https://doi.org/10.1016/j.jinf.2008.05.006. Epub
83. Haslam IS, Jones K, Coleman T, Simmons 2008 Jun 16
NL. Rifampin and digoxin induction of MDR1 94. Pau AK, Penzak SR, Boyd SD, McLaughlin M,
expression and function in human intestinal Morse CG. Impaired maraviroc and raltegravir
(T84) epithelial cells. Br J Pharmacol. 2008;154: clearance in a human immunodeficiency virus-­
246–55. infected patient with end-stage liver disease
84. Kim J, Nam WS, Kim SJ, Kwon OK, Seung EJ, Jo and renal impairment: a management dilemma.
JJ, Shresha R, Lee TH, Jeon TW, Ki SH, Lee HS, Lee Pharmacotherapy. 2012;32(1):e1–6. https://doi.
S. Mechanism investigation of Rifampicin-induced org/10.1002/PHAR.1003. PMID: 22392831;
liver injury using comparative toxicoproteomics in PMCID: PMC4518450
mice. Int J Mol Sci. 2017:18(1417):1–13. 95. Zheng Y, Lui G, Boujaafar S, Aboura R, Bouazza
85. Akiyoshi T, Ito M, Murase S, Miyazaki M, N, Foissac F, Treluyer JM, Benaboud S, Hirt D,
Guengerich FP, Nakamura K, Yamamoto K, Gana I. Development of a simple and rapid method
Ohtani H. Mechanism-based inhibition pro- to determine the unbound fraction of dolutegravir,
files of Erythromycin and Clarithromycin with raltegravir and darunavir in human plasma using
Cytochrome P450 3A4 genetic variants. Drug Metab ultrafiltration and LC-MS/MS. J Pharm Biomed
Pharmacokinet. 2013;28(5):411–5. Anal. 2021;196:113923. https://doi.org/10.1016/j.
86. Boetsch C, Parrott N, Fowler S, Poirier A, Hainzl D, jpba.2021.113923. Epub 2021 Jan 25
Banken L, Martin-Facklam M, Hofmann C. Effects 96. Sulkowski MS. Drug-induced liver injury associ-
of Cytochrome P450 3A4 inhibitors–Ketoconazole ated with antiretroviral therapy that includes HIV-1
and Erythromycin–on bitopertin pharmacokinet- protease inhibitors. Clin Infect Dis. 2004;38(Suppl
ics and comparison with physiologically based 2):S90–7. https://doi.org/10.1086/381444.
modelling predictions. Clin Pharmacokinet. 97. Parant F, Miailhes P, Brunel F, Gagnieu
2016;55:237–47. MC. Dolutegravir-related neurological adverse
87. Tonyushkina K, Nichols JH. Glucose meters: events: a case report of successful management
a review of technical challenges to obtain- with therapeutic drug monitoring. Curr Drug Saf.
ing accurate results. J Diabetes Sci Technol. 2018;13(1):69–71. https://doi.org/10.2174/1574886
2009;3(4):971–80. Published 2009 Jul 1. https://doi. 313666180116124046.
org/10.1177/193229680900300446. 98. Nakaharai K, Miyajima M, Kobayashi H, Shimizu
88. Haliassos A, Drakopoulos I, Katritsis D, Chiotinis A, Hosaka Y, Horino T, Hori S. Severe thrombo-
N, Korovesis S, Makris K. Measurement of glycated cytopenia during Dolutegravir-containing antiret-
hemoglobin (HbA1c) with an automated POCT roviral therapy. Intern Med. 2017;56(16):2229–32.
instrument in comparison with HPLC and auto- https://doi.org/10.2169/internalmedicine.8377-­16.
mated immunochemistry method: evaluation of the Epub 2017 Aug 1. PMID: 28781310; PMCID:
influence of hemoglobin variants. Clin Chem Lab PMC5596289
Med. 2006;44(2):223–7. https://doi.org/10.1515/ 99. Schalkwijk S, Greupink R, Colbers AP, Wouterse
CCLM.2006.041. AC, Verweij VG, van Drongelen J, Teulen M, van
42 G. Fishberger et al.

den Oetelaar D, Burger DM, Russel FG. Placental plasma and cerebrospinal fluid samples. Biomed
transfer of the HIV integrase inhibitor dolutegravir Chromatogr. 2018;32(2) https://doi.org/10.1002/
in an ex vivo human cotyledon perfusion model. J bmc.4058. Epub 2017 Aug 21
Antimicrob Chemother. 2016;71(2):480–3. https:// 102. Duthaler U, Berger B, Erb S, Battegay M, Letang E,
doi.org/10.1093/jac/dkv358. Epub 2015 Nov 3 Gaugler S, Natamatungiro A, Mnzava D, Donzelli
100. Mohan H, Lenis MG, Laurette EY, Tejada O, Sanghvi M, Krähenbühl S, Haschke M. Using dried blood
T, Leung KY, Cahill LS, Sled JG, Delgado-Olguín P, spots to facilitate therapeutic drug monitoring of
Greene NDE, Copp AJ, Serghides L. Dolutegravir in antiretroviral drugs in resource-poor regions. J
pregnant mice is associated with increased rates of Antimicrob Chemother. 2018;73(10):2729–37.
fetal defects at therapeutic but not at supratherapeu- https://doi.org/10.1093/jac/dky254.
tic levels. EBio Med. 2021;63:103167. https://doi. 103. Wecker L, Taylor DA, Theobald RJ. Chapter 66:
org/10.1016/j.ebiom.2020.103167. Epub 2020 Dec Antiretroviral drugs for HIV. In: Brody's human
18. PMID: 33341441; PMCID: PMC7753150 pharmacology: mechanism-based therapeutics.
101. Tsuchiya K, Ohuchi M, Yamane N, Aikawa H, Essay Mosby/Elsevier; 2019. p. 570–1.
Gatanaga H, Oka S, Hamada A. High-performance 104. Sax PE. Patient monitoring during HIV antiretro-
liquid chromatography-tandem mass spectrometry viral therapy. UpToDate. 2020, June 15. Retrieved
for simultaneous determination of raltegravir, dolute- from https://www.uptodate.com/contents/patient-­
gravir and elvitegravir concentrations in human monitoring-­during-­hiv-­antiretroviral-­therapy#H4
Dried Blood Spots in Therapeutic
Drug Monitoring and Toxicology
4
Raphael N. Alolga, Qun Liu, and Qi Lian-Wen

Abstract and weaknesses associated with its use, the


various presumptions that need to be consid-
In the quest for suitable surrogates to veni- ered in the use of same for TDM, and other
puncture and conventional biological matrices pertinent issues are herein addressed. Due
(plasma and serum), dried blood spot (DBS) consideration has also been devoted to dis-
sampling has emerged as a very credible can- cussing the implications of blood hematocrit
didate with good analytical prospects. The use variations (known as the hematocrit factor)
of DBS for qualitative and quantitative pur- and ways to tackle or cope with this pitfall.
poses has garnered much attention from the Finally, this chapter delineates the procedural
scientific community over the past 60 years. steps in DBS sampling, recent innovations in
This chapter details the applicability of DBS DBS sampling, and high-throughput applica-
in therapeutic drug monitoring (TDM) and tion of same.
toxicology. Specifically, this chapter high-
lights topical issues including but not limited Keywords
to the application of DBS sampling in phar-
macokinetics (PK), toxicokinetics (TK), Dried blood spots · Therapeutic drug
forensic toxicology, and biomonitoring of monitoring · Toxicology · Hematocrit ·
environmental contaminants. The strengths Biological matrices

R. N. Alolga (*) · Q. Lian-Wen 4.1 Introduction


State Key Laboratory of Natural Medicines, School
of Traditional Chinese Pharmacy, Department of
Pharmacognosy, China Pharmaceutical University, The gold standard for TDM has been serum or
Nanjing, China plasma, but the emergence of DBS in the early
Clinical Metabolomics Center, Department of 1960s makes it an alternative that is worth con-
Pharmacognosy, China Pharmaceutical University, sidering. Interest in DBS for TDM was ignited by
Nanjing, China the work of Guthrie and Susi, who reported the
e-mail: alolgara@cpu.edu.cn
utility of DBS in the screening of phenylketon-
Q. Liu (*) uria in newborns [1]. Their work paved way for
State Key Laboratory of Natural Medicines, School
numerous newborn screening programs using
of Traditional Chinese Pharmacy, Department of
Pharmacognosy, China Pharmaceutical University, DBS [2–4]. The scope of applicability for DBS
Nanjing, China has increased considerably over the years, from

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 43


S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_4
44 R. N. Alolga et al.

newborns to adults, animals, and even for post- • Successful and meaningful application of this
mortem purposes. DBS has also been applied for technique requires substantial validation.
the analyses of proteins, DNA, trace elements, Factors such as the influence of hematocrit
and small molecules [5]. (Hct) variations on the size and homogeneity
DBS sampling is a relatively simple and of the blood spot, impact of blood volume and
convenient method compared to venipuncture. spot homogeneity, and the type of filter paper
Generally, in DBS sampling, an individual used all require due consideration and
disinfects his/her finger and pricks same with validation.
a sterile lancet. The first drop of blood is usu- • There is need to take into consideration the
ally discarded since it contains mainly tissue fact that capillary blood (which is what is used
fluid. Subsequent blood drop is then deposited in DBS) concentration of an analyte is essen-
on a filter paper with premarked circle, tially different from venous blood, and appro-
allowed to dry at room temperature, and then priate translational corrections/calculations
sent for laboratory analysis. Laboratory analy- done.
sis is hence conducted using a punched circu-
lar disc (representing a defined volume) of the Undoubtedly, the most notable challenge in
blood spot with sensitive analytical technique the use of DBS remains the “hematocrit factor.”
[6, 7]. The advantages of this technique over This is a double-sided problem that is both ana-
the conventionally established venous sam- lytical and physiological in nature [8].
pling include [8]: Analytically, the influence of hematocrit altera-
tions affects the accuracy and precision of the
• Ease of sampling and minimal invasiveness of analytical method used to determine the level(s)
technique. This is a technique (which basi- of the analyte(s). The physiological effect of the
cally involves a finger prick with a lancet) that hematocrit factor relates to the plasma/analyte
can be performed by any adult without the ratio of the analyte under investigation [8].
need for a phlebotomist. Details of these problems and how to possibly
• A small amount of blood is required. circumvent them are discussed in this chapter.
• Enhanced stability of analytes in the DBS than This chapter also discusses the applicability of
frozen samples. DBS in TDM, toxicology, and innovations made
• Storage and transportation convenience to the to improve upon the potential of DBS toward
laboratory for analysis. possible adoption by clinical biochemistry
• Risk of infection from dried sample is reduced. laboratories.
• Use of DBS leads to simplification in the pro-
cessing of samples for analysis.
4.2 Therapeutic Drug
The weaknesses or challenges encountered in Monitoring (TDM)
the use of DBS include:
TDM basically entails measurement of the levels
• The small sample volumes which are charac- (concentrations) of drugs in the bloodstream of
teristic of DBS call for sensitive analytical individuals (patients) at specified time intervals
techniques. with the ultimate aim of establishing optimal
• Only one assay per spot can be performed at a dosage regimens. It is mainly employed for
time. drugs with the following characteristics: (1)
• There are usually no spare DBS samples for drugs with narrow therapeutic indices, (2) drugs
confirmatory analysis. that exhibit significant variabilities in their phar-
• Independent sampling by non-health workers macokinetic (PK) profiles, (3) drugs whose sys-
requires adequate training and even that does temic concentrations are not easy to monitor,
not automatically guarantee success by first-­ and (4) drugs with known or predicted to have
time users. serious adverse effects. TDM operates on the
4 Dried Blood Spots in Therapeutic Drug Monitoring and Toxicology 45

premise that a reasonably established relation- DBS sampling a suitable technique for multi-
ship exists between the dose and plasma or blood center studies in low-resource settings where mul-
concentration of a drug and the concentration tiple sampling is required and cold-­chain facilities
and therapeutic effect(s) of same. A plethora of are unavailable [10]. DBS has hence been vari-
variables i­nfluence the interpretation of drug ously used for the PK studies of different classes
concentration data, from dose and dosage form of drugs including but not limited to antimicrobi-
to sampling time, handling of blood samples, als (antibiotics and antifungals), immunosuppres-
analytical method, health status of individual sants, analgesics, antidepressants, antiretrovirals,
(healthy, sick, presence of comorbid conditions, anticonvulsants, etc. Table 4.1 summarizes the
etc.), and reliability of PK models used. TDM classes of drugs, names of drugs, and the analyti-
generally involves factors and variables that cal techniques applied for the PK studies by vari-
influence the PK and pharmacodynamic (PD) ous research groups.
monitoring of drugs. These variables therefore
need to be taken into consideration during TDM
for the data to be clinically useful. Clinically, 4.2.2 Factors to Consider in PK
TDM aims at enhancing patient care by modify- Studies Using DBS Sampling
ing the dosages of drugs to suit individual/spe-
cific patient needs [9]. In order to fully tap from the benefits of DBS
sampling in PK studies as well as other strategies
of TDM, certain inherent correlational establish-
4.2.1 DBS in PK Monitoring ments and assumptions need to be duly recog-
nized [11]. Due consideration should be given to
In a much as venipuncture remains the mainstay the relationships that exist between the total
blood sampling technique for PK studies, DBS plasma concentration, total whole blood concen-
sampling is gradually attaining recognition as a tration, and the unbound concentration of the
credible surrogate or complementary technique. analyte as shown in Eqs. 4.1 and 4.2.
The invasiveness of venipuncture constitutes a
Cu
major limitation that accounts considerably for Ct =
poor volunteer recruitments in such studies. Also fu
(4.1)
multiple withdrawal of large volumes of blood
samples from experimental animals constitutes 1  
Cb       Cu
another limitation for venipuncture. Under such  fu  (4.2)
circumstances, meeting the ethical requirements
of the “3Rs” (replacement, reduction, and refine- Ct is the total plasma concentration; Cu is the
ment) becomes a strenuous task. Blood sampling unbound concentration; Cb is the total blood con-
by DBS particularly meets the latter two of the centration; fu is the unbound plasma fraction; H is
“3Rs,” thus reduction and refinement. DBS sam- the hematocrit and ρ is the blood cell-to-unbound
pling allows for a reduction in the number of ani- plasma concentration ratio. It is evident from
mals (rodents) needed. This comes with reduced these equations that the plasma concentration of
cost of experiments particularly when knockout an analyte is proportional to its unbound concen-
or transgenic animal models are used. The need tration if fu is constant. The blood concentration
for refinement is met in the way and manner blood of the analyte is proportional to its unbound con-
is sampled from the animals. Aside from the gen- centration when fu, H, and ρ are constant. Under
eral strengths and advantages of DBS as a sam- such ideal circumstances, accurate determina-
pling technique earlier outlined, the analytes are tions of the analyte concentration can either be
subjected to relatively fewer processing stages done using plasma or whole blood, since the
prior to analysis and do not need to be stored result of using any of these accurately reflects the
under strict cold-chain conditions. This makes unbound concentration of same.
46 R. N. Alolga et al.

However, under non-ideal conditions these consider. Strong cellular binding of the drug
general assumptions do not hold validity. For (especially to the RBCs) than to plasma protein
instance, for drugs that do not have any binding (ρ > 1/fu) changes Eq. 4.4 to:
affinity for plasma proteins (in this case fu = 1)
Cb     Cu
such as the aminoglycosides, total plasma con- (4.5)
centration (Ct) is approximately equal to the con- As noted from Eq. 4.5, the blood concentration of
centration of the unbound drug (Cu). Plasma (or an analyte becomes directly proportional to its
even whole blood) could therefore be appropriate unbound concentration and is not prone to altera-
matrix for the analysis of these drugs. In the tions in plasma protein binding, thereby making
instance where the drug binds to plasma proteins blood superior to plasma as an analytical matrix.
(as is the case for most drugs), fu becomes a cru- This ideal situation does not play out all the time
cial parameter. The value of fu is influenced by because the blood concentrations of most drugs
disease (e.g., kidney diseases), burns, pregnancy, are usually sensitive to changes in ρ. Also drugs
age, species, etc. Under these circumstances, it that fall under this category possess high blood-­
advisable to directly measure the unbound drug to-­plasma concentration ratios (Cb/Ct). Blood-to-­
concentration and calculate the fu value there- plasma concentration ratio therefore provides a
from. There are also instances where the hemato- useful guide to the significance of either plasma
crit (H) and the blood cell-to-unbound plasma binding or blood cell binding in the use of DBS
concentration ratio (ρ) are additional confound- for PK studies. For instance, blood cell binding
ing parameters. Though relatively constant, the becomes the predominant issue when the
hematocrit of the blood can fall drastically below Cb/Ct > 1.5.
the average value of 0.45 to about 0.2 under ane- In brief, under ideal conditions, the blood con-
mic conditions and should therefore be taken into centration of a drug is the sum of its concentra-
account in blood analysis and data interpretation. tions in the RBCs (predominantly) and plasma
For polar, hydrophilic drugs that cannot permeate [10]. Preferential and differential binding of a
the blood cells, the blood cells tend to act as a drug to other components of the blood and other
diluent for such drugs. The blood concentration tissues result in variations in the concentrations
of the analytes (Cb) is thus represented by Eq. 4.3: of same in the plasma and RBCs and invariably
the blood concentration. To this end, for a com-
1   parison between the amount of a drug in the
Cb    Cu
 fu  (4.3) plasma or serum and the DBS to hold any valid-
ity, the hematocrit (H), RBC to plasma partition-
Such drugs also exhibit either poor or no binding ing (KRBC/plasma), and blood-plasma partitioning
affinity to plasma proteins (fu = 1), thereby chang- (KBlood/plasma) need to be determined. To calculate
ing Eq. 4.3 to: the hematocrit of blood, a packed RBC volume is
divided by total blood volume. The packed RBC
Cb  1   Cu (4.4) volume is simply determined by centrifuging
heparinized blood in microhematocrit tubes at
For these drugs (e.g., the aminoglycosides), use specified speed and time. The KRBC/Plasma or KBlood/
of blood is a good matrix to monitor their plasma could be estimated by spiking a drug in
unbound levels. For drugs such as caffeine or plasma or plasma with suspended RBCs, equili-
alcohol that freely permeate the blood cells and brating the mixture before centrifugation. The
do not bind to the plasma proteins, their Cb is amount of the drug in the plasma and RBCs can
almost equal to Cu. In this instance, blood again is then be quantified [12]. Determinations of these
a good matrix to determine their levels. parameters make it possible for the plasma con-
For drugs that permeate the blood cells as well centration of the drug to be estimated from the
as bind to same, ρ becomes a critical factor to DBS results based on Eq. 4.6:
4 Dried Blood Spots in Therapeutic Drug Monitoring and Toxicology 47

Drug Conc.in DBS 4.2.3.1 Toxicokinetics


Estimated plasma conc. 
(1  H)  H + K RBC/ plasma  DBS sampling has also been applied for toxico-
(4.6)
kinetic studies. Toxicokinetics, which is similar
For a drug with KRBC/plasma or KBlood/plasma values to PK, is mainly focused on toxicants – the
greater than 1, the amount of that drug will be absorption, distribution, and elimination of toxi-
higher in the DBS than plasma, hence making cants in a living organism. In this respect, the ear-
DBS a superior matrix for PK analysis. KRBC/plasma lier stated advantages, weaknesses, assumptions,
or KBlood/plasma values equal to 1 lead to comparable and correlational evaluations with the use of DBS
amounts of the drug in plasma or serum and the for PK studies apply for toxicokinetics. Details of
DBS. Use of DBS under such circumstance does specific toxicokinetic studies are summarized in
not offer any obvious benefit over plasma or Table 4.1.
serum. However, when the KRBC/plasma or KBlood/
plasma values are less than 1, the amount of the drug 4.2.3.2 Forensic Toxicology
in the DBS will be lower than in plasma or serum, Due to the inherent strengths of DBS sampling,
making DBS not the recommended method for it has been suggested for use in forensic toxico-
PK studies [13]. DBS is best suited for the analy- logical analysis as either an alternative or main-
sis of drugs with higher RBC binding affinities stay technique. Forensic toxicology generally
relative to plasma. deals with the application of the principles and
knowledge of toxicology to investigate issues
that are of interest to the law enforcement
4.2.3 Toxicology authorities [86]. In this regard, the testimonies
of forensic toxicologists in the delivery of fair
Aside from the relevance of DBS in TDM, its and just judgments by the law courts cannot be
application in various aspects of toxicological overstated. This branch of science mainly con-
assessments such as toxicokinetics, forensic, sists of three major disciplines: postmortem
environmental, and clinical toxicology is gaining toxicology, forensic drug testing, and human
traction. In neonatal screening programs, DBS performance toxicology. Broadly, forensic toxi-
samples are used not only to determine the pres- cology refers to the qualitative as well as the
ence or otherwise of inborn errors of metabolism quantitative evaluation of biological samples for
but serve as useful matrices to monitor the effects both legal and illegal drugs. Hence, drugs or
of certain prenatal exposure conditions, notably substances of abuse including the benzodiaze-
exposure to potentially harmful and toxic com- pines, opiates, cannabinoids, cocaine, amphet-
pounds. In this respect, the ability of these com- amines, gamma-­ hydroxybutyric acid, and
pounds to cross the fetoplacental barrier is alcohol (including its metabolites, ethyl gluc-
assessed, albeit with caution since postpartum uronide and ethyl sulfate, phosphatidyl ethanol)
exposure via the mother’s milk needs to be taken fall under this category. Table 4.2 summarizes
into consideration. The information obtained some drugs or compounds of interest for which
therefrom could retrospectively reveal the extent DBS sampling was performed. Despite the mer-
of exposure to these chemicals. Since early expo- its of DBS sampling, it is still used with caution.
sure to these harmful chemicals could have debil- For instance, in the determination of a DUI case
itating effects on the child later in life, the (driving under the influence of alcohol), the
toxicological information obtained could be used accepted convention is to determine the amount
for specific follow-up studies and possible inter- of alcohol in the exhaled breath of the person
ventions [14]. using a breathalyzer. Also, for substance abuse
cases, urine usually serves as the main biologi-
cal matrix.
48 R. N. Alolga et al.

Table 4.1 PK and TK studies of drugs for which sampling was done by DBS
Type of
Class and name of drug study Analytical platform References
Antivirals
Amprenavir PK LC-MS [15]
Atazanavir PK and LC-MS [15]
TDM
Darunavir PK and LC-MS/MS [15, 16]
TDM
Saquinavir PK and LC-MS/MS [15]
TDM
Amprenavir PK and LC-MS/MS [15]
TDM
Lopinavir PK and LC-MS/MS [15]
TDM
Ritonavir PK and LC-MS/MS [15]
TDM
Etravirine PK and LC-MS/MS [15]
TDM
Efavirenz PK and LC-MS/MS [15]
TDM
Nevirapine PK and LC-MS/MS [15]
TDM
Tenofovir and emtricitabine PK LC-MS/MS [17]
Oseltamivir PK LC-MS/MS [18]
Zidovudine TDM RIA [19]
Raltegravir TDM LC-MS/MS [20]
Nevirapine and efavirenz TDM LC-MS/MS [21]
Etravirine TDM LC-MS/MS [15]
Efavirenz TDM RP-HPLC-UV [22]
Anticonvulsants/antidepressants
Levetiracetam TDM HPLC [23]
Lamotrigine TDM HPLC [23–25]
Phenobarbital TDM HPLC [23]
Carbamazepine TDM HPLC [23, 25]
Gabapentin PK LC-MS/MS [26]
Rufinamide TDM LC-MS/MS [27]
Topiramate TDM FPIA and LC-MS/MS [28]
Fluoxetine TDM and NICI-MS-MS [29]
PK
Norfluoxetine TDM and NICI-MS-MS [29]
PK
Reboxetine TDM and NICI-MS-MS [29]
PK
Paroxetine TDM and NICI-MS-MS [29]
PK
Clozapine TDM HPLC [30, 31]
Diazepam PK LC-MS/MS [32]
Donepezil PK LC-MS/MS [33]
Midazolam PK LC-MS/MS [34]
Valproic acid TDM LC-MS/MS [25]
Phenytoin PK LC-MS/MS [35]
Antimicrobials
Actinomycin-D PK LC-MS/MS [36]
(continued)
4 Dried Blood Spots in Therapeutic Drug Monitoring and Toxicology 49

Table 4.1 (continued)


Type of
Class and name of drug study Analytical platform References
Gemifloxacin PK HILIC with fluorescence detection [37]
Linezolid TDM LC-MS/MS [38]
Moxifloxacin TDM LC-MS/MS [39]
Posaconazole PK and LC-MS/MS [40, 41]
TDM
Fluconazole TDM LC-MS/MS [41]
Voriconazole TDM LC-MS/MS [41]
Rifampicin TDM and HPLC-UV; LC-MS/MS [42, 43]
PK
Pyrazinamide PK LC-MS/MS [43]
Ethambutol PK LC-MS/MS [43]
Rifaximin TDM and LC-MS [44]
TK
Sisomicin and netilmicin TDM Post-column HPLC with [45]
fluorescence detection
Mycophenolic acid TDM LC-MS/MS [46]
Ertapenem TDM and LC-MS/MS [47]
PK
Metronidazole PK HPLC [48, 49]
Ampicillin PK LC-MS/MS [50]
Benzathine penicillin PK LC-MS/MS [51]
Solithromycin PK LC-MS/MS [52, 53]
Ceftriaxone PK LC-MS/MS [54]
Piperacillin-tazobactam PK LC-MS/MS [55]
Analgesics
Acetaminophen TK LC-MS/MS [56]
Flurbiprofen PK LC-MS/MS [34]
Methadone TDM HPLC [57]
Naproxen PK LC-MS/MS [58]
Anticancers
Busulfan TDM LC-MS/MS [59]
Paclitaxel PK LC-MS/MS [60, 61]
Vincristine TDM and LC-MS/MS [62]
PK
Larotrectinib PK LC-MS/MS [63]
Abiraterone and delta(4)-abiraterone TDM LC-MS/MS [64]
Radotinib TDM LC-MS/MS [65]
Immunosuppressants
Tacrolimus TDM LC-MS/MS [66]
Sirolimus TDM LC-MS/MS [66, 67]
Everolimus TDM LC-MS/MS [66, 67]
Cyclosporin A TDM LC-MS/MS [66]
Antihypertensives
Clonidine PK LC -MS/MS [68]
Atenolol TDM and LC-TOF-HRMS [69]
PK
Bisoprolol TDM LC-HRMS [70]
Ramipril TDM LC-HRMS [70]
Simvastatin TDM LC-HRMS [70]
Bosentan PK LC-MS/MS [71]
(continued)
50 R. N. Alolga et al.

Table 4.1 (continued)


Type of
Class and name of drug study Analytical platform References
Losartan PK LC-MS/MS [72]
Metoprolol PK LC-MS/MS [73]
Propranolol PK LC-MS/MS [74]
Guanfacine PK LC-MS/MS [75]
Antimalarials
Tafenoquine TDM HILIC with fluorescence detection [76]
Sulfadoxine and sulfamethoxazole TDM HPLC-UV [77]
Sulfadoxine and pyrimethamine TDM HPLC-UV [78]
Quinine and 3-hydroxyquinine PK HILIC with fluorescence detection [79]
Monodesethylchloroquine, chloroquine, cycloguanil, PK HPLC [80]
and proguanil
Amodiaquine, chloroquine, and chlorthalidone PK LC-MS/MS [81]
Chloroquine and desethylchloroquine PK HPLC-UV [82]
Antidiabetics
Metformin TDM HPLC-UV [83]
Metformin and sitagliptin PK LDTD-MS/MS [84]
Pioglitazone TK LC-MS/MS [85]
Rosiglitazone PK LC-MS/MS [34]
LC-MS liquid chromatography-mass spectrometry; LC-MS/MS liquid chromatography-tandem mass spectrometry;
LC-HRMS liquid chromatography-high-resolution mass spectrometry; LC-TOF-HRMS liquid chromatography-time-­
of-flight high-resolution mass spectrometry; NICI-MS/MS negative-ion chemical ionization-tandem mass spectrometry;
LDTD-MS/MS laser diode thermal desorption tandem mass spectrometry; FPIA fluorescence polarization immunoas-
say; RIA radioimmunoassay

4.2.3.3 Screening for Environmental animals at the top of the food chain. This strategy
Contaminants has been applied to monitor the level of exposure
DBS has served as the matrix of choice for the of the bottlenose dolphin to the biotoxins of
screening of various environmental contaminants Pseudo-nitzschia and Karenia brevis, thus
in humans and animals. These contaminants domoic acid and brevetoxins, respectively [123].
range from heavy metals, benzene oxide (metab- These toxins have also been detected in other
olite of benzene), perfluoroalkyl compounds, experimental animals (mice, rats) using DBS
perchlorate, organochlorine pesticides, polychlo- sampling. Analysis of these biotoxins in the DBS
rinated biphenyls, to polybrominated diphenyl samples was accomplished using radioimmuno-
esters. Exposure of newborns to toxic compounds assay [124], receptor-binding assay [125], or
such as dichlorodiphenyldichloroethylene competitive ELISA [126].
(metabolite of DDT), perfluorooctane sulfonate,
perfluorooctanoate, etc. has been assessed using
DBS sampling [120–122]. Animals intoxicated 4.3 The Hematocrit Factor
with biohazard materials can be monitored using
the DBS sampling technique. The levels of cho- The hematocrit factor remains by far the single
linesterase inhibitors such as the carbamate and most relevant and discussed issue in DBS sam-
organophosphate insecticides, in birds for pling. Since the viscosity of whole blood is
instance, can be assessed by DBS sampling. As determined by its hematocrit, it invariably
earlier indicated, the utility of this technique affects the spreadability of same on a filter
allows for samples to be collected in secluded paper. Hence, aside from the type of filter paper
areas where no special equipment are available. It used, the content of an analyte in punched discs
is also possible to monitor the levels of biohazard from the DBS card obtained using blood of dif-
compounds in the environment by targeting the ferent hematocrit values tends to differ and ulti-
4 Dried Blood Spots in Therapeutic Drug Monitoring and Toxicology 51

Table 4.2 List of some drugs or markers of drugs of Several strategies have been advanced with
abuse for forensic purposes
the aim of determining how to either minimize
Name of drug (marker Analytical the impact of hematocrit variations or tackle
compounds) platform References
(avoid) it entirely. To avoid the hematocrit prob-
Fentanyl and its metabolites LC-MS/MS [87–89]
(norfentanyl and lem, researchers tend to use volumetrically gen-
despropionyl fentanyl) and erated DBS and analyze the DBS punches
analogs holistically. Others use dried plasma spots (DPS)
Ethylglucuronide LC-MS/MS [90] in place of DBS. The impact of hematocrit can be
− ethylsulfate
minimized by spotting the blood on special filter
Phosphatidylethanol LC-MS/MS [91–93]
Cotinine LC-MS/MS [94–96] substrates, using calibrators with hematocrit val-
Tramadol and LC-MS/MS; [97, 98] ues within the range of the target population and
O-desmethyltramadol LC-HRMS estimating (measuring) the hematocrit of the
Tetrahydrocannabinol and LC-MS/MS [99–101] DBS [8].
metabolites
Methadone and metabolites LC-MS/MS [102, 103]
Buprenorphine and LC-MS/MS [104]
metabolites 4.3.1 Holistic Analysis
Ketamine and norketamine LC-MS/MS [104] of Volumetrically Spotted DBS
Opiates and metabolites LC-MS/MS [105, 106]
Gamma-hydroxybutyric GC-MS [107–109] A convenient way to tackle the hematocrit issue
acid
is to avoid it by analyzing whole-cut volumetri-
Benzodiazepines LC-MS/MS [110, 111]
Zolpidem CE-MS [112]
cally applied DBS. It involves mainly two
Cocaine, benzoylecgonine, LC-MS/MS [113] approaches. The first approach involves the use
and other metabolites of the entire DBS punched out of their respective
Methamphetamine LC-MS/MS [105] filter papers after specific volumes of blood have
a
MDEA, MDMA, MDA LC-MS/MS [114] been applied. In the other approach, volumetri-
Amphetamine LC-MS/MS [115] cally generated blood is spotted on pre-punched
Anabolic steroids LC-MS/MS; [116–119]
GC-MS
discs, dried, and the whole discs analyzed. This
approach at tackling the hematocrit problem
LC-MS liquid chromatography-mass spectrometry; LC-­
MS/MS liquid chromatography-tandem mass spectrome- requires that the volume of blood used for spot-
try; LC-HRMS liquid chromatography-high-resolution ting be accurately and precisely delivered.
mass spectrometry; GC-MS gas chromatography-mass Accurate and precise volumes of blood are
spectrometry
obtained using specialized microcapillary sam-
a
MDEA 3,4-methylenedioxyethylamphetamine; MDMA
3,4-methylenedioxymethamphetamine; MDA pling devices that are best used by trained per-
3,4-methylenedioxyamphetamine sons. This obviously tends to be a limitation since
this approach cannot be readily used outside a
specialized environment (e.g., at home) by non-­
mately introduces significant assay biases and trained individuals, thus defeating one of the
errors. The hematocrit of blood is the volume main strengths of DBS sampling.
ratio of red blood cells to the total blood vol-
ume. The typical hematocrit range for men is
0.41–0.50, while that for women is 0.36–0.44 4.3.2 Use of DPS in Place of DBS
[127]. Elevated values are generally observed
for people living in areas of high altitude, new- An alternative strategy for circumventing the
borns, and persons with primary polycythemia hematocrit problem is the use of dried plasma
and chronic obstructive pulmonary disease, spots (DPS) instead of DBS. By virtue of the fact
while anemic persons, immunocompromised that plasma is used instead of whole blood as the
individuals, and persons receiving chemother- matrix, RBCs are excluded – ultimately avoiding
apy have lower hematocrit values [8]. the hematocrit problem. Many a researcher have
52 R. N. Alolga et al.

reported an almost excellent association between lish an acceptable hematocrit range prior to anal-
the concentrations of various drugs (such as par- ysis and validation of the analytical method.
oxetine, nevirapine, triazole antibiotics) in DPS Since hematocrit could vary based on specific
and plasma [128–131]. Most researchers obtained target groups, its impact could hence be reduced
plasma from blood using various methods that by using blood samples from such groups as cali-
eventually involved centrifugation, a process that brators. In line with this, when quantitatively
cannot be readily carried out at home. Other monitoring the level of an analyte in newborns
researchers have however devised various prepa- (healthy) who usually have hematocrit values
ratory steps that are independent of centrifuga- above 0.50, the same calibration line cannot be
tion. A study worth mentioning is that of Li et al. used for immunocompromised patients, typically
who, in the biomonitoring of guanfacine, used a with lower than the 0.30 average hematocrit
device that was capable of separating plasma value [132]. In the situation whereby the target
from whole blood [13]. The DPS was obtained population is heterogeneous, the calibrators can
from blood drops applied onto a filter membrane be prepared using blood with approximate aver-
(multilayered and polymeric) with collection and age hematocrit value of 0.4 [133]. On the other
separation membranes, and a top layer that is hand, one could also use two calibration graphs,
removable. The plasma seeped through the sepa- one constructed within the upper limit and the
ration membrane to the collection membrane at other within the lower limit of acceptable hema-
the bottom, while the solid residue (RBCs) tocrit range.
remained on top and could then be peeled off
after about 5 min [13].
4.3.5 Estimation or Prediction
of Hematocrit
4.3.3 Use of Special Filter
Substrates Different analytical strategies have been pro-
pounded or applied to cope with the hematocrit
As a means of minimizing the hematocrit effect, factor [8]. These strategies essentially allow for
special types of filter material or paper have been confirmation of the hematocrit of DBS samples
manufactured by several companies. Examples and acquisition of a correction factor to compen-
of these special filter materials include Bond Elut sate for the impact of the hematocrit. It has been
Dried Matrix (a noncellulose filter paper devel- suggested that extra venous blood sample be
oped by Agilent Technologies), HemaForm™, taken for hematocrit estimation in addition to the
HemaSpot™ (developed by Spot on Sciences capillary blood used for the DBS. This however
Inc.), glass paper filters, chitosan and alginate is feasible under controlled conditions with the
forms, etc. These special materials exhibit unique aid of trained personnel. Also with this approach,
strengths and weaknesses compared to the con- one wonders the essence of using capillary blood
ventional cellulose filter paper and therefore for the DBS if venous blood is available except
require validation before use [8]. for issues of stability. Differences in the hemato-
crit of capillary blood relative to venous blood
make this approach quite problematic. Ideally,
4.3.4 Use of Hematocrit Calibrators the hematocrit of the capillary blood used for the
DBS should be determined. To achieve this, a
To better minimize the impact of hematocrit on calibrated microcapillary coated with an antico-
any DBS-based analytical assay, it is advisable to agulant can used to deliver a minute volume of
consider the target population (e.g., healthy per- capillary blood and the hematocrit read after the
sons, newborns, the elderly, anemic individuals, sample is centrifuged. Here again, the ease of
immunocompromised persons, etc.) and estab- sampling which is central to DBS is eventually
4 Dried Blood Spots in Therapeutic Drug Monitoring and Toxicology 53

lost. Though costly to the patient, a more conve- tions; (4) the compound should be stable in both
nient and practical option is to rely on point-of-­ new and old DBS, for its measurement to be pos-
care tests (e.g., those developed by HemoCue®) sible; and (5) determination of the level of such
that are comparatively fast, easy to handle, and compound should be relatively easy to accom-
require minimal amount of blood (10 μl) to deter- plish. On the basis of this criterion, potassium
mine its hematocrit [134–136]. totally fulfilled all requirements and has therefore
Some researchers have also suggested that the been variously recommended by researchers
hematocrit be estimated on the basis of certain [138, 139].
physical characteristics of the DBS including the Hemoglobin seems an obvious candidate to
surface area, diameter, frustum volume, and color accurately estimate the hematocrit of blood.
[39, 127, 137]. Needless to mention, but this Hemoglobin is an iron-binding protein exclu-
approach also has its shortfalls and consequently sively found in the red blood cells (RBCs) and
introduces certain errors and biases in the deter- a transporter of oxygen through the body. As
mination of hematocrit. Notably its reliance on earlier outlined, the calculation of hematocrit
the exactitude of the volume of blood spotted of fresh blood is already done using hemoglo-
ranks topmost since the characteristics of the bin. However, the use of hemoglobin to calcu-
DBS that depend on its dimensions such as area late the hematocrit of DBS is met with some
(diameter) of spread, color of spot, and frustum challenges that consequently compromise the
volume are in turn dependent on the amount of accuracy and precision of the method. It is
blood spotted. For instance, using the photo- worth noting that hemoglobin changes with
graphs of DBS, Denniff and Spooner sought to time as evidenced by the change in color of
measure the spot area of blood on different kinds DBS. This implies that for routine measure-
of filter paper [127]. This proved difficult to ment of hematocrit using analyzers that only
accomplish since the differences in the type of measure absorption at specific wavelengths,
filter paper affected the homogeneous spread of there is a tendency to obtain false readings
the spot, making it difficult to determine the exact (lower than fresh samples) especially for older
edges of the spot for the frustum volume to be DBS samples. This could be the result of con-
calculated. They however found that the frustum version of hemoglobin to another form that
volume was linearly correlated with hematocrit. does not absorb at that wavelength but not nec-
Other authors established a correction factor in a essarily due to lower amount of hemoglobin per
bid to eliminate biases resulting from the punch se. Calibration curves should therefore be con-
diameter, blood volume, and DBS area. That not- structed using the appropriate samples. Hence,
withstanding, the biases were not totally quantification of hemoglobin in fresh DBS sam-
eliminated. ples should be based on calibration curves
A more reliable and resilient alternative to obtained with fresh samples and not using cali-
measure or estimate the hematocrit is to rather bration curves of aged samples.
measure the level of an endogenous compound
that has a strong correlation with it (hematocrit).
According to De Kesel et al., for a compound to 4.4 General Steps in DBS
be considered as a hematocrit marker, it needs to Sampling
satisfy a five-point criterion [8]: (1) the com-
pound should have a strong correlation with As summarized in Fig. 4.1, the main procedural
hematocrit; (2) the compound should be univer- steps in the analysis of various compounds (ana-
sally applicable, in the sense that, it should be lytes) in the matrix of blood via DBS sampling
easily measurable in any population regardless of include selection of filter material, collection of
factors such as age, sex, or race; (3) the com- specimen, drying of spotted cards, packaging of
pound should exhibit minimal interindividual DBS for storage and transport, and extraction and
variation, irrespective of even diseased condi- analysis of analyte.
54 R. N. Alolga et al.

4.4.1 Selection of Filter Material for analysis and/or storage. Moreover, the DMPK
cards denature and activate enzymes such as the
DBS cards consist of mainly cellulose (paper) or esterases in the blood, thereby preventing enzy-
noncellulose matrix with varied pore sizes and matic breakdown of drugs such as aspirin (acetyl-
thickness. Commercially available DBS cards salicylic acid) and procaine, leading to their
include the Whatman fast transient analysis increased stability. The DMPK-C type is not pre-
(FTA) elute cards, the FTA-drug metabolism and treated with any special chemical and therefore
pharmacokinetic, FTA-DMPK cards (types A, B, does not denature proteins or produce any chemi-
C), and Whatman 903® cards. The quality speci- cal that could interfere with sample analysis.
fications of these cards define their respective Another type of card that exhibits the same prop-
applications. For routine screening of newborns, erties as the DMPK-C type is the Ahlstrom 226
the Whatman 903® cards are used. The FTA-­ card (ID Biological Systems, Greenville, SC).
DMPK cards are suited for PK/TK studies, while These two are therefore recommended for
the FTA elute cards are designed to accurately protein-­based biomolecular analysis. There are
collect and purify DNA for subsequent analysis also commercially available noncellulose-based
[10]. DBS cards for drug monitoring and PK studies
The DMPK cards come in two main forms, such as the Bond Elut DMS card by Agilent
regular and indicating. When colorless samples Technologies. They are deemed to be superior in
such as plasma, urine, synovial fluid, etc. are to quality, require less punching effort, enhance
be analyzed, the indicating cards are the most mass spectrometry signals of the analyte, and are
appropriate type. Types A and B of the DMPK not affect by hematocrit variations [10].
cards are pretreated chemically such that they
cause cells to lyse and proteins to denature, as
well as inactivate enzymes and inhibit bacteria 4.4.2 Collection of Specimen
growth on contact with the biosample (blood,
plasma, urine, etc.). These precoated cards also In humans, whole blood is sampled from a finger,
allow for cellular and nuclear membranes to lyse toe, or heel after a prick with a sterile and dispos-
so that nucleic acids that are very stable remain able lancet. Blood can be collected from rats and

Fig. 4.1 Schematic representation of the main steps in DBS sampling (b), capillary blood from a finger prick is
venous blood sampling (a) and DBS sampling (b). In spotted on the DBS card, dried under room temperature,
venous blood sampling (a), blood is collected via veni- and transported as a parcel by normal courier to the labo-
puncture into special sample tubes (for plasma or serum) ratory for analysis. In the laboratory, the frozen sample is
and centrifuged and the supernatant (plasma or serum) thawed, centrifuged, and prepared for analysis, while the
collected and stored under strict cold-chain conditions till analyte in the DBS sample is extracted, centrifuged, and
analysis or for transport to the laboratory for analysis. For prepared for analysis
4 Dried Blood Spots in Therapeutic Drug Monitoring and Toxicology 55

mice for the purposes of PK and TK studies, from 4.4.4 Packaging of DBS for Storage
the caudal vein. For the purposes of qualitative and Transport
determination, drops of blood from the finger,
toe, or heel are cautiously spotted within defined DBS sampling offers a very convenient avenue
areas on the DBS card. For quantitative analyses, for the storage and transportation of the DBS
accurate volumes of blood are spotted on the cards. Compared to traditional biological matri-
DBS cards using a micropipette or capillary tube. ces (plasma, serum, urine, etc.), the need for strict
It is common in such instances to use anticoagu- adherence to cold-chain during storage or trans-
lants such as heparin or ethylenediaminetetraace- port is almost totally eliminated with DBS. One
tic acid (EDTA), though they need to be used does not require to use bulky equipment such as
with caution in quantitative polymerase chain freezers or even dry ice for the storage or trans-
(qPCR)-based analysis since they affect telomere portation of DBS. Once humidity is controlled,
length measurements [140]. When the samples DBS cards can be stored or transported at ambi-
are analyzed by MS (i.e., LC-MS), EDTA inter- ent temperature. To guard against the influence of
feres with the spectrum of the analyte of interest. humidity from the environment, the cards can be
That notwithstanding, it is preferred to heparin packed in sealable plastic bags with a desiccant
for its drying effects and inhibition of ester and humidity indicators to monitor when to
hydrolases and phospholipases (calcium-­replace the desiccant. For compounds that are
dependent) [141–143]. light-sensitive, it is advisable to use an aluminum-­
Since the usability of the DBS is dependent on coated bag [145]. The DBS cards can be trans-
the volume of blood spotted and the accuracy and ported safely using normal postal mail or courier
sensitivity of the analytical platform, it is advisable without threat of exposure of handlers to infec-
that the sampling process be done correctly. tion. It is however recommended that the pack-
Improperly spotted cards and DBS cards that are aged cards be labeled to clearly reflect the
discolored, supersaturated, dark in color, clotted, or biohazardous nature of sealed content before
contaminated cannot be used for any purpose, be it transportation to the analytical laboratory for
qualitative or quantitative. As earlier indicated, the analysis.
differential hematocrit values of blood also need to
be duly considered and corrected in order to accu-
rately determine the content of the analyte. 4.4.5 Extraction and Analysis
of Analyte

4.4.3 Drying of Spotted Cards In the analytical laboratory, the spotted portions
of the DBS cards are punched out and used
Drying of the spotted cards could be a crucial directly or analyzed after extraction using appro-
step particularly for unstable, heat-sensitive ana- priate solvents. A crucial step in the extraction
lytes or metabolically unstable analytes. The process is the addition of internal standard (IS).
cards need to be completely dried prior to trans- The IS could be directly added in the extracting
portation, storage, or analysis so as to avoid solvent or spotted on the DBS card subsequent to
microbial growth and other quality defects. the spotting of the blood. For the sake of homo-
Drying time is mainly dependent on the type of geneity and reproducibility, the IS can be sprayed
filter material used and the volume of blood spot- on the card [145]. Regardless of the method used
ted. Generally, the cards are dried at room tem- to introduce the IS, the extraction efficiencies of
perature for 2–3 h or under controlled humidity various solvents and their combinations for the
using a gentle stream of nitrogen [144, 145]. analyte need to be assessed and the best solvents
Exposure to direct sunlight, dust, and insects used and extraction conditions optimized. After
should be avoided in order not to hamper the optimized extraction, the analyte(s) can be sub-
integrity of the DBS specimen [145]. jected to either qualitative or quantitative analy-
56 R. N. Alolga et al.

sis under various analytical platforms such as make sure that the blood is evenly spread on the
high-performance liquid chromatography card, and larger than the defined punch area.
(HPLC), gas chromatography-mass spectrometry Depending on the type of DBS card and the phys-
(GC-MS), liquid chromatography-tandem mass icochemical properties of the analyte, differences
spectrometry (LC-MS/MS), matrix-assisted laser in spot volume could be critical to the attainment
desorption mass spectrometry (MALDI-MS), of reproducible and accurate results from quanti-
inductively coupled plasma mass spectrometry tative analyses. A series of spot volumes ranging
(ICP-MS), enzyme-linked immunosorbent assay from 15 to 40 μL is usually recommended for the
(ELISA), polymerase chain reaction (PCR), etc. validation process. Triplicate analyses of at least
With the advent of automated online high-­ two concentrations (low and high) are recom-
throughput equipment, the sampling process and mended for all spot volumes used. Finally, the
analysis of DBS cards have become easier, faster, spot volumes used for the calibration curves and
and more accurate and precise [145]. quality control samples should be within the
range of the clinical samples under study, so that
the effect of differential spot volumes of the sam-
4.4.6 Validation of DBS-Based ples on the analyte can be controlled [147].
Analytical Processes Sample homogeneity using the liquid matrices
could be achieved by thorough mixing (vortex-
As is required of every quantitative analytical ing) and centrifugation. This is not possible with
process, DBS-based quantitative methods need to the DBS samples since the analytes within the
be validated. The guidelines by various estab- blood are spotted and spread on the card. The
lished authorities such as the US FDA, the EMA, need for spot homogeneity becomes paramount
and the ICH for the validation of bioanalytical when a specific part within the spotted area is
methods could be referenced. However, since used for analysis rather than the whole spot. In
these guidelines pertain particularly to quantita- such cases, the blood spots need to be evenly dis-
tive analyses in liquid matrices like plasma, tributed on the card, so that the spot size could be
serum, urine, etc., the solid matrix of DBS pres- equated to a fixed volume. The influence of spot
ents certain challenges that require additional homogeneity on the outcome of quantitative
validation. In addition to the known validation analysis is also dependent on the DBS card type
parameters, certain parameters applicable to only and analyte under investigation. Differences in
DBS-based samples need to be considered. The analyte content in punches gotten from the center
following parameters need special attention in and periphery of the DBS card need to be evalu-
DBS-based quantitative analysis: spot volume, ated at least three times at two concentration lev-
hematocrit (already discussed), spot homogene- els (high and low) during validation. Marked
ity, spot-to-spot carry-over, recovery, matrix differences should guide in the choice of punches
effects, stability under transport and storage con- to use (i.e., bigger or smaller) and the need to
ditions, and dilution integrity. Acceptance criteria conduct whole-spot analysis.
for these parameters similar to those of the recog- Spot-to-spot carry-over basically concerns the
nized regulatory authorities need to be met (e.g., device used to punch out the spotted DBS. This is
±15 for accuracy and precision) [146]. a source of preanalytical error that could be
Spot volume is usually controlled when cali- reduced to the barest minimum by performing
bration standards or quality control samples are “between-punches” using blank DBS cards [147].
prepared by spotting exact volumes of whole That is, after punching out the defined diameter
blood on DBS cards. In the case of clinical sam- on the DBS sample, the same device should be
ples however, this cannot be controlled since the used to perform two punches on blank DBS cards,
blood from a pricked finger or heel is applied so that the carry-over of the analyte from one
directly on the DBS cards and could differ. The punch to another could be minimized or avoided
DBS samples need to be visually checked to totally. According to the EMA guidelines, the
4 Dried Blood Spots in Therapeutic Drug Monitoring and Toxicology 57

spot-to-spot carry-over should not be more than (ion suppression) of the analyte. It is a normal
20% of the lower limit of quantification. practice to assess the matrix effects using at least
Per the guidelines of the US FDA, EMA, and six different batches of whole blood at two con-
ICH, the recovery of an analytical method deter- centrations (high and low). Hence, the following
mined at different concentration levels needs to solutions can used: (1) neat solutions at high and
be precise, reproducible, and consistent. These low analyte concentrations (100% analyte recov-
requirements hold true for DBS-based analyses. ery) and (2) blank extracts of DBS sample pre-
The addition of IS to the spot prior to extraction pared with neat solutions [146]. The influence of
could commensurate somewhat for variations in the matrix could then be calculated as follows:
the extraction efficiencies of the solvent(s) as ear- Content of analyte
lier indicated. Aging and hematocrit are known to in neat solution
affect the extraction recovery of some analytes in Matrix factor =
Content of analyte in blanck
DBS. It is recommended that a method that BDSextract in neat solutions
results in at least 85% extraction recovery of the (4.8)
analyte be used [146]. Similar to the require- Stability of the analyte at every stage of handling
ments for matrix effect evaluation during valida- from sampling, storage, transport to analytical
tion, it is recommended that the recovery be laboratories is a major quality requirement. With
assessed using a minimum of six batches of DBS respect to storage and transport, the DBS samples
cards, with different hematocrit values and at need to be stored under controlled temperature
least two concentrations. It is also advisable to and humidity. The time and route of transport of
use an accurate volume of whole blood that pro- the samples should also be considered. If the
duces a spot size within the punch size of the samples are to be transported in mailboxes via
punching instrument used so as to ensure whole-­ regular postal service, potential temperature fluc-
spot extraction. Analyses can then be performed tuations would therefore be a significant factor to
on three different types of samples, thus the DBS consider. This is particularly crucial when the
samples at low and high concentrations (here storage and transport of the samples are done
referred to as “A”), solutions that represent 100% during seasons of very high (summer) and very
recovery (i.e., neat solutions of the analyte at low low (winter) temperatures [146, 147].
and high concentrations, “B”), and solutions of Another essential validation parameter worthy
blank DBS sample prepared with neat solutions of attention is dilution integrity. It should be
(“C”). The recovery can then be calculated using assessed at concentrations that foreseeably fall
Eq. 4.7: within that of the clinical sample (DBS sample).
Content of analyte in A
This can be achieved in one of two main ways:
Recovery  %   100% (1) by diluting the final DBS extract with the final
Content of analyte in C
(4.7) blank extract that contains the IS or (2) the ana-
Matrix effect is an essential validation parameter lyte can be processed using a high concentration
in mass spectrometric analysis of biosamples. of the IS to obtain a final extract solution. This
Results of matrix effects also need to be consis- final extract is then diluted with the final extract
tent and reproducible. Following the laid down of the blank solution (obtained with the same
protocols by recognized authorities, matrix dilution factor as the final analyte extract) that
effects can be investigated and validated in DBS-­ does not contain the IS. This method is known as
based samples. Particular attention needs to be the IS track dilution method. A minimum of five
paid to the DBS samples spotted on precoated determinations for every dilution factor is
cards since these could introduce additional com- ­recommended regardless of the methodological
ponents that could adversely affect the detection approach [146].
58 R. N. Alolga et al.

4.5 Recent Innovative DBS been made by different researchers to nib this
Sampling Alternatives problem in the bud. For instance, Genk and core-
searchers recently devised a microfluidic sam-
In recent times, various innovative approaches pling device consisting of a combination of DBS
have been made in respect of improving the paper, plastic foils, and a thin dissolvable film
extraction efficiency of DBS as well as address- [150]. This device operates on capillary forces,
ing its shortfalls. An example of such was intro- while the dissolvable film valves function as a
duced by Damon and company called, the timer that allows for automation of volume
three-dimensional (3D) dried blood spheroids metering. The metering chip of this device was
[148]. Instead of the conventional hydrophilic able to collect and store microliter quantities of
cellulose-based DBS cards, they used a function- whole blood on the DBS paper substrate in 20 s.
alized hydrophobic paper substrate that was Neto et al. established an accurate and precise
obtained via gas phase silanization of blood collection and metering system capable of
trichloro(3,3,3-trifluoropropyl)silane on a preparing DBS with as low as 3 μL of whole
triangular-­shaped filter paper. The silanization blood [151]. Their proposed methodological
process of the filter paper led to a reduction in its approach proved superior to traditional microvol-
surface energy, resulting in the maintenance of ume collection systems such as using a micropi-
the shape of the blood droplets, consequently pette or an analytical syringe and also almost
forming the 3D dried blood spheroids. Using this completely solved the hematocrit problem. A
analytical strategy, they were able to tackle group of researchers from Japan, Nakahara and
known systemic challenges in traditional DBS colleagues in 2018 developed the volumetric
sampling such as the volcanic and chromato- absorptive paper disc (VAPD) and its scaled-­
graphic effects and the hematocrit factor. Due to down version (VAPDmini) for accurate and sim-
minimal interactions of the analytes with the sur- ple volumetric collection of blood for DBS [152].
face of the paper, its extraction efficiency was The VAPD is made up of a filter paper disc and a
enhanced as evidenced by extreme low LOD val- filter paper sheet with holes slightly larger than
ues. Also, the spheroidal shape of the blood drop- the disc. With the addition of whole blood to the
lets tends to protect the analyte(s) from the effects disc, saturation of same allows for the surround-
of ambient air such as oxidative degradation – ing filter sheet to absorb the excess blood. The
enhancing the collective stability of the analytes. accuracies and precisions of their devices were
In a bid to determine the content of a drug tested using clozapine and its metabolites and
with a narrow therapeutic index (carbamazepine) found to be within acceptable standards. Also,
by capillary electrophoresis, Nuchtavorn et al. the analytes’ recoveries were found to be
coated a Whatman® Grade 1 filter paper with a hematocrit-­independent (not influenced by blood
hybrid of homogenous polystyrene and silica gel with different hematocrit values).
polymer to form what they termed molecularly
imprinted-interpenetrating networks (MI-IPN)
[149]. Using the MI-IPN for extraction of the 4.6 High-Throughput
analyte, they were able to eliminate proteins and Application of DBS Sampling
other matrices that could affect the results of the
capillary electrophoresis instrument. The MI-IPN A very notable application of DBS sampling is in
provided better on-spot extraction efficiencies various metabolomics studies. The advantages of
than conventional DBS cards (Whatman® 903 this microsampling technique have been vari-
protein saver card and GenCollect™ 2.0 card). ously applied to characterize and aid in the
As early on indicated, the analyte variability ­diagnosis or identification of potential biomark-
in punched spots of DBS due to hematocrit dif- ers for different diseases. Metabolomics, which
ferences poses a major analytical challenge dur- is generally defined as the holistic qualitative and
ing its quantification. Numerous attempts have quantitative analyses of as much as practicable
4 Dried Blood Spots in Therapeutic Drug Monitoring and Toxicology 59

all metabolites present within a biological system 4.7 Conclusion and Future
under defined conditions, is an ever-evolving Perspectives
field in systems biology. It is an analytical tech-
nique that can provide a sneak peek into meta- The increased interest in DBS sampling by the
bolic processes that underlie various scientific community over the past 60 years not
pathophysiological conditions. Also with metab- only points to its gradual acceptance but also its
olomics, unique chemical fingerprints of specific potential applicability in research areas that hith-
metabolic processes can be obtained and provide erto received little to no attention. Its application
useful clues on diagnosis and treatment options in various fields under TDM with the potential
[153–155]. The metabolomes of diverse diseased for large-scale clinical adoption is worth appreci-
conditions including but not limited to the fol- ating. Among the numerous advantages associ-
lowing have been explored using DBS sampling; ated with the use of DBS sampling, the ease of
Diamond-Blackfan anemia [156], cystic fibrosis sample (whole blood) collection and the stabiliz-
[157, 158], acute lymphoblastic leukemia [159], ing effect DBS stand out. This assertion holds
biliary atresia [160], pediatric acute myeloid leu- true for both animal and human studies. For
kemia [161], colorectal cancer [162], ischemic instance, DBS sampling in animal studies allows
and hemorrhagic stroke [163], breast cancer for adherence to the principles of the 3Rs, such
[164], pyruvate kinase deficiency [165], autism that the number of animals required is reduced
[166, 167], small cell lung cancer [168], etc. and there is marked refinement in respect of the
DBS sampling has also been applied for the collection of blood without the need to warm the
detection and quantification of proteins and pep- animals (rodents) to ease blood flow. The stabi-
tides, particularly in immunoassays. The use of lizing effect conferred on the analyte by the DBS
DBS sampling for enzyme-linked immunosor- largely prevents its degradation ex vivo and/or de
bent assay (ELISA) is characterized by high novo formation of other analytes. These and
selectivity, specificity, and reproducibility of data other advantages notwithstanding, a few points
[169]. Of note, this sampling technique has been are worth noting in the use of DBS for both quali-
successfully used for the detection of antibodies tative and quantitative purposes. Due to the pos-
against various viruses such as rubella virus sibility of sample contamination and the quality
[170], Epstein-Barr virus [171], hepatitis C virus implications therefrom, much attention needs to
[172], and dengue virus [173]. The surge in the be devoted to the process of blood collection,
use of high-throughput analytical platforms such spotting, drying of the DBS cards, and transpor-
as LC-MS, LC-Q-TOF-MS, MALDI-TOF-MS, tation of the dried cards. Second, the influence of
etc. has made combinations of these platforms factors such as hematocrit variation and its atten-
with DBS sampling for both targeted and untar- dant issues as well as volume of blood spotted
geted analyses of proteins and peptides possible. and site of punching need to be addressed (as ear-
Using such combinations, it has been possible to lier outlined). Third, for the findings of any DBS-­
determine the level of ceruloplasmin during neo- based study to possess any potential clinical
natal screening for the Wilson’s disease [174, significance and be reproducible, it is essential
175], quantify C-peptide as a measure of normal that the entire analytical process be validated in
beta cell function [176], quantify amino acids accordance with the guidelines of recognized
and acylcarnitines as part of a newborn screening standard authorities.
strategy [177, 178], predict major depressive dis- As researchers work at providing innovative
order based on proteomics data [179], quantify ways to improve on this microsampling tech-
metanephrines to aid in the diagnosis of pheo- nique and tackle inherent challenges with its
chromocytoma and paragangliomas [180], screen application, the aim ultimately is to establish a
for primary immunodeficiency disorders in new- robust analytical system that could form part of
borns [181], and screen for sickle cell disease in routine tests in the clinic. As most of the pro-
neonates [182], to mention a few. cesses are being automated, in the foreseeable
60 R. N. Alolga et al.

future, it might be possible to perform all the ana- in different conditions. J Pharm Biomed Anal.
2010;52(5):774–80.
lytical steps, from DBS sampling through to 16. Ramisetti NR, Arnipalli MS, Nimmu NV. Dried
analysis using a simple but fully automated blood spot analysis of (+) and (−) darunavir
equipment. enantiomers on immobilized amylose tris-(3,
5-­dimethylphenylcarbamate) LC and its applica-
tion to pharmacokinetics. Biomed Chromatogr.
2015;29(12):1878–84.
References 17. Zheng JH, et al. Quantitation of tenofovir and emtric-
itabine in dried blood spots (DBS) with LC-MS/
1. Guthrie R, Susi A. A simple phenylalanine method MS. J Pharm Biomed Anal. 2014;88:144–51.
for detecting phenylketonuria in large populations of 18. Hooff GP, et al. Dried blood spot UHPLC-MS/MS
newborn infants. Pediatrics. 1963;32:338–43. analysis of oseltamivir and oseltamivircarboxylate-
2. Seashore MR, Seashore CJ. Newborn screening -a validated assay for the clinic. Anal Bioanal Chem.
and the pediatric practitioner. Semin Perinatol. 2011;400(10):3473–9.
2005;29(3):182–8. 19. Mei JV, et al. Radioimmunoassay for monitoring
3. Garg U, Dasouki M. Expanded newborn screening zidovudine in dried blood spot specimens. Clin
of inherited metabolic disorders by tandem mass Chem. 1998;44(2):281–6.
spectrometry: clinical and laboratory aspects. Clin 20. Ter Heine R, et al. Quantification of the HIV-­
Biochem. 2006;39(4):315–32. integrase inhibitor raltegravir and detection of its
4. Chace DH. Mass spectrometry in newborn and met- main metabolite in human plasma, dried blood spots
abolic screening: historical perspective and future and peripheral blood mononuclear cell lysate by
directions. J Mass Spectrom. 2009;44(2):163–70. means of high-performance liquid chromatography
5. Stove CP, et al. Dried blood spots in toxicology: tandem mass spectrometry. J Pharm Biomed Anal.
from the cradle to the grave? Crit Rev Toxicol. 2009;49(2):451–8.
2012;42(3):230–43. 21. Kromdijk W, et al. Use of dried blood spots for
6. Wilhelm AJ, den Burger JC, Swart EL. Therapeutic the determination of plasma concentrations of
drug monitoring by dried blood spot: progress to nevirapine and efavirenz. J Antimicrob Chemother.
date and future directions. Clin Pharmacokinet. 2012;67(5):1211–6.
2014;53(11):961–73. 22. Hoffman JT, et al. Determination of efavirenz in
7. Robijns K, Koster RA, Touw DJ. Therapeutic human dried blood spots by reversed-phase high-­
drug monitoring by dried blood spot: progress to performance liquid chromatography with UV detec-
date and future directions. Clin Pharmacokinet. tion. Ther Drug Monit. 2013;35(2):203–8.
2014;53(11):1053. 23. Shah NM, et al. A simple bioanalytical method
8. De Kesel PM, et al. Hemato-critical issues in quan- for the quantification of antiepileptic drugs
titative analysis of dried blood spots: challenges and in dried blood spots. J Chromatogr B Analyt
solutions. Bioanalysis. 2013;5(16):2023–41. Technol Biomed Life Sci. 2013;923-924:65–73.
9. Kang JS, Lee MH. Overview of therapeu- 24. Dodin YI, et al. Population pharmacokinetics mod-
tic drug monitoring. Korean J Intern Med. eling of Lamotrigine in Jordanian epileptic patients
2009;24(1):1–10. using dried blood spot sampling. Drug Res (Stuttg).
10. Sharma A, et al. Dried blood spots: concepts, present 2021;71(8):429–37.
status, and future perspectives in bioanalysis. Drug 25. Linder C, et al. Comparison between dried blood
Test Anal. 2014;6(5):399–414. spot and plasma sampling for therapeutic drug mon-
11. Rowland M, Emmons GT. Use of dried blood spots itoring of antiepileptic drugs in children with epi-
in drug development: pharmacokinetic consider- lepsy: a step towards home sampling. Clin Biochem.
ations. AAPS J. 2010;12(3):290–3. 2017;50(7–8):418–24.
12. Hinderling PH. Red blood cells: a neglected com- 26. Kolocouri F, Dotsikas Y, Loukas YL. Dried plasma
partment in pharmacokinetics and pharmacodynam- spots as an alternative sample collection technique for
ics. Pharmacol Rev. 1997;49(3):279–95. the quantitative LC-MS/MS determination of gaba-
13. Li Y, et al. The use of a membrane filtration device pentin. Anal Bioanal Chem. 2010;398(3):1339–47.
to form dried plasma spots for the quantitative 27. la Marca G, et al. Rapid assay of rufinamide in dried
determination of guanfacine in whole blood. Rapid blood spots by a new liquid chromatography-tandem
Commun Mass Spectrom. 2012;26(10):1208–12. mass spectrometric method. J Pharm Biomed Anal.
14. Searles Nielsen S, et al. Newborn screening archives 2011;54(1):192–7.
as a specimen source for epidemiologic studies: 28. la Marca G, et al. Rapid assay of topiramate in dried
feasibility and potential for bias. Ann Epidemiol. blood spots by a new liquid chromatography-tandem
2008;18(1):58–64. mass spectrometric method. J Pharm Biomed Anal.
15. D'Avolio A, et al. HPLC-MS method for the quan- 2008;48(5):1392–6.
tification of nine anti-HIV drugs from dry plasma 29. Déglon J, et al. Use of the dried blood spot sampling
spot on glass filter and their long term stability process coupled with fast gas chromatography and
4 Dried Blood Spots in Therapeutic Drug Monitoring and Toxicology 61

negative-ion chemical ionization tandem mass spec- tial method for therapeutic drug monitoring. J Pharm
trometry: application to fluoxetine, norfluoxetine, Biomed Anal. 2007;44(4):963–9.
reboxetine, and paroxetine analysis. Anal Bioanal 43. Martial LC, et al. Evaluation of dried blood spot sam-
Chem. 2010;396(7):2523–32. pling for pharmacokinetic research and therapeutic
30. Saracino MA, et al. Rapid assays of clozapine and drug monitoring of anti-tuberculosis drugs in chil-
its metabolites in dried blood spots by liquid chro- dren. Int J Antimicrob Agents. 2018;52(1):109–13.
matography and microextraction by packed sorbent 44. Rao RN, et al. Rapid determination of rifaximin
procedure. J Chromatogr A. 2011;1218(16):2153–9. on dried blood spots by LC-ESI-MS. Biomed
31. Geers LM, et al. Dried blood spot analysis for Chromatogr. 2011;25(11):1201–7.
therapeutic drug monitoring of Clozapine. J Clin 45. Tawa R, Hirose S, Fujimoto T. Determination
Psychiatry. 2017;78(9):e1211–8. of the aminoglycoside antibiotics sisomicin and
32. Alfazil AA, Anderson RA. Stability of benzodiaz- netilmicin in dried blood spots on filter discs, by
epines and cocaine in blood spots stored on filter high-performance liquid chromatography with pre-­
paper. J Anal Toxicol. 2008;32(7):511–5. column derivatization and fluorimetric detection. J
33. Meier-Davis SR, et al. Dried blood spot analy- Chromatogr. 1989;490(1):125–32.
sis of donepezil in support of a GLP 3-month 46. Heinig K, et al. Determination of mycophenolic
dose-range finding study in rats. Int J Toxicol. acid and its phenyl glucuronide in human plasma,
2012;31(4):337–47. ultrafiltrate, blood, DBS and dried plasma spots.
34. Lad R. Validation of individual quantitative methods Bioanalysis. 2010;2(8):1423–35.
for determination of cytochrome P450 probe sub- 47. la Marca G, et al. Development of an UPLC-MS/MS
strates in human dried blood spots with HPLC-MS/ method for the determination of antibiotic ertape-
MS. Bioanalysis. 2010;2(11):1849–61. nem on dried blood spots. J Pharm Biomed Anal.
35. Baldo MN, et al. Liquid chromatography tandem-­ 2012;61:108–13.
mass spectrometry (LC-MS/MS) and dried blood 48. Suyagh MF, et al. Development and validation of a
spot sampling applied to pharmacokinetics studies dried blood spot-HPLC assay for the determination
in animals: correlation of classic and block design. of metronidazole in neonatal whole blood samples.
Lab Anim. 2018;52(2):125–34. Anal Bioanal Chem. 2010;397(2):687–93.
36. Damen CW, et al. Application of dried blood spots 49. Cohen-Wolkowiez M, et al. Determining popu-
combined with high-performance liquid chromatog- lation and developmental pharmacokinetics of
raphy coupled with electrospray ionisation tandem metronidazole using plasma and dried blood spot
mass spectrometry for simultaneous quantification samples from premature infants. Pediatr Infect Dis
of vincristine and actinomycin-D. Anal Bioanal J. 2013;32(9):956–61.
Chem. 2009;394(4):1171–82. 50. Le J, et al. Comparative analysis of ampicillin
37. Nageswara Rao R, et al. Determination of gemiflox- plasma and dried blood spot pharmacokinetics in
acin on dried blood spots by hydrophilic interaction neonates. Ther Drug Monit. 2018;40(1):103–8.
liquid chromatography with fluorescence detector: 51. Page-Sharp M, et al. Penicillin dried blood spot
application to pharmacokinetics in rats. Biomed assay for use in patients receiving intramuscular
Chromatogr. 2012;26(12):1534–42. Benzathine Penicillin G and other Penicillin prepara-
38. la Marca G, et al. Rapid and sensitive LC-MS/ tions to prevent rheumatic fever. Antimicrob Agents
MS method for the analysis of antibiotic line- Chemother. 2017;61(8):e00252-17
zolid on dried blood spot. J Pharm Biomed Anal. 52. Beechinor RJ, et al. A dried blood spot analysis
2012;67-68:86–91. for Solithromycin in adolescents, children, and
39. Vu DH, et al. Determination of moxifloxacin infants: a short communication. Ther Drug Monit.
in dried blood spots using LC-MS/MS and the 2019;41(6):761–5.
impact of the hematocrit and blood volume. J 53. Gonzalez D, et al. Solithromycin pharmacokinet-
Chromatogr B Analyt Technol Biomed Life Sci. ics in plasma and dried blood spots and safety
2011;879(15–16):1063–70. in adolescents. Antimicrob Agents Chemother.
40. Reddy TM, Tama CI, Hayes RN. A dried blood spots 2016;60(4):2572–6.
technique based LC-MS/MS method for the analy- 54. Page-Sharp M, et al. Validation and application of
sis of posaconazole in human whole blood samples. a dried blood spot Ceftriaxone assay. Antimicrob
J Chromatogr B Analyt Technol Biomed Life Sci. Agents Chemother. 2016;60(1):14–23.
2011;879(30):3626–38. 55. Cohen-Wolkowiez M, et al. Developmental
41. van der Elst KC, et al. Dried blood spot pharmacokinetics of piperacillin and tazo-
analysis suitable for therapeutic drug moni- bactam using plasma and dried blood spots
toring of voriconazole, fluconazole, and from infants. Antimicrob Agents Chemother.
posaconazole. Antimicrob Agents Chemother. 2014;58(5):2856–65.
2013;57(10):4999–5004. 56. Barfield M, et al. Application of dried blood spots
42. Allanson AL, et al. Determination of rifampicin in combined with HPLC-MS/MS for the quantifica-
human plasma and blood spots by high performance tion of acetaminophen in toxicokinetic studies. J
liquid chromatography with UV detection: a poten- Chromatogr B Analyt Technol Biomed Life Sci.
2008;870(1):32–7.
62 R. N. Alolga et al.

57. Saracino MA, et al. A novel test using dried blood 71. Ganz N, et al. Development and validation of a fully
spots for the chromatographic assay of methadone. automated online human dried blood spot analysis
Anal Bioanal Chem. 2012;404(2):503–11. of bosentan and its metabolites using the Sample
58. Youhnovski N, et al. Determination of naproxen Card And Prep DBS System. J Chromatogr B Analyt
using DBS: evaluation & pharmacokinetic com- Technol Biomed Life Sci. 2012;885-886:50–60.
parison of human plasma versus human blood 72. Rao RN, et al. Liquid chromatography-mass spec-
DBS. Bioanalysis. 2010;2(8):1501–13. trometric determination of losartan and its active
59. Ansari M, et al. A simplified method for busulfan metabolite on dried blood spots. J Chromatogr B
monitoring using dried blood spot in combina- Analyt Technol Biomed Life Sci. 2012;902:47–54.
tion with liquid chromatography/tandem mass 73. Liang X, Jiang H, Chen X. Human DBS sampling
spectrometry. Rapid Commun Mass Spectrom. with LC-MS/MS for enantioselective determination
2012;26(12):1437–46. of metoprolol and its metabolite O-desmethyl meto-
60. Nageswara Rao R, et al. LC-ESI-MS/MS determi- prolol. Bioanalysis. 2010;2(8):1437–48.
nation of paclitaxel on dried blood spots. Biomed 74. Della Bona ML, et al. A rapid liquid chromatogra-
Chromatogr. 2012;26(5):616–21. phy tandem mass spectrometry-based method for
61. Xie F, et al. A dried blood spot assay for pacli- measuring propranolol on dried blood spots. J Pharm
taxel and its metabolites. J Pharm Biomed Anal. Biomed Anal. 2013;78-79:34–8.
2018;148:307–15. 75. Li Y, et al. Dried blood spots as a sampling technique
62. Agu L, et al. Simultaneous quantification of vin- for the quantitative determination of guanfacine in
cristine and its major M1 metabolite from dried clinical studies. Bioanalysis. 2011;3(22):2501–14.
blood spot samples of Kenyan pediatric cancer 76. Römsing S, Lindegardh N, Bergqvist
patients by UPLC-MS/MS. J Pharm Biomed Anal. Y. Determination of tafenoquine in dried blood spots
2021;203:114143. and plasma using LC and fluorescence detection.
63. Tripathy HK, et al. A dried blood spot assay Bioanalysis. 2011;3(16):1847–53.
with HPLC-MS/MS for the determination of 77. Lindkvist J, Malm M, Bergqvist Y. Straightforward
larotrectinib in mouse blood and its application and rapid determination of sulfadoxine and sulfa-
to a pharmacokinetic study. Biomed Chromatogr. methoxazole in capillary blood on sampling paper
2020;34(12):e4953. with liquid chromatography and UV detection. Trans
64. Dillenburg Weiss TL, et al. Evaluation of dried blood R Soc Trop Med Hyg. 2009;103(4):371–6.
spots as an alternative matrix for therapeutic drug 78. Green MD, Mount DL, Nettey H. High-performance
monitoring of abiraterone and delta(4)-abiraterone liquid chromatographic assay for the simultane-
in prostate cancer patients. J Pharm Biomed Anal. ous determination of sulfadoxine and pyrimeth-
2021;195:113861. amine from whole blood dried onto filter paper. J
65. Lee J, et al. Development of a dried blood spot sam- Chromatogr B Analyt Technol Biomed Life Sci.
pling method towards therapeutic monitoring of 2002;767(1):159–62.
radotinib in the treatment of chronic myeloid leukae- 79. Jansson A, Gustafsson LL, Mirghani RA. High-­
mia. J Clin Pharm Ther. 2020;45(5):1006–13. performance liquid chromatographic method for
66. Koster RA, et al. Fast LC-MS/MS analysis of tacro- the determination of quinine and 3-­hydroxyquinine
limus, sirolimus, everolimus and cyclosporin A in in blood samples dried on filter paper. J
dried blood spots and the influence of the hematocrit Chromatogr B Analyt Technol Biomed Life Sci.
and immunosuppressant concentration on recovery. 2003;795(1):151–6.
Talanta. 2013;115:47–54. 80. Lejeune D, et al. Simultaneous determination of
67. Veenhof H, et al. Clinical application of a dried blood monodesethylchloroquine, chloroquine, cyclogua-
spot assay for sirolimus and everolimus in transplant nil and proguanil on dried blood spots by reverse-­
patients. Clin Chem Lab Med. 2019;57(12):1854–62. phase liquid chromatography. J Pharm Biomed Anal.
68. Li F, et al. LC-MS/MS sensitivity enhancement using 2007;43(3):1106–15.
2D-SCX/RPLC and its application in the assessment 81. Kurawattimath V, et al. A modified serial blood
of pharmacokinetics of clonidine in dried blood sampling technique and utility of dried-blood spot
spots. Bioanalysis. 2011;3(14):1577–86. technique in estimation of blood concentration:
69. Lawson G, Cocks E, Tanna S. Quantitative deter- application in mouse pharmacokinetics. Eur J Drug
mination of atenolol in dried blood spot samples by Metab Pharmacokinet. 2012;37(1):23–30.
LC-HRMS: a potential method for assessing medi- 82. Cheomung A, Na-Bangchang K. HPLC with ultra-
cation adherence. J Chromatogr B Analyt Technol violet detection for the determination of chloroquine
Biomed Life Sci. 2012;897:72–9. and desethylchloroquine in whole blood and finger-­
70. Lawson G, Cocks E, Tanna S. Bisoprolol, ramipril prick capillary blood dried on filter paper. J Pharm
and simvastatin determination in dried blood Biomed Anal. 2011;55(5):1031–40.
spot samples using LC-HRMS for assessing 83. Aburuz S, Millership J, McElnay J. Dried blood spot
medication adherence. J Pharm Biomed Anal. liquid chromatography assay for therapeutic drug
2013;81-82:99–107. monitoring of metformin. J Chromatogr B Analyt
Technol Biomed Life Sci. 2006;832(2):202–7.
4 Dried Blood Spots in Therapeutic Drug Monitoring and Toxicology 63

84. Swales JG, et al. Simultaneous quantitation of met- Δ9-tetrahydrocannabinol and its main metabolites.
formin and sitagliptin from mouse and human dried J Chromatogr A. 2013;1271(1):33–40.
blood spots using laser diode thermal desorption 100. Kyriakou C, et al. Identification and quantification of
tandem mass spectrometry. J Pharm Biomed Anal. psychoactive drugs in whole blood using dried blood
2011;55(3):544–51. spot (DBS) by ultra-performance liquid chromatog-
85. Turpin PE, et al. Application of the DBS methodol- raphy tandem mass spectrometry. J Pharm Biomed
ogy to a toxicokinetic study in rats and transferabil- Anal. 2016;128:53–60.
ity of analysis between bioanalytical laboratories. 101. Gelmi TJ, Weinmann W, Pfäffli M. Impact of smok-
Bioanalysis. 2010;2(8):1489–99. ing cannabidiol (CBD)-rich marijuana on driving
86. Adatsi FK. Forensic toxicology. In: Wexler P, edi- ability. Forensic Sci Res. 2021;6(3):195–207.
tor. Encyclopedia of toxicology. 3rd ed. Oxford: 102. Davari B, et al. A sensitive LC-MS/MS assay for
Academic Press; 2014. p. 647–52. the quantification of methadone and its metabolites
87. Clavijo CF, et al. A low blood volume LC-MS/MS in dried blood spots: comparison with plasma. Ther
assay for the quantification of fentanyl and its major Drug Monit. 2020;42(1):118–28.
metabolites norfentanyl and despropionyl fentanyl 103. Odoardi S, Anzillotti L, Strano-Rossi S. Simplifying
in children. J Sep Sci. 2011;34(24):3568–77. sample pretreatment: application of dried blood spot
88. Seymour C, et al. Determination of fentanyl analog (DBS) method to blood samples, including postmor-
exposure using dried blood spots with LC-MS-MS. J tem, for UHPLC-MS/MS analysis of drugs of abuse.
Anal Toxicol. 2019;43(4):266–76. Forensic Sci Int. 2014;243:61–7.
89. Shaner RL, et al. Quantitation of fentanyl analogs in 104. Thomas A, et al. On-line desorption of dried blood
dried blood spots by flow-through desorption cou- spots coupled to hydrophilic interaction/reversed-­
pled to online solid phase extraction tandem mass phase LC/MS/MS system for the simultaneous anal-
spectrometry. Anal Methods. 2017;9:3876–83. ysis of drugs and their polar metabolites. J Sep Sci.
90. Kummer N, et al. Quantification of EtG in hair, 2010;33(6–7):873–9.
EtG and EtS in urine and PEth species in capillary 105. Saussereau E, et al. On-line liquid chromatography/
dried blood spots to assess the alcohol consumption tandem mass spectrometry simultaneous determi-
in driver's licence regranting cases. Drug Alcohol nation of opiates, cocainics and amphetamines in
Depend. 2016;165:191–7. dried blood spots. J Chromatogr B Analyt Technol
91. Luginbühl M, et al. Quantitative determina- Biomed Life Sci. 2012;885-886:1–7.
tion of phosphatidylethanol in dried blood spots 106. Protti M, et al. Determination of oxycodone and its
for monitoring alcohol abstinence. Nat Protoc. major metabolites in haematic and urinary matri-
2021;16(1):283–308. ces: comparison of traditional and miniaturised
92. Nguyen VL, Fitzpatrick M. Should phosphatidyl- sampling approaches. J Pharm Biomed Anal.
ethanol be currently analysed using whole blood, 2018;152:204–14.
dried blood spots or both? Clin Chem Lab Med. 107. Sadones N, et al. Do capillary dried blood spot con-
2019;57(5):617–22. centrations of gamma-hydroxybutyric acid mirror
93. Beck O, et al. Measurement of the alcohol bio- those in venous blood? A comparative study. Drug
marker phosphatidylethanol (PEth) in dried blood Test Anal. 2015;7(4):336–40.
spots and venous blood-importance of inhibition of 108. Ingels AS, Lambert WE, Stove CP. Determination
post-­sampling formation from ethanol. Anal Bioanal of gamma-hydroxybutyric acid in dried blood
Chem. 2021;413(22):5601–6. spots using a simple GC-MS method with direct
94. Spector LG, et al. Prenatal tobacco exposure and "on spot" derivatization. Anal Bioanal Chem.
cotinine in newborn dried blood spots. Pediatrics. 2010;398(5):2173–82.
2014;133(6):e1632–8. 109. Forni S, et al. Quantitation of gamma-­hydroxybutyric
95. Yang J, et al. Levels of cotinine in dried blood acid in dried blood spots: feasibility assessment for
specimens from newborns as a biomarker of mater- newborn screening of succinic semialdehyde dehy-
nal smoking close to the time of delivery. Am J drogenase (SSADH) deficiency. Mol Genet Metab.
Epidemiol. 2013;178(11):1648–54. 2013;109(3):255–9.
96. Ladror D, Pitt B, Funk W. Quantification of cotinine 110. Moretti M, et al. Determination of benzodiazepines
in dried blood spots as a biomarker of exposure to in blood and in dried blood spots collected from
tobacco smoke. Biomarkers. 2018;23(1):44–50. post-mortem samples and evaluation of the sta-
97. Salamin O, et al. Is pain temporary and glory for- bility over a three-month period. Drug Test Anal.
ever? Detection of tramadol using dried blood 2019;11(9):1403–11.
spot in cycling competitions. Drug Test Anal. 111. Déglon J, et al. Rapid LC-MS/MS quantification
2020;12(11–12):1649–57. of the major benzodiazepines and their metabo-
98. Luginbühl M, et al. Automated high-throughput anal- lites on dried blood spots using a simple and
ysis of tramadol and O-desmethyltramadol in dried cost-effective sample pretreatment. Bioanalysis.
blood spots. Drug Test Anal. 2020;12(8):1126–34. 2012;4(11):1337–50.
99. Mercolini L, et al. Dried blood spots: liquid 112. Świądro M, et al. Development of a new method for
chromatography-mass spectrometry analysis of drug detection based on a combination of the dried
64 R. N. Alolga et al.

blood spot method and capillary electrophoresis. J 128. Barfield M, Wheller R. Use of dried plasma spots
Chromatogr B Analyt Technol Biomed Life Sci. in the determination of pharmacokinetics in clini-
2020;1157:122339. cal studies: validation of a quantitative bioanalytical
113. Ambach L, Stove C. Determination of cocaine method. Anal Chem. 2011;83(1):118–24.
and metabolites in dried blood spots by LC-MS/ 129. Hagan AS, Jones DR, Agarwal R. Use of dried
MS. Methods Mol Biol. 2019;1872:261–72. plasma spots for the quantification of iothala-
114. Ambach L, et al. Rapid and simple LC-MS/MS mate in clinical studies. Clin J Am Soc Nephrol.
screening of 64 novel psychoactive substances using 2013;8(6):909–14.
dried blood spots. Drug Test Anal. 2014;6(4):367–75. 130. Calcagno A, et al. Influence of CYP2B6 and
115. Teran RA, et al. Stimulant use and study protocol ABCB1 SNPs on nevirapine plasma concentra-
completion: assessing the ability of men who have tions in Burundese HIV-positive patients using
sex with men to collect dried blood spots for labo- dried sample spot devices. Br J Clin Pharmacol.
ratory measurement of HIV viral load. Arch Sex 2012;74(1):134–40.
Behav. 2020;49(1):195–209. 131. Baietto L, et al. Development and validation of a
116. Jing J, et al. Automated online dried blood spot new method to simultaneously quantify triazoles
sample preparation and detection of anabolic ste- in plasma spotted on dry sample spot devices and
roid esters for sports drug testing. Drug Test Anal. analysed by HPLC-MS. J Antimicrob Chemother.
2022;14(6):1040–1052. 2012;67(11):2645–9.
117. Yuan Y, Xu Y, Lu J. Dried blood spots in doping 132. Sadilkova K, et al. Clinical validation and implemen-
analysis. Bioanalysis. 2021;13(7):587–604. tation of a multiplexed immunosuppressant assay in
118. Chang WC, et al. Determination of anabolic ste- dried blood spots by LC-MS/MS. Clin Chim Acta.
roids in dried blood using microsampling and gas 2013;421:152–6.
chromatography-tandem mass spectrometry: appli- 133. Ingels AS, et al. Dried blood spot punches for confir-
cation to a testosterone gel administration study. J mation of suspected γ-hydroxybutyric acid intoxica-
Chromatogr A. 2020;1628:461445. tions: validation of an optimized GC-MS procedure.
119. Salamin O, et al. Steroid profiling by UHPLC-MS/ Bioanalysis. 2011;3(20):2271–81.
MS in dried blood spots collected from healthy 134. Skelton VA, et al. Evaluation of point-of-care hae-
women with and without testosterone gel adminis- moglobin measuring devices: a comparison of
tration. J Pharm Biomed Anal. 2021;204:114280. Radical-7™ pulse co-oximetry, HemoCue(®) and
120. Funk WE, et al. Hemoglobin adducts of benzene laboratory haemoglobin measurements in obstetric
oxide in neonatal and adult dried blood spots. Cancer patients*. Anaesthesia. 2013;68(1):40–5.
Epidemiol Biomark Prev. 2008;17(8):1896–901. 135. Nkrumah B, et al. Hemoglobin estimation by
121. Kato K, et al. Analysis of blood spots for the HemoCue® portable hemoglobin photom-
polyfluoroalkyl chemicals. Anal Chim Acta. eter in a resource poor setting. BMC Clin Pathol.
2009;656(1–2):51–5. 2011;11(1):5.
122. Otero-Santos SM, et al. Analysis of perchlo- 136. Adam I, et al. Comparison of HemoCue®
rate in dried blood spots using ion chromatogra- hemoglobin-­meter and automated hematology ana-
phy and tandem mass spectrometry. Anal Chem. lyzer in measurement of hemoglobin levels in preg-
2009;81(5):1931–6. nant women at Khartoum hospital, Sudan. Diagn
123. Twiner MJ, et al. Concurrent exposure of bottle- Pathol. 2012;7(1):30.
nose dolphins (Tursiops truncatus) to multiple algal 137. O'Mara M, et al. The effect of hematocrit and punch
toxins in Sarasota Bay, Florida, USA. PLoS One. location on assay bias during quantitative bio-
2011;6(3):e17394. analysis of dried blood spot samples. Bioanalysis.
124. Woofter R, et al. Measurement of brevetoxin levels 2011;3(20):2335–47.
by radioimmunoassay of blood collection cards after 138. De Kesel PM, et al. Current strategies for coping
acute, long-term, and low-dose exposure in mice. with the hematocrit problem in dried blood spot
Environ Health Perspect. 2003;111(13):1595–600. analysis. Bioanalysis. 2014;6(14):1871–4.
125. Fairey ER, et al. Biomonitoring brevetoxin exposure 139. Capiau S, et al. Prediction of the hematocrit of
in mammals using blood collection cards. Environ dried blood spots via potassium measurement on
Health Perspect. 2001;109(7):717–20. a routine clinical chemistry analyzer. Anal Chem.
126. Maucher JM, et al. Optimization of blood collec- 2013;85(1):404–10.
tion card method/enzyme-linked immunoassay 140. Zanet DL, et al. Blood and dried blood spot telomere
for monitoring exposure of Bottlenose Dolphin length measurement by qPCR: assay considerations.
to Brevetoxin-producing red tides. Environ Sci PLoS One. 2013;8(2):e57787.
Technol. 2007;41(2):563–7. 141. Demirev PA. Dried blood spots: analysis and appli-
127. Denniff P, Spooner N. The effect of hematocrit cations. Anal Chem. 2013;85(2):779–89.
on assay bias when using DBS samples for the 142. Sadagopan NP, et al. Investigation of EDTA anti-
quantitative bioanalysis of drugs. Bioanalysis. coagulant in plasma to improve the through-
2010;2(8):1385–95. put of liquid chromatography/tandem mass
4 Dried Blood Spots in Therapeutic Drug Monitoring and Toxicology 65

spectrometric assays. Rapid Commun Mass 158. DiBattista A, et al. Metabolic signatures of cystic
Spectrom. 2003;17(10):1065–70. fibrosis identified in dried blood spots for newborn
143. Li W, Zhang J, Tse FL. Strategies in quantita- screening without carrier identification. J Proteome
tive LC-MS/MS analysis of unstable small mol- Res. 2019;18(3):841–54.
ecules in biological matrices. Biomed Chromatogr. 159. Petrick LM, et al. Metabolomics of neonatal blood
2011;25(1–2):258–77. spots reveal distinct phenotypes of pediatric acute
144. Li W, Tse FL. Dried blood spot sampling in com- lymphoblastic leukemia and potential effects of
bination with LC-MS/MS for quantitative analy- early-life nutrition. Cancer Lett. 2019;452:71–8.
sis of small molecules. Biomed Chromatogr. 160. Xiao Y, et al. Targeted metabolomics reveals birth
2010;24(1):49–65. screening biomarkers for biliary atresia in dried
145. Edelbroek PM, van der Heijden J, Stolk LM. Dried blood spots. J Proteome Res. 2021;21:721.
blood spot methods in therapeutic drug monitor- 161. Petrick L, et al. Untargeted metabolomics of new-
ing: methods, assays, and pitfalls. Ther Drug Monit. born dried blood spots reveals sex-specific associa-
2009;31(3):327–36. tions with pediatric acute myeloid leukemia. Leuk
146. Jager NG, et al. Procedures and practices for Res. 2021;106:106585.
the validation of bioanalytical methods using 162. Jing Y, et al. Rapid differentiating colorectal cancer
dried blood spots: a review. Bioanalysis. and colorectal polyp using dried blood spot mass
2014;6(18):2481–514. spectrometry metabolomic approach. IUBMB Life.
147. Timmerman P, et al. EBF recommendation on the 2017;69(5):347–54.
validation of bioanalytical methods for dried blood 163. Hu Z, et al. Rapid and sensitive differentiating isch-
spots. Bioanalysis. 2011;3(14):1567–75. emic and Hemorrhagic strokes by dried blood spot
148. Damon DE, et al. Dried blood spheroids for dry-state based direct injection mass spectrometry metabolo-
room temperature stabilization of microliter blood mics analysis. J Clin Lab Anal. 2016;30(6):823–30.
samples. Anal Chem. 2018;90(15):9353–8. 164. Wang Q, et al. A dried blood spot mass spectrometry
149. Nuchtavorn N, Dvořák M, Kubáň P. Paper-based metabolomic approach for rapid breast cancer detec-
molecularly imprinted-interpenetrating polymer net- tion. Onco Targets Ther. 2016;9:1389–98.
work for on-spot collection and microextraction of 165. Van Dooijeweert B, et al. Untargeted metabolic pro-
dried blood spots for capillary electrophoresis deter- filing in dried blood spots identifies disease finger-
mination of carbamazepine. Anal Bioanal Chem. print for pyruvate kinase deficiency. Haematologica.
2020;412(12):2721–30. 2021;106(10):2720–5.
150. Lenk G, et al. A disposable sampling device to collect 166. Barone R, et al. A subset of patients with autism
volume-measured DBS directly from a fingerprick spectrum disorders show a distinctive metabolic
onto DBS paper. Bioanalysis. 2015;7(16):2085–94. profile by dried blood spot analyses. Front Psych.
151. Neto R, et al. Precise, accurate and user-­ 2018;9:636.
independent blood collection system for dried 167. Courraud J, et al. Studying autism using untargeted
blood spot sample preparation. Anal Bioanal Chem. metabolomics in newborn screening samples. J Mol
2018;410(14):3315–23. Neurosci. 2021;71(7):1378–93.
152. Nakahara T, et al. Development of a hematocrit-­ 168. Yu L, et al. Metabolomic profiling of dried blood
insensitive device to collect accurate volumes of spots reveals gender-specific discriminant models
dried blood spots without specialized skills for mea- for the diagnosis of small cell lung cancer. Aging
suring clozapine and its metabolites as model ana- (Albany NY). 2020;12(1):978–95.
lytes. J Chromatogr B Analyt Technol Biomed Life 169. Malsagova K, et al. Dried blood spot in labora-
Sci. 2018;1087-1088:70–9. tory: directions and prospects. Diagnostics (Basel).
153. Tobin NH, et al. Comparison of dried blood spot 2020;10(4):248.
and plasma sampling for untargeted metabolomics. 170. Helfand RF, et al. Dried blood spots versus sera for
Metabolomics. 2021;17(7):62. detection of rubella virus-specific immunoglobu-
154. Li K, et al. Improved dried blood spot-based metab- lin M (IgM) and IgG in samples collected during
olomics: a targeted, broad-spectrum, single-injection a rubella outbreak in Peru. Clin Vaccine Immunol.
method. Meta. 2020;10(3):82. 2007;14(11):1522–5.
155. Ward C, et al. Nontargeted mass spectrom- 171. Fachiroh J, et al. Dried-blood sampling for epstein-­
etry of dried blood spots for interrogation of the barr virus immunoglobulin G (IgG) and IgA serol-
human circulating metabolome. J Mass Spectrom. ogy in nasopharyngeal carcinoma screening. J Clin
2021;56(8):e4772. Microbiol. 2008;46(4):1374–80.
156. van Dooijeweert B, et al. Dried blood spot metabo- 172. Tuaillon E, et al. Dried blood spot for hepatitis C
lomics reveals a metabolic fingerprint with diagnos- virus serology and molecular testing. Hepatology.
tic potential for Diamond Blackfan Anaemia. Br J 2010;51(3):752–8.
Haematol. 2021;193(6):1185–93. 173. Balmaseda A, et al. Evaluation of immunological
157. Al-Qahtani W, et al. Dried blood spot-based markers in serum, filter-paper blood spots, and saliva
Metabolomic profiling in adults with cystic fibrosis. for dengue diagnosis and epidemiological studies. J
J Proteome Res. 2020;19(6):2346–57. Clin Virol. 2008;43(3):287–91.
66 R. N. Alolga et al.

174. Kroll CA, et al. Retrospective determination of of carnitines in dried blood spots. Anal Chim Acta.
ceruloplasmin in newborn screening blood spots 2018;1037:177–87.
of patients with Wilson disease. Mol Genet Metab. 179. Cooper JD, et al. Schizophrenia-risk and urban
2006;89(1–2):134–8. birth are associated with proteomic changes in
175. Jung S, et al. Quantification of ATP7B protein in neonatal dried blood spots. Transl Psychiatry.
dried blood spots by peptide immuno-SRM as a 2017;7(12):1290.
potential screen for Wilson's disease. J Proteome 180. Richard VR, et al. An LC-MRM assay for the quanti-
Res. 2017;16(2):862–71. fication of metanephrines from dried blood spots for
176. Johansson J, et al. C-peptide in dried blood spots. the diagnosis of pheochromocytomas and paragan-
Scand J Clin Lab Invest. 2010;70(6):404–9. gliomas. Anal Chim Acta. 2020;1128:140–8.
177. Thompson JW, et al. Extraction and analysis of car- 181. Collins CJ, et al. Multiplexed proteomic analysis for
nitine and acylcarnitines by electrospray ionization diagnosis and screening of five primary immuno-
tandem mass spectrometry directly from dried blood deficiency disorders from dried blood spots. Front
and plasma spots using a novel autosampler. Rapid Immunol. 2020;11:464.
Commun Mass Spectrom. 2012;26(21):2548–54. 182. Hachani J, et al. MALDI-TOF MS profiling as the
178. Han J, et al. Isotope-labeling derivatization with first-tier screen for sickle cell disease in neonates:
3-nitrophenylhydrazine for LC/multiple-reaction matching throughput to objectives. Proteomics Clin
monitoring-mass-spectrometry-based quantitation Appl. 2011;5(7–8):405–14.
The Role of Artificial Intelligence 5
in Therapeutic Drug Monitoring
and Clinical Toxicity

Surovi Saikia, Jinga B. Prajapati,


Bhupendra G. Prajapati, Vijaya V. Padma,
and Yashwant V. Pathak

Abstract such as PCR plus support vector machines


(SVMs), naïve Bayes, random forest, neural
The core concept of drug design is data analy- networks and recursive partitioning are quite
sis and the methods used in such analysis. The often used for the generation of ML models by
angle of artificial intelligence (AI) in drug dis- correlating descriptors with experimental
covery is mainly concentrated towards the activity. Therapeutic drug monitoring and
methodological aspect of computation such as clinical toxicity are faced by new challenges
deep learning. With the growth in computing every day as the problem-solving approaches
power and advancement in AI methods, this are fetched with more complex causes in regu-
field including machine learning/deep learn- latory and health technology to assist the
ing (ML/DL) has moved away from theoreti- healthcare system. Drug monitoring for the
cal to real-world application. The generation patient is enhancing its functioning by various
of AI and its subset learning models are based technical tools supported with numerous treat-
on properties of the training data sets such as ment models. There exist many platforms to
physiochemical properties, quantum mechani- improve disease management by digitally
cal, 2D properties, 3D descriptors, molecular leveraging the best patient care. In this chap-
patterns, molecular finger prints, etc. Methods ter, we have discussed the various key points
of AI along with its role in monitoring drug
toxicity. Additionally, implementing AI in
S. Saikia · V. V. Padma (*) drug discovery and obtaining medical big data
Translational Research Laboratory, Department of
Biotechnology, Bharathiar University, Coimbatore, is also discussed. The pipeline which is criti-
Tamil Nadu, India cal in monitoring diseases and the implemen-
J. B. Prajapati tation of AI during pharmacovigilance are
Acharya Motibhai Patel Institute of Computer described. Other topics such as systematic
Studies, Ganpat University, Kherva, Mahesana, collection of adverse drug reactions (ADR)
Gujarat, India through spontaneous logical methods and the
B. G. Prajapati impact of AI during pharmacovigilance and
Shree S K Patel College of Pharmaceutical Education toxicity are also discussed. Finally, recent
and Research, Ganpat University, Mahesana, Gujarat,
India advances in AI for drug safety and post-­market
surveillance along with the illusions, reality
Y. V. Pathak
USF Health Taneja College of Pharmacy, University and future of AI use in drug design and dis-
of South Florida, Tampa, FL, USA covery were also mentioned.

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 67


S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_5
68 S. Saikia et al.

Keywords of data with increased automation [6]. The


advanced tools and methods used in AI can
Artificial intelligence · Machine learning · mimic human intelligence without replacing the
Deep learning · Drug design · Toxicity · physical presence of humans [7]. In this chapter,
Pharmacovigilance · Adverse drug reaction we have discussed the various key points of AI
along with its role in monitoring drug toxicity.

5.1 Introduction
5.2 AI Terms
The interdisciplinary nature of the drug discovery
field has always been on wheels fuelled by new 5.2.1 Machine Learning (ML)
ideas and development in sciences (biological or
physical) along with the application of computers Machine learning (ML) is a subset of AI which
and algorithms such as computer-aided drug enables a software, system or application to
design (CADD), chemoinformatics, machine function more accurately to predict outcomes.
learning and artificial intelligence. The core con- Machine learning algorithms use trained data
cept in all the aforementioned is data analysis and sets as input to predict new rules considering the
the methods utilized for such analysis. The artifi- underlying understanding and reasoning. Data
cial intelligence angle in drug discovery is mainly classification is a common process but when the
concentrated towards the methodological aspect data set becomes complicated with n-numbers
of computation such as deep learning [1]. The of attributes, machine learning comes into the
Food and Drug Administration (FDA) recently picture. Broadly machine learning is supervised
has been promoting the term real-world learning, unsupervised learning, semi-­
data (RWD) to be used in drug discovery which supervised learning and reinforcement learning.
means data collected from other conventional Each type has its own structure to predict differ-
research settings like billing data, administrative ent results.
claims, electronic health records, etc. [2]. The
status of treatment, disease, adherence to treat- Supervised learning: In this type of machine
ment, comorbidities, outcomes, concurrent treat- learning labelled training data are used. It
ments, etc. which can be tracked are present in defines the variables to the different labels to
such RWD. This RWD information can provide associate correlations with the algorithm.
critical evidence (real world) to develop good Unsupervised learning: In this type of machine
patient care, safety surveillance, therapeutic learning function with unlabelled data are
development, and comparative studies of effec- used. The possible useful connection is
tiveness. [3]. scanned in the data sets to process further.
The field of artificial intelligence (AI) includ- Semi-supervised learning: Semi-supervised
ing machine learning/deep learning (ML/DL) has learning is the combination of supervised
moved away largely from theoretical to real-­ learning and unsupervised learning. The algo-
world application due to the growth in computer rithm is mostly fed with labelled and training
systems and advancement in AI methods [3]. AI data. There is the freedom to model and
is used in target identification, understanding dis- explore the data on its own understanding of
ease mechanism, biomarker development and the data set.
stages of drug discovery [4]. Digitization of the
pharmaceutical sector has given birth to various
challenges such as acquiring these data and scru- 5.2.2 Deep Learning (DL)
tinizing and applying them for solving clinical
problems [5]. These challenges have brought Deep learning is a subset of machine learning
about the use of AI as it can handle large volume techniques which enables systems, software or
5 The Role of Artificial Intelligence in Therapeutic Drug Monitoring and Clinical Toxicity 69

application to operate naturally. It learns from 5.3 Implementing AI in Drug


various examples based on labelled data only. Design: Early Stages
The labelled data can be classified as images,
text, sound or other relevant sources. The com- Artificial intelligence (AI) may work as humans
puter model associated with deep learning per- and involves artificially incorporated human-like
forms classification tasks directly from labelled intelligence built using complex algorithms and
data. Associated models are using a large set of mathematical functions [8]. AI is a technology-­
labelled data in multi-layer neural network based system with advanced tools and networks,
architectures. models and algorithms to function as human
Deep learning applications are popular in intelligence without replacing the physical pres-
aerospace, defence, automated car driving, elec- ence of humans completely. AI provides such
tronics, healthcare, etc. To detect the particular system which learns from the input data to make
disease cell automatically, high-dimensional data independent decisions for completing specific
set of concerned diseases are classified as per objectives [9].
need. The trained data is modelled to identify the Drug designing involves huge expenditure,
concerned disease cell. time and high cost along with many existing fac-
tors that affect the success and failure of drug dis-
Reinforcement Learning Here the machine is covery, development and marketing. It is a slow
given the complete multi-step process based on process with high investment and is quite chal-
earlier defined rules which is reward-based learn- lenging due to rapid changes in drug demand and
ing that performs on the principle of feedback. technological developments [10]. Pharmaceutical
Tasks are completed with positive or negative companies and academic institutions spend a lot
cues in the flow but majority of the algorithms for commercial potential and societal benefit. A
decide the next steps. In disease cell identifica- huge amount of experimental data has been accu-
tion processes, the feedback from various sources mulated from the past decades, including the bio-
will be fitted into the desired reinforcement chemical backups and clinical trials which may
model. The model will then predict cell legacy become a valuable source for learning and under-
belonging to a particular disease or not based on standing the success patterns and failures of com-
rewards and feedback fitted in the data set. pounds in the entire drug discovery process. Data
collection, analysis and digitalization for drug
design are rapidly increasing in the pharmaceuti-
5.2.3 Artificial Neural Network cal sector. Digitalization of data has many chal-
(ANN) lenges such as appropriate data collection,
scrutinizing relevant attributes and then applying
ANN is similar to the human brain that consists that knowledge to selected data frames to solve
of connected neurons which communicate via complex clinical problems [11].
signals. It works with three layers which are Many subsets exist in AI such as ML-DL, neu-
input layer, hidden layer and output layer. Data ral network, expert system, robotics, machine
are fed to the input layers which forward them to vision, speech recognition, etc. Such technology
the hidden layer. The main operations are per- provides a machine with the capability to learn
formed at hidden layers, and after completion, from data and experience through algorithms [8].
results are forwarded to output layer. Generally, The integration of various tools enables AI to
ANN works with all nodes interconnected to serve as an end-to-end point in drug design and
each other with output node value and the asso- development with millions of experimental data
ciated weight with each link. The ANNs are available in the public domain. The experimental
based on making the right connections with sili- data includes multidimensional attributes with a
con and wires as live neurons and dendrites to wide range of elements. Properly curated data
provide the best output. should be considered for the generation of pre-
70 S. Saikia et al.

cise AI learning models. Different types of AI leveraging on the best patient care. Such systems
methods are widely used in small molecule drug also supplements the physician’s information
designing using either supervised or unsuper- processing. AI is amongst one of the most appro-
vised learning methods. Unsupervised methods priate ways to provide solutions for personalized
cluster the molecules based on chemical similar- drug monitoring [2]. Figure 5.1 represents the
ity. Clustering methods are useful to identify the functioning of AI in drug monitoring and
nearest neighbours and have a greater application toxicity.
in repurposing the off-target prediction.
Clustering methods with shifting, Gaussian mix-
ture can be applied to yield great results. The 5.4 Essentials for Implementing
models for data sets, having the experimental AI in Drug Discovery
activity, are predictive methods, with either quan-
titative continue data or qualitative categorical The current computational drug discovery pro-
based data on the experimental activity of the cess has certain weakness which exhibits com-
training data. Generating models for each protein plex challenges when AI is applied. These
or type of disease when applied may classify the challenges include the basic chemical and bio-
unexplored data more precisely to identify new logical differences which are both crucial for the
hits. However, the quality of input data mainly application of AI in drug designing and monitor-
dictates the precision with many models and this ing. Chemistry has been well described from data
can accelerate the drug design process. available from drug assays which can be used for
Supervised learning methods will have a major modelling. However, biology has its own diffi-
role to identify the druggability of compounds culty in setting its own finite parameters for
based on ADMET data. The most druggable which uncertainty exists. Some of the chemical
compounds for biological studies can be obtained and biological differences are discussed under
by building a highly predictive model for the following subheadings [5].
absorption, distribution, metabolism, excretion and
toxicity; using adequate samples and filtering
the compounds during the screening process 5.4.1 Data Describing the System
[12]. Physiochemical properties, quantum
mechanics, 2D properties, 3D descriptors, molec- In the chemical information field, small molecule
ular patterns and molecular finger prints of the in its entirety (crystal packing) can be described
training data sets are used to generate the AI and by chemical structures; however, dynamic chem-
its subset learning models. Methods such as PCR ical states such as tautomeric ones remain
plus support vector machines (SVMs), naïve unclear. For biological information, the signal
Bayes, random forest, neural networks and recur- type may be unclear and thus the information is
sive partitioning are often used for the generation unclear [14].
of ML models by correlating descriptors with
experimental activity. AI associates along with
their role in drug discovery, design or toxicity 5.4.2 Integration of Data
monitoring are listed below in Table 5.1.
Therapeutic drug monitoring and clinical tox- Once the chemical structures are determined,
icity face new challenges every day. Problem-­ their representation remains static across
solving approaches are required in regulatory and experiments. On the other hand, due to the dif-
health technology to assist the healthcare system ferences in bioactivity across various assays
[13]. Drug monitoring for the patient can be and laboratories, performances of models dif-
enhanced with various technical tools supported fer [15]. Biological data are often not repro-
with numerous treatment models [11]. There are ducible as the results are highly dependent on
many platforms to improve disease management, assay or system used for the experiment.
5
Table 5.1 Artificial intelligence in drug discovery, design and development
AI associates AI involvement URL of AI associates
BenevolentAI, To identify new drug candidates for chronic https://www.benevolent.com/news/
AstraZeneca kidney disease and idiopathic pulmonary astrazeneca-­starts-­artificial-­intelligence-­collaboration-­to-­accelerate-­drug-­discovery
fibrosis
Insitro, Gilead To identify new drug targets for non-alcoholic https://www.gilead.com/news-­and-­press/press-­room/press-­releases/2019/4/
steatohepatitis gilead-­and-­insitro-­announce-­strategic-­collaboration-­to-­discover-­and-­develop-­novel-­therapies-­
for-­nonalcoholic-­steatohepatitis
Exscientia, Rallybio To discover small-molecule drugs for rare https://www.biospace.com/article/
diseases exscientia-­enters-­broad-­rare-­disease-­drug-­discovery-­collaboration-­with-­rallybio/
Microsoft, Novartis To identify and develop therapeutics https://news.microsoft.com/transform/
novartis-­empowers-­scientists-­ai-­speed-­discovery-­development-­breakthrough-­medicines/
ZebiAI Therapeutics, To discover small-molecule drug candidates https://www.globenewswire.com/news-­release/2021/04/16/2211478/0/en/Relay-­Therapeutics-­
Google Accelerated Extends-­Leadershipin-­Integrating-­Computational-­and-­Experimental-­Approaches-­to-­Create-­
Science Precision-­Medicines-­by-­Acquiring-­ZebiAI.html
Exscientia, Bayer To discover cardiovascular and oncology drug https://media.bayer.com/baynews/baynews.nsf/id/
candidates Bayer-­Exscientia-­collaborate-­leverage-­potential-­artificial-­intelligence-­cardiovascular-­
oncology
BioSymetrics, Sema4, To predict onset and severity of COVID-19 https://www.prnewswire.com/news-­releases/biosymetrics-­collaborates-­with-­janssen-­and-­
Johnson & Johnson among different populations, with a goal of sema4-­to-­predict-­the-­onset-­of-­covid-­19-­301114823.html
developing new treatments and vaccines
Recursion In drug development, to deal with new https://media.bayer.com/baynews/baynews.nsf/ID/
Pharmaceuticals, Bayer small-molecule therapies to treat fibrotic Bayer-­collaborates-­Recursion-­strengthen-­digital-­discovery-­advance-­therapies-­fibrotic-­diseases
diseases
Insitro, Bristol Myers To identify potential drug targets by developinghttps://www.businesswire.com/news/home/20201028005276/en/
Squibb predictive models of amyotrophic lateral insitro-­Announces-­Five-­Year-­Discovery-­Collaboration-­with-­Bristol-­Myers-­Squibb-­to-­
sclerosis and frontotemporal dementia Discover-­and-­Develop-­Novel-­Treatments-­for-­Amyotrophic-­Lateral-­Sclerosis-­and-­
Frontotemporal-­Dementia
Genesis Therapeutics, To identify drug candidates for a range of https://www.businesswire.com/news/home/20201019005182/en/
Genentech disorders Genesis-­Therapeutics-­Enters-­AI-­driven-­Multi-­Target-­Drug-­Discovery-­Partnership-­with-­
The Role of Artificial Intelligence in Therapeutic Drug Monitoring and Clinical Toxicity

Genentech
Roivant, Silicon In silico small-molecule drug design https://silicontx.com/news/press-­releases/
Therapeutics roivant-­grows-­computational-­drug-­discovery-­engine-­with-­acquisition-­of-­silicon-­therapeutics/
Exscientia, University of Collaborate to develop treatments for Alzheimer https://www.ndm.ox.ac.uk/about/news/
Oxford disease exscientia-­and-­the-­university-­of-­oxford-­announce-­partnership-­to-­develop-­treatments-­for-­
alzheimer2019s-­disease
Iktos, Pfizer De novo design software to a number of Pfizer’s https://www.businesswire.com/news/home/20210302005501/en/
small-molecule programmes Iktos-­Announces-­Collaboration-­With-­Pfizer-­in-­AI-­for-­Drug-­Design
71
72 S. Saikia et al.

Naive Bayes Out put

Clinical SVM

Decision Tree
Dosimetric
AI K Nearest Neighbor Drug Monitoring

Radiomic Linear regression Drug Toxicity

Random Forest.

Fig. 5.1 Functioning of AI in drug monitoring and toxicity

Corrections required for batch effects, normal- 5.4.5 Field Knowledge


ization and integration of data measured in
different scales, time points and resolutions Proper understanding of the chemical field such
are needed, viz. histopathology images, as physical, chemical and thermodynamics is rel-
and single cell RNA-seq data integration. [16]. evant for the display of the chemical information.
The biological field is still not well understood
and with limited knowledge. A relevant signal for
5.4.3 System Stability certain types of biological endpoints remains
unclear; this includes the pathways/molecule for
A chemical structure needs to be highly stable a given disease [21, 22].
when used as a drug so that it can be easily syn-
thesized. Significant compound degradation does
not occur when stored for a long time because deg- 5.5 AI or Ligand Discovery:
radation may lead to a problem of compound Which One Is Critical in Drug
libraries [17]. Cell plasticity and heterogeneity Discovery?
present difficulty to biological systems because
this leads to different drug responses within sub- Significant attention has been given to de novo
population of cell lines and tumours [18]. drug design wherein computation devises
and novel structures are desired [23]. Fields of
retrosynthesis prediction and forward prediction
5.4.4 Dimension [24] which screens the chemical worth of experi-
and Interdependency mental investigation are also relevant. The area of
ligand binding to target has been revived recently
Chemical structures can be represented with with large-scale comparisons [25], applying
high dimension, while no dependency is methods such as matrix factorization [26] and
observed among the dimensions [19]. deep learning [27]. AI usage in the chemical have
Biological information has medium to high been of research interest in recent times. As suf-
dimension and sometimes even higher interde- ficiently well-labelled data prevail in these areas,
pendency among data types is observed. This data mining and computational analyses have
can be observed among protein interactions, positive impact in protein-ligand interactions.
gene expressions, etc. [20]. Deep learning has however some impact in
5 The Role of Artificial Intelligence in Therapeutic Drug Monitoring and Clinical Toxicity 73

increasing the performance measure [28] but ated with the emerging medical technologies.
recent large-scale studies have not corroborated Examples of such big data integration from large
this data [29]. When special emphasis is paid on population for helping drug discovery process are
the metrics of model performances, practical and Alzheimer’s Disease Neuroimaging
significant tweaks in model quality are identified, Initiative (ADNI), UK Biobank, Osteoarthritis
which are were practical and relevant [30]. Even Initiative (OAI), The Cancer Genome
though the efforts for identifying disease driver Atlas (TCGA), etc. In the near future, millions of
genes are of great interest, this alone does not such patient data will be integrated across multi-
offer an easy path for target identification [31]. ple diseases and with exponential growth of data
An integrated approach is currently required compiled within biomedical databases such as
for AI in drug discovery including factors such as those maintained by the US National Center for
protein-ligand activity and PK properties of the Biotechnology Information (NCBI) and
compound. The steps involved in quantitative European Bioinformatics Institute (EBI).
systems pharmacology (QSP) include interaction When such huge medical data are captured,
modelling of small molecules with the interact- one demerit remains to be the proper availability
ing partners, understanding the expression of tar- and selection of standard data sets in machine-­
get in diseased tissue and its role in disease readable forms. Data complexity, scarcity and
modulation, the PK behaviour in the molecule in heterogeneity further add to the problem, making
relation to the in vivo system and considerations data capturing a huge challenging task. In the
for safety and efficacy [32]. data life-cycle management step, the integration
The application of AI in drug discovery is of multimodal massive data produced by differ-
rather more fragmented in the current situation. ent technologies poses a significant difficulty
AI is capable of designing ligands as per the with respect to reliability and consistency of
interested protein making the synthesis much using attributes. Availability to curated and accu-
easier, thus helping in the discovery of ligands. rate data is a critical aspect when it comes to
Technical facilities as those offered by IBM improving machine learning (ML) repeatability.
(https://www.ibm.com/in) and other robotic plat- However, good computer hardware architecture
forms [33] to achieve multi-objective optimiza- settings in relation to the requirements of life sci-
tion endpoints have been around for some time ences can solve these issues as most of the infor-
[34]. These technical facilities however do not mation is deported to cloud. For example, FAIR
enable us to design an efficacious drug in vivo. guiding principles [35] and Clinical Data
Thus, we need better decisions on considering Interchange Standards Consortium (CDISC) [36]
the type of molecule for further studies but very are such efforts made in this line. The implemen-
little of such data is provided by the proxy data tation of coherent and functional data control is
we possess in practice. The question remains on imposed by agencies such as the US Health
how these decisions can be made and how AI can Information Technology for Economic and
help in decision making and to what length. Clinical Act and European General Data
When these fundamental questions are answered, Protection Regulation (GDPR) for regulatory
AI can be of great help in the drug discovery demands for storage, sharing and access to sensi-
sector. tive and personal data of patients [37].
Collaboration between academic labs and
pharmaceutical companies such as Drug Target
5.6 AI in Drug Monitoring: Commons [38] or MELLODY provides novel
Gathering Medical Big Data initiatives to gather, curate and share huge data
for ML-based algorithm development. Several
The physiology and pathophysiology of a disease drug companies were brought under the same
can now be characterized at an unprecedented umbrella to share their individual chemical librar-
level along with the environmental risk associ- ies for training predictive algorithms (multitask
74 S. Saikia et al.

ones) by MELLODY. The challenge is to stan- to the incorporation of genomic data, biochemi-
dardize the proposed reference data set for bench- cal data and target manageability [46]. “Open
marking and testing new algorithms by multiple Targets” is one such platform for target predic-
crowd-source challenges such as PrecisionFDA tion which consists of gene-disease association.
[39], Kaggle [40] and Dream [41] in order to This has shown that animal models exhibiting a
address complex medical problems. disease-related phenotype with a neural network
classifier greater than 71% makes the most accu-
rate prediction [47]. Five new RNA-binding pro-
5.7 Pipeline for Disease teins (RBP) were identified by IBM’s Watson AI
Monitoring drug discovery platform related to the pathogen-
esis of amyotrophic lateral sclerosis (ALS) [42].
Disease monitoring has been in existence for Computational methods are being developed
about a decade and remains to be a critical aspect to identify disease-related proteins or genes of
in detecting outbreaks and epidemics. Traditional which deregulated molecular pathways of a cer-
disease monitoring relied on time-consuming lab- tain disease can be constructed from protein-­
oratory diagnosis, while the computer-based sys- protein interaction [48] or reconstructed from
tem is significantly fast to alert hospitals, state and correlation or Bayesian network [49], also known
medical officers. The latter relies on databases, as knowledge graphs. Further computational
intelligent systems, advanced informatics and analysis of the congenital topology in such graphs
advanced analytical techniques such as text min- leads to identification of nodes [50, 51]. Nodes
ing, social-network analysis, time-series analysis, exhibiting little or no evidence of disease link are
monitoring, visualization and analysis. Thus, with amplified using network propagation algorithm
these advances real-time or near to that identifica- [52], with the challenge of multi-layer network
tion of serious diseases and future exposure to integration [53] and their large-scale representa-
bioterrorism agents can be detected [42]. tion [54].
The merging of biotechnologies and AI has pro-
vided us with the opportunity to create disease
models which can help to position therapies to tar- 5.7.2 Design, Optimization of Drug
geted subpopulations. Extensive molecular profil- Design
ing of patients in comparison with normal
individuals is required for such models. Multi-­ The use of chemical space is required in the utili-
omics technologies are used to represent subtypes zation of AI for the discovery of small drug-like
of a disease based on the underlying pathophysio- molecules. Novel and high-quality molecules are
logical conditions [43]. This type of information is discovered from this stage of chemical space due
generated by the follow-up of large groups of peo- to computational cataloging of probable organic
ple via public-private partnership through patient compounds [43]. Also, the identification of vir-
arrangement using supervised and unsupervised tual molecules specifically designed for targets is
learning methods. Such clustering is based on clini- made possible by the predictive software model
cal phenotypes which aid precision medicine [44]. and ML along with the optimization of safety and
Conventional bioinformatics cannot integrate mas- efficacy data.
sive and multimodal data for which comprehensive Repurposing drugs is possible using the prox-
modelling by AI is required [45]. imity analysis of network-based methods used
for drug-target interactions [55] such as deepDT-
net algorithm [55]. Deep learning and neural net-
5.7.1 Target Selection, Validation works together with better versions of algorithms
and higher computational power have addressed
Currently, AI has changed the way pathway or the gradient problem and overfitting of quantita-
targets are identified for a particular disease due tive structure-activity relationships (QSAR) anal-
5 The Role of Artificial Intelligence in Therapeutic Drug Monitoring and Clinical Toxicity 75

ysis of the last decade [56]. Drug interactions cognitive function, sleep patterns, motricity,
with targets in large ligand-based virtual screens symptoms, pain, etc. can be mined using AI [63].
are monitored using trained ML-based neural AI and ML are used to analyse data from both
networks which are also used to predict absorp- successful and failed studies to generate models
tion, distribution, metabolism, excretion and tox- capable of predicting multiple clinical parame-
icity (ADMET) or repurpose [57]. Bioactivity ters of multimodal nature [64]. All these analyses
and ADME prediction can also be performed are essential for biomarker prediction for sever-
using deep learning as it allows multitask predic- ity, progression, and response to treatment [65].
tion. Besides accuracy, multitask prediction The acceptance of such virtual trials by regula-
enhances drug monitoring as compared to classi- tory agencies remains a major challenge in the
cal ML methods such as support vector machine application of AI in clinical studies for synthetic
and random forest [58]. patients, virtual trials, validation of algorithms
and digital endpoints. Few real-world clinical tri-
als will still be needed which may be simple and
5.7.3 Clinical Studies (Virtual) better guided by AI.

Recognition of disease in patients, gene target


identification and prediction of molecular effi- 5.8 AI in Pharmacovigilance
cacy along with the on-and off-target effects are
the assistances provided by AI in clinical trials. It Monitoring the safety of drug applications is a
was observed in AiCure, an AI-based mobile common issue in today’s era [10]. The systematic
application, that it increased adherence by 25% collection of adverse drug reactions (ADR)
when compared to the traditional way of medica- through spontaneous logical methods and analy-
tion adherence [59]. In Phase II and III clinical sis of adverse events associated with the use of
trials, AI approaches are developed for identifica- drugs are very essential to solve emerging prob-
tion and prediction of human-relevant biomark- lems, record signals and communicate to mini-
ers, thus helping in the recruitment of certain mize or prevent harm. Safety is always a major
specific patient pool which has the possibility to concern in any drug design process. During cer-
increase the success rate of clinical trials [60]. tain clinical trials, the use of the drug may be a
The efficacy and success rate of a drug can be major source of erosion is unpredictable toxici-
improved by using AI during the design, execu- ties which may cause morbidity [9]. To manage
tion and monitoring [61]. Disease understanding such unpredictable toxicities, the etiquette data
and patient heterogeneity helps in the recruitment will play a vital role with AI.
of patients for trials. Real-world evidence or AI has a wide scope of covering heteroge-
health records are mined using natural language neous sectors such as medical diagnosis, clinical
processing to examine the eligibility of patients situations, trial treatment, and toxicity detection
for clinical trials [62]. The selection and identifi- and prediction. It is used in various healthcare
cation of patients is done by automated text min- sectors to monitor health problems. AI provides
ing which fulfils the inclusion criteria such as different subsets as machine learning and deep
specific target organs, disease severity and back- learning with many algorithms to extract feature
ground therapies. The design of innovative preci- attributes from a large data [66].
sion medicine trials is possible by incorporating There are different stages in license holder
huge imaging (medical), biological and clinical ADR data collector to authenticate pharmaceuti-
data that define patient’s specificity. Trial moni- cal companies and local drug regulators. The
toring is done by AI in remote fashion for patients same data set will be recreated with feature
using wearable devices or sensors. Therapeutic abstraction, feature alignment, noise removal,
or symptomatic decisions are easy to make for splitting and labelling desired attributes to imple-
the physician when critical information such as ment relevant AI-ML algorithms. AI can shorten
76 S. Saikia et al.

the process of detection, reporting and labelling lance chain value and this was to amplify it by
technical terms of ADRs associated with indi- involving the contextual analysis and cognitive
vidual reports and their relationship with the sus- load theory. The investigators recognized 51
pected drug. AI has introduced the minimum contending decision points and validated the
human interface during the collection of ADRs. process to develop clarity about the information
Big data cannot be analysed manually, however, of pharmacovigilance. Further, the SMEs were
AI an be very helpful. integrated and acceptability was validated by
AI techniques play a significant role in phar- employing quality inspections. The results of
macovigilance by predicting the related ADRs validation of such 126 cognitive services were
from the well-collected data set. The authenti- interesting; however, the acceptability of the
cated data set can be fitted into various working proposed use of AI needs more confirmation
models to predict the desired effect. The reposi- [68].
tory of ADRs is analysed with numerical and cat- Danysz et al. discussed the present status of
egorical data. The data set is classified as per the pharmacovigilance and drug safety profession-
selected model. Various algorithms like linear als, along with an investigation carried out at
regression, logistic regression, decision tree, Celgene’s Global Drug Safety and Risk
SVM, naïve Bayes algorithm, kNN, K-means, Management (GDSRM). The outcomes of the
random forest, dimensionality reduction algo- aforementioned investigation indicated that phar-
rithms, etc. are used on the pre-processed data set macovigilance professionals are in the favour of
to the derived prediction of ADRs and their utilizing their knowledge and expertise at the full
effects. Figure 5.2 depicts AI impact in pharma- extreme for imparting value to their activities. It
covigilance/ADR repositories. was suggested that machine learning algorithms
AI and machines are utilized in enhancing may facilitate the improvement in decision mak-
the understanding of the science of drug safety. ing with regard to drug safety and better control
The impact of pharmacovigilance starts with the over the processing of cases [69]. The in-depth
development process employing in vitro and knowledge of artificial intelligence in pharmaco-
in vivo studies and proceeds into clinical trials vigilance will prove to be beneficial for setting up
and post-marketing surveillance [67]. Mockute the methods, action plans and data sets [70].
et al. proposed an idea about the pharmacovigi-

PV Report ADR Repositories

Case
ADR Data
Data classification

narrative
AI Techniques Implementation
License
authority
Casualty
ADR Prediction
Predication
Data Duplicate
entry search.

Triage

Fig. 5.2 Artificial intelligence and its impact in pharmacovigilance


5 The Role of Artificial Intelligence in Therapeutic Drug Monitoring and Clinical Toxicity 77

Although the technological advancements models for envisaging the probability that an
related to artificial intelligence and machine individual will develop an adverse outcome of
learning have shown a lot of potential in the phar- acute coronary syndrome by using their drug
macovigilance and drug safety field, its imple- history and comorbidities as inputs. The explain-
mentation in everyday pharmacovigilance has able artificial intelligence (XAI) was employed
been problematic. The implementation should in quantifying the role of each drug and a
take place readily. Enhancing patient safety and method for incorporating the principles of
synchronization of technologies with current XAI. The research comprised of using multiple
pharmacovigilance processes is also a challenge models to check whether the individuals aged
[71]. Routray et al. designed a method based on over 65 (administered with musculo-skeletal
augmented intelligence for recognizing the system or cardiovascular system drugs) between
adverse drug reactions from the extemporaneous, 1993 and 2009 were recognized and the drug
supplicated and medical reports. The study history, comorbidities and other characteristics
involved the development of three neural net- were taken out from Western Australian data
works for approaching recognition of the severity sets. It was found out that adverse reactions
of adverse reactions, like classifiers for a level of related to acute coronary syndrome had some
adverse events, classifier for severity categoriza- linkages with dispensing attributes for rofecoxib
tion of adverse events and annotator for recogniz- and celecoxib and prediction of the adverse
ing the severity criteria. The outcomes of the results was 72% accurate. Moreover, the local
study showed that the strategy of using neural interpretable model-agnostic explanations
networks has sufficient accuracy and scalability (LIME) and shapely additive explanations
[72]. (SHAP) were able to recognize essential and
The assessment of textual information from non-essential characteristics, although SHAP
various sources may be evident for pharmaco- performed slightly better than LIME [12].
vigilance professionals to get a better under-
standing of the use and effects of medicinal
compounds in individuals. Text mining strategy 5.9 AI in Toxicity
can be improvised to fit in various fields by
using machine learning approach which is char- The study of drug toxicity is one of the key
acterized by experts who employ guidelines of parameters for regulatory approvals. Almost
annotation. Thompson et al. put forward a con- 90% of new drug moieties failed, of which
noted body of work from about 597 abstracts around 40% of failure was due to safety [12]. In
from MEDLINE, and PHAEDRA, which the last decade, AI is extensively studied for
gave evidence of drug effects and interactions. drug discovery, diagnosis, toxicity studies and
The literature has furnished a preliminary illus- clinical research which reduce time, and
tration of mining textual information by training increase productivity and quality [75]. AI in tox-
baseline classifiers for the named entities and icity studies also improves accuracy and multi-
events. This occurs by employing available tools level correlations [76]. In the developing world,
which can also be implemented to other infor- the demand to reduce animal studies at laborato-
mation sources as well [73]. Segura-Bedmar ries attracts the focus of regulatory agency to
et al. highlighted the implementation of the nat- replace the traditional animal study with AI and
ural language processing and text mining tech- ML-based in silico models [77]. Figure 5.3 indi-
niques to pharmacovigilance for resolving the cates an analysis of the different types of toxici-
impenetrable problems and as a prospect for ties using AI.
future research [74]. Ward et al. designed the
78 S. Saikia et al.

Skin &
other Neuro Haemolytic Cardio Cytotoxic Kidney Liver
Phototoxicity

Fig. 5.3 Analysis of different types of toxicity using AI

cial neural networks (ANN), decision trees and


5.10 Structural Toxicity support vector machines (SVM) can be
for Endpoints used [83]. Recent years have seen deep learning
with rectified linear unit (ReLU) activation
Various kinds of toxicity studies are typically function and architectures such as recurrent
used to investigate specific adverse reactions or neural networks (RNN) and convolutional neu-
endpoints of the drug. The common classes of ral networks (CNN). These have emerged as the
toxicities are liver toxicity, cancer/mutagenicity, most important tools for in silico toxicity
cardiotoxicity, cytotoxic effects, neurotoxicity, [84–87].
radiotoxicity, skin irritation/sensitization, and In 2016, Mayr et al. created a DeepTox pipe-
phytotoxicity [78]. The studies usually conclude line for predicting toxicity using artificial intel-
the mortality, behaviour, reproductive status or ligence. The model was ascertained by a large
physiological and biochemical changes. In gen- number of data inputs of important chemical
eral toxicity studies are divided into in vivo, features representing particular compounds.
in vitro and in silico approaches [79]. With the They proved that DL outshined other AI
increasing cost of experiments and ethical issues approaches such as naïve Bayes, SVM and RF
over animal studies, gradually such studies are [88]. Allen et al. demonstrate that adverse out-
drifting away from in vivo to in silico [80]. come pathway uses a logical sequence of trials
The structure-activity relationship-based within its biological system that can understand
models can provide both qualitative and quanti- toxicity. The same framework is applied in
tative toxicity endpoint data by molecular examining the molecular initiating event (MIE)
descriptors and making predictive algorithms drugs. Studies proved that MIEs can be identi-
[81, 82]. For this purpose, linear method-based fied and characterized despite lack of detailed
models such as multiple linear regression reports, even for some of the most studied mol-
(MLR), partial least squares (PLS) and linear ecules in toxicology [89].
discriminant analysis (LDA) to non-linear meth- In 2018, Ambe et al. used DL, RF and SVM
ods, such as k-nearest neighbours (KNN), artifi- to create predictive classification models of
5 The Role of Artificial Intelligence in Therapeutic Drug Monitoring and Clinical Toxicity 79

hepatocellular hypertrophy extracting data on ficient for each feature [91]. Chang et al. evalu-
rats from two toxicological databases. The study ated digoxin (low therapeutic window drug)
revealed SVM model using the Hazard cardiotoxicity using AI-based electrocardiogra-
Evaluation Support System Integrated Platform phy (deep learning model constructed on ECG
data set, trained with 251 chemicals and pre- manifestations). The study of 61 ECGs from
dicted 214 test chemicals exclusive to the appli- patients suffering from digoxin toxicity and
cability domain. The model has a prediction 177,066 ECGs from various patients indicated
accuracy of 0.76, sensitivity of 0.90 and area that the proposed model is possibly more appli-
under the curve of 0.81 [90]. cable to patients with heart failure (HF) and
Recently, Liu et al. performed Critical without atrial fibrillation (AF) than those with-
Assessment of Massive Data Analysis (CAMDA) out HF and with AF [92].
using chemical structure-based descriptors such Allergic or hypersensitive reactions are the
as structural fingerprints and predicted protein most common type of skin toxicities, due to addi-
targets by utilizing binary DILIrank annotations tives of cosmeceuticals or topical preparations.
for prediction of drug-induced liver injury (DILI). Peiwen et al. studied and proved that an ML-based
They concluded that perception based on pro- prediction model can be used to forecast the skin
teins and their pathways mathematically allied to sensitizing probability and effectiveness of com-
DILI [89]. Usually, acute oral toxicity (AOT) is pounds. They created and compare various binary
specified by median lethal dose, LD50. Xu et al. and ternary classification models based on a
studied various kinds of in silico methods mechanism using the OECD’s eChemPortal data-
designed for AOT prediction intending to base and the NICEATM databases [93]. Verma
decrease cost and time. The team developed an and Matthews investigated possible ocular irrita-
upgraded molecular graph encoding convolu- tion and safety concerning household and cosme-
tional neural network (MGE-CNN) architecture ceutical chemicals using the in silico method as
to notion three sorts of superior AOT models: the rabbit Draize test is not permissible as per EU
regression model (deepAOT-R), multi-­regulations. The research included 21 ANN
classification model (deepAOT-C) and multitask c-QSAR models (QSAR-21) to envisage ocular
model (deepAOT-CR) [90]. The study utilized toxicity by means of the ADMET Predictor pro-
two exterior data sets comprising 1673 (test set I) gramme and an assorted training data set of 2928
and 375 (test set II) composites, the R2 and mean compounds. The findings indicated that the com-
absolute errors (MAEs) of deepAOT-R on the test bined quantitative structure-toxicity relationship
set I were 0.864 and 0.195, and the prediction (QSTR) models by ANN resulted in 88% sensi-
accuracies of deepAOT-C were 95.5% and 96.3% tivity and 82% specificity for EI and 96% sensi-
on test sets I and II, respectively. The two exter- tivity and 91% specificity for eye corrosion (EC).
nal calculation accuracies of deepAOT-CR were The developed in silico models for EI/EC using
95.0% and 94.1%, while the R2 and MAE were ML approaches and molecular fingerprints
0.861 and 0.204 for test set I, respectively. This ­collected data manually from X-Mol (http://
novel architecture has proven to be superior as www.x-­mol.com) and ChemIDplus performed a
compared to all available models for qualitative 95% accuracy for EI and 96% for EC [94].
toxicity prediction.
A recent investigation by Tokarz et al. used
computer-assisted image analysis algorithm, 5.10.1 Toxicity for Multiple-Time
enabled by an entirely convolutional network Point Assays
deep learning method, to sense and measure the
microscopic structures of progressive cardiomy- The early preclinical stages of drug discovery are
opathy (PCM) in the hear of a rat. The algo- in need of predictive models using technologies
rithms which are trained attained high values for which are better in assessing drug toxicity. Cells
accuracy, intersection over union and dice coef- representing specific tissues can be utilized for
80 S. Saikia et al.

predicting the toxic effects of drugs. Image anal- model for HDAC1 (histone deacetylase) inhibitor
ysis techniques such as high-content microscopy [99]. Ensemble learning methods combine many
are used to observe the drug-induced structural ML methods into one predictive model which is
toxicity wherein intracellular structures are less prone to bias and overfitting – the two most
labelled. These approaches are comparatively common challenges of QSARs.
more accurate and precise than human inspection DL, on the other hand, is an extension of arti-
as last minute structural changes may be missed ficial neural network (ANN), which uses a hierar-
by the human eye with no option for change chy of ANNs to gather useful information from
under live-cell bright-field imaging. AI-based raw data. DL was introduced to drug design in
tools are developed for calculating such struc- 2012 by a competition named Kaggle sponsored
tural changes in cell-based models using cellular by Merck [100]. Deep convolutional neural net-
images taken during multiple doses of a drug works (CNNs) are used to predict toxicity from
(reference with vehicle). As input and the output pretreated cells with drugs which predicted a
show a metric of changes for each drug dose at broad spectrum of toxicity mechanisms, cell lines
the structural level, the result is a high level ofand nuclear stains [101]. Generative adversarial
sensitivity [95]. networks (GANs) [102], long short-term mem-
ory (LSTM) [103] and autoencoders [104] are
some of the other DL methods for predicting
5.11 AI for Drug Safety: Recent drug toxicity.
Advances and Post-Market The FDA maintains FAERS containing com-
Surveillance plaints about product quality, adverse events and
medication error reports, which is a critical source
AI techniques have shown promise in pre-­ for post-marketing data mining. Venulet algo-
marketing drug safety, particularly in toxicity rithm [105], Naranjo algorithm [106] and the
evaluation. Toxic drugs can be prevented from WHO-UMC system are the classical methods to
moving to clinical trials by making preclinical evaluate causality. System pharmacology applica-
evaluations robust. High toxicity remains a tion in detecting adverse events leads to identifi-
major cause of drug failures and this leads to cation of off-target effects and their clinical
about two-­third of post-marketing withdrawals observations being the most data-rich source for
and one-fifth of clinical trial failures. Animal in silico adverse drug reaction mining. Due to
studies remain the most conventional way for availability of a number of open databases, sys-
toxicity estimation. This can, however, be con- tem pharmacology has methods to choose; net-
strained by factors such as time, cost and ethical work approaches and integrate many features.
issues [96]. In silico approaches and computa- This includes modular assembly of drug safety
tional techniques have utility in such cases to subnetworks using knowledge base from litera-
predict toxicity by taking into account various ture mining; genome-wide association study data,
features of the drug. DrugBank and ChEMBL (which assigns pheno-
Several ML approaches have been used for type target); and finally training random forest
QSAR analysis which rely on regression for models to predict drugs causing adverse reactions
chemical properties of drug candidates [97], but [107]. Integration of drug-gene interaction can be
assumptions on linearity as well the issue of made for different scales by mining literature on
dimensionality affect most QSARs. So, the most drug-drug interactions (DDI) and further training
common alternative to regression is ensemble or random forest models for prediction of DDIs
SVM ways which has easy interpretation, high [108]. Thus, all these methods heavily rely on
accuracy and robustness [98]. It has been found molecular features linked directly to drugs with
that SVM outperformed k-NN (k-nearest neigh- open-source data but with limited clinical
bour), RF and naїve Bayes algorithms in a QSAR information.
5 The Role of Artificial Intelligence in Therapeutic Drug Monitoring and Clinical Toxicity 81

5.12 Illusions and Reality of AI Halicin, a new antibiotic, has been identified
in Drug Design within a short period of time using AI mining
from the existing molecules [113].
We need to make proper decisions relating to Experts are of the belief that AI has the poten-
compounds while taking them through clinical tial to change the pharmaceutical industry and
studies. In order to use AI in drug design, often the drug discovery process. Efficient drug discov-
times the data availability may not be sufficient. ery using AI requires human input to design algo-
So, we need better molecules with right dosing/ rithms and with domain expertise. A closed
PK to attain therapeutic efficacy. Usually, 3D workspace between medicinal chemist and AI is
models with better predictability are useful in essential for analysis of huge data sets, algorithm
picking safety relevant endpoints [109], which training and optimization [114]. Advances in AI
can be later validated in animal models [110]. methods particularly DL have prompted consid-
Again for considering the molecule of interest, its eration of heterogeneous data types and sources
in vivo mode of action is essential so that we can in one model such as imaging, clinical data,
have validated targets [111]. In most complex knowledge bases, omics data, etc. AI has poten-
diseases, chemical data are available in large tial use in clinical trial simulation studies. This
scale which has been successfully used in ligand can be used to test trial design on virtual popula-
design and synthesis. These data are very useful tion prior to actual clinical trial [115].
for target validation but a lot is needed for AI
application rather than limiting it to ligand dis-
covery alone. Total advancement in the use of AI 5.14 Conclusions
in drug discovery is possible only when we
understand biology and produce single interest-­ The challenges faced by pharmaceutical compa-
driven data with better efficacy and endpoints. nies affect the whole drug development process
Thus, advancing better candidates to clinics, hav- and the overall life of the product; the progress in
ing better target validation, improving patient AI and its tools aim to reduce these challenges.
requirement and advancement in clinical trials AI has been included in the manufacturing pro-
are essential in reflecting the biological aspect ofcess of pharmaceutical products, expected dose
drug discovery upon which AI can be used [5]. in personalized medicines and other individual-
ized patient needs [77]. Implementing the latest
AI-based technologies will help to bring drugs
5.13 Future of AI in Drug Design or molecules to the market with improved quality
and better safety (128).
Drug development involves successful decision In this chapter, we have reviewed the various
making such as correct selection of target, drug, aspects of drug design and the use of AI in drug
patient, dosages, etc., in which AI can support toxicity prediction and monitoring. The early
such decision making by taking into account stage challenges in drug design were discussed
massive multimodal data to predict models. An along with essentials for implementing AI. The
unparalleled revolution can be expected in the importance of ligand discovery and the process
near future by AI and ML as they are capable of of gathering big medical data have been dis-
making the costly and complex process of drug cussed. Disease monitoring and the pharmaco-
discovery cheaper, effective and less time-­ vigilance aspect of AI have been explained in
consuming. This can be applied in the current detail. The predictions of toxicity endpoints using
health industry as seen in the exponential increase AI and post-marketing surveillance together with
of using AI in drug development such as Iktos, the future of AI have been briefly discussed.
Exscientia, Schrödinger, Exscientia, etc. [112]. Thus, herein we highlighted several key points to
The first AI designed drug from immune-­ be taken into consideration when using AI in the
oncology entered Phase I clinical trials in 2020, fields of medicine, drug discovery and public
after 1 year of research as compared to 5–7 years. health.
82 S. Saikia et al.

Acknowledgements We thank the Vice Chancellor, dent cytotoxicity measurements. ChemMedChem.


Bharathiar University, Coimbatore-641046, Tamilnadu, 2016;11:57–71.
for providing the necessary facilities. We also thank UGC- 15. Tran HTN, Ang KS, Chevrier M, et al. A benchmark
New Delhi for Dr. D S Kothari Fellowship (No.F-2/2006 of batch-effect correction methods for single-cell
(BSR)/BL/20-21/0396). RNA sequencing data. Genome Biol. 2020;21(1):12.
16. Kozikowski BA, Burt TM, Tirey DA, et al. The effect
of freeze/thaw cycles on the stability of compounds
Conflict of Interest The authors have no finan- in DMSO. J Biomol Screen. 2003;8(2):210–5.
cial or non-financial interests to declare. 17. Kinker GS, Greenwald AC, Tal R, et al. Pan-­
cancer single-cell RNA-seq identifies recurring
programs of cellular heterogeneity. Nat Genet.
2020;52(11):1208–18.
18. Bohacek RS, McMartin C, Guida WC. The art
References and practice of structure-based drug design: a
molecular modeling perspective. Med Res Rev.
1. LeCun Y, Bengio Y, Hinton G. Deep learning. 1996;16(1):3–50.
Nature. 2015;521(7553):436–44. 19. Ghandi M, Huang FW, Jané-Valbuena J, et al. Next-­
2. Yu K-H, Beam AL, Kohane IS. Artificial intelligence generation characterization of the cancer cell line
in healthcare. Nat Biomed Eng. 2018;2(10):719–31. Encyclopedia. Nature. 2019;569:503–8.
3. Van Drie JH. Computer-aided drug design: the next 20. Campbell PJ, Getz G, Korbel JO. Pan cancer analysis
20 years. J Comput Aid Mol Des. 2007;21:591–601. of whole genomes. Nature. 2020;578(7793):82–93.
4. JKim J, Hu C, Moufawad El Achkar C, et al. Patient-­ 21. Magill SL, Davoli T, Mamie ZL, et al. Profound
customized Oligonucleotide therapy for a rare genetic tissue specificity in proliferation control under-
disease. N Engl J Med. 2019;381(17):1644–52. lies cancer drivers and aneuploidy patterns. Cell.
5. Bender A, Cortés-Ciriano I. Artificial intelligence in 2018;173(2):499–514.
drug discovery: what is realistic, what are illusions? 22. Schneider G, Clark DE. Automated de novo drug
Part 1: ways to make an impact, and why we are not design: are we nearly there yet? Angew Chem Int
there yet. Drug Discov Today. 2021;26) 2:511–24. Ed. 2019;58:10792–803.
6. Bender A, Cortes-Ciriano I. Artificial intelligence in 23. Davies IW. The digitization of organic synthesis.
drug discovery: what is realistic, what are illusions? Nature. 2019;570:175–81.
Part 2: a discussion of chemical and biological data 24. Mayr A, Klambauer G, Unterthiner T, et al. Large-­
used for AI in drug discovery. Drug Discov Today. scale comparison of machine learning methods
2021;26(4):1040–52. for drug target prediction on ChEMBL. Chem Sci.
7. Brown N, Ertl P, Lewis R, Luksch T, Reker D, 2018;9(24):5441–51.
Schneider N. Artificial intelligence in chemis- 25. Trapotsi M-A, Mervin LH, Afzal AM, et al.
try and drug design. J Comput Aid Mol Des. Comparison of chemical structure and cell morphol-
2020;34:709–15. ogy information for multitask bioactivity predic-
8. Mathea M, Klingspohn W, Baumann tions. J Chem Inf Model. 2021;61(3):1444–56.
KJ. Chemoinformatic classification methods 26. Lenselink EB, Dijke NT, Bongers B, et al. Beyond
and their applicability domain. Mol Inform. the hype: deep neural networks outperform estab-
2016;35(5):160–80. lished methods using a ChEMBL bioactivity bench-
9. Shinde A, Pawar D, Sonawane K. Int J Basic Clin mark set. J Cheminform. 2017;9(1):45.
Pharm. 2021;10(7):863–9. 27. Lane TR, Foil DH, Minerali E, Urbina F, Zorn
10. Basile AO, Yahi A, Tatonetti NP. Artificial intelli- KM, Ekins S. Bioactivity comparison across mul-
gence for drug toxicity and safety. Trends Pharmacol tiple machine learning algorithms using over
Sci. 2019;40(9):624–35. 5000 datasets for drug discovery. Mol Pharm.
11. Wirtz BW, Weyerer JC, Geyer C. Artificial intelli- 2021;18(1):403–15.
gence and the public sector—applications and chal- 28. Nathanson DA, Gini B, Mottahedeh J, et al. Targeted
lenges. Int J Public Adm. 2019;42(7):596–615. therapy resistance mediated by dynamic regulation
12. Ramesh AN, Kambhampati C, Monson JRT, Drew of extrachromosomal mutant EGFR DNA. Science.
PJ. Artificial intelligence in medicine. Ann R Coll 2014;343(6166):72–6.
Surg Engl. 2004;86(5):334–8. 29. Davies M, Jones RDO, Grime K, et al. Improving
13. Lamberti MJ, Wilkinson M, Donzanti BA, the accuracy of predicted human pharmacokinet-
Wohlhieter GE, Parikh S, Wilkins RG, et al. A ics: lessons learned from the AstraZeneca drug
study on the application and use of artificial intel- pipeline over two decades. Trends Pharmacol Sci.
ligence to support drug development. Clin Ther. 2020;6:390–408.
2019;41(8):1414–26. 30. Coley CW, Thomas DA, Lummiss JAM, et al.
14. Corte’s-Ciriano I, Bender A. How consistent are A robotic platform for flow synthesis of organic
publicly reported cytotoxicity data? Large-­scale sta- compounds informed by AI planning. Science.
tistical analysis of the concordance of public indepen- 2019;365(6453):eaax1566.
5 The Role of Artificial Intelligence in Therapeutic Drug Monitoring and Clinical Toxicity 83

31. Nicolaou CA, Brown N. Multi-objective optimiza- 49. Cowen L, Ideker T, Raphael BJ, Sharan R. Network
tion methods in drug design. Drug Discov Today propagation: a universal amplifier of genetic associa-
Technol. 2013;10:e427–35. tions. Nat Rev Genet. 2017;18(9):551–62.
32. FAIR Principles. GO FAIR. https://www.go-­fair.org/ 50. Cantini L, Medico E, Fortunato S, Caselle
fair-­principles/. Accessed 14 Dec 2021. M. Detection of gene communities in multi-networks
33. CDISC | Clear Data. Clear Impact. https://www. reveals cancer drivers. Sci Rep. 2015;5:17386.
cdisc.org/. Accessed 14 Dec 2021. 51. Raue A, Schilling M, Bachmann J, et al. Lessons
34. General Data Protection Regulation (GDPR) – learned from quantitative dynamical modeling in
Official Legal Text. General Data Protection systems biology. PLoS One. 2013;8(9):e74335.
Regulation (GDPR). https://gdpr-­info.eu/. Accessed 52. Reymond J-L, van Deursen R, Blum LC, Ruddigkeit
19 Nov 2021. L. Chemical space as a source for new drugs. Med
35. Tang J, Tanoli Z-u-R, Ravikumar B, et al. Drug target Chem Comm. 2010;1:30–8.
commons: a community effort to build a c­ onsensus 53. Katsila T, Spyroulias GA, Patrinos GP, Matsoukas
knowledge base for drug-target interactions. Cell. MT. Computational approaches in target identifica-
Chem Biol. 2018;25(2):224–9. tion and drug discovery. Comput Struct Biotechnol
36. PrecisionFDA Truth Challenge – precisionFDA. J. 2016;14:177–84.
https://precision.fda.gov/challenges/truth. Accessed 54. Duch W, Swaminathan K, Meller J. Artificial intel-
20 Sept 2021. ligence approaches for rational drug design and dis-
37. Kaggle: Your Machine Learning and Data Science covery. Curr Pharm Des. 2007;13(14):1497–508.
Community. https://www.kaggle.com/. Accessed 22 55. Emig D, Ivliev A, Pustovalova O, et al. Drug tar-
Dec 2021. get prediction and repositioning using an inte-
38. DREAM Challenges. https://dreamchallenges.org/. grated network-­ based approach. PLoS One.
Accessed 12 Oct 2021. 2013;8(4):e60618.
39. Chen H, Zeng D, Buckeridge DL, et al. AI for 56. Bain EE, Shafner L, Walling DP, et al. Use of a novel
global disease surveillance. IEEE Intell Syst. artificial intelligence platform on mobile devices to
2009;24:66–82. assess dosing compliance in a phase 2 clinical trial
40. García Del Valle EP, Lagunes García G, Lucía Prieto in subjects with Schizophrenia. JMIR Mhealth
Santamaría L, et al. Disease networks and their con- Uhealth. 2017;5(2):e18.
tribution to disease understanding: a review of their 57. Deliberato RO, Celi LA, Stone DJ. Clinical note cre-
evolution, techniques and data sources. J Biomed ation, binning, and artificial intelligence. JMIR Med
Inform. 2019;94:103–206. Inform. 2017;5(3):e24.
41. Dugger SA, Platt A, Goldstein DB. Drug develop- 58. Galbusera F, Niemeyer F, Seyfried M, et al.
ment in the era of precision medicine. Nat Rev Drug Exploring the potential of generative adversarial
Discov. 2018;17(3):183–96. networks for synthesizing radiological images of
42. Lee L-H, Loscalzo J. Network medicine in pathobi- the spine to be used in in Silico trials. Front Bioeng
ology. Am J Pathol. 2019;189(7):1311–26. Biotechnol. 2018;6:53.
43. Wang Q, Feng Y, Huang J, Wang T, Cheng G. A 59. Ryu JY, Kim HU, Lee SY. Deep learning
novel framework for the identification of drug tar- improves prediction of drug-drug and drug-
get proteins: combining stacked auto-encoders food interactions. Proc Natl Acad Sci U S A.
with a biased support vector machine. PLoS One. 2018;115(18):E4304–11.
2017;12(4):e0176486. 60. Swan AL, Stekel DJ, Hodgman C, et al. A machine
44. Ferrero E, Dunham I, Sanseau P. In silico prediction learning heuristic to identify biologically relevant
of novel therapeutic targets using gene-disease asso- and minimal biomarker panels from omics data.
ciation data. J Transl Med. 2017;15(1):182. BMC Genomics. 2015;16 Suppl 1(Suppl1):S2.
45. Bakkar N, Kovalik T, Lorenzini I, et al. Artificial 61. Krittanawong C. The rise of artificial intelligence
intelligence in neurodegenerative disease research: and the uncertain future for physicians. Eur J Intern
use of IBM Watson to identify additional RNA-­ Med. 2018;48:e13–4.
binding proteins altered in amyotrophic lateral scle- 62. Powles J, Hodson H. Google DeepMind and health-
rosis. Acta Neuropathol. 2018;135(2):227–47. care in an age of algorithms. Health Technol.
46. Stelzl U, Worm U, Lalowski M, et al. A human 2017;7:351–67.
protein-protein interaction network: a resource for 63. Mahnaz M, Kevin L, Alexandre R. Artificial intelli-
annotating the proteome. Cell. 2005;122(6):957–68. gence enables structural toxicity testing for endpoint
47. Needham CJ, Bradford JR, Bulpitt AJ, Westhead and multiple-timepoint assays. J Pharmacol Toxicol
DR. Inference in Bayesian networks. Nat Biotechnol. Methods. 2020;9(105):106852.
2006;24(1):51–3. 64. Onakpoya IJ, Heneghan CJ, Aronson JK. Worldwide
48. Barabási AL, Gulbahce N, Loscalzo J. Network withdrawal of medicinal products because of adverse
medicine: a network-based approach to human dis- drug reactions: a systematic review and analysis.
ease. Nat Rev Genet. 2011;12(1):56–68. Crit Rev Toxicol. 2016;46(6):477–89.
84 S. Saikia et al.

65. Segall MD, Barber C. Addressing toxicity risk when 80. Plewczynski D, Spieser SA, Koch U. Assessing dif-
designing and selecting compounds in early drug ferent classification methods for virtual screening. J
discovery. Drug Discov Today. 2014;19:688–93. Chem Inf Model. 2006;46(3):1098–106.
66. Mockute R, Desai S, Perera S, Assuncao B, Danysz 81. Dudek ZA, Arodz T, Galvez J. Computational meth-
K, Tetarenko N, et al. Artificial intelligence within ods in developing quantitative structure-activity
pharmacovigilance: a means to identify cognitive relationships (QSAR): a review. Comb Chem High
services and the framework for their validation. Throughput Screen. 2006;9(3):213–28.
Pharm Med. 2019;33(2):109–20. 82. Wu Y, Wang G. Machine learning based toxicity
67. Danysz K, Cicirello S, Mingle E, Assuncao B, prediction: from chemical structural description
Tetarenko N, Mockute R, et al. Artificial intelligence to transcriptome analysis. WIRES Compt Mol Sci
and the future of the drug safety professional. Drug 2018;19(8):2358.
Saf. 2019;42(4):491–7. 83. Xu Y, Dai Z, Chen F, Gao S, Pei J, Lai L. Deep learn-
68. Hauben M, Hartford CG. Artificial intelligence in ing for drug-induced liver injury. J Chem Inf Model.
pharmacovigilance: scoping points to consider. Clin 2015;55(10):2085–93.
Ther. 2021;43(2):372–9. 84. Hughes TB, Miller GP, Swamidass SJ. Modeling
69. Bate A, Hobbiger SF. Artificial intelligence, real-­ epoxidation of drug-like molecules with a deep
world automation and the safety of medicines. Drug machine learning network. ACS Central Sci.
Saf. 2021;44(2):125–32. 2015;1(4):168–80.
70. Routray R, Tetarenko N, Abu-Assal C, Mockute 85. Forghani M, Khachay M. Convolutional neural net-
R, Assuncao B, Chen H, et al. Application of aug- work based approach to in Silico non-anticipating
mented intelligence for pharmacovigilance case seri- prediction of antigenic distance for influenza virus.
ousness determination. Drug Saf. 2020;43(1):57–66. Viruses. 2020;12(9):1019.
71. Thompson P, Daikou S, Ueno K, Batista-Navarro R, 86. Mayr A, Klambauer G, Unterthiner T, Hochreiter
Tsujii J, Ananiadou S. Annotation and detection of S. DeepTox: toxicity prediction using deep learning.
drug effects in text for pharmacovigilance. J Chem. Front Environ Sci. 2016;3:80
2018;10(1):37. 87. Allen TEH, Goodman JM, Gutsell S, Russell
72. Segura-Bedmar I, Martínez P. Pharmacovigilance PJ. Defining molecular initiating events
through the development of text mining and natural in the adverse outcome pathway frame-
language processing techniques. J Biomed Inform. work for risk assessment. Chem Res Toxicol.
2015;58:288–91. 2014;27(12):2100–12.
73. Ward IR, Wang L, Lu J, Bennamoun M, Dwivedi 88. Ambe K, Ishihara K, Ochibe T, Ohya K, Tamura
G, Sanfilippo FM. Explainable artificial intelligence S, Inoue K, et al. In Silico prediction of chemical-­
for pharmacovigilance: what features are impor- induced hepatocellular hypertrophy using molecular
tant when predicting adverse outcomes? Comput descriptors. Toxicol Sci. 2017;162(2):667–75.
Methods Prog Biomed. 2021;212:106415. 89. Liu A, Walter M, Wright P, Bartosik A, Dolciami D,
74. Vo AH, Van Vleet TR, Gupta RR, Liguori MJ, Rao Elbasir A, et al. Prediction and mechanistic analysis
MS. An overview of machine learning and big data of drug-induced liver injury (DILI) based on chemi-
for drug toxicity evaluation. Chem Res Toxicol. cal structure. Biol Direct. 2021;16(1):6.
2020;33(1):20–37. 90. Xu Y, Pei J, Lai L. Deep learning based regression
75. Pérez Santín E, Rodríguez Solana R, González and multiclass models for acute oral toxicity predic-
García M, García Suárez MDM, Blanco Díaz GD, tion with automatic chemical feature extraction. J
Cima Cabal MD, et al. Toxicity prediction based on Chem Inf Model. 2017;57(11):2672–85.
artificial intelligence: a multidisciplinary overview. 91. Tokarz DA, Steinbach TJ, Lokhande A, Srivastava
WIRES Compt Mol Sci 2021;11(5):e1516. G, Ugalmugle R, Co CA, et al. Using artificial intel-
76. Paul D, Sanap G, Shenoy S, Kalyane D, Kalia K, ligence to detect, classify, and objectively score
Tekade RK. Artificial intelligence in drug dis- severity of rodent cardiomyopathy. Toxicol Pathol.
covery and development. Drug Discov Today. 2021;49(4):888–96.
2021;26(1):80–93. 92. Chang DW, Lin CS, Tsao TP, Lee CC, Chen JT,
77. Setzer RW, Kimmel CA. Use of NOAEL, benchmark Tsai CS, et al. Detecting digoxin toxicity by artifi-
dose, and other models for human risk assessment of cial intelligence-assisted electrocardiography. Int J
hormonally active substances %. J Pure Appl Chem. Environ Res Public Health. 2021;18(7):3839.
2003;75(11–12):2151–8. 93. Di P, Yin Y, Jiang C, Cai Y, Li W, Tang Y, et al.
78. Krewski D, Acosta D Jr, Andersen M, Anderson H, Prediction of the skin sensitising potential and
Bailar JC 3rd, Boekelheide K, et al. Toxicity testing potency of compounds via mechanism-based binary
in the 21st century: a vision and a strategy. J Toxicol and ternary classification models. Toxicol in vitro
Environ Health B Crit Rev. 2010;13(2–4):51–138. :an international journal published in association
79. Saito T, Rehmsmeier M. The precision-recall plot is with BIBRA. 2019;59:204–14.
more informative than the ROC plot when evaluating 94. Verma RP, Matthews EJ. Estimation of the chemical-­
binary classifiers on imbalanced datasets. PLoS One. induced eye injury using a weight-of-evidence
2015;10(3):e0118432. (WoE) battery of 21 artificial neural network (ANN)
5 The Role of Artificial Intelligence in Therapeutic Drug Monitoring and Clinical Toxicity 85

c-QSAR models (QSAR-21): part I: irritation poten- 105. Lorberbaum T, Nasir M, Keiser MJ, Vilar S,
tial. Regul Toxicol Pharm. 2015;71(2):318–30. Hripcsak G, Tatonetti NP. Systems pharmacology
95. Choi SM, Kang CY, Lee BJ, Park JB. In vitro-in vivo augments drug safety surveillance. Clin Pharmacol
correlation using in Silico modeling of physiological Ther. 2015;97(2):151–8.
properties, metabolites, and intestinal metabolism. 106. Raja K, Patrick M, Elder JT, Tsoi LC. Machine
Curr Drug Metab. 2017;18(11):973–82. learning workflow to enhance predictions of Adverse
96. Algamal Z, Lee M. A new adaptive l1-norm for opti- Drug Reactions (ADRs) through drug-gene interac-
mal descriptor selection of high-dimensional QSAR tions: application to drugs for cutaneous diseases.
classification model for anti-hepatitis c virus activ- Sci Rep. 2017;7(1):3690.
ity of thiourea derivatives. SAR QSAR Environ. 107. Horvath P, Aulner N, Bickle M, et al. Screening out
2017;Res. 28:75–90. irrelevant cell-based models of disease. Nat Rev
97. Ma J, Sheridan RP, Liaw A, Dahl GE, Svetnik Drug Discov. 2016;15(11):751–69.
V. Deep neural nets as a method for quantitative 108. Leenaars CHC, Kouwenaar C, Stafleu FR, et al.
structure-activity relationships. J Chem Inf Model. Animal to human translation: a systematic scoping
2015;55(2):263–74. review of reported concordance rates. J Transl Med.
98. Shi J, Zhao G, Wei Y. Computational QSAR model 2019;17(1):223.
combined molecular descriptors and fingerprints to 109. Lin A, Giuliano CJ, Palladino A, et al. Off-target
predict HDAC1 inhibitors. Med Sci (Paris). 2018;34 toxicity is a common mechanism of action of can-
Focus issue F1:52–8. cer drugs undergoing clinical trials. Sci Transl Med.
99. Torng W, Altman RB. 3D deep convolutional neu- 2019;11(509):eaaw8412.
ral networks for amino acid environment similarity 110. Moingeon P, Kuenemann M, Guedj M. Artificial
analysis. BMC Bioinform. 2017;18:302. intelligence-enhanced drug design and devel-
100. Kadurin A, Nikolenko S, Khrabrov K, Aliper A, opment: toward a computational preci-
Zhavoronkov A. druGAN: an advanced generative sion medicine. Drug Discov Today. 2021;
adversarial autoencoder model for de novo genera- S1359-6446(21)00396-2
tion of new molecules with desired molecular prop- 111. Stokes JM, Yang K, Swanson K, Jin W, et al. A
erties in Silico. Mol Pharm. 2017;14(9):3098–104. deep learning approach to antibiotic discovery. Cell.
101. Altae-Tran H, Ramsundar B, Pappu AS, Pande 2020;180(4):688–702.
V. Low data drug discovery with one-shot learning. 112. Mak KK, Pichika MR. Artificial intelligence in drug
ACS Cent Sci. 2017;3(4):283–93. development: present status and future prospects.
102. Hu Q, Feng M, Lai L, Pei J. Prediction of drug-­ Drug Discov Today. 2019;24(3):773–80.
likeness using deep autoencoder neural networks. 113. Perez-Gracia JL, Sanmamed MF, Bosch A, et al.
Front Genet. 2018;9:585. Strategies to design clinical studies to identify pre-
103. Venulet J, Ciucci AG, Berneker GC. Updating dictive biomarkers in cancer research. Cancer Treat
of a method for causality assessment of adverse Rev. 2017;53:79–97.
drug reactions. Int J Clin Pharmacol Ther Toxicol. 114. Rantanen J, Khinast J. The future of pharma-
1986;24(10):559–68. ceutical manufacturing sciences. J Pharm Sci.
104. Naranjo CA, Busto U, Sellers EM, et al. A method 2015;104(11):3612–38.
for estimating the probability of adverse drug reac- 115. Ja¨msa¨-Jounela S-L. Future trends in process auto-
tions. Clin Pharmacol Ther. 1981;30(2):239–45. mation. Annu Rev Control. 2007;31:211–20.
Therapeutic Drug Monitoring
and Optimal Pharmacotherapy
6
with Medicines of Narrow
Therapeutic Index

Anthony Kwaw, Arnold Forkuo Donkor,


and Kwame Ohene Buabeng

Abstract optimizing therapy can only be attained if the


method is adequately unified with treatment.
Therapeutic drug monitoring (TDM) remains TDM processes assume that there is a signifi-
an under-utilized approach in giving life to the cant relationship between dose and plasma
practice of personalized medicine, as it is the drug concentration and between the latter and
medical practice of measuring certain medica- pharmacodynamic properties of the drug.
tions at intervals to ensure a constant plasma TDM takes into consideration the patient indi-
drug concentration and to optimize dosage cation and other factors such as weight, age,
regimens. It can be used to enhance patient concomitant drug therapy, and organ function
treatment outcomes due to clear dose-response in defining an appropriate initial dosage regi-
relationships. TDM also reduces the overall men. Also, patient compliance to treatment,
cost of healthcare. Unfortunately, most drugs pregnancy, and drug interactions are to be
cannot be monitored with TDM due to the cost considered as they are major sources of phar-
and turnaround times, but rather those with a macokinetic variability and may have to be
narrow therapeutic indices (e.g. digoxin, lith- considered when interpreting TDM results. In
ium, phenytoin, vancomycin, etc.) which have the interpretation of TDM measurements,
a high risk of causing adverse effects. Also, sampling time with drug dose, dosage history,
drugs with significant pharmacokinetic vari- patient response, and the desired medicinal
ability as well as those with difficult-to-­ targets need to be established. Genetic poly-
monitor clinical endpoints are TDM morphisms, smoking, drug formulation, and
candidates. TDM and its associated benefits in methodology used in testing may also affect
the results. TDM also aims to optimize clini-
cal outcomes in patients with various clinical
scenarios in terms of appropriate concentra-
A. Kwaw (*) · A. Forkuo Donkor tions of difficult-to-manage medications.
Department of Pharmacology, Faculty of Pharmacy TDM is thus a multidisciplinary discipline
and Pharmaceutical Sciences, College of Health
Sciences, Kwame Nkrumah University of Science involving scientists, pharmacists, clinicians,
and Technology, Kumasi, Ghana and nurses, and collaboration is recommended
K. O. Buabeng to ensure that best practice is attained to
Department of Pharmacy Practice, Faculty of patients’ benefit.
Pharmacy and Pharmaceutical Sciences, College of
Health Sciences, Kwame Nkrumah University of
Science and Technology, Kumasi, Ghana

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 87


S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_6
88 A. Kwaw et al.

Keywords often used during therapy with such medications to


optimize therapy and reduce toxicity whilst main-
Drug assay · Pharmacokinetics · Plasma taining the medicine concentrations within estab-
concentration · Therapeutic index lished target ranges that are considered safe [6].
Some criteria that are considered in TDM:

6.1 Background • When clinical effects and other pharmacody-


namic responses are hard to establish.
Medicines are used therapeutically based on • The association between drug concentration
safety and efficacy, as well as the benefit of the in plasma and biological effects should be
medicines to patient health, which should out- likely. Phenytoin seems to be the only anti-­
weigh their potential adverse effects. However, epileptic drug with an exceptional positive
that may not always be the case with some medi- association between its concentration in
cines even if the best evidence suggests that like- plasma and biological effects.
liness [1]. There may be uncertainties to patients’ • The therapeutic window is narrow or has
safety and clinical outcomes with some individu- concentration-­dependent pharmacokinetics.
als as they are exposed to certain medicines, due An idea about the appropriate dosage alone
to individual variability in drug pharmacokinet- should be sufficient to forecast whether the
ics and pharmacodynamics [2]. For this reason, drug concentration in plasma is within the
whenever possible, plasma or serum concentra- therapeutic range [7].
tion should be measured for medicines of that
nature, and treatment outcomes monitored to TDM is not supposed to be limited only to
assess safety, tolerability, and therapeutic impact. establishing plasma drug concentrations, but more
Monitoring the effects of medicines with narrow importantly the interpretation of the outcome in a
therapeutic indices is attainable in diverse sce- clinical sense. Therefore, the drug pharmacokinet-
narios. It is possible subjectively when the patient ics, sampling time, previously taken medication(s),
talks about the effects observed following drug and the disease condition are crucial [6].
use. However, a more objective approach may be
to measure the actual biological effect (e.g. blood
pressure control for anti-hypertensives, blood 6.3 Medicines That Are Known
glucose control for anti-diabetic medicines, etc.) to Require Therapeutic Drug
and the adverse effects experienced following Monitoring
exposure to various dosing schedules [3]. Other
alternatives may be to use a biological marker Medicines that may require TDM to assure safety
that acts as a proxy for the therapeutic outcome and efficacy include digoxin, used in cardiovascu-
or adverse effect [4, 5]. If the direct drug effect lar health therapeutics, and carbamazepine and
appears complex or inaccessible, then use can be sodium valproate used in the management of epi-
made of the measured plasma drug concentration lepsy and other neurological disorders. Some other
and how it is closely related to the effect observed, medicines used in cardiovascular therapeutics for
whether adverse or beneficial [6, 7]. arrhythmias that may also require TDM include
amiodarone, flecainide, and disopyramide. An
aminoglycoside such as gentamicin, kanamycin,
6.2 Therapeutic Drug amikacin, etc. is documented to be used safely and
Monitoring efficiently with TDM for dosage adjustments in
the elderly, paediatrics, cardiovascular, and renal
TDM refers to the monitoring of the biological impaired patients with infectious diseases includ-
effects of medicines with a narrow therapeutic win- ing multidrug-resistant infections where the patho-
dow using the plasma drug concentration. TDM is gens are susceptible [9, 10].
6 Therapeutic Drug Monitoring and Optimal Pharmacotherapy with Medicines of Narrow Therapeutic… 89

It has also been established that based on the Table 6.1 Medicines that require therapeutic drug moni-
toring to optimize pharmacotherapy and the target range
clinical pharmacology of the class of medicines
are considered to be safe
below, it may be appropriate to do TDM to guide
Drugs monitored frequently in clinical practice
appropriate dosing and use and to minimize the
Drug Target range (may vary between
risk of toxicity and achieve improved therapeutic laboratories)
outcomes when used in depression, in epilepsy, Phenytoin 10–20 mg/L
in asthma, and for cancer or autoimmune disor- Digoxin 0.8–2 microgram/L
ders, respectively. These are: <0.01 microgram/L in refractory
heart failure
Lithium – acute 0.8–1.2 mmol/L
• Antidepressants such as lithium, imipramine/ mania 0.4–1.0 mmol/L
clomipramine, etc.  
• Antiepileptic medications such as phenytoin,    –
clonazepam, and ethosuximide, among others maintenance
Tacrolimus 5–20 microgram/L (whole
• Bronchodilators like theophylline in asthma blood)
management Drugs for which TDM may be beneficial
• Immunosuppressants for autoimmune disor- Vancomycin 150–300 mg/L
ders and medicines for cancer chemotherapy Amiodarone 1–2.5 mg/L
such as cyclosporine, methotrexate, tacroli- Salicylate 150–300 mg/L
mus, etc. (Table 6.1) Sodium valproate 50–100 mg/L
Carbamazepine 5–12 mg/L
Vancomycin Trough 10–20 mg/L
Unfortunately, it appears resources in terms of
equipments/machineries and trained human
resources are not readily available in the Ghanaian vancomycin helps increase efficacy; that of ami-
Health System, and in most countries in sub-­ noglycosides, cyclosporine, and paracetamol to
Saharan Africa, so the use of TDM to optimize reduce toxicity; and salicylates to help in salicy-
therapy and improve patient safety and outcomes late poisoning. TDM can be key in establishing
is not common, if not very rare. Our consulta- toxicity in undifferentiated clinical syndrome
tions suggest that most of the teaching hospitals such as a patient on digoxin experiencing unex-
in Ghana have seen the need to have the infra- plained nausea [12, 13]. Drugs with an unpredict-
structure and trained staff to initiate the use of able association between a given dose and
TDM efficiently to improve the safe use of the concentration in plasma and saturable metabo-
medicines listed in Table 6.1. These medicines lism like phenytoin will benefit from TDM as
are often used in patients with complex health well as those with steep dose-response curves
issues and require monitoring to ensure that they like theophylline. Drugs with poorly defined end
are not harmed, nor worsen their state with phar- point (e.g. immunosuppressants) and narrow
macotherapy but with improved health status therapeutic windows (e.g. digoxin, phenytoin,
[11]. lithium) require TDM permit adjustment in dose
The indication for a drug may also play a role to avoid toxicity [6]. Impairment in renal func-
in determining the target concentration as exem- tion may also play a part in the alteration in the
plified in the case of digoxin in Table 6.1. association between drug concentration in plasma
and administered dose as in the case of lithium,
gentamicin, and digoxin which requires dose
6.4 Medication Therapy That adjustment.
Requires TDM for Optimal Therapeutic drug monitoring is useful after
Outcomes dose adjustments usually when a steady state is
realized, and for establishing a suitable loading
Due to the costly nature of most drug assays, the dose (after initiating therapy with phenytoin).
reason for monitoring, as well as other benefits, TDM can be used to monitor patient compliance
should be well defined. For example, TDM of to anticonvulsant medications, drug treatment
90 A. Kwaw et al.

failure, and side effects that mimic the underlying this may need diverse sampling times as well as
pathology. Prophylactic drugs such as immuno- different numbers of samples. For instance, a
suppressants and anticonvulsants can be used to single sample may be used to answer suspicion of
diagnose undertreatment [14]. toxicity (e.g. lithium, carbamazepine), whereas
establishing a half-life may necessitate not less
than two samples drawn at the appropriate time
6.5 Guidelines for Sample after drug administration [15].
Collection

Blood is the most commonly used specimen for 6.6 Timing of Plasma Sample
TDM, and its use is recommended when definite for TDM
information is essential. To ensure adequate drug
absorption and accurate therapeutic levels, there Samples for TDM are to be taken at a steady
must be ample time between drug administration state, which is about 4–5 half-lives after begin-
and taking of sample. Samples for TDM espe- ning pharmacological therapy. However, for con-
cially blood can be taken during the peak level or cerns about toxicity or loading dose, it can be
trough level, which should fall within the thera- taken earlier before steady-state concentration is
peutic range. Drug trough levels mostly correlate attained. Sampling time is essential in TDM
with therapeutic levels, whereas peak levels of because the serum drug concentration may alter
drugs correlate commonly with toxic side effects. along with the dosing interval with the least vari-
The best time to take a blood sample for TDM is able point in the dosing interval being just before
just before the next dose irrespective of the route the next dose. Specimens can, therefore, be taken
[8]. at any point in the dosage interval for drugs with
There are usually significant fluctuations in long half-lives like amiodarone and phenobarbi-
peak and trough levels in patients who receive the tal [6].
drug at a dosing interval longer than the half-life Plasma drug concentrations may peak
of the drug and vice versa. Peak and trough drug 1–2 hours after oral administration.
levels for chronically administered oral medica- Notwithstanding, certain factors like erratic
tions typically ensue 1–2 hours after administra- absorption and distribution can delay the time to
tion and soon after the dose is administered reach peak plasma concentrations. For instance,
respectively. However, digoxin peak level is TDM for digoxin can only be performed 6 hours
determined after it has had enough time to equili- after a dose, since it will be undergoing
brate within the tissue, usually 6–10 hours post-­ distribution.
administration orally. It is regarded as correct if In the case of aminoglycoside antibiotics, the
the trough level is taken at the time the dose is time for taking a specimen is occasioned by the
given [7]. technique used for the TDM if it’s a once-daily
For intravenously administered medications, dosage regimen. The sample is therefore taken
peak and trough drug levels can be established by after 6–14 hours when using a nomogram, or two
sampling ½–1 hour post-administration and after samples within the dosing interval to compute the
dose administration, respectively. For gentami- area under the drug concentration-time curve.
cin, a blood sample can be taken 30 mins post-­ However, when given in divided doses in the
peak intravenous administration to establish the treatment of enterococcal endocarditis, the sam-
peak level. This principle applies to intramuscu- ples for trough levels are to be measured to limit
larly and intravenously administered medications toxicity and evaluate if serum concentrations are
in an area with enough blood perfusion. It is within the acceptable therapeutic range [6].
therefore imperative that the clinician knows Below is the sampling time for certain
what questions are to be answered by TDM as medications:
6 Therapeutic Drug Monitoring and Optimal Pharmacotherapy with Medicines of Narrow Therapeutic… 91

• Carbamazepine: trough and peak level col- to take into consideration the time for taking a
lected just after a dose 3 hours later respec- sample, the dosage regimen, and the reason for
tively since its half-life is 48 hours after a drug monitoring [6].
single dose.
• Theophylline: sampling time is not important
for slow-release formulations. 6.8 Therapeutic Drug
• Gentamicin: pre-dose peak 0.5 hours (intrave- Monitoring
nous) and 1 hour (intramuscular). and Interpretation of Results
• Phenytoin: due to its long half-life (10–
15 hours for IV and 22 hours for PO), TDM Therapeutic ranges of drugs are usually obtained
may not be useful as it is commonly taken by measuring the clinical responses of the drug in
once daily. a limited number of patients. The lower limit of
the therapeutic range is set to elicit ~50% of the
maximal therapeutic response, whilst the upper
6.7 Request for Therapeutic limit is well-defined by toxicity. Expert clinical
Drug Monitoring interpretation of the result is necessary as thera-
peutic windows are not absolutes. Background
Assaying of medications can be requested for such as the drug therapy duration and time of the
TDM or clinical toxicology purposes (Fig. 6.1). last dosage is imperative though it is the unbound
In interpreting the result, the clinician may have form of the drug that interacts with the effector

TDM REQUEST FORM


Patient Name……………………………………………………… Date……………………….
Age…………… Sex………….. Weight……………. Height…………….
Ward……………….. Request by………………………. Phone No………………...
REQUESTED DRUG LEVEL…………………………………………………………………..
REASON FOR REQUISITION:
( ) Compliance ( ) Suspected toxicity
( ) Absence of therapeutic response ( ) Therapeutic confirmation
Please indicate when needed:
( ) stat. ( ) within 1-2 hours ( ) within 24 hours ( ) Others………………..........
Date………………… Route: ( ) IV ( ) IM ( ) SC ( ) PO ( ) Others………………...
Time………………… Dose………………………………. Frequency……………...
DRUG LEVEL USE: SAMPLING TIME:
( ) Trough or pre-dose level Date…………….. Time………………...
( ) Peak level Date…………….. Time…………………
ORGAN-SYSTEM DYSFUNCTION PRESENT?
( ) Cardiac ( ) Hepatic ( ) Renal ( ) GI ( ) Endocrine ( ) Others……………………..
CONCOMITANT DRUG(S):
…………..………………………………………………………………………………………..
DRUG LEVEL AND USUAL THERAPEUTIC RANGE………………………………………
INTERPRETATION OF RESULT………………………………………………………………

Fig. 6.1 Sample request form for TDM


92 A. Kwaw et al.

Table 6.2 Factors that account for low or high drug as phenylethylmalonamide is also therapeutically
concentration
active [20–22].
Lower than anticipated Higher than anticipated
Reduce plasma binding Increase in plasma protein
Timing of sample binding
Rapid elimination Rapid bioavailability 6.9 Medications That May
Poor bioavailability Decrease in renal or hepatic Benefit from Therapeutic
Changing hepatic blood function Drug Monitoring
flow Slow elimination
Patient non-adherence Error in dosage elimination
Error in dosage regimen TDM may not be suitable for the listed medica-
tions for unique reasons:

site to produce a response [16, 17]. Also, the tim- • Medications with a wide margin of safety or
ing of the sample, when steady state was achieved, therapeutic index.
and patient compliance to treatment should all be • Medications whose clinical responses are
considered. True therapeutic drug monitoring obtained with practical investigations:
testing considers all factors that affect results and warfarin.
interpretation likewise. Some of these factors are • Medications with plasma concentration not
enumerated in Table 6.2. associated with a clinical response: warfarin.
Drug concentration needs to be understood in • Medications like penicillin whose toxicity is
the context of each patient without necessarily not a realistic concern except for anaphylactic
adhering to the target range. For instance, a reactions in those who are hypersensitive to
patient on an anticonvulsant with serum drug the drug.
concentration just below the target range and not • The association between the plasma concen-
having seizures requires no increase in dose. In tration and clinical response is not clearly
the case of digoxin, serum potassium levels defined: antidepressants.
should be monitored when interpreting results as • Hit and run drugs: Monoamine oxidase inhibi-
toxic since toxicity can as well occur at therapeu- tors and the proton pump inhibitor, omepra-
tic concentrations in the presence of hypokalae- zole [23].
mia [3, 6].
In TDM, plasma or serum may be used to
measure both bound and unbound drugs. 6.10 Techniques for Measurement
Therefore, in patients with altered binding capac- of Therapeutic Drug
ity due to disease states like renal failure, drug Monitoring
interactions, or non-linear binding, the protein
binding effect on drug concentration needs to be Several separation techniques are available for
considered when interpreting results. This is rel- use in TDM as described in Table 6.3 [24, 25].
evant for phenytoin such that should its unbound
portion double from 10% to 20%, the target ther-
apeutic range as a result of total phenytoin con- 6.11 Practical Use of Therapeutic
centration needs to be halved, which if not Drug Monitoring
adhered to may lead to toxic adverse effects [6,
18, 19]. Drug assays ought to be done within a clinically
Some drugs whose metabolites may be active beneficial timeframe with the appropriate bio-
are not measured, though they add to the thera- logical specimen to be able to make clinical rel-
peutic effect of the parent drug. Therapy with evant interpretations and decisions. Absorption
primidone may be monitored by determining after oral drug administration delays some drugs,
phenobarbital, an active metabolite; nonetheless, and this requires the specimen to be taken in the
primidone together with other metabolites such elimination phase rather than in the absorption or
6 Therapeutic Drug Monitoring and Optimal Pharmacotherapy with Medicines of Narrow Therapeutic… 93

Table 6.3 Separation techniques for therapeutic drug distribution phases. Slow drug absorption among
monitoring
others may affect the time to reach peak drug lev-
Technique Description els after an oral dose. The measured drug concen-
High-pressure liquid It is a technique based on trations can then be compared to published
chromatography the relative distribution of
(HPLC) the constituents of a mixture therapeutic ranges, so as relate them to the phar-
between a mobile and macodynamic responses. However, drugs with
stationary phase virtually no individual variability in plasma con-
Liquid chromatography-­ The mobile phase moves the centration will not benefit from TDM assays. For
mass spectrometry sample along with the
(LCMS) stationary phase, which
drugs with therapeutically active metabolites,
leads to the separation of the both parent drug and active metabolites should be
sample constituents determined to establish an inclusive representa-
Gas chromatography-­ A technique that uses very tion of the association between the total plasma
mass spectrometry high temperatures to cause
concentration of all the active molecules and the
(GC/MS) vaporization of the sample,
which is then passed through biological response, but it is advisable in routine
an electrical field therapeutic monitoring. The results should be
The molecules can be ready if possible within 24 hours of sample
separated based on
receipt. The most vital consideration in interpret-
molecular weight since the
pattern of separation is ing the plasma concentration of a drug is the indi-
unique to each drug vidualization of therapy. The clinician, therefore,
Enzyme immunoassay A non-radioactive enzyme should consider the patient’s clinical presentation
(EIA) label technique with assays which may adversely affect the association
performed in a single step
Radioactivity (RIA) It uses radioactivity to detect
between plasma drug concentration and clinical
the existence of an analyte response [8, 15].
Particle-enhanced A technique that separates There must be cost-effectiveness in measuring
turbidimetric inhibition sample constituents by the drug levels in clinical samples. The use of clini-
immunoassay creation of light scattering
cal pharmacokinetic principles in offering TDM
(PETINIA) particles to quantify drug
levels services offers enormous benefits as significant
Enzyme-multiplied Separation technique based reduction in adverse reactions, shorter stay in the
immunoassay technique on competition for the target ICU, and shorter overall hospital stay [24, 25].
(EMIT) analyte antibody binding
sites
Fluorescence A technique that makes use
polarization of a fluorescent molecule as References
immunoassay (FPIA) the label and is very
sensitive 1. Tamargo J, Le Heuzey JY, Mabo P. Narrow therapeutic
Chemiluminescence: A chemical reaction index drugs: a clinical pharmacological consideration
produces light, which when to flecainide. Eur J Clin Pharmacol. 2015;71(5):549–
combined with 67. https://doi.org/10.1007/s00228-­015-­1832-­0.
immunoassay technology 2. Alshammari TM. Drug safety: the concept, inception
specifies the amount of and its importance in patients’ health. Saudi Pharm
analyte in a test sample J. 2016;24(4):405–12. https://doi.org/10.1016/j.
Affinity chrome-­ A technique for measuring jsps.2014.04.008.
mediated immunoassay plasma drug concentrations 3. Kang JS, Lee MH. Overview of therapeutic drug
(ACMIA) where both bound and monitoring. Korean J Intern Med. 2009;24(1):1–10.
unbound drug-antibody-­ https://doi.org/10.3904/kjim.2009.24.1.1.
enzyme conjugates are 4. FDA Facts: Biomarkers and Surrogate Endpoints
separated using magnetic | FDA. https://www.fda.gov/about-­fda/innovation-­
particles called chrome fda/fda-­facts-­biomarkers-­and-­surrogate-­endpoints.
Cloned enzyme donor A technique that uses Accessed 9 Mar 2022.
immunoassay (CEDIA) recombinant DNA 5. Luis García-Giménez J, et al. Biomarkers in stress
technology related diseases/disorders: diagnostic, prognos-
tic, and therapeutic values. Front Mol Biosci
94 A. Kwaw et al.

| w
­ wwfrontiersinorg. 2019;6:91. https://doi. 16. Cooney L, et al. Overview of systematic reviews
org/10.3389/fmolb.2019.00091. of therapeutic ranges: methodologies and rec-
6. Ghiculesco R. Abnormal laboratory results: therapeu- ommendations for practice. BMC Med Res
tic drug monitoring: which drugs, why, when and how Methodol. 2017;17(1) https://doi.org/10.1186/
to do it. Aust Prescr. 2008;31(2):42–4. https://doi. s12874-­017-­0363-­z.
org/10.18773/austprescr.2008.025. 17. What is a Therapeutic Range? - Definition from
7. Therapeutic drug monitoring | Pharmacology WorkplaceTesting. https://www.workplacetesting.
Education Project. https://www.pharmacologyedu- com/definition/1357/therapeutic-­range. Accessed 9
cation.org/clinical-­pharmacology/therapeutic-­drug-­ Mar 2022.
monitoring. Accessed 9 Mar 2022. 18. Kidd JM, Asempa TE, Abdelraouf K. Therapeutic
8. Therapeutic Index and Therapeutic Range in drug monitoring. Remington. 2021:243–62. https://
Pharmacology Tutorial 09 March 2022 - Learn doi.org/10.1016/B978-­0-­12-­820007-­0.00013-­1.
Therapeutic Index and Therapeutic Range in 19. Johnson-Davis KL, Dasgupta A. Special issues in
Pharmacology Tutorial (13907) | Wisdom Jobs therapeutic drug monitoring in patients with Uremia,
India. https://www.wisdomjobs.com/e-­university/ liver disease, and in critically ill patients. Clin
pharmacology-­tutorial-­128/therapeutic-­index-­and-­ Challenges Ther Drug Monit Spec Popul Physiol
therapeutic-­range-­13907.html. Accessed 9 Mar Cond Pharmacogenomics. 2016:245–60. https://doi.
2022. org/10.1016/B978-­0-­12-­802025-­8.00011-­8.
9. Therapeutic Drug Monitoring: MedlinePlus Medical 20. Hvidberg EF, Dam M. Clinical pharmacokinetics of anti-
Test. https://medlineplus.gov/lab-­tests/therapeutic-­ convulsants. Clin Pharmacokinet. 1976;1(3):161–88.
drug-­monitoring/. Accessed 9 Mar 2022. https://doi.org/10.2165/00003088-­197601030-­00001.
10. Flower RJ, Gavins F. Dexamethasone. xPharm Compr 21. Leal KW, Rapport RL, Wilensky AJ, Friel PN. Single-­
Pharmacol Ref. 2008:1–6. https://doi.org/10.1016/ dose pharmacokinetics and anticonvulsant efficacy of
B978-­008055232-­3.61572-­7. primidone in mice. Ann Neurol. 1979;5(5):470–4.
11. Azevedo MJ. Historical perspectives on the state of https://doi.org/10.1002/ana.410050512.
health and health systems in Africa, volume II, vol. 22. Bentué-Ferrer D, Verdier MC, Tribut O. Therapeutic
II; 2017. drug monitoring of primidone and phenobarbi-
12. Cauffield JS, Gums JG, Grauer K. The serum tal. Therapie. 2012;67(4):381–90. https://doi.
digoxin concentration: ten questions to ask. Am Fam org/10.2515/THERAPIE/2012036.
Physician. 1997;56(2):495–503. 23. Boucher BA, Wood GC, Swanson
13. Do therapeutic doses of acarbose alter the pharmaco- JM. Pharmacokinetic changes in critical illness.
kinetics of digoxin? - PubMed. https://pubmed.ncbi. Crit Care Clin. 2006;22(2):255–71. https://doi.
nlm.nih.gov/12389338/. Accessed 9 Mar 2022. org/10.1016/j.ccc.2006.02.011.
14. Gibson TP. Renal disease and drug metabolism: an 24. Charbe NB, Zacconi FC, Amnerkar N, Ramesh
overview. Am J Kidney Dis. 1986;8(1):7–17. https:// B, Tambuwala MM, Clementi E. Bio-analytical
doi.org/10.1016/S0272-­6386(86)80148-­2. assay methods used in therapeutic drug monitor-
15. ISMAAP.org: Why it is so important to be in the ing of antiretroviral drugs-a review. Curr Drug Ther.
therapeutic range. https://www.ismaap.org/important-­ 2018;14(1):16–57.
to-­know-­detail/why-­it-­is-­so-­important-­to-­be-­in-­the-­ 25. Handbook of drug monitoring methods therapeu-
therapeutic-­range/. Accessed 9 Mar 2022. tics and drugs of abuse. https://lotsofbook.club/sl-­
S1B8Q-­D7256/signup. Accessed 9 Mar 2022.
Therapeutic Drug Monitoring
(TDM) and Toxicological Studies
7
in Alternative Biological Matrices

Biswajit Basu, Bhupendra G. Prajapati,


Swarupananda Mukherjee, Tapas Kumar Roy,
Arnab Roy, Chowdhury Mobaswar Hossain,
Jigna B. Prajapati, and Jayvadan Patel

Abstract urine can provide supplementary evidence


about drug exposure and analytical benefits.
Recent technology advancements have Several analytical techniques such as laser
sparked renewed interest in individualized diode thermal desorption-tandem mass spec-
medicine. Some approaches in attaining indi- trometry (LDTD-MS-MS), ultra-high perfor-
vidualized therapy such as therapeutic drug mance liquid chromatography-tandem mass
monitoring (TDM) have, on the other hand, spectrometry, liquid chromatography-­
remained underappreciated. TDM has the electrospray mass spectrometry, enzyme
potential to enhance patient outcomes while immunoassay and gas chromatography/mass
also lowering healthcare expenditures due to spectrometry, gas chromatography-surface
its unambiguous dose-response correlations. ionization organic mass spectrometry, chemi-
Two new approaches to TDM have emerged in cal ionization mass spectrometry with ammo-
recent years: target concentration interven- nia, chromatography coupled with
tion (TCI) and a priori TDM (by combining electrospray-ionization mass spectrophotom-
TDM with pharmacogenomics). Alternative etry (LC-ESI-MS), etc. have been used for the
matrices are increasingly being used in toxico- detection of drugs of exploitation in the above-
logical analyses in clinical and forensic con- mentioned biosamples. This chapter will pro-
texts. Alternative specimens to blood and vide an overview of TDM, including its

A. Roy
B. Basu Department of Pharmacology and Toxicology,
Bengal School of Technology (A College of NIPER, Hyderabad, India
Pharmacy), Sugandha, Hooghly, West Bengal, India
C. M. Hossain
B. G. Prajapati (*) Department of Pharmaceutical Science &
Department of Pharmaceutics and Pharmaceutical Technology, Maulana Abul Kalam Azad University of
Technology, Shree S K Patel College of Technology, Haringhata, Nadia, West Bengal, India
Pharmaceutical Education and Research, Ganpat
J. B. Prajapati
University, Kherva, Mahesana, Gujarat, India
Acharya Motibhai Patel Institute of Computer
S. Mukherjee Studies, Ganpat University, Kherva, Mahesana,
NSHM Knowledge Campus, Kolkata – Group of Gujarat, India
Institutions, Kolkata, India
J. Patel
T. K. Roy Nootan Pharmacy College, Faculty of Pharmacy,
Ocular Pharmacology and Pharmacy Division, Dr. Sankalchand Patel University, Visnagar, Gujarat,
R.P Centre, AIIMS, New Delhi, India India

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 95


S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_7
96 B. Basu et al.

history, current developments, and prospects, could be reduced [10]. The development of clini-
as well as toxicological research using various cal pharmacokinetic monitoring was supported
analytical techniques in various biological by increasing awareness of drug concentration-­
matrices. response relationships, the mapping of drug phar-
macokinetic properties, the introduction of
Keywords high-throughput computerization, and develop-
ments in analytical technology [11].
Therapeutic drug monitoring · A priori TDM
· Thermal desorption-tandem mass spectrom- Historical Perspectives on TDM
etry (LDTD-MS-MS) · Alternative matrices · The first PK study was published in the 1960s,
Liquid chromatography coupled with and the significance of PK was proven [12].
electrospray-ionization mass spectrophotom- Another historical milestone was a 1965 publica-
etry (LC-ESI-MS) · Target concentration tion [13] that was the first formal review on the
intervention (TCI) · Biochips importance of “drug monitoring.” With advance-
ments in chromatographic techniques, these
inquiries received even more traction. The years
7.1 Introduction that followed were a golden age for drug surveil-
lance. Gas chromatography (GC), HPLC, and
Therapeutic drug monitoring (TDM) is the mea- mass spectrometry were used to determine the
surement of a chemical parameter in a clinical concentrations of various medications (MS). The
laboratory that, when combined with competent development in the field of immunoassays
medical interpretation, directly impacts drug pre- changed the notion by enhancing the viability of
scribing procedures [1]. TDM, also refers to the the execution of assays [14], which was another
individualization of drug dosage by keeping drug significant milestone. Simultaneously, previously
concentrations in the plasma or blood within a proposed concepts like the relationship between
therapeutic range or window [2]. TDM combines dose and toxicity, PK, and drug-protein binding
pharmaceutics, pharmacokinetics, and pharma- were thoroughly studied, leading to improve-
codynamics knowledge to analyze a medication’s ments in sampling procedures and analysis.
efficacy and safety in a variety of clinical scenar- The trend in chromatographic methods during
ios [3–7]. This process tries to tailor therapy regi- the 1990s shifted to software programs for estab-
mens for the best possible outcomes for patients. lishing dosage regimens, the idea of noninvasive
TDM is typically described as the measurement and reductions in invasive therapies, wearable
of drug concentrations in various biological flu- sensors, and feedback-controlled smart devices.
ids and their interpretation in terms of therapeuti-
cally relevant parameters. Clinical pharmacists
and pharmacologists employ pharmacokinetic 7.2 Therapeutic Drug
ideas to assess these interpretations. In the 1960s, Monitoring (TDM)
the science of TDM brought a new dimension to
clinical practice with the publication of the first TDM is the clinical technique of monitoring ther-
pharmacokinetic studies linking mathematical apeutic agents in patients. The drug concentra-
theories to patient outcomes [3]. In the late 1960s tion is determined at fixed intervals to find out the
and early 1970s, clinical pharmacokinetics desirability and consistency of the amount of
became a topic. Pioneers in drug monitoring in drug in blood circulation, allowing individual
the 1970s focused on adverse drug reactions, dose treatments to be optimized [7]. TDM is
proving that by constructing therapeutic ranges, applied by most pharmaceutical companies to
the incidence of toxicity to drugs such as digoxin monitor drugs with limited therapeutic ranges,
[8], phenytoin, lithium, and theophylline [9] high pharmacokinetic inconsistency, and difficult
7 Therapeutic Drug Monitoring (TDM) and Toxicological Studies in Alternative Biological Matrices 97

target concentrations to monitor and document approach can detect digitalis sensitivity in
therapeutic and adverse effects [11]. digitalis-­treated individuals with toxicity associ-
ated with digitalis plasma values less than 2.0 ng/
mL. Aronson and Hardman [19] discovered that
7.2.1 Purpose of Therapeutic Drug selecting a dosage regimen after careful monitor-
Monitoring (TDM) ing of plasma concentration can reduce digitalis
toxicity to less than 4%. This approach is not
TDM necessitates an interdisciplinary approach. commonly used currently. Plasma digoxin con-
The TDM team consists of experts like scientists, centrations should be measured and monitored in
doctors, nurses, and pharmacists. They can prop- digitalis-treated patients with marginal renal
erly monitor the therapeutically meaningful drug function, the elderly, and patients with quick
concentrations at the site achieved or not. To atrial fibrillation who require higher digitalis dos-
guarantee that the best effective monitoring the ages for heart rate control [20]. When a digoxin-­
communication between various team members treated patient requires a loading dose of oral
is necessary, as represented in Fig. 7.1 [15, 16]. amiodarone, the digoxin dose should be decreased
Parameters like efficacy, plasma drug concen- first, and digoxin therapy should be adjusted
tration, compliance, drug-drug interactions, tox- based on any symptoms and signs of digoxin tox-
icity avoidance, and therapy discontinuation icity [21].
observation were used as the signs for monitoring
purposes [17, 18], though each parameter may
not apply equally to every agent. In many cases, 7.2.2 TDM: Estimating Plasma Drug
physicians monitor the drug concentration then Concentration
customize the dosage form for an individual dur-
ing various therapies. This method is particularly In general, finding the drug concentration at a
crucial in the case of drugs with limited therapeu- steady-state and altering the dose to attain the
tic ranges, i.e., narrow therapeutic window, such desired concentration is known to be related to
as lithium, cyclosporine, and aminoglycoside the efficacy of the drug. In this process, the over-
antibiotics. It may be highly valuable in cases all contribution of pharmacokinetic unpredict-
where changes in dosage regimen are suggested ability to changes in quantity needs can be
at a later stage, like patines having renal compli- discovered. However, there are significant inter-
cations. Drug toxicity can be detected clinically individual pharmacodynamic differences at a
in many circumstances. For example, acute phe- specified plasma concentration [22]; it is more
nytoin toxicity is rather straightforward to spot, common to target instead of a single threshold, a
and testing plasma concentrations is not essential concentration range. The assessment of drug con-
for judgment of disease, but it can be useful in centration in plasma or blood has become a good
correcting dosage later. Digoxin toxicity can replacement gauge of drug exposure in the body
have similar symptoms to heart disease; thus, for a restricted number of drugs where better
monitoring the plasma levels in subjects where association is found among plasma or blood con-
toxicity is suspected might assist confirm the centration response than dose response [23].
diagnosis. The extent of plasma digoxin content Therapeutic drug measurement is just one
in 260 subjects treated with Digitalis lanata aspect of TDM, which also includes expert clini-
preparations (digoxin, lanatoside C, beta methyl-­ cal interpretation of drug concentrations and
digoxin) allowed Aronson and Hardman to track pharmacokinetic evaluation. To obtain a maximal
some outcomes that would not have been visible clinical benefit, expert interpretation of a medica-
otherwise. The method’s applicability in the tion concentration measurement is required.
diagnosis of digitalis toxicity is limited by the Clinicians take a look at physiological indices of
significant connection among “toxic” as well as healing responses which include lipid concentra-
“nontoxic” plasma concentration levels [19]. The tions, blood glucose, blood strain, and coagula-
98 B. Basu et al.

Patient
Design of dosage assessment
schedule to reach a Drug
Diagnosis Selection of drug
target plasma administration A pharmacokinetic
concentration If dosage
Determination of model is applied &
adjustment
drug concentration clinical judgement
necessary
is used
A

Breath
Minutes to 1-2 Days Blood

Urine Several hrs to 1-3 Days Detection Window


Sweat
Weeks
Oral Fluid
Hair

Fig. 7.1 (a) Process for dosage decision with TDM. (b) Detection window for drugs in biological matrices

tion to monitor drug pharmacodynamics. Many interpreting the data are most likely due to timing
medications have no problems in measuring issues in the sample [28]. The blood sample can
motion, or the approach is insufficiently sensitive be collected into a heparinized tube or left to clot
[23]. As a result, TDM is predictable totally from for most medicines [29, 30].
the assumption that there is a distinct relationship
among dose and plasma or blood concentration,
as well as between blood drug concentration and 7.2.4 Practical Issues in TDM
pharmacodynamic properties [23]. There is no
reason to monitor plasma concentrations regu- In an ideal world, a high-quality drug assay
larly except when there is a precise cause [24]. would be completed in a therapeutically useful
amount of time. Many doctors believe that reports
from big chemical pathology laboratories, which
7.2.3 TDM Analytical Issues are operated by skilled scientists and equipped
with cutting-edge automated analyzers, will be
As previously stated, TDM necessitates the col- correct [23]. As a result, analytical laboratories
laboration of various disciplines of sciences, should confirm that methods are in place to get
including pharmacokinetics, pharmacodynamics, any missing details from the drug assay request
and laboratory analysis. The method of analysis that may be needed for proper clinical interpreta-
has a significant impact on the determination of tion of the results, such as dosage regimen and
pharmacokinetic parameters, which is often over- blood sampling time, and that each assay’s accu-
looked [25]. The analytical goals established in racy, precision, acuity, and specificity are docu-
TDMTherapeutic drug monitoring (TDM) are mented and measured regularly. Because the
assessing the nature of the issue to be resolved, most essential uses of the measures are during
choosing the suitable matrixMatrices and scien- dose changes and in detecting toxicity, when
tific method for addressing the problem [26, 27]. prompt judgments must be made, the assay out-
The important factors taken into consideration are comes should be provided rapidly, rather than
the timing of blood sampling, the type of blood within 1 day of receipt of the sample [24, 31].
sample, measuring technology, and data interpre- TDM involves a multidisciplinary approach
tation to make it more expressive. First and fore- that includes pharmacological, pharmacokinetic,
most, obtaining a blood sample for evaluating and pharmacodynamic approaches and analyses.
medication concentration at the proper time after TDM takes more than just a simple measurement
the dose is critical. The majority of problems in of a patient’s blood drug content and a compari-
7 Therapeutic Drug Monitoring (TDM) and Toxicological Studies in Alternative Biological Matrices 99

son to a target range to be effective. TDM, on the 7.2.6 Recent Advances in TDM
other hand, is critical in the creation of safe and Practice
effective therapeutic drugs, as well as their indi-
vidualization [13, 32]. TDM can also aid in the 7.2.6.1 Chromatographic Methods
detection of medication compliance issues in Even though chromatography has been effec-
noncompliant patient instances. The sample tively applied in clinical research, there are still
period about the dose, the dosage history, the several issues in liquid chromatography with tan-
patient’s response, and the planned clinical tar- dem mass spectrometry (LC-MS/MS) procedures
gets are all elements to consider when interpret- that need to be solved. Matrix interference,
ing drug concentration values [33, 34]. This despite its high specificity, can lead to falsely low
information can be utilized to determine the best or high readings in MS [44]. This is because the
dosing regimen for achieving the best response matrix and co-eluting chemicals can interfere
with the least amount of toxicity [35–39]. with the ionization process (through ion suppres-
sion/enhancement). Furthermore, LC-MS/MS
technologies have a lower throughput than tradi-
7.2.5 TDM’s Translational tional immunoassay platforms. Recent research
Challenges has focused on either fixing these limitations [45,
46] or leveraging or improving the inherent ben-
Because medication concentration in the blood efits of this strategy [47, 48].
is linked to pharmacological action, concentra- As a result, a substantial effort has been made
tion is a better indicator of effectiveness or toxic- to boost the throughput of chromatographic pro-
ity than dosage. TDM is the clinical practice of cedures [49]. Pioneering multiplex techniques
determining the concentration of drugs in blood, were reported have been all reported for antiret-
plasma, or other body fluids that are related to roviral medicines (ARVs) [50], antifungals [51],
blood drug concentrations. The medication con- antineoplastics [52], antibiotics [45, 46, 53], anti-
centration is then used to change the dosing regi- depressants [54], and immunosuppressive medi-
men by looking for a therapeutic range, which is cations [55] in the previous decade. Recent
a collection of concentrations or exposure inter- studies have employed ultra-performance liquid
vals. As a result, the analysis method’s specific- chromatography (UPLC)-MS/MS for simultane-
ity and sensitivity have a significant impact on ous measurement of antibiotics [56] and ARVs in
TDM reliability. These tests are now performed plasma [50] and breast milk samples [57].
in clinical TDM using either chromatographic Another area of attention is the use of TDM
procedures with specific detectors (typically research to nontraditional samples and sampling.
mass spectrometers) or immunoassays. Hair [50, 57], dried blood spots [58], urine [59],
Traditional approaches, on the other hand, have sweat [60], saliva [61, 62], and tissue biopsies
several practical drawbacks for the envisioned [63] have all been studied using LC-MS/MS.
large-scale, distributed TDM practice, such as a To overcome the limitation of LC-MS/MS,
lack of workflow uniformity, long turnaround like matrix effects, high costs of equipment and
times, and high instrumentation costs due to analysis, tedious development and optimization,
extensive sample preparation. In this sense, and requirement of a highly skilled operator, one
recent advances in sensing technology provide a can opt for HPLC methods in combination with
once-in-a-lifetime opportunity to overcome simpler detectors such as UV, flame ionization
these constraints and fully exploit TDM’s prom- detection (FID), or diode-array detection (DAD).
ise. Several recent evaluations using biosensing However, in such instances, overlapping peaks
technology [40–43] have properly addressed across analytes with similar retention periods
recent improvements and the current capabilities may be detected, which is exacerbated through
of such applications. the inclusion of unknown components in
100 B. Basu et al.

c­omplicated sample matrices. Chemometric reaction into a quantifiable signal [73]. These
techniques, which allow the removal of unantici- recognition elements have a high binding affinity
pated signal interferences from the total signal by for a given analyte and can be natural (antibodies,
mathematical modeling [64, 65], are one solution enzymes, membranes) or artificial (molecularly
to this problem. imprinted polymers or aptamers) [74, 75]. The
interaction between the analyte and the biorecep-
7.2.6.2 Immunoassays tor causes modification in local physicochemical
Immunoassays have been extensively studied for parameters, which translated into a readable sig-
the analysis of drugs in laboratories for many nal. Theoretically, this interaction can be utilized
years owing to their high affinity, small sample to track the concentrations of both the medicine
volume requirements, ease of working, good and the biomarker (protein/metabolite) [76].
compliance to high-throughput, and cost-­ Biosensors are characterized by their detect-
effectiveness. The underlying premise is that an ing method (optical, electrochemical, thermo-
analyte is detected by binding to analyte-specific metric, magnetic, or mechanical), sensing
antibodies. For analytes with large and small mechanism (direct/label-free or indirect/labeled),
molecular weights, the competitive and sandwich functionality (single- or multi-use), or degree of
techniques of assay design are most typically invasiveness. Recent review articles [43, 77] pro-
used. The majority of TDM assays use a competi- vide greater detail on material selection, identifi-
tive design, in which limited target drug mole- cation elements, signal detection algorithms,
cules in the material interact with labeled rivals amplification approaches, and various sensor
for a restricted number of binding sites. To put it application domains. Another fascinating point
another way, the signal produced is inversely pro- of view is the use of no-wash biosensors for real-­
portional to the analyte concentration in the time monitoring of tiny compounds [78, 79].
tested sample. However, as biopharmaceuticals Optical sensing is one of the most widely used
and biosimilars have become more prevalent, the methods for drug treatment monitoring. These
noncompetitive model has been instigated to play sensors may function in a variety of spectral
a greater role [66]. bands ranging from ultraviolet to near-infrared,
Multi-center evaluation studies [42, 67], the and they can be used with sophisticated analyti-
development of new reagents, immobilization cal techniques like NMR and Raman spectros-
techniques [68], and signal augmentation copy. Surface plasmon resonance (SPR) and
approaches to overcoming specificity challenges localized surface plasmon resonance (LSPR) are
have all received a lot of attention in recent years. two potent optical sensor technologies that rely
A detailed examination of claimed interferences on changes in the local refractive indices (RIs) of
for digoxin, immunosuppressant, anticonvulsant, metal films or nanoparticles. Because molecules
and tricyclic antidepressant immunoassays has of interest in TDM applications are typically tiny,
been provided [42]. Recent reviews [44, 69, 70] their impact on local RI can be negligible,
have compared immunoassay techniques used depending on the analyte size. This is why, in
for diverse TDM applications, particularly immu- recent SPR research, indirect approaches for sig-
nosuppressive medications. Studies comparing nal augmentation, such as those involving
the effectiveness of commonly available immu- nanoparticles, have been adopted. Biosensor-­
noassays to the performance of LC-MS/MS have based TDM investigations for anticancer medi-
also been conducted [71, 72]. cines [79, 80], antibiotics [81–84], and therapeutic
drug antibodies [85, 86] have all used these
7.2.6.3 Biosensors sophisticated approaches. SPR has also been
The term “biosensor” refers to a device that ana- used in conjunction with lateral flow tests to
lyzes biological samples that use molecular rec- assess the therapeutic immunogenicity of inflix-
ognition and bioreceptors to turn a biological imab [87].
7 Therapeutic Drug Monitoring (TDM) and Toxicological Studies in Alternative Biological Matrices 101

Surface-enhanced Raman spectroscopy is one TDM is critical in the management of trans-


of the innovative optical approache utilized in planted patients because it allows for the accurate
development anticancer drugs [88], antibiotics assessment of drug dosages with restricted thera-
[89], and antiepileptic drugs [90]. Although opti- peutic windows. TDM of immunosuppressants
cal sensors have fast turnaround times and great usually entails drawing blood before the next
sensitivity, they are generally plagued by large dose to establish the lowest drug concentration,
background signals, analyte signal attenuation known as the trough level [104]. Preliminary
through the matrix, high instrument costs, and tests on the bioassay implementation for drug
inadequate specificity due to nonspecific biore- detection in transplanted patients were success-
ceptor binding [91]. ful, and a novel therapeutic drug monitoring
Another prominent method for TDM is (TDM) point-of-care testing (POCT) biochip for
electrochemical sensing [92]. Anticancer med- immunosuppressant detection in transplanted
icines [93], antibiotics [94–97], and antifun- patients was created [105].
gals have recently been detected using
electrochemical biosensors combined with
C-nanotubes [98], nanoparticles/doped elec- 7.3 Science of Toxicology
trodes [99], and various bioreceptors such as
DNA [93], antibodies [94, 95], membranes The study of chemicals that affect living organ-
[100], and aptamers [96, 97]. Because of the isms is known as toxicology. Toxic exposure can
fundamental nature of bioreceptors, the issue happen transdermally through skin contact,
of nonspecific binding still exists in electro- orally, or through inhalation [106]. Toxicity test-
chemical sensors, as it does in optical sensors, ing is required to establish a foundation for the
and it must be addressed. control of substances with which humans and
other living things may come into contact,
7.2.6.4 Biochips whether intentionally or unintentionally.
Computer science, electronics, and biology have Cosmetics, pharmaceuticals, food additives, pes-
combined to create biochip, the most fascinating ticides, chemicals, additives, and consumer prod-
future technology. The potential applications are ucts are all tested for safety. A toxic impact can
numerous, both for research and therapeutic be caused by natural or man-made material, and
usage, with a sizable market [101]. One of the it can cause a wide range of symptoms, both short
most important parts of immunosuppression in and long term [107]. As a result, toxicity testing
transplanted patients is the proper dosage of employs a variety of procedures and rates of
immunosuppressants, which have the key role of exposure to the test organism to develop a more
preventing transplant rejection by partially inhib- precise estimate of the risk of harm that the test
iting the body’s immunological reaction to the material may pose to human health and the envi-
donated organ [102]. ronment. Animal research provides the majority
According to recent clinical investigations, the of human information about the toxicity of vari-
area under the concentration-time curve (AUC) ous compounds, albeit it is primarily used to
of immunosuppressant concentrations correlates extrapolate expected human physiological
better with immunosuppressive medication effi- responses [108, 109].
ciency and adverse effects than the classic TDM Serum, blood, and urine are used as specimens
of trough concentrations before the next dosage. in the great majority of toxicological tests done in
Clinicians altering immunosuppressant doses for clinical laboratories. Blood is frequently
patients in the initial phase after transplantation employed as a surrogate for measuring a drug’s
could be interested in a unique POCT (point-of-­ concentration at the site of action; however, it
care testing) equipment that allows for drug AUC may not accurately reflect the concentration at
monitoring [103]. the site of action [110]. Drugs’ exact sites of
102 B. Basu et al.

action range from nerve endings to receptors on variety of samples obtained from the persons
cells all over the body. Blood is chosen as a moni- under inquiry. Specimens such as blood, urine,
toring specimen because sampling from these nails, hair, bile, gastric contents, liver, and
areas is rarely possible. Blood is usually the brain tissue can all be valuable [115, 116]. The
transporter of the medicine from the place of demand for advanced analytical techniques
absorption to the site of action, according to used in toxicology to resolve conflicts is gradu-
logic. Because most medications are water-­ ally increasing. For toxicological analysis,
soluble, monitoring plasma or serum without the many procedures are employed to determine
influence of red cells is a sensible choice [111]. numerous medicines from a variety of biologi-
Other specimens may, however, be tested and, in cal materials [117]. Table 7.1 outlines the ana-
certain situations, may be required to provide the lytical methodologies utilized in various drug
desired clinical information. Because of the analysis investigations, as well as the types of
increased sensitivity of contemporary technol- biological matrices used.
ogy, most of this testing has only become achiev-
able in recent years. Many of these alternative 7.3.1.1 Reasons for Testing
specimens have extremely low quantities of phar- Toxicology screening (safety assessment) is a
maceuticals, drug metabolites, or other poisons way of figuring out how a drug of the issue has a
that are undetectable using traditional methods negative impact on the biological activity of an
[112, 113]. Liquid chromatography-tandem mass organism, given a certain duration, route of the
spectrometry (LC-MS) has been an analytical exposure, and concentration. Various reasons for
technique that has opened the door for the testing toxicological screening are described below.
of alternative specimens, particularly in the drug
industry. This review looks at these specimens 7.3.1.2 Pharmacologic Reasons
but does not examine them for infectious dis- Saliva was once one of the additional specimens
eases, which is another rationale for looking at evaluated to determine the “free” portion of a
different specimen matrices [114]. therapeutic medication or hormone. The amount
of drug that could bind to the receptor was better
reflected when saliva-free drug concentrations
7.3.1 Toxicology: Biological were used [14]. Alternative specimens are fre-
Sampling and Use of Different quently utilized in drug misuse testing and thera-
Analytical Techniques peutic drug management testing. Saliva was first
used to monitor free drugs like phenytoin, primi-
Toxicology is a sophisticated scientific area done, and ethosuximide [14, 144].
that makes use of a wide range of analytical
methods like laser diode thermal desorption- 7.3.1.3 Pharmacokinetic Reasons
tandem mass spectrometry (LDTD-MS-MS) The length of time a medicine stays in the body
[115], hyphenated liquid chromatographic varies depending on the samples. The amount of
techniques [116], chromatography using sil- medicine that circulates throughout the body is
ica-gel chromatograms [117], ultra-high per- reflected in the blood, which is a decent indica-
formance liquid chromatography-­tandem mass tion of the period when the drug reaches its
spectrometry [118], DNA typing [119], and intended target for treatments [145]. Urine mea-
capillary electrophoresis [120] Pesticides, surements can assist in defining the elimination
pharmaceuticals, natural products, industrial parameters and measuring the duration of action.
chemicals, metals, and contaminants are For some medications, other specimens like
among the forensic discoveries that can be saliva, sweat, hair, nails, etc. may offer particular
made using these procedures [119]. concentrations at the site of action or metabolism
Toxicological testing can be performed on a data [146].
7 Therapeutic Drug Monitoring (TDM) and Toxicological Studies in Alternative Biological Matrices 103

Table 7.1 Different biological samples and analytical techniques for toxicological studies
Sr.
no. Samples Method used References
1 Urine and blood Laser diode thermal desorption-tandem mass spectrometry (LDTD-MS-MS) [115]
Ultra-high performance liquid chromatography-tandem mass spectrometry [118]
Liquid chromatography-electrospray mass spectrometry [121]
Enzyme immunoassay and gas chromatography/mass spectrometry [122]
Liquid chromatography-mass spectrometry [91]
2 Biological samples Gas chromatography-surface ionization organic mass spectrometry [123]
Chemical ionization mass spectrometry with ammonia [124]
High-resolution mass spectrometry [125]
Liquid chromatography-mass spectrometry [126]
3 Hair Gas chromatography-negative chemical ionization tandem mass [127]
spectrometry
Matrix-assisted laser desorption/ionization (MALDI) combined with [128–131]
imaging mass spectrometry, gas chromatography-mass spectrometry
(GC-MS)
Liquid chromatography coupled with electrospray-ionization mass [130]
spectrophotometry (LC-ESI-MS)
4 Blood, plasma, Liquid chromatography-mass spectrometry [132]
serum, or urine
5 Nails Chromatography coupled with electrospray-ionization mass [133]
spectrophotometry (LC-ESI-MS)
6 DNA, biological Polymerase chain reaction (PCR) [134]
sample
7 Blood samples HPLC and capillary zone electrophoresis (CZE) techniques [135]
8 Sweat GC-MS, LC-MS, enzyme-linked immunosorbent assay (ELISA) [136, 137]
10 Oral fluid (GC-MS), liquid chromatography coupled to mass spectrometry (LC-MS) [133–135]
11 Vitreous humor GC-MS, gas chromatography with nitrogen-phosphorus detector (GC-NPD), [138–140]
(VH) gas chromatography coupled to tandem ion trap mass spectrometry
(GC-MS/MS)
12 Meconium ELISA, GC-MS, LC-MS, enzyme-multiplied immunoassay technique [141–143]
(EMIT)

7.3.1.4 Availability of Specimens 7.4 Alternative Matrices


The types of specimens available in clinical toxi- in Toxicology
cology are frequently limited by case circum-
stances. The most appropriate samples for detecting Alternative biological matrices have been studied
recent oral ingestions are gastric contents, although in toxicological testing for many years, owing to
this sample may not be readily obtainable due to their advantages over traditional matrices [151–
patient condition constraints or treatment. The abil- 154]. These benefits include the ability to gather
ity to get a urine sample may be hampered by the specimens more easily and with less invasiveness,
patient’s medical state [147, 148]. as well as bigger detection windows in some cir-
cumstances [153]. Furthermore, these matrices
7.3.1.5 Ease of Collection can be employed when blood samples are unavail-
Hair, saliva, and perspiration are examples of able, deteriorated, or influenced by postmortem
noninvasive specimens that can be collected by redistribution, as well as when drug consumption
non-skilled workers. Samples gathered like this is delayed [155]. However, drug levels in some of
are used in instances when obtaining patient or these matrices may be lower than in urine or blood
subject approval is more difficult. Biomonitoring due to inherent features and toxicokinetics. As a
and family toxicology studies are two recent result, contemporary instrument technology has
instances of this [149, 150]. enabled the investigation and analysis of other
104 B. Basu et al.

matrices; sophisticated and sensitive instruments more advanced instruments and better techniques
are allowing the identification of lower amounts of are required for the analysis of alternative matri-
medicines and toxins in some of these alternative ces to detect the lower concentrations of drugs in
specimens [151]. Saliva and hair are already well- the sample of interest [151].
known and have been used by several labs for drug
testing. Oral fluid, hair, sweat, meconium, breast
milk, and vitreous humor are some common alter- 7.5.1 Microextraction Techniques
native matrices in drug testing, based on their fea- in Different Biological
tures, advantages, and limitations [135]. Matrices
In toxicological assessments, selecting speci-
mens is a crucial step. To pick the best biological Microextraction procedures concede high recov-
fluid/tissue for the investigation, it’s critical to ery of target analytes with independence of the
understand the qualities of both the target analyte matrices. Preparation of the samples is the most
and the matrix. Biological fluids/tissues are alter- critical step in the analysis of analytes of interest.
native biological matrices that can provide extra Conventional procedures for preparing samples
information and advantages over blood and urine like solid-phase extraction (SPE) and liquid-­
testing in a variety of ways, including sample col- liquid extraction (LLE) are used over the years.
lection/preparation/analysis complexity and For the last few decades, the microextraction
detection window [134]. Furthermore, when technique has taken the attention due to the pres-
blood and urine are not accessible, these matrices ence of various drawbacks with these classical
can be collected and examined. However, each of sample preparation techniques. The major clas-
these alternate matrices has its own set of quali- sification of microextraction techniques is liquid-­
ties, benefits, and drawbacks that must be evalu- phase microextraction (LPME), solid-phase
ated. According to toxicology, alternative microextraction (SPME), microextraction by
matrices will be examined more frequently in the packed sorbent (MEPS), and fabric-phase sorp-
future [156]. tive extraction (FPSE) [158, 159].
SPME is the simple, minimized number of
steps involved in sample preparation, reduced
7.5 Different Approaches volume solvent, and cost-effective process with
in Various Biological reduced analysis time [152, 160]. Microextraction
Matrices, with an Emphasis methods were developed to predict cannabinoids
on Toxicology in various matrices by LPDE or SPME [161].
LPME is beneficial for water analysis of several
The application of alternative matrices has been pesticide groups in the environmental analysis
an upsurge in forensic toxicology. Conventional [162]. Moreover, these techniques are useful for
biological fluids like blood, plasma, serum, and pharmacokinetics study, direct in vivo sampling,
urine specimens are normally used for drug test- and chip-based microfluidic systems.
ing in forensic toxicology [151]. For the last few
years, enormous research work has been done in
this field to investigate different types of drug and 7.5.2 Other Green Extraction
their metabolites in biological samples [157]. Technique Application
Alternative biological matrices are more compel-
ling in toxicological testing for their benefits over Besides SPME and LPME, other important green
conventional matrices in terms of larger detection extraction techniques are used in the specimen
windows and the requirement of fewer samples preparation of alternative biosamples.
[158, 159]. However, insufficient drug levels Amphetamines and methadone in saliva matrices
have been found in some of these alternative were evaluated by Meng and Yang with the appli-
matrices used for toxicological studies. Thus, cation of small-volume LPE techniques [163].
7 Therapeutic Drug Monitoring (TDM) and Toxicological Studies in Alternative Biological Matrices 105

FPSE is first introduced in 2014. These tech- drug discovery is the identification of new chemi-
niques integrate principles typical of SPME and cal entities, which have desired biological activ-
SPE and use a flexible fabric surface of different ity. For the treatment of a particular disease, the
materials [164]. active learning algorithms can identify the poten-
tial lead compound. For preclinical safety evalu-
ation and toxicity, the quantitative-structure
7.5.3 Applications of Mass activity relationship (QSAR) method is used,
Spectrometry Techniques which creates a link between chemical structure
in Alternative Biological and biological activities [169]. AI has an impor-
Matrices tant role in omics study, which are helpful to
identify disease pathogenesis by finding the new
There are different types of mass spectrometers biomarkers and integrating drugs with the dis-
available for different applications. Coupling of ease. AI-based models have been developed for
different chromatography techniques to mass COVID-19 drug discovery and vaccine develop-
spectrometry is used for the identification and ment [170]. Drug target interaction and pharma-
quantification of metabolites from an unknown cological properties of the potential drug
drug, i.e., LC-MS and GC-MS [165, 166]. compound can be predicted by the use of AI
Looking forward to the precise estimation and (Fig. 7.2a) [171].
analysis of compounds for their pure structural Chemical toxicity is defined as the adverse
information, one would opt for MALDI (matrix-­ effect produced by exposure to chemical agents,
assisted laser desorption ionization) coupled to which can be referred to by LD50, moderate,
time of flight (TOF) or quadrupole coupled to and high toxicity [171]. Evaluation of toxicity
time of flight [167]. Endogenous matrix compo- of the chemical agents can be done in animal
nents, metabolites, degradation products, exoge- models. The first principle (replacement) of
nous xenobiotics, and other interfering chemicals three “R” principles (replacement, reduction,
could be present in the alternative biological and refinement), refers to the use of alternative
matrix. Samples spiked with calibration (refer- methods over the use of animal models [172].
ence) standards are used to analyze drugs in the Most of the newly discovered drug molecules
biological matrix. Furthermore, hallucinogens are excluded based on toxicological studies. In
such as LSD and its metabolites are quantified terms of ethical concern and result reproducibil-
using an ultrafast and sensitive microflow liquid ity, the researcher found more reliable findings
chromatography-MS (MFLC-MS/MS) [167]. with the use of AI-based models compared to
exchange traditional in vivo toxicity models. In
food technology, to determine the concentration
7.6 Artificial Intelligence (AI) of harmful compounds, spectroscopic tech-
in Toxicity and Drug niques include hyperspectral imaging, fluores-
Discovery cence spectroscopy, near-infrared (IR)
spectroscopy, Fourier transform IR (FTIR), and
“Artificial intelligence” was first coined by John Raman spectroscopies; biosensors are used
McCarthy in 1956 [168]. AI is the set of theories which are based on AI algorithms, i.e., machine
and techniques used to create machines that are learning (ML), deep learning (DL), artificial
capable of stimulating intelligence and perform- neural networks (ANNs), deep neural networks
ing functions, executed by humans, and that can (DNNs), and convolutional neural networks
mimic human intelligence [169]. The first step in (CNNs) (Fig. 7.2b) [171, 172].
106 B. Basu et al.

Fig. 7.2 (a) Artificial intelligence in drug discovery. (b) Stages making up in silico toxicology

7.7 Promises and Pitfalls the optimal sample matrix for the application.
of Alternative Matrices Whether it’s urine, blood, hair, oral fluid, breath,
or sweat, each matrix has its own set of benefits
Many labs are increasingly analyzing fit-for-­ and drawbacks, as well as unique information
purpose alternative sample matrices such as oral that can be utilized to interpret drug usage and
fluid, hair, and exhaled breath to detect an the cause of impairment or death. One factor to
expanding range of drugs faster and with greater consider is the detection window, or how long
certainty, as well as to make sample collection as evidence of drug usage may be discovered in a
easy, noninvasive, and trustworthy as possible given matrix. Analysts can, for example, deter-
[173]. These analyses are becoming more com- mine use days, weeks, and even months in the
mon because of advances in toxicokinetics (TKS) past based on hair length (Fig. 7.1b). Urine and
research and new analytical technology [174]. blood offer data on drug usage over a signifi-
cantly shorter period [176].
Window of Detection Recent advancements in sample preparation,
The “detection window” for a drug and its metab- chromatography, and MS technologies are assist-
olites in distinct biological matrices is a crucial ing labs in overcoming the challenges of adopt-
concept in PKS and TKS. A drug’s detection ing alternate sample matrix analysis. Alternative,
window must be long enough for sampling to purpose-built sample matrices are attracting a lot
occur to identify it [175]. Today it tends to look at of attention because they have the potential to
7 Therapeutic Drug Monitoring (TDM) and Toxicological Studies in Alternative Biological Matrices 107

make sample collection more convenient, nonin- tem pharmacology which focuses on target
vasive, and dependable [177]. Though interpret- effects and clinical adverse events. Furthermore,
ing drug concentrations in alternative sample in silico ADR mining contains a massive data
matrices is still a hot research area, advances in pool for medication safety. Databases like
analytical technologies have made the testing of DrugBank, SIDER, ChEMBL, ChEBI, PubChem,
alternative, fit-for-purpose sample matrices a fea- Reactome, and KEGG are available for data min-
sible reality. Few detection methods were sensi- ing. Recently, researchers proposed modular
tive enough to measure the target medicines and assembly of drug safety subnetworks in which
metabolites at the levels required when other using knowledge bases connected to literature
matrices were introduced [174]. searches, genome data generated a network of
proteins. Further, it is compiled into network
metrics for the prediction of adverse effects in
7.8 Artificial Intelligence (AI) new drugs [185]. Another approach is based on
in Toxicology and TDM drug-drug interaction by literature mining and
integration of drug-gene interaction and creating
7.8.1 Artificial Intelligence a model for prediction of drug-drug interaction
for Clinical Toxicity focused on skin diseases as represented in Fig. 7.3
and Patient Safety [186].

Bringing novel medications to market poses a


significant challenge in terms of drug safety. To 7.8.2 Artificial Intelligence (AI)
solve the problem of drug safety there are two in the Prediction of Adverse
systems. Before the approval of the drug for Drug Reaction (ADR)
patient use, clinical trials are performed to ensure and a Case Study
the safety and efficacy of its intended use. AI
approaches are increasingly routinely optimized Adverse drug reactions are pharmacological
to enhance patient care, diagnosis, therapy, and occurrences caused by the interaction between
patient follow-up. Machine learning (ML) and drugs which can be caused by a variety of factors
deep learning (DL) are two AI subfields. After the like drug-drug interaction. Anticipating and
drug is marketed, the agencies collect the adverse reducing ADRs upfront in the drug development
event reports for performing analyses on follow-­ process can improve drug safety while also low-
ups but can be complicated for rare events like ering costs. Few researchers developed an inno-
drug-drug interaction [178]. Researchers thus vative and effective computational methodology
now focus on various statistical and computa- for accurately predicting adverse drug reactions
tional tools to minimize the loopholes and act as (ADRs) for trial drugs by combining protein-­
an add-on to the pharmacovigilance toolbox protein interaction (PPI) networks, drug target
[179, 180]. Individual case safety reports data, and gene ontology (GO) annotations with
(ICSRs), which are self-reported, have been a clinical observation data, and the results also
primary data collection method for post-­ depict the importance of including prior
marketing drug safety research. The Naranjo ­knowledge of gene annotations and gene net-
algorithm and Venulet algorithm are the most works, for better estimation of ADR [187].
well-known methods for determining causation Deep learning (DL) is an artificial neural net-
[181, 182]. Consequently, dataData mining tech- work (ANN) extension that learns relevant attri-
niques were able to detect statistical connections butes from unprocessed data using a cascade of
between medications and adverse events using ANNs. To predict activity levels for distinct com-
spontaneous reporting systems (SRSs) such as FDA pounds, DL is used with a set of varied QSAR
Adverse Event Reporting System (FAERS) [183, datasets [188]. Recently researchers employed
184]. Another approach is the application of sys- DL to develop drug-induced liver injury predic-
108 B. Basu et al.

TYPES OF AI

Machine learning Deep learning


 Make prediction directly form data  Prepared to handle very large,
Preclinical Clinical toxicity and
 Less complex unstructured, and complex feature-rich
toxicity  e.g., survey vector machine, toxicity data patient safety
random forests, logistic regression  More complex
 e.g., artificial neural network,
convolutional neural network

 Drug toxicity predication  Market monitoring


 Diverse model of patient profile  Safety comparison base analyses
 Give insights on MOA  Precision modelling of patients
 Helps in drug development and clinical  Helps in regulatory affairs and clinical
trials decisions

Fig. 7.3 Role of artificial intelligence (AI) in drug toxicity and patient safety

tion models with structural data for measuring ies [192]. Toxicologists, clinicians, and AI
hepatotoxicity [189]. CNNs are a type of deep programmers should work together to reduce the
convolutional neural network that generates adverse effects with the help of AI.
interpretations of raw pictures from pixel data as AI has been also used to interpret plasma con-
a stack of images from which attributes may be centration in anti-retroviral for TDM. The objec-
retrieved and then used to identify complicated tive of the study was to create a software-based
patterns. Images of cells that were pre-treated system to interpret plasma concentration in anti-
with a series of medications were used to train retrovirals for therapeutic drug monitoring [193].
CNNs to estimate toxicity. In this method, differ- From 199 HIV-positive patients in a TDM
ent medications, nuclear stains, and cell lines can study (CCTG 578), data were extracted.
all be used to predict a wide range of toxicity Pharmacokinetic parameters of lopinavir and efa-
pathways [190]. Another group of researchers virenz were modeled using a Bayesian approach
created machine learning algorithms, along with and interpreted by a specialized committee hav-
a deep learning model, that can anticipate ADRs ing expertise in HIV and pharmacologists pro-
and detect the chemical substructures linked with vided TDM suggestions. The pharmacokinetic
those ADRs without having to define the sub- models acted as the base to form the artificial
structures ahead of time. The authors evaluated intelligence (AI) system which could predict the
the performance of the model with ten other fin- exposure of drugs and data interpretation of the
gerprint models. It was discovered that neural pharmacokinetic parameters and initiate TDM
fingerprints performed the best among all meth- suggestions. The recommendations from the
ods of predicting ADRs [191]. Some researchers expert committee and modeled pharmacokinetic
utilized a selection strategy to find important fea- exposure proven to be optimal for validation of
tures and machine learning algorithms to create the results obtained from the AI system [193].
computational approaches that could predict neu- It has been seen that among all the patients,
rological ADRs before preclinical toxicity stud- lopinavir was administered to 67 patients and
7 Therapeutic Drug Monitoring (TDM) and Toxicological Studies in Alternative Biological Matrices 109

efavirenz was administered to 46 patients and 3 As a result, rather than automatized TDM per-
patients were administered with both the drugs. formed by a computer, we propose computer-­
After 4 hours of administration dose concentra- assisted TDM by practitioners. TDM’s
tion was estimated and a high correlation of lopi- development, diffusion, and oversight will con-
navir and efavirenz between the estimate through tinue to necessitate the involvement of competent
AI and PK modeled values was obtained clinical pharmacologists who are available to
(r > 0.79; P < 0.0001). Considerable difference advise in difficult circumstances. Automatically
was seen in the mean predicted 4-hour concentra- collecting monitoring data, on the other hand,
tion of lopinavir (7.99 μg/ml against 8.79 μg/ml; will amass large datasets suitable for intriguing
P < 0.001) and efavirenz (4.16 μg/ml against new sorts of medical research [197]. Finally, the
3.89 μg/ml; P = 0.02). 53 out of 69 lopinavir global trend toward patient empowerment, aided
cases were agreed by the AI and specialized com- by appropriate mobile apps, will encourage
mittee TDM suggestions, whereas 47 out of 49 patients to take an active role in their therapeutic
were in the case of efavirenz [193]. The study monitoring. Most patients will appreciate being
concluded that the AI system correctly calculated able to see the circulation exposure caused by
lopinavir and efavirenz through concentrations their medications, and an increasing number of
and was in good agreement with TDM recom- them want to be in charge of self-monitoring.
mendations for efavirenz- and lopinavir-treated
patients from the specialized committee [193].
7.10 Challenges Faced
in Managing Patient
7.9 Importance and Future Health Data
Perspective of TDM
Patient health data management is a complex pro-
There are some reasons to be optimistic about cess. The majority of healthcare businesses only
TDM’s future. Analytical methods are improving work with digital data. The amount continues to
in terms of simplicity, operability, and automation, rise, laws are continuously changing, and trading it
allowing for the use of different matrices such as remains difficult. Because standardization is still
saliva, interstitial fluid, or dried blood spots, which absent, the path to healthcare interoperability is
makes sample collection easier [194]. In a differ- riddled with potholes. Fragmentation persists in
ent perspective, fresh analytical methods are being the absence of interconnected systems.
used to produce point-of-care TDM methods Organizations should create a strategic plan to
[195]. TDM research in other areas is also on the achieve internal interoperability at the very least.
rise, with an increasing number of scientists from There might be a lot of duplicate information in
other disciplines, particularly biomedical engi- the system, or it could be erroneous and old and it
neers, uncovering this wide field of study. cannot be even deleted due to the various regula-
Therapeutic monitoring in its broadest sense will tions. The more data in the system increases the
become a mainstay of tomorrow’s precision medi- strain on the software leading to an increase in the
cine, encompassing not only drug concentrations cost of software hosting. There is also the problem
but also a variety of efficacy and tolerability bio- of data analytics since the huge data takes time for
markers [84]. Progress in medical information analysis which makes the process cumbersome.
technology will soon integrate all of our healthcare
systems, removing the challenges of communica-
tion and medical use of concentration measure- 7.11 Conclusions
ments. TDM pharmacological interpretation will
benefit from new computer tools with increased TDM is required for just a tiny percentage of
user-friendliness and performance, which are cur- medications used in pharmacotherapy, but these
rently being developed [196]. drugs must achieve maximal efficacy and avoid
110 B. Basu et al.

drug toxicity. TDM can also help avoid bad side measurement of serum levels. Ann Intern Med.
1974;80(4):516–9.
effects. The old approach of TDM for avoiding 9. Atkinson AJ Jr, Nordstrom K. The challenge of
drug toxicity needs to be revised due to new in-hospital medication use: an opportunity for
developments in information technology, new clinical pharmacology. Clin Pharmacol Ther.
analytical procedures for less frequently moni- 1996;60(4):363–7.
10. Shenfield GM. Therapeutic drug monitoring beyond
tored drugs, and new clinical pharmacological 2000. Br J Clin Pharmacol. 2001;52 Suppl 1(Suppl
expert opinions in the presentation of laboratory 1):3S–4S.
medicine results. Rather than establishing the 11. Ensom MH, Davis GA, Cropp CD, Ensom
cause of an existing adverse drug reaction, RJ. Clinical pharmacokinetics in the 21st century.
Does the evidence support definitive outcomes? Clin
TDM can now be utilized to avoid an adverse Pharmacokinet. 1998;34(4):265–79.
drug reaction. TDM has several advantages, 12. Nelson E. Kinetics of drug absorption, distribu-
including significant cost savings due to the tion, metabolism, and excretion. J Pharm Sci.
patient’s shorter stay in the hospital and the 1961;50:181–92.
13. Finney DJ. The design and logic of a monitor of drug
avoidance of costly identification and treatment use. J Chronic Dis. 1965;18:77–98.
of an adverse medication event. TDM’s success 14. Horning MG, Brown L, Nowlin J, Lertratanangkoon
is mostly dependent on physicians, clinical phar- K, Kellaway P, Zion TE. Use of saliva in thera-
macologists, pharmacists, analytical laboratory peutic drug monitoring. Clin Chem. 1977;23(2 pt.
1):157–64.
scientists, and nurses working together to provide 15. Gross AS. Best practice in therapeutic drug moni-
a coordinated and integrated approach to patient toring. Br J Clin Pharmacol. 2001;52 Suppl 1(Suppl
care. Alternative matrices are increasingly being 1):5S–10S.
used in clinical and forensic toxicological testing. 16. Dipiro JT, Spruill WJ, Blouin RA. Lesson 1: intro-
duction to pharmacokinetics and pharmacodynam-
A new study is needed to develop more analytical ics. In: Concepts in clinical pharmacokinetics. 3rd
tools and a better knowledge of drug behavior in ed. Bethesda: ASPS; 2002. p. 1–11.
various matrices. 17. Bochner F, Tonkin A. The clinician and thera-
peutic drug monitoring in the 1990s. Med J Aust.
1993;158(6):422–6.
18. Reynolds DJ, Aronson JK. ABC of monitoring drug
References therapy. Making the most of plasma drug concentra-
tion measurements. BMJ. 1993;306(6869):48–51.
1. Touw DJ, Neef C, Thomson AH, Vinks AA. Cost-­ 19. Aronson JK, Hardman M. ABC of monitoring drug
effectiveness of therapeutic drug monitoring: a sys- therapy. Measuring plasma drug concentrations.
tematic review. Ther Drug Monit. 2005;27(1):10–7. BMJ. 1992;305(6861):1078–80.
2. Birkett DJ. Pharmacokinetics made easy: therapeu- 20. Cristodorescu R, Deutsch G, Drăgan S. Clinical util-
tic drug monitoring. Aust Prescr. 1997;20:9–11. ity of plasma digoxin measurements. Med Interne.
3. Tange SM, Grey VL, Senécal PE. Therapeutic drug 1989;27(1):25–32.
monitoring in pediatrics: a need for improvement. J 21. DeVore KJ, Hobbs RA. Plasma digoxin con-
Clin Pharmacol. 1994;34(3):200–14. centration fluctuations associated with timing of
4. Reed MD, Blumer JL. Therapeutic drug monitoring plasma sampling and amiodarone administration.
in the pediatric intensive care unit. Pediatr Clin N Pharmacotherapy. 2007;27(3):472–5.
Am. 1994;41(6):1227–43. 22. Levy G. Pharmacologic target-mediated drug dispo-
5. Kearns GL, Moss MM, Clayton BD, Hewett sition. Clin Pharmacol Ther. 1994;56(3):248–52.
DD. Pharmacokinetics and efficacy of digoxin 23. Vozeh S. Cost-effectiveness of therapeutic drug mon-
specific Fab fragments in a child following itoring. Clin Pharmacokinet. 1987;13(3):131–40.
massive digoxin overdose. J Clin Pharmacol. 24. Spector R, Park GD, Johnson GF, Vesell
1989;29(10):901–8. ES. Therapeutic drug monitoring. Clin Pharmacol
6. Ohning BL. Neonatal pharmacodynamics--basic Ther. 1988;43(4):345–53.
principles. II: drug action and elimination. Neonatal 25. McInnes GT. The value of therapeutic drug monitor-
Netw. 1995;14(2):15–9. ing to the practising physician--an hypothesis in need
7. Ohning BL. Neonatal pharmacodynamics--ba- of testing. Br J Clin Pharmacol. 1989;27(3):281–4.
sic principles. I: drug delivery. Neonatal Netw. 26. Sjoqvist F. Interindividual differences in drug
1995;14(2):7–12. responses: an overview. In: Variability in drug ther-
8. Duhme DW, Greenblatt DJ, Koch-Weser apy: description, estimation, and control. New York:
JAN. Reduction of digoxin toxicity associated with Raven Press; 1985. p. 1–9.
7 Therapeutic Drug Monitoring (TDM) and Toxicological Studies in Alternative Biological Matrices 111

27. Bowers LD. Analytical goals in therapeutic drug drug monitoring of β-lactam antibiotics: a structured
monitoring. Clin Chem. 1998;44(2):375–80. review. Int J Antimicrob Agents. 2015;46(4):367–75.
28. Smith PE. Third international conference on harmo- 43. Meneghello A, Tartaggia S, Alvau MD, Polo F,
nization of technical requirements for registration Toffoli G. Biosensing technologies for thera-
of pharmaceuticals for human use—a toxicologist’s peutic drug monitoring. Curr Med Chem.
perspective. Toxicol Pathol. 1996;24(4):519–28. 2018;25(34):4354–77.
29. Winter ME. Part 1: interpretation of plasma drug 44. Mika A, Stepnowski P. Current methods of the
concentration. In: Winter ME, editor. Basic clinical analysis of immunosuppressive agents in clini-
pharmacokinetics. 3rd ed. Philadelphia: Lippincott cal materials: a review. J Pharm Biomed Anal.
Williams & Wilkins; 2004. p. 73–96. 2016;127:207–31.
30. Patteet L, Cappelle D, Maudens KE, Crunelle CL, 45. Decosterd LA, Widmer N, André P, Aouri M, Buclin
Sabbe B, Neels H. Advances in detection of anti- T. The emerging role of multiplex tandem mass
psychotics in biological matrices. Clin Chim Acta. spectrometry analysis for therapeutic drug monitor-
2015;441:11–22. ing and personalized medicine. TrAC Trends Anal
31. Glazko AJ. Phenytoin, chemistry and methods Chem. 2016;84:5–13.
of determination. In: Antiepileptic drugs. 3rd ed. 46. Sime FB, Roberts MS, Roberts JA, Robertson
New York: Raven; 1989. p. 159–76. TA. Simultaneous determination of seven β-lactam
32. Steijns LS, Bouw J, van der Weide J. Evaluation of antibiotics in human plasma for therapeutic
fluorescence polarization assays for measuring val- drug monitoring and pharmacokinetic studies. J
proic acid, phenytoin, carbamazepine and phenobar- Chromatogr B Analyt Technol Biomed Life Sci.
bital in serum. Ther Drug Monit. 2002;24(3):432–5. 2014;960:134–44.
33. Patel JA, Clayton LT, LeBel CP, McClatchey 47. Lindner JM, Vogeser M, Sorg K, Grimm SH. A
KD. Abnormal theophylline levels in plasma by flu- semi-automated, isotope-dilution high-resolution
orescence polarization immunoassay in patients with mass spectrometry assay for therapeutic drug moni-
renal disease. Ther Drug Monit. 1984;6(4):458–60. toring of antidepressants. Clin Mass Spectrom.
34. Hicks JM, Brett EM. Falsely increased digoxin con- 2019;14 Pt B:89–98.
centrations in samples from neonates and infants. 48. Bhatnagar A, McKay MJ, Crumbaker M, Ahire K,
Ther Drug Monit. 1984;6(4):461–4. Karuso P, Gurney H, et al. Quantitation of the anti-
35. Frank EL, Schwarz EL, Juenke J, Annesley TM, cancer drug abiraterone and its metabolite Δ(4)-
Roberts WL. Performance characteristics of four abiraterone in human plasma using high-­resolution
immunoassays for antiepileptic drugs on the mass spectrometry. J Pharm Biomed Anal.
IMMULITE 2000 automated analyzer. Am J Clin 2018;154:66–74.
Pathol. 2002;118(1):124–31. 49. Veringa A, Sturkenboom MGG, Dekkers BGJ,
36. Wu SL, Li W, Wells A, Dasgupta A. Digoxin-like Koster RA, Roberts JA, Peloquin CA, et al. LC-MS/
and digitoxin-like immunoreactive substances in MS for therapeutic drug monitoring of anti-infective
elderly people. Impact on therapeutic drug monitor- drugs. TrAC Trends Anal Chem. 2016;84:34–40.
ing of digoxin and digitoxin concentrations. Am J 50. Bollen PDJ, de Graaff-Teulen MJA, Schalkwijk S,
Clin Pathol. 2001;115(4):600–4. van Erp NP, Burger DM. Development and vali-
37. Steimer W, Müller C, Eber B. Digoxin assays: fre- dation of an UPLC-MS/MS bioanalytical method
quent, substantial, and potentially dangerous inter- for simultaneous quantification of the antiretro-
ference by spironolactone, canrenone, and other viral drugs dolutegravir, elvitegravir, raltegra-
steroids. Clin Chem. 2002;48(3):507–16. vir, nevirapine and etravirine in human plasma. J
38. Somerville AL, Wright DH, Rotschafer Chromatogr B Analyt Technol Biomed Life Sci.
JC. Implications of vancomycin degradation prod- 2019;1105:76–84.
ucts on therapeutic drug monitoring in patients 51. Yoon SJ, Lee K, Oh J, Woo HI, Lee SY. Experience
with end-stage renal disease. Pharmacotherapy. with therapeutic drug monitoring of three antifungal
1999;19(6):702–7. agents using an LC-MS/MS method in routine clini-
39. Sym D, Smith C, Meenan G, Lehrer M. Fluorescence cal practice. Clin Biochem. 2019;70:14–7.
polarization immunoassay: can it result in an overes- 52. van Nuland M, Venekamp N, de Vries N, de Jong
timation of vancomycin in patients not suffering from KAM, Rosing H, Beijnen JH. Development and
renal failure? Ther Drug Monit. 2001;23(4):441–4. validation of an UPLC-MS/MS method for the
40. Shafiee A, Ghadiri E, Kassis J, Atala A. Nanosensors therapeutic drug monitoring of oral anti-hormonal
for therapeutic drug monitoring: implica- drugs in oncology. J Chromatogr B Analyt Technol
tions for transplantation. Nanomedicine (Lond). Biomed Life Sci. 2019;1106–1107:26–34.
2019;14(20):2735–47. 53. Parker SL, Pandey S, Sime FB, Lipman J, Roberts
41. McKeating KS, Aubé A, Masson JF. Biosensors and JA, Wallis SC. A validated LC-MSMS method
nanobiosensors for therapeutic drug and response for the simultaneous quantification of merope-
monitoring. Analyst. 2016;141(2):429–49. nem and vaborbactam in human plasma and renal
42. Carlier M, Stove V, Wallis SC, De Waele JJ, replacement therapy effluent and its application
Verstraete AG, Lipman J, et al. Assays for therapeutic
112 B. Basu et al.

to a pharmacokinetic study. Anal Bioanal Chem. P-glycoprotein in single human renal core biopsies.
2019;411(29):7831–40. Ther Drug Monit. 2018;40(3):292–300.
54. Weber J, Oberfeld S, Bonse A, Telger K, Lingg R, 64. Vosough M, Tehrani SM. Development of a fast
Hempel G. Validation of a dried blood spot method HPLC-DAD method for simultaneous quantitation
for therapeutic drug monitoring of citalopram, mir- of three immunosuppressant drugs in whole blood
tazapine and risperidone and its active metabolite samples using intelligent chemometrics resolving
9-hydroxyrisperidone using HPLC-MS. J Pharm of coeluting peaks in the presence of blood interfer-
Biomed Anal. 2017;140:347–54. ences. J Chromatogr B Analyt Technol Biomed Life
55. Krnáč D, Reiffová K, Rolinski B. A new HPLC-MS/ Sci. 2018;1073:69–79.
MS method for simultaneous determination 65. Sun XD, Wu HL, Liu Z, Chen Y, Chen JC, Cheng
of Cyclosporine A, Tacrolimus, Sirolimus and L, et al. Target-based metabolomics for fast and
Everolimus for routine therapeutic drug monitoring. sensitive quantification of eight small molecules in
J Chromatogr B Analyt Technol Biomed Life Sci. human urine using HPLC-DAD and chemometrics
2019;1128:121772. tools resolving of highly overlapping peaks. Talanta.
56. Magréault S, Leroux S, Touati J, Storme T, Jacqz-­ 2019;201:174–84.
Aigrain E. UPLC/MS/MS assay for the simultane- 66. Li M, An W, Wang L, Zhang F, Li J, Zhang Y, et al.
ous determination of seven antibiotics in human Production of monoclonal antibodies for measuring
serum-application to pediatric studies. J Pharm Avastin and its biosimilar by Sandwich ELISA. J
Biomed Anal. 2019;174:256–62. Immunol Methods. 2019;469:42–6.
57. Ramírez-Ramírez A, Sánchez-Serrano E, Loaiza-­ 67. Shipkova M, Vogeser M, Ramos PA, Verstraete AG,
Flores G, Plazola-Camacho N, Rodríguez-Delgado Orth M, Schneider C, et al. Multi-center analytical
RG, Figueroa-Damián R, et al. Simultaneous quan- evaluation of a novel automated tacrolimus immuno-
tification of four antiretroviral drugs in breast milk assay. Clin Biochem. 2014;47(12):1069–77.
samples from HIV-positive women by an ultra-high 68. Broto M, McCabe R, Galve R, Marco MP. A
performance liquid chromatography tandem mass high-specificity immunoassay for the therapeutic
spectrometry (UPLC-MS/MS) method. PLoS One. drug monitoring of cyclophosphamide. Analyst.
2018;13(1):e0191236. 2019;144(17):5172–8.
58. Velghe S, Deprez S, Stove CP. Fully automated 69. Zhang Y, Zhang R. Recent advances in analyti-
therapeutic drug monitoring of anti-epileptic drugs cal methods for the therapeutic drug monitor-
making use of dried blood spots. J Chromatogr A. ing of immunosuppressive drugs. Drug Test Anal.
2019;1601:95–103. 2018;10(1):81–94.
59. Naicker S, Guerra Valero YC, Ordenez Meija 70. Freudenberger K, Hilbig U, Gauglitz G. Recent
JL, Lipman J, Roberts JA, Wallis SC, et al. A advances in therapeutic drug monitoring of immu-
UHPLC-MS/MS method for the simultaneous nosuppressive drugs. TrAC Trends Anal Chem.
determination of piperacillin and tazobactam in 2016;79:257–68.
plasma (total and unbound), urine and renal replace- 71. Juenke JM, Miller KA, Ford MA, McMillin
ment therapy effluent. J Pharm Biomed Anal. GA, Johnson-Davis KL. A comparison of two
2018;148:324–33. FDA approved lamotrigine immunoassays with
60. Xing Y, Fuss H, Lademann J, Huang MD, Becker-­ ultra-high performance liquid chromatogra-
Ross H, Florek S, et al. A new concept of efficient phy tandem mass spectrometry. Clin Chim Acta.
therapeutic drug monitoring using the high-resolution 2011;412(19–20):1879–82.
continuum source absorption spectrometry and the 72. Hashi S, Masuda S, Kikuchi M, Uesugi M, Yano I,
surface enhanced Raman spectroscopy. Spectrochim Omura T, et al. Assessment of four methodologies
Acta Part B At Spectrosc. 2018;142:91–6. (microparticle enzyme immunoassay, chemilumines-
61. Ventura S, Rodrigues M, Pousinho S, Falcão A, cent enzyme immunoassay, affinity column-­mediated
Alves G. Determination of lamotrigine in human immunoassay, and flow injection assay-tandem mass
plasma and saliva using microextraction by packed spectrometry) for measuring tacrolimus blood con-
sorbent and high performance liquid chromatogra- centration in Japanese liver transplant recipients.
phy–diode array detection: an innovative bioanalyti- Transplant Proc. 2014;46(3):758–60.
cal tool for therapeutic drug monitoring. Microchem 73. Kazemi-Darsanaki R, Azizzadeh A, Nourbakhsh
J. 2017;130:221–8. M, Raeisi G, AzizollahiAliabadi M. Biosensors:
62. Ghareeb M, Gohh RY, Akhlaghi F. Tacrolimus functions and applications. J Biol Today’s World.
concentration in saliva of kidney transplant recipi- 2013;2(1):53–61.
ents: factors influencing the relationship with 74. Lazcka O, Del Campo FJ, Muñoz FX. Pathogen detec-
whole blood concentrations. Clin Pharmacokinet. tion: a perspective of traditional methods and bio-
2018;57(9):1199–210. sensors. Biosens Bioelectron. 2007;22(7):1205–17.
63. Krogstad V, Vethe NT, Robertsen I, Hasvold G, 75. Choi YE, Kwak JW, Park JW. Nanotechnology for
Ose AD, Hermann M, et al. Determination of early cancer detection. Sensors (Basel, Switzerland).
tacrolimus concentration and protein expression of 2010;10(1):428–55.
7 Therapeutic Drug Monitoring (TDM) and Toxicological Studies in Alternative Biological Matrices 113

76. Bohunicky B, Mousa SA. Biosensors: the new using surface-enhanced Raman scattering of
wave in cancer diagnosis. Nanotechnol Sci Appl. 2-­mercaptobenzothiazole labeled gold@silver
2010;4:1–10. nanoparticles. Anal Chim Acta. 2014;817:33–41.
77. Dincer C, Bruch R, Costa-Rama E, Fernández-­ 90. Yamada K, Yokoyama M, Jeong H, Kido M, Ohno
Abedul MT, Merkoçi A, Manz A, et al. Disposable Y. Paper-based surfaced enhanced Raman spectros-
sensors in diagnostics, food, and environmental copy for drug level testing with tear fluid. Optical
monitoring. Adv Mater. 2019;31(30):e1806739. Society of America, Bellingham, USA; 2015.
78. Huang X, Liu Y, Yung B, Xiong Y, Chen 91. Maurer HH. Current role of liquid chromatography-­
X. Nanotechnology-enhanced no-wash biosen- mass spectrometry in clinical and forensic toxicol-
sors for in vitro diagnostics of cancer. ACS Nano. ogy. Anal Bioanal Chem. 2007;388(7):1315–25.
2017;11(6):5238–92. 92. Dincer C, Bruch R, Wirth S, Schumann S, Urban
79. Zhang Y, Chen X. Nanotechnology and GA. Biosensors and personalized drug therapy: what
nanomaterial-­based no-wash electrochemical bio- does the future hold?. Expert Rev Precis Med Drug
sensors: from design to application. Nanoscale. Dev. 2017 Nov 2;2(6):303–5.
2019;11(41):19105–18. 93. Tajik S, Taher MA, Beitollahi H, Torkzadeh-Mahani
80. Yockell-Lelièvre H, Bukar N, Toulouse JL, Pelletier M. Electrochemical determination of the anticancer
JN, Masson JF. Naked-eye nanobiosensor for thera- drug taxol at a ds-DNA modified pencil-graphite
peutic drug monitoring of methotrexate. Analyst. electrode and its application as a label-free electro-
2016;141(2):697–703. chemical biosensor. Talanta. 2015;134:60–4.
81. Losoya-Leal A, Estevez MC, Martínez-Chapa SO, 94. Bruch R, Chatelle C, Kling A, Rebmann B, Wirth
Lechuga LM. Design of a surface plasmon reso- S, Schumann S, et al. Clinical on-site monitoring of
nance immunoassay for therapeutic drug monitoring ß-lactam antibiotics for a personalized antibiother-
of amikacin. Talanta. 2015;141:253–8. apy. Sci Rep. 2017;7(1):3127.
82. Cappi G, Spiga FM, Moncada Y, Ferretti A, 95. Kling A, Chatelle C, Armbrecht L, Qelibari E,
Beyeler M, Bianchessi M, et al. Label-free detec- Kieninger J, Dincer C, et al. Multianalyte antibiotic
tion of tobramycin in serum by transmission-­ detection on an electrochemical microfluidic plat-
localized surface plasmon resonance. Anal Chem. form. Anal Chem. 2016;88(20):10036–43.
2015;87(10):5278–85. 96. Dauphin-Ducharme P, Yang K, Arroyo-Currás N,
83. Spiga FM, Maietta P, Guiducci C. More DNA-­ Ploense KL, Zhang Y, Gerson J, et al. Electrochemical
aptamers for small drugs: a capture-SELEX coupled aptamer-based sensors for improved therapeutic drug
with surface plasmon resonance and high-­throughput monitoring and high-precision, feedback-­controlled
sequencing. ACS Comb Sci. 2015;17(5):326–33. drug delivery. ACS Sens. 2019;4(10):2832–7.
84. Tenaglia E, Ferretti A, Decosterd LA, Werner D, 97. Li F, Yu Z, Han X, Lai RY. Electrochemical aptamer-­
Mercier T, Widmer N, et al. Comparison against based sensors for food and water analysis: a review.
current standards of a DNA aptamer for the label-­ Anal Chim Acta. 2019;1051:1–23.
free quantification of tobramycin in human sera 98. Sengiz C, Congur G, Eksin E, Erdem A. Multiwalled
employed for therapeutic drug monitoring. J Pharm carbon nanotubes-chitosan modified single-use bio-
Biomed Anal. 2018;159:341–7. sensors for electrochemical monitoring of drug-DNA
85. Lu J, Spasic D, Delport F, Van Stappen T, Detrez interactions. Electroanalysis. 2015;27(8):1855–63.
I, Daems D, et al. Immunoassay for detection of 99. Lin M-J, Chen Y-M, Li C-Z, Wu C-C. Electrochemical
infliximab in whole blood using a fiber-optic sur- sandwich immunoassay for quantification of thera-
face plasmon resonance biosensor. Anal Chem. peutic drugs based on the use of magnetic nanopar-
2017;89(6):3664–71. ticles and silica nanoparticles. J Electroanal Chem.
86. Beeg M, Nobili A, Orsini B, Rogai F, Gilardi D, 2019;849:113381.
Fiorino G, et al. A surface plasmon resonance-based 100. Fahem DK, El Houssini OM, Abd El-Rahman MK,
assay to measure serum concentrations of therapeu- Zaazaa HE. Screen printed potentiometric sensor for
tic antibodies and anti-drug antibodies. Sci Rep. therapeutic monitoring of rocuronium at the point of
2019;9(1):2064. care. Talanta. 2019;196:137–44.
87. Van Stappen T, Bollen L, Vande Casteele N, 101. Oyebola B. Study on biochips technology. J Ind
Papamichael K, Van Assche G, Ferrante M, et al. Technol. 2017;2(1):29–37.
Rapid test for infliximab drug concentration 102. Andrews LM, Li Y, De Winter BCM, Shi YY, Baan
allows immediate dose adaptation. Clin Transl CC, Van Gelder T, et al. Pharmacokinetic consid-
Gastroenterol. 2016;7(12):e206. erations related to therapeutic drug monitoring of
88. Hidi IJ, Mühlig A, Jahn M, Liebold F, Cialla D, tacrolimus in kidney transplant patients. Expert Opin
Weber K, et al. LOC-SERS: towards point-of-­ Drug Metab Toxicol. 2017;13(12):1225–36.
care diagnostic of methotrexate. Anal Methods. 103. Brandenburg A, Curdt F, Sulz G, Ebling F, Nestler
2014;6(12):3943–7. J, Wunderlich K, et al. Biochip readout system for
89. Zengin A, Tamer U, Caykara T. Extremely point-of-care applications. Sensors Actuators B
sensitive sandwich assay of kanamycin Chem. 2009;139(1):245–51.
114 B. Basu et al.

104. Morris RG, Russ GR, Cervelli MJ, Juneja R, 119. Chaturvedi AK, Vu NT, Ritter RM, Canfield
McDonald SP, Mathew TH. Comparison of trough, DV. DNA typing as a strategy for resolving issues
2-hour, and limited AUC blood sampling for moni- relevant to forensic toxicology. J Forensic Sci.
toring cyclosporin (Neoral) at day 7 post-renal 1999;44(1):189–92.
transplantation and incidence of rejection in the first 120. Bertaso A, Sorio D, Vandoros A, De Palo EF,
month. Ther Drug Monit. 2002;24(4):479–86. Bortolotti F, Tagliaro F. Use of finger-prick dried
105. Giannetti A, Adinolfi B, Berneschi S, Berrettoni C, blood spots (fpDBS) and capillary electrophore-
Chiavaioli F, Tombelli S, et al. Optical sensing in sis for carbohydrate deficient transferrin (CDT)
POCT: the contribution of the Institute of Applied screening in forensic toxicology. Electrophoresis.
Physics of the Italian CNR. LaboratoriumsMedizin. 2016;37(21):2867–74.
2017;41(5):251–6. 121. Dienes-Nagy A, Rivier L, Giroud C, Augsburger M,
106. Zurlo J, Rudacille D, Goldberg AM. Animals and Mangin P. Method for quantification of morphine
alternatives in testing: history, science, and ethics. and its 3- and 6-glucuronides, codeine, codeine gluc-
New York: Mary Ann Liebert; 1994. uronide and 6-monoacetylmorphine in human blood
107. Andersen ME, Thomas RS, Gaido KW, Conolly by liquid chromatography-electrospray mass spec-
RB. Dose-response modeling in reproductive toxi- trometry for routine analysis in forensic toxicology.
cology in the systems biology era. Reprod Toxicol. J Chromatogr A. 1999;854(1–2):109–18.
2005;19(3):327–37. 122. Fenton J, Schaffer M, Chen NW, Bermes EW Jr. A
108. Edwards SW, Preston RJ. Systems biology and comparison of enzyme immunoassay and gas chro-
mode of action based risk assessment. Toxicol Sci. matography/mass spectrometry in forensic toxicol-
2008;106(2):312–8. ogy. J Forensic Sci. 1980;25(2):314–9.
109. Waters MD, Olden K, Tennant RW. Toxicogenomic 123. Ishii A, Watanabe-Suzuki K, Seno H, Suzuki O,
approach for assessing toxicant-related disease. Katsumata Y. Application of gas chromatography-­
Mutat Res. 2003;544(2–3):415–24. surface ionization organic mass spectrometry to
110. Olelsiak MF. Toxicogenomics. In: Smart RC, forensic toxicology. J Chromatogr B Analyt Technol
Hodgson E, editors. Molecular and biochemical Biomed Life Sci. 2002;776(1):3–14.
toxicology. Hoboken: Wiley; 2008. p. 25–39. 124. Kauert G, Drasch G, von Meyer L. Possibilities of
111. Plant N. Can systems toxicology identify com- using chemical ionization mass spectrometry with
mon biomarkers of non-genotoxic carcinogenesis? ammonia as the selective reaction gas in forensic
Toxicology. 2008;254(3):164–9. toxicology. Beitr Gerichtl Med. 1979;37:329–35.
112. Hodgson E, Smart RC. Molecular and biochemical 125. Ojanperä I, Kolmonen M, Pelander A. Current
toxicology: definition and scope. In: Molecular and use of high-resolution mass spectrometry in drug
biochemical toxicology. New Jersey, United States: screening relevant to clinical and forensic toxi-
Wiley. 2008. p. 1–4. cology and doping control. Anal Bioanal Chem.
113. Stone EA, Nielsen DM. Bioinformatics. In: Smart 2012;403(5):1203–20.
RC, Hodgson E, editors. Molecular and biochemical 126. Van Bocxlaer JF, Clauwaert KM, Lambert WE,
toxicology. Hoboken: Wiley; 2008. p. 81–107. Deforce DL, Van den Eeckhout EG, De Leenheer
114. Waring JF, Ciurlionis R, Jolly RA, Heindel M, AP. Liquid chromatography-mass spectrom-
Ulrich RG. Microarray analysis of hepatotoxins etry in forensic toxicology. Mass Spectrom Rev.
in vitro reveals a correlation between gene expres- 2000;19(4):165–214.
sion profiles and mechanisms of toxicity. Toxicol 127. Kharbouche H, Sporkert F, Troxler S, Augsburger M,
Lett. 2001;120(1–3):359–68. Mangin P, Staub C. Development and validation of
115. Bynum ND, Moore KN, Grabenauer M. Evaluation a gas chromatography-negative chemical ionization
of laser diode thermal desorption-tandem mass spec- tandem mass spectrometry method for the determi-
trometry (LDTD-MS-MS) in forensic toxicology. J nation of ethyl glucuronide in hair and its applica-
Anal Toxicol. 2014;38(8):528–35. tion to forensic toxicology. J Chromatogr B Analyt
116. Bogusz MJ. Hyphenated liquid chromatographic Technol Biomed Life Sci. 2009;877(23):2337–43.
techniques in forensic toxicology. J Chromatogr B 128. Muhammad U, Abid N, Yawar B, Tahir J, Muhammad
Biomed Sci Appl. 1999;733(1–2):65–91. S, Shazia K. Forensic toxicological analysis of hair:
117. Canale M, Bistarini S, Merler M. Chromatography a review. Egypt J Forensic Sci. 2019;9(1):1–12.
by silica-gel chromatobars. Prospects of its applica- 129. Kuwayama K, Miyaguchi H, Kanamori T,
tion in the field of forensic toxicology. Arch Toxicol. Tsujikawa K, Yamamuro T, Segawa H, et al.
1977;37(2):143–7. Development of the “selective concentration” ana-
118. Carlier J, Guitton J, Romeuf L, Bévalot F, Boyer B, lytical method for drug-containing hair regions
Fanton L, et al. Screening approach by ultra-high based on micro-­segmental analysis to identify a
performance liquid chromatography-tandem mass trace amount of drug in hair: hair analysis follow-
spectrometry for the blood quantification of thirty-­ ing single-dose ingestion of midazolam. Forensic
four toxic principles of plant origin. Application to Toxicol. 2021;39(1):156–66.
forensic toxicology. J Chromatogr B Analyt Technol
Biomed Life Sci. 2015;975:65–76.
7 Therapeutic Drug Monitoring (TDM) and Toxicological Studies in Alternative Biological Matrices 115

130. Kintz P. Hair analysis in forensic toxicology: an 147. Farnaud SJ, Kosti O, Getting SJ, Renshaw D. Saliva:
updated review with a special focus on pitfalls. Curr physiology and diagnostic potential in health and
Pharm Des. 2017;23(36):5480–6. disease. Sci World J. 2010;10:434–56.
131. Gomez-Roig MD, Marchei E, Sabra S, Busardò 148. Dawes C. Salivary flow patterns and the health
FP, Mastrobattista L, Pichini S, et al. Maternal hair of hard and soft oral tissues. J Am Dent Assoc.
testing to disclose self-misreporting in drinking 2008;139 Suppl:18s–24s.
and smoking behavior during pregnancy. Alcohol 149. O’Neal CL, Crouch DJ, Rollins DE, Fatah A,
(Fayetteville, NY). 2018;67:1–6. Cheever ML. Correlation of saliva codeine con-
132. Marquet P. Progress of liquid chromatography-mass centrations with plasma concentrations after
spectrometry in clinical and forensic toxicology. oral codeine administration. J Anal Toxicol.
Ther Drug Monit. 2002;24(2):255–76. 1999;23(6):452–9.
133. Pufal E, Sykutera M, Piotrowski P. Development of a 150. Kidwell DA, Holland JC, Athanaselis S. Testing for
method for determining antidepressant drugs in nails drugs of abuse in saliva and sweat. J Chromatogr B
and its usefulness in forensic toxicology. Arch Med Biomed Sci Appl. 1998;713(1):111–35.
Sadowej Kryminol. 2008;58(4):167–70. 151. Frederick DL. Toxicology testing in alternative spec-
134. Bellagambi FG, Lomonaco T, Salvo P, Vivaldi F, imen matrices. Clin Lab Med. 2012;32(3):467–92.
Hangouët M, Ghimenti S, et al. Saliva sampling: 152. Manousi N, Samanidou V. Green sample prepara-
methods and devices. An overview. TrAC Trends tion of alternative biosamples in forensic toxicology.
Anal Chem. 2020;124:115781. Sustain Chem Pharm. 2021;20:100388.
135. Costa B, Santos Júnior WJR, Maximiano IF, Gomes 153. Gallardo E, Queiroz JA. The role of alternative speci-
NC, Freitas BT, De Martinis BS. Application of mens in toxicological analysis. Biomed Chromatogr.
microextraction techniques in alternative biological 2008;22(8):795–821.
matrices with focus on forensic toxicology: a review. 154. Moriya F. Pitfalls and cautions in analysis of drugs
Bioanalysis. 2021;13(1):45–64. and poisons. InDrugs and Poisons in Humans.
136. Jadoon S, Karim S, Akram MR, Kalsoom Khan A, 2005:17–24. Springer, Berlin, Heidelberg.
Zia MA, Siddiqi AR, et al. Recent developments in 155. Bévalot F, Cartiser N, Bottinelli C, Guitton J, Fanton
sweat analysis and its applications. Int J Anal Chem. L. State of the art in bile analysis in forensic toxicol-
2015;2015:164974. ogy. Forensic Sci Int. 2016;259:133–54.
137. De Giovanni N, Fucci N. The current status of sweat 156. Wille SM, Raes E, Lillsunde P, Gunnar T, Laloup
testing for drugs of abuse: a review. Curr Med Chem. M, Samyn N, et al. Relationship between oral fluid
2013;20(4):545–61. and blood concentrations of drugs of abuse in driv-
138. Jenkins AJ, Oblock J. Phencyclidine and can- ers suspected of driving under the influence of drugs.
nabinoids in vitreous humor. Legal Med. Ther Drug Monit. 2009;31(4):511–9.
2008;10(4):201–3. 157. Khajuria H, Nayak BP. Hair: a bio-marker for
139. Kugelberg FC, Jones AW. Interpreting results of eth- detection of drugs of abuse. Asian J Psychiatr.
anol analysis in postmortem specimens: a review of 2017;30:208–9.
the literature. Forensic Sci Int. 2007;165(1):10–29. 158. Hansen FA, Pedersen-Bjergaard S. Emerging extrac-
140. Pigaiani N, Bertaso A, De Palo EF, Bortolotti F, tion strategies in analytical chemistry. Anal Chem.
Tagliaro F. Vitreous humor endogenous compounds 2020;92(1):2–15.
analysis for post-mortem forensic investigation. 159. Pourshamsi T, Amri F, Abniki M. A comprehensive
Forensic Sci Int. 2020;310:110235. review on application of the syringe in liquid- and
141. Concheiro M, Huestis MA. Drug exposure during solid-phase microextraction methods. J Iran Chem
pregnancy: analytical methods and toxicological Soc. 2021;18(2):245–64.
findings. Bioanalysis. 2018;10(8):587–606. 160. Kataoka H. Recent developments and applications
142. Clark DA. Times of first void and first stool in 500 of microextraction techniques in drug analysis. Anal
newborns. Pediatrics. 1977;60(4):457–9. Bioanal Chem. 2010;396(1):339–64.
143. Bakdash A, Burger P, Goecke TW, Fasching PA, 161. Jain R, Singh R. Microextraction techniques for
Reulbach U, Bleich S, et al. Quantification of fatty analysis of cannabinoids. TrAC Trends Anal Chem.
acid ethyl esters (FAEE) and ethyl glucuronide 2016;80:156–66.
(EtG) in meconium from newborns for detection of 162. Lambropoulou DA, Albanis TA. Liquid-phase micro-­
alcohol abuse in a maternal health evaluation study. extraction techniques in pesticide residue analysis. J
Anal Bioanal Chem. 2010;396(7):2469–77. Biochem Biophys Methods. 2007;70(2):195–228.
144. Langman LJ. The use of oral fluid for therapeutic 163. Meng P, Wang Y. Small volume liquid extrac-
drug management: clinical and forensic toxicology. tion of amphetamines in saliva. Forensic Sci Int.
Ann N Y Acad Sci. 2007;1098:145–66. 2010;197(1–3):80–4.
145. Siegel IA. The role of saliva in drug monitoring. Ann 164. Kumar R, Gaurav, Heena, Malik AK, Kabir A,
N Y Acad Sci. 1993;694:86–90. Furton KG. Efficient analysis of selected estrogens
146. Aps JK, Martens LC. Review: the physiology of using fabric phase sorptive extraction and high
saliva and transfer of drugs into saliva. Forensic Sci performance liquid chromatography-fluorescence
Int. 2005;150(2–3):119–31. detection. J Chromatogr A. 2014;1359:16–25.
116 B. Basu et al.

165. Shah I, Petroczi A, Uvacsek M, Ránky M, Naughton drug reactions. Int J Clin Pharmacol Ther Toxicol.
DP. Hair-based rapid analyses for multiple drugs in 1986;24(10):559–68.
forensics and doping: application of dynamic 183. Sakaeda T, Tamon A, Kadoyama K, Okuno
multiple reaction monitoring with LC-MS/ Y. Data mining of the public version of the FDA
MS. Chem Cent J. 2014;8(1):73. adverse event reporting system. Int J Med Sci.
166. Amirav A. Fast heroin and cocaine analysis by GC– 2013;10(7):796–803.
MS with cold EI: the important role of flow pro- 184. Banda JM, Evans L, Vanguri RS, Tatonetti NP,
gramming. Chromatographia. 2017;80(2):295–300. Ryan PB, Shah NH. A curated and standardized
167. Partridge E, Trobbiani S, Stockham P, Scott T, adverse drug event resource to accelerate drug safety
Kostakis C. A validated method for the screen- research. Sci Data. 2016;3:160026.
ing of 320 forensically significant compounds in 185. Lorberbaum T, Nasir M, Keiser MJ, Vilar S,
blood by LC/QTOF, with simultaneous quanti- Hripcsak G, Tatonetti NP. Systems pharmacology
fication of selected compounds. J Anal Toxicol. augments drug safety surveillance. Clin Pharmacol
2018;42(4):220–31. Ther. 2015;97(2):151–8.
168. Rajaraman V. JohnMcCarthy — father of artificial 186. Raja K, Patrick M, Elder JT, Tsoi LC. Machine
intelligence. Resonance. 2014;19(3):198–207. learning workflow to enhance predictions of Adverse
169. Lavecchia A. Deep learning in drug discovery: Drug Reactions (ADRs) through drug-gene interac-
opportunities, challenges and future prospects. Drug tions: application to drugs for cutaneous diseases.
Discov Today. 2019;24(10):2017–32. Sci Rep. 2017;7(1):3690.
170. Keshavarzi Arshadi A, Webb J, Salem M, Cruz E, 187. Huang LC, Wu X, Chen JY. Predicting adverse
Calad-Thomson S, Ghadirian N, et al. Artificial side effects of drugs. BMC Genom. 2011;12 Suppl
intelligence for COVID-19 drug discovery and vac- 5(Suppl 5):S11.
cine development. Front Artif Intell. 2020;3:65. 188. Ma J, Sheridan RP, Liaw A, Dahl GE, Svetnik
171. Raies AB, Bajic VB. In silico toxicology: com- V. Deep neural nets as a method for quantitative
putational methods for the prediction of chemical structure-activity relationships. J Chem Inf Model.
toxicity. Wiley Interdiscip Rev Comput Mol Sci. 2015;55(2):263–74.
2016;6(2):147–72. 189. Xu Y, Dai Z, Chen F, Gao S, Pei J, Lai L. Deep learn-
172. Vinardell Martínez-Hidalgo MP. Alternatives to ani- ing for drug-induced liver injury. J Chem Inf Model.
mal experimentation in toxicology: current situation. 2015;55(10):2085–93.
Acta Bioeth. 2007;13:41–52. 190. Jimenez-Carretero D, Abrishami V, Fernández-­
173. Pappas AA, Massoll NA, Cannon DJ. Toxicology: de-­Manuel L, Palacios I, Quílez-Álvarez A, Díez-­
past, present, and future. Ann Clin Lab Sci. Sánchez A, et al. Tox_(R)CNN: deep learning-based
1999;29(4):253–62. nuclei profiling tool for drug toxicity screening.
174. Madea B. History of forensic medicine. In: PLoS Comput Biol. 2018;14(11):e1006238.
Handbook of forensic medicine. Hoboken: Wiley; 191. Dey S, Luo H, Fokoue A, Hu J, Zhang P. Predicting
2014. p. 1–14. adverse drug reactions through interpretable
175. Welling PG. Differences between pharmaco- deep learning framework. BMC Bioinform.
kinetics and toxicokinetics. Toxicol Pathol. 2018;19(Suppl 21):476.
1995;23(2):143–7. 192. Jamal S, Goyal S, Shanker A, Grover A. Predicting
176. Capture traces of tomorrow’s drugs today identify neurological adverse drug reactions based on bio-
rapidly evolving novel psychoactive drug substances logical, chemical and phenotypic properties of
faster. Ebook. Thermo Scientific; 2017. drugs using machine learning models. Sci Rep.
177. Huestis M. Advances, benefits and challenges of 2017;7(1):872.
oral fluid testing in forensic toxicology. Webinar. 193. Goicoechea M, Vidal A, Capparelli E, Rigby A,
Forensic Science Magazine. November 2017. Kemper C, Diamond C, et al. A computer-based
178. Marengoni A, Onder G. Guidelines, polypharmacy, system to aid in the interpretation of plasma con-
and drug-drug interactions in patients with multi- centrations of antiretrovirals for therapeutic drug
morbidity. BMJ. 2015;350:h1059. monitoring. Antivir Ther. 2007;12(1):55–62.
179. Tatonetti NP, Fernald GH, Altman RB. A novel sig- 194. Gallay J, Prod’hom S, Mercier T, Bardinet C,
nal detection algorithm for identifying hidden drug-­ Spaggiari D, Pothin E, et al. LC-MS/MS method for
drug interactions in adverse event reports. J Am Med the simultaneous analysis of seven antimalarials and
Inform Assoc. 2012;19(1):79–85. two active metabolites in dried blood spots for appli-
180. Tatonetti NP, Ye PP, Daneshjou R, Altman RB. Data-­ cations in field trials: analytical and clinical valida-
driven prediction of drug effects and interactions. Sci tion. J Pharm Biomed Anal. 2018;154:263–77.
Transl Med. 2012;4(125):125ra31. 195. Sanavio B, Krol S. On the slow diffusion of point-of-­
181. Naranjo CA, Busto U, Sellers EM, Sandor P, Ruiz I, care systems in therapeutic drug monitoring. Front
Roberts EA, et al. A method for estimating the prob- Bioeng Biotechnol. 2015;3:20.
ability of adverse drug reactions. Clin Pharmacol 196. Tucker GT. Personalized drug dosage – closing the
Ther. 1981;30(2):239–45. loop. Pharm Res. 2017;34(8):1539–43.
182. Venulet J, Ciucci AG, Berneker GC. Updating 197. Horwitz RI, Hayes-Conroy A, Caricchio R, Singer
of a method for causality assessment of adverse BH. From evidence based medicine to medicine
based evidence. Am J Med. 2017;130(11):1246–50.
Analyzing Data from Therapeutic
Drug Monitoring,
8
Pharmacokinetics, and Clinical
Toxicology Studies

Abdul Malik Sulley

Abstract 8.1 Introduction

Both individual- and population-based data Therapeutic drug monitoring (TDM) takes place
are useful for optimizing the efficacy of drugs during routine clinical care as well as in clinical
with narrow therapeutic indices, while at the research settings. The aim of data analysis in
same time, limiting the frequency and severity TDM is to answer research questions about the
of adverse effects. Pharmacokinetic, pharma- safety and/or efficacy/effectiveness of a drug
codynamic, descriptive, and inferential analy- with a narrow or not-well-established therapeutic
ses all provide valuable information by which index.
safe dosage regimens can be established for This chapter introduces key concepts and
patients at individual and population levels. issues surrounding planning, handling, and ana-
However, to draw reliable conclusions from lyzing data in TDM. Some sections are relevant
such analyses requires an understanding of the to routine care for individual patients only, clini-
best endpoints to assess and what types of data cal trials only, or both. The chapter is organized
to collect. Furthermore, data needs to be han- in such a way as to address the following con-
dled with such high standards that the integ- cepts and issues:
rity of conclusions drawn from it cannot be
disputed. This chapter describes key concepts • Endpoints – what are our measures of safety
and provides practical examples on planning, and efficacy/effectiveness?
handling, and analyzing data in therapeutic • Types of data – how do we tell what type(s) of
drug monitoring, pharmacokinetics and clini- data we have?
cal toxicology studies. • Data handling – how do we treat our data prior
to and during analysis?
• Pharmacokinetics – how does the drug “move”
Keywords
through the body and how do we monitor an
optimal plasma drug concentration?
Analysis · Endpoint · Data · Pharmacokinetics • Pharmacodynamics – how does the body
· Steady state respond to the drug and how do we know the
drug is working?
A. M. Sulley (*) • Sample size – how many participants we need
IQVIA RDS Inc., Kirkland, QC, Canada to test a hypothesis?
e-mail: abdul.sulley@iqvia.com

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 117
S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_8
118 A. M. Sulley

• Analysis methods – how do we summarize when it is run. Those that occur on the lines
our data and what statistical methods do we below or above this will be evaluated.
use in hypothesis tests?
#This is a comment

8.1.1 Software Setup Other packages used in examples that need to


be installed are car [4], PKNCA [5], and
Analysis examples are provided throughout the DescTools [6]. These can be installed using the
chapter. The examples are written in R code [1]. commands below.
R is a free statistical programming language and
environment. There are 18,000 packages that can > install.packages("car")
be installed to enable conduct of several types of > install.packages("PKNCA")
analyses and generate graphics. The examples in > install.packages("DescTools")
this chapter were run using version 4.1.3. Further
information about installation and packages can After installing packages, please make sure
be found through the R-Project homepage each package is loaded into the programming
(https://www.r-­project.org/). environment using the commands below.
R-Studio/Posit is a free coding environment
that makes coding with R much easier and effi- > library(car)
cient [2]. The examples in this chapter were run > library(PKNCA)
using R-Studio version 2022.02.0 Build 443 and > library(DescTools)
can be obtained from https://www.rstudio.com/.
Posit is the new name for R-Studio (https://www. To find out more information about any pack-
posit.co). age or command, type “?” followed by the pack-
The code examples in this chapter use three age or command name. For example, to learn
dummy datasets from the qwickr package [3], about the stats package which is automatically
namely: installed along with R, run the following line of
code:
• catdata – a dummy categorical dataset
• pkdata – a dummy pharmacokinetics dataset > ?stats
• rmdata – a dummy repeated measures dataset

Qwickr package can be installed using the 8.2 Endpoints


code:
An endpoint is a parameter, measure, or phenom-
> remotes::install_github("qwickmalik/ enon by which the safety, efficacy, or effective-
qwickr") ness of an intervention (or drug) is measured. At
the individual patient level, the decision of
For all examples, command lines begin with whether an intervention is safe, efficacious, or
>, while output lines do not. Commands that span effective is based on that patient’s data, compris-
multiple lines are identified by + after the first ing diagnostic and lab tests and clinical examina-
line. However, when running your code, do not tion. At the population level, it is usually based
include > or + at the beginning of your command on a test of hypothesis. The null hypothesis is
lines. Comments begin with #. Code that appears usually that there is no significant difference
after # on the same line will not be evaluated between two interventions (placebo could be
8 Analyzing Data from Therapeutic Drug Monitoring, Pharmacokinetics, and Clinical Toxicology Studies 119

considered as an intervention), with respect to can also be referred to as continuous data.


that endpoint. Categorical and continuous data consist of sub-
Efficacy can be described as the ability of an types as listed below.
intervention to produce an intended desired effect (a) Qualitative data
in a controlled experimental setting. Here, par- (a) Unstructured data (free text)
ticipants are carefully selected to limit the effects (b) Structured or categorical data
of external factors on the outcome of the inter- (i) Binary – only two categories or responses
vention. In effectiveness studies, an intervention often coded 0 and 1, e.g., coin (tail,
is administered in real-life settings where a drug head), mortality (dead, alive)
may be used in the presence of other drugs and/or (ii) Nominal – more than two categories, with
diseases. no specific order between them, e.g., blood
In TDM several types of endpoints can be type (A, B, AB, O) or eye color (amber,
assessed. Examples of these include the brown, gray, green, blue, etc.)
following: (iii) Ordinal – more than two categories with
(a) Pharmacokinetic profile (including plasma order, although the difference between
drug concentration) [7–9] adjacent categories cannot be quantified,
(b) Pharmacodynamic profile [8] e.g., education level (primary school,
(c) Incidence of an event, e.g., clinical failure, high school, bachelor’s degree, master’s
adverse event (skin rash), mortality [9, 10] degree, doctorate degree)
(d) Biomarker levels, e.g., c-reactive protein (b) Quantitative or continuous data (numeric)
(CRP), procalcitonin [10] (a) Discrete – whole numbers often repre-
(e) Time to event, e.g., time to infection site neg- senting counts, e.g., number of cancer
ative culture [10] patients in a ward.
(b) Interval – there are order and a meaning-
ful difference between two values with
8.3 Types of Data no meaningful zero (0), e.g., Celsius
temperature scale,
Data (singular: datum, although data is used as (c) Ratio – interval data type with a mean-
both singular and plural in some publications) ingful zero (0) i.e., zero means an
can be described as a collection of information absence of the phenomenon being mea-
that is usually organized to facilitate analysis and sured, e.g., hemoglobin levels, plasma
decision-making. The pieces of information can drug concentration, and Kelvin temper-
be classified as qualitative or quantitative. ature scale (0 Kelvin is absence of
Qualitative data often describe types or qualities heat).
of a subject or phenomenon. For example, Joe
has some money in his wallet, consisting of a $50
note, a $10 note, and a 25¢ coin. Here we are 8.4 Data Handling
describing the denominations of money.
Quantitative data, on the other hand, are numeri- Vast amounts of data are often collected during
cal measurements by which properties or attri- clinical trials or routine clinical care. Before
butes of a subject or phenomenon are quantified. analysis of clinical data can be done, data needs
For example, the total amount of money in Joe’s to be collated (grouped together usually in an
wallet is $60.25. Joe’s friends have $55, $80.15, analysis-ready dataset) and validated (checked
and $77.50 each. Here, we are measuring amount for quality). Quality checks are be conducted to
or quantities. confirm that the analysis dataset is not different
In clinical research, qualitative data can be of from the source, be it case report forms, paper
two main types: structured (categorical) or charts, lab reports, electronic medical records, or
unstructured (free text), while quantitative data other datasets.
120 A. M. Sulley

As much as possible, data editing rules should For such drugs, there is a small dose range of
be prespecified in study protocols, statistical effectiveness below which the drug is ineffective,
analysis plans (SAPs), or standard operating pro- and above which the drug can be toxic.
cedures (SOPs) and should be done in the source Furthermore, a fixed dose may not be effective or
document and/or the analysis dataset. For exam- safe in every patient; therefore, the ideal dose
ple, a plasma drug concentration measurement depends on the individual patient. Finding the
that is below the level of quantification (BLQ) appropriate dose not only helps to prevent serious
may be treated as missing, a zero (0), or half the adverse events (SAEs) but can also help to
lower limit of quantification (LLQ), and this improve compliance to treatment. For example, if
needs to be specified. When plasma concentra- a drug causes unbearable adverse drug reaction
tions are above the upper limit of quantification (ADR) or needs to be taken frequently, patients
(ULQ), samples need to be diluted and re-­ would be less likely to comply with treatment
analyzed to ensure that data is accurately and therefore not benefit from the drug. Therefore,
captured. using PK information, healthcare teams and
The Clinical Data Interchange Standards researchers can find ideal dosages for individual
Consortium (CDISC) has developed a series of patients or develop controlled-release or
standards for organizing, formatting, and sharing extended-release formulations to reduce the fre-
clinical trial data between and among sponsors, quency of administering the drug. Medical
contract research organizations (CROs), laborato- devices can also be developed to automatically
ries, and regulatory authorities. Of these standards, administer the drug as needed.
the Study Data Tabulation Model (SDTM) and Pharmacokinetic (PK) parameters are proper-
Analysis Data Model (ADaM) are most often used ties of a drug by which its absorption, distribu-
by biostatisticians. Further information is avail- tion, and elimination are assessed. Table 8.1 lists
able on the CDISC website (www.cdisc.org). and describes key parameters commonly used in
TDM, although other PK parameters may be
explored as well. In Table 8.1, although reference
8.5 Pharmacokinetics is made to concentrations of drug or metabolite in
the body, in practice, concentrations are mostly
Pharmacokinetics (PK) is often described sim- measured in plasma.
plistically as “what the body does to a drug.”
However, there is more to this. We are interested
in changes across time, in drug concentrations 8.5.1 Single-Dose PK Parameters
from the time it enters the body, moves through
the body as an intact molecule or a metabolite, After a single dose of a drug is administered, its
and ending with its removal from the body. By concentration typically rises to a peak in the body
examining PK properties of a drug, we can under- and then drops over time as it is eliminated. Non-­
stand how it is absorbed, distributed, metabo- compartmental analysis (NCA) assumes that the
lized, and eliminated from the body. Common whole body is one compartment, and this pro-
routes of administration in PK studies are intra- vides for simpler methods to calculate PK param-
venous (IV), intramuscular (IM), and oral. eters. All calculations in this chapter are based on
Depending on the route of administration, there this assumption.
may not be an absorption phase, e.g., IV Figure 8.1 illustrates how the plasma concen-
administration. tration changes over time after a single dose of an
In TDM, PK is used in routine clinical care to unknown drug is administered orally. The accom-
find drug doses that are optimal for individual panying data is provided in Table 8.2. The
patients. Here, the key emphasis is on finding an concentration-­ time curve and data are very
appropriate balance between safety and effective- important in understanding what PK parameters
ness of a drug with a narrow therapeutic range. are and in calculating them.
8 Analyzing Data from Therapeutic Drug Monitoring, Pharmacokinetics, and Clinical Toxicology Studies 121

Table 8.1 Pharmacokinetic parameters


PK parameter Description
Cmax Maximum (peak) observed concentration of drug in the body
tmax Time to reach maximum (peak) drug concentration after drug administration
AUC0–t or AUCt Area under the concentration-time curve from time zero to time t
AUClast Area under the concentration-time curve from time zero to the last quantifiable concentration
AUC0–∞ or AUC∞ Area under the concentration-time curve from time zero to infinity
λ Elimination rate constant
t1/2 Terminal half-life
CL Apparent total clearance of drug from the body
CL/F Apparent total clearance of drug from the body after non-intravenous administration
F Bioavailability – fraction or percentage of drug systemically available after administration
Vd Apparent volume of distribution after non-­intravenous drug administration
Vd/F Apparent volume of distribution after non-­intravenous drug administration
Clast Last observed quantifiable drug concentration
Css Steady-state plasma concentration of drug during constant rate infusion
Cav,ss Average steady-state plasma concentration of drug during multiple dose administration
Cmax,SS Maximum (peak) steady-state drug concentration within a dosage interval
Ctrough Trough plasma concentration, at the end of a dosing interval, immediately before next dose
administration
τ Dosing interval

Given that it is impossible and harmful to Example 8.1


patients to collect blood at every minute, it is
important to plan blood sampling times based on
#Cmax
existing information of the drug’s profile. This
> pk.calc.cmax(conc=patient1$CONC)
information can come from animal or human
>
studies, depending on the stage of drug
[1] 0.644
development.
>
#tmax
8.5.1.1 Cmax and tmax > pk.calc.tmax(conc=patient1$CONC,
The maximum or peak concentration and nomi- time=patient1$TIME)
nal time at which this occurs are the easiest >
parameters to estimate, as is evident from Fig. 8.1 [1] 1
and Table 8.2. Careful planning of blood s­ ampling
times is critical to obtaining the highest possible
blood concentration. 8.5.1.2 AUCt
When analyzing data involving several par- The area under the concentration-time curve
ticipants, it is more efficient to determine Cmax gives an indication of how much of a drug was in
and tmax without having to graph each partici- the body, after administration from time zero (0)
pant’s concentration-time curve. Example 8.1 to time t. It is measured units of concentration-­
illustrates how to do this using the PKNCA time, e.g., ng.h/mL. There are several ways of
package in R. Cmax is 0.644 ng/mL, while tmax calculating this, of which the most common is the
is 1 h. trapezoidal method or linear trapezoidal rule. By
122 A. M. Sulley

Fig. 8.1 (a) Concentration-time curve for an unknown drug. (b) Semilogarithmic curve for the same data

this rule, the concentration-time curve is divided areas summed to give AUCt. The trapezoidal rule
into a series of adjacent trapezoids. The area for is represented by Eq. 8.3, which is obtained by
each trapezoid is calculated and all the calculated combining Eqs. 8.1 and 8.2:
8 Analyzing Data from Therapeutic Drug Monitoring, Pharmacokinetics, and Clinical Toxicology Studies 123

Table 8.2 How to calculate AUC by hand using a spreadsheet


Sample Time (h) Concentration (ng/mL) Ci + Ci − 1 Ti−Ti − 1 AUC48
1 0 0 – – –
2 0.25 0.129 0.129 0.25 0.016
3 0.5 0.308 0.437 0.25 0.055
4 1 0.644 0.952 0.5 0.238
5 2 0.632 1.276 1 0.638
6 4 0.553 1.185 2 1.185
7 5 0.494 1.047 1 0.524
8 7 0.402 0.896 2 0.896
9 9 0.33 0.732 2 0.732
10 12 0.258 0.588 3 0.882
11 24 0.12 0.378 12 2.268
12 48 0.03 0.15 24 1.800
Total 9.233

#Step 2 calculate the AUC using trape-


C2  C1 
Area of atrapezoid  A   .  t2  t1  (8.1) zoid method
2 > AUC(x=patient1$TIME, y=patient1$CONC,
method="trapezoid")
>
AUCt  A1  A2  A3  At (8.2)
[1] 9.23325

T
Ci  Ci 1  A more accurate method for calculating AUC
AUCt   .  ti  ti 1  (8.3)
i 0 2 is the integration method, which can be done
using the PKNCA package as follows:
where:
A is the area of a single trapezoid. Example 8.3
C is the concentration at a given point in time.
ti is the ith time point.
T is the last sampling time point and is equal to t > pk.calc.auc.last(conc=patient1$CONC,
in AUCt. patient1$TIME)
The AUCt can be calculated by hand using a >
spreadsheet (Table 8.2) or in R using the PKNCA [1] 8.874462
(Example 8.2).
8.5.1.3 λ and t1/2
Example 8.2 The elimination rate constant (λ) is a value that
describes the rate at which a drug leaves the body.
Assuming the entire body is a single compart-
#Step 1 extract data for patient 1 for ment, deriving the elimination rate constant
treatment A assumes that there is a direct proportional rela-
#This data will be used for many of the tionship between the rate of drug elimination and
examples that follow drug concentration. Given that the elimination
> patient1 <- subset(pkdata, phase of the curve (terminal/descending arm) is
+ pkdata$SUBJECTNUM == 1 & exponential in nature (Fig. 8.1a), a natural log
+ pkdata$GROUPING == "Group A") transformation of plasma drug concentrations is
>
necessary to illustrate this linear relationship,
124 A. M. Sulley

which appears as a straight line through points on pling is often stopped at a prespecified time point
the descending arm. The terminal slope of the t and AUCt reflects how much of the drug was in
resulting semilogarithmic graph (Fig. 8.1b) is the the body up till that time point. In this case, the
elimination rate constant. last observed plasma concentration is usually
The terminal elimination phase must not be quantifiable or nonzero. To get an estimate of
influenced by absorption; therefore, a simple way how much drug would have been in the body if
to identify it is that there will be a straight line sampling was not stopped, AUC∞ is calculated
from its starting point to the last measured con- by extrapolating the concentration-time curve to
centration [11]. This phase must have at least infinity. The ratio AUCt/AUC∞ gives an indica-
three (3) points to estimate the elimination rate tion of the extent to which this extrapolation
constant, although more points will increase the went. AUC∞ is calculated using the formula
reliability of estimates. below (12), where Clast is the last measured
Half-life (t1/2) is the amount of time it takes for concentration:
half of a drug to be eliminated from the body. It is
Clast
related to the elimination rate constant (λ) accord- AUC  AUCt  (8.5)
ing to the formula below: 
In Example 8.5, AUC∞ is calculated in R using
ln 2
t1/ 2  (8.4) λ calculated in the previous example. The same
 result will be obtained by hand using Eq. 8.5.
Both can be calculated in one step using the
PKNCA package in R using our patient1 data. Example 8.5
The λ is 0.059, while t1/2 is 11.65 h.

Example 8.4 > pk.calc.auc.inf(conc=patient1$CONC,


patient1$TIME, lambda.z = 0.05948419)
>
> pk.calc.half.life( [1] 9.378798
+ conc=patient1$CONC,
+ time=patient1$TIME, 8.5.1.5 CL/F and Vd/F
+ tmax=pk.calc.tmax(conc=patient1$CONC, Clearance (CL) is the rate at which a drug is
time=patient1$TIME), removed from a reference fluid in the body, which
+ tlast=48) in most cases is plasma or blood. Its unit of mea-
> surement is volume of reference fluid cleared of
lambda.z r.squared adj.r.squared
drug per unit time, e.g., mL/min.
lambda.z.time.first
Volume of distribution (Vd) is the fluid volume
0.05948419 0.9993719 0.9987438 12
in which a drug circulates in the body.
lambda.z.n.points clast.pred half.life
Clearance is related to the elimination rate
span.ratio
constant and volume of distribution according to
3 0.02969165 11.65263 3.089431
the equation below:
CL
8.5.1.4 AUC∞  (8.6)
For drugs that are eliminated relatively quickly, Vd
it is possible to measure drug concentrations
until the drug is eliminated from the body com- Clearance is independent of Vd; therefore, if
pletely; therefore, the AUCt provides a picture of clearance increases or decreases, Vd will not
the entire exposure. However, for many drugs, change, although the elimination rate constant
elimination is quite slow and nonlinear, and it will. Based on this, and comparing Eqs. 8.5 and
may be impractical to continue sampling blood 8.6, it is evident that CL and Vd directly deter-
over long periods of time. Therefore, blood sam- mine half-life (t1/2). In other words, a drug that is
8 Analyzing Data from Therapeutic Drug Monitoring, Pharmacokinetics, and Clinical Toxicology Studies 125

cleared fast will have a shorter half-life compared over a period. One of the reasons for this is to
to a drug that is cleared slowly. ensure that organs or pathogens are exposed to an
Clearance is also related to AUC according to optimal concentration of the drug over the period,
Eq. 8.7, where Xo is the drug dose and F is bio- to provide the desired effect. In TDM, an added
availability (fraction of drug that is systemically concern is that plasma concentrations (Cp) should
available). If the drug is administered intrave- not exceed minimum toxic levels (Cmin,tox).
nously, then all of it will be systemically avail- Steady-state is a situation where the rate at
able so F = 1. which a drug enters the body is equal to the rate
at which it is eliminated. The amount of time it
X o .F
CL = (8.7) takes to achieve steady state is determined by the
AUC elimination half-life of the drug. Approximately
With Eqs. 8.6 and 8.7, it is possible to deter- 50% steady state is achieved after one half-life. If
mine Vd as follows: a dose is administered every one half-life, then
75% steady state is achieved after two half-lives,
X o .F
Vd  (8.8) 85.5% after three half-lives, and about 97% after
AUC. five half-lives. Therefore, as a rule of thumb,
After obtaining Vd, this can be plugged into steady state is assumed to be achieved after five
Eq. 8.6 to obtain CL. half-lives. Figure 8.2 shows concentration-time
For non-intravenously administered drugs curve for a drug at steady state after multiple oral
where F is not known, apparent clearance (CL/F) doses.
and apparent volume of distribution (Vd/F) can be
presented. 8.5.2.1 Cav,SS and CSS
Assuming a dose of 10 ng was administered, At steady state achieved through multiple dose
CL/F and Vd/F can be obtained using the lines of administration (e.g., oral, intravenous bolus, or
code in Example 8.6. intravenous infusion), plasma concentrations
rise to a peak (Cmax,SS) after drug administration
Example 8.6 at the beginning of each dosing interval (τ) and
drop to a trough level (Ctrough) immediately
before the next dose is administered. Therefore,
> # Cl/F a more useful measure of drug exposure is the
> pk.calc.cl(10, pk.calc.auc. average plasma concentration (Cav,SS). One rule
last(conc=patient1$CONC, to note is that the AUC for each dosing interval
patient1$TIME)) at steady state (AUCτ) is equal to AUC∞. Since
> AUCτ is the total amount of drug in plasma
[1] 1.126829 during a dosing interval, we can find the aver-
> age concentration per unit time according to
> # Vd/F
Eq. 8.10:
> pk.calc.vd(dose=10, aucinf = 9.378798,
lambda.z = 0.05948419) AUC
Cav,SS  (8.10)
> 
[1] 17.92467

Example 8.7 In this example, auclast is the


AUC∞ calculated from the single acute dose PK
8.5.2 Steady-State PK Parameters profile. Start and end indicate when a steady-state
dosing interval starts and ends:
The PK parameters discussed so far are based on
a single administered dose of a drug. However,
most drugs are administered at regular intervals > # Cav,ss
126 A. M. Sulley

Fig. 8.2 Steady-state concentration-time graph for multiple oral doses of an unknown drug

> pk.calc.cav(auclast = 9.378798, start > # Ctrough


= 23.2, end =34.8) > pk.calc.ctrough(conc=pkssdata$CONC,
> time = pkssdata$TIME, end = 92.8)
[1] 0.8085171 >
[1] 0.65

During constant rate infusion, the infusion


rate (Ro) can be set based on the rate of clearance With the PK parameters described above, it is
(CL), so at steady state, plasma concentration at possible to calculate other measures such as con-
any point in time would be constant (CSS). centration to dose ratio (Ctrough/Dose) and metab-
Equation 8.11 illustrates this relationship. olite to parent compound ratio [13].
Ro
CSS = (8.11)
CL 8.6 Pharmacodynamics

8.5.2.2 Ctrough and Cmax,SS Broadly, pharmacodynamics (PD) refers to the


The trough plasma concentration (Ctrough) is the effects of a drug on the body. Specifically, it is the
last concentration measured at steady state, effect of drug concentration at the site of action.
immediately before the next dose administration. Since is it not always possible to determine drug
As its name implies, Cmax,SS is the maximum concentration at the site of action, plasma con-
plasma concentration at steady state. centrations are usually used as a proxy. Where
endpoints are blood biomarkers, they can be
Example 8.8 In this example, conc and time measured from the same blood samples used to
are the variables for plasma concentrations and measure drug concentrations for PK analysis.
time, respectively. End is the time for a steady- Here, it is quite easy to correlate effects with drug
state dosing interval. In this example 92.8 h is concentrations. On the other hand, if pharmaco-
chosen. logical effects are assessed using other means,
8 Analyzing Data from Therapeutic Drug Monitoring, Pharmacokinetics, and Clinical Toxicology Studies 127

e.g., cognitive tests, it is very important that these based on data from a sample taken from the pop-
assessments are timed to coincide with blood ulation. Inferential statistics usually involves
draws for PK analysis. hypothesis testing and confidence intervals.

8.6.1 PK/PD 8.7.1 Statistical Hypothesis Testing

PK/PD analysis involves exploring relationships Hypothesis testing is a means of assessing


between drug concentrations or PK parameters whether results from one group are different from
and PD measurements, or calculating indices another group or a prespecified value. It involves
based on PK and PD parameters [14]. For exam- stating a null hypothesis (Ho) and an alternative
ple, in TDM of antibiotics, the minimal inhibi- hypothesis (Ha). The Ho is generally considered
tory concentration (MIC) is a PD parameter the status quo, while Ha is the result we suspect
representing the minimum amount of drug expo- and want to confirm or conclude. The Ha is usu-
sure that prevents visible microbial growth. PK/ ally the opposite of Ho. For example, if Ho is that
PD indices such as Cmax/MIC and AUC/MIC give the groups or values being compared are not dif-
an idea of how effective a dose might be [15]. ferent from each other, Ha will be that the groups
or values being compared are different from each
other. Similarly, if Ho is that X is smaller than or
8.7 Descriptive and Inferential equal to Y, Ha will be that X is greater than Y.
Statistics There are two types of errors to be aware of in
hypothesis testing – Type I and Type II errors.
Broadly, two types of analyses can be performed
on group-level data or data from multiple study 8.7.1.1 Type I Error
participants: descriptive (summary statistics) and Type I error or alpha (α) is the probability of hav-
inferential. Often, both are presented for group-­ ing a false-positive result in a hypothesis test.
level data. Type I error is committed when the null hypoth-
Descriptive analyses provide an overview of esis is rejected although it is in fact true. Type I
what our data looks like, i.e., describing the data. error must be kept low to increase reliability of
For categorical data in clinical research frequen- results. It is usually set at 5% or 0.05. The smaller
cies and percentages for each category are usu- the alpha, the larger the sample size.
ally. For continuous data, summary statistics
include arithmetic mean, geometric mean, 8.7.1.2 Type II Error
median, minimum-maximum range, standard Type II error or beta (β) is the probability of not
deviation (SD), and inter-quartile range (IQR). rejecting the null hypothesis when the null
Descriptive statistics also includes graphical pre- hypothesis is false (false-negative).
sentation of the summaries mentioned above, for Power is the probability of rejecting Ho cor-
both categorical and continuous data. Sometimes, rectly and is calculated as 1 − β. Type II error
categorical data are stored as numbers in a data- must be kept low to increase reliability of results.
set for simplicity and ease of analysis; however, It is usually set at 10% or 20%, i.e., power is usu-
this does not make them continuous data. For ally 90% or 80%, respectively. The smaller the β,
example, mortality variable may be stored as 1 the larger the power and the larger the sample
for “alive” and 0 for “dead” in a database of 100 size.
patients. However, a mean of 0.73 will be mean-
ingless for this variable. Instead, 73% alive and 8.7.1.3 Significance Testing
27% dead are more meaningful. and p-Value
Inferential statistics provide a means to make Tests of significance are statistical methods used
inferences or predictions about the population, to assess the strength of evidence against the null
128 A. M. Sulley

hypothesis. It enables us to determine if Ho 8.8 Sample Size and Power


should be rejected in favor of Ha or not.
Tests of significance generally produce a Sample size calculation enables us to determine
p-value, which is the probability that results as how many participants need to be studied to reli-
extreme as those from a sample could be obtained ably answer the research question. The most
when the null hypothesis is true. important endpoint is used to formulate the pri-
The p-value is usually compared to a prespeci- mary objective of the study, while the others can
fied significance level or alpha (α). If the p-value be grouped into secondary and/or exploratory
is smaller than or equal to α, Ho is rejected, in objectives. Sample size calculations are always
favor of Ha. If the p-value is larger than α, there is based on the primary objective.
not enough evidence to reject Ho, and therefore To calculate sample size, significance level
we fail to reject it. (alpha) and power need to be set.
The p-value is influenced by sample size; The sample size for any clinical study is based
therefore, for a given endpoint, if the sample size on the type of data collected for the primary end-
is large enough, it is possible to reject the null point. For example, if the data is continuous and
hypothesis. Therefore, rather than “accepting” the null hypothesis involves a comparison of
the null hypothesis, it is better to say we “fail to means, the sample size calculation will be based
reject the null hypothesis” or “the null hypothesis on the t-test. Using the power.t.test command in
is supported.” R, the sample size for a hypothetical study is 34
The p-value is not an indicator of the magni- per arm as calculated below:
tude of effect, but rather the strength of
evidence. Example 8.9

8.7.2 Confidence Interval (CI) > Sample size calculation for differ-
ence in means
A confidence interval is a range within which we > p_sd = q.pooled_sd(sds=c(2.41, 2.21),
are confident that the population measure falls. A + ns=c(20, 20))
level of confidence needs to be set, and this is > power.t.test(n = NULL,
usually 1 − α expressed as a percent. Therefore, + delta = 1.6,
if α is 5%, our level of confidence will be + sd = p_sd,
+ sig.level = 0.05,
(1 − 0.05) × 100 = 95%. This CI will be called a
+ power = 0.8,
95% CI.
+ type = “two.sample”,
The CI gives us an idea of how close results
+ alternative = “two.sided”)
from a sample from the population are to the pop-
>
ulation. Therefore, CIs are a means of assessing
>
if results from samples are generalizable to the Two-sample t test power calculation
population. n = 33.76509
Methods for calculating CIs depend on the delta = 1.6
type of inferential statistics being conducted and sd = 2.312
are beyond the scope of this chapter. However, in sig.level = 0.05
analysis examples provided later in the chapter, power = 0.8
they include how to interpret CIs. alternative = two.sided
NOTE: n is number in *each* group
8 Analyzing Data from Therapeutic Drug Monitoring, Pharmacokinetics, and Clinical Toxicology Studies 129

where: p2 = 0.3
q.pooled_sd is a command from the qwickr pack- sig.level = 0.05
power = 0.8
age that calculates pooled standard deviations
alternative = two.sided
(SD).
NOTE: n is number in *each* group
sds contains the SDs of 2.41 and 2.21 for the two
study groups (for calculating pooled SD
p_sd).
ns contains the number of participants in each 8.9 Analysis Methods
group 20 (for calculating pooled SD).
power.t.test is a command from the stats package Descriptive and inferential statistics can be
that calculates sample size for comparison of applied to pharmacokinetic and pharmacody-
means. namic parameters, as well as the other endpoint
delta is the difference in mean (0.6). types listed at the beginning of this chapter. In the
sd is the pooled SD (p_sd). remaining sections of this chapter, a selection of
n is the number of participants per group (equal analysis methods is presented, with simple exam-
to NULL because we want the power.t.test ples using R.
command to calculate it for us).
sig.level is the significance level or Type I error
probability. 8.9.1 Categorical Endpoints/
power is the power of test (1 − β). Comparison of Proportions
type is the type of t test, here “two.sample” for
comparison of two independent groups. 8.9.1.1 Chi-Square Test
alternative is the alternative hypothesis, here The chi-square test is used to assess the relation-
“two.sided” for a two-sided comparison. ship between two categorical variables. It tests
Sample size calculations for comparison of the null hypothesis that there is no statistically
means can also be based on other statistical tests significant relationship between the two categori-
such as ANOVA or repeated measures. However, cal variables. The main assumption is that for
these are more advanced and will not be the focus each combination of exposure (or treatment) and
of this chapter. outcome variables, there are at least five observa-
For a categorical endpoint where the expected tions. This can be checked using the table com-
proportion of events (e.g., bleeding adverse mand. The frequencies summarized by the
event) in group 1 is 50% and group 2 is 30%, the command can be supplied to the chisq.test com-
sample size required to detect such a difference mand to run the test.
in proportions at 80% power and 5% significance
level is 93 per arm as calculated below: Example 8.11 In the example below, we want to
find out if there is a significant relationship
Example 8.10 between Group and Sex.

With a p-value of 0.201, we fail to reject the


> Sample size calculation for differ- null hypothesis. Therefore, we conclude that
ence in proportions based on our data, there is no significant relation-
> power.prop.test(n = NULL, p1 = 0.5, ship between study Group and Sex. This is
p2 = 0.3, power = 0.8, sig.level = 0.05) expected in a randomized trial if randomization
> was done effectively.
Two-sample comparison of proportions
power calculation > # Chi-square test
n = 92.99884 > ## Cross-tabulate the data
p1 = 0.5
130 A. M. Sulley

> tab <- table(catdata$GROUPING, >


catdata$SEX) Fisher’s Exact Test for Count Data
> print(tab)
Female Male data: pain
A 11 9 p-value = 0.2351
B 6 14 alternative hypothesis: true odds ratio
> # Run Chi-square test is not equal to 1
> chisq.test(tab) 95 percent confidence interval:
0.02308176 1.80076397
Pearson’s Chi-squared test with Yates’ sample estimates:
continuity correction odds ratio
0.2680041

data: tab
X-squared = 1.6368, df = 1, p-value = 8.9.1.3 McNemar Test
0.2008 The McNemar test is used to test the consistency
of an outcome when assessed at two different
time points (e.g., before and after an interven-
8.9.1.2 Fisher’s Exact Test tion) or assessed using two different methods
When the main assumption in the chi-square test (e.g., COVID-19 using polymerase chain reac-
fails to hold, i.e., there is at least one combination tion (PCR) versus a rapid diagnostic test). The
of exposure and outcome variables with less than key requirement is that the same individual is
five observations, Fisher’s exact test is used. assessed twice; therefore, the observations are
paired. As a result, McNemar test is sometimes
Example 8.12 In this example, we test the null referred to as the paired chi-square test. Another
hypothesis that there is no association between requirement is that the outcomes are both binary.
treatment (grouping) and improvement in pain. A
cross-tabulation of the two variables shows that Example 8.13 In this example, we use the
one cell has two observations; therefore, chi-­ repeated measures dataset rmdata from the
square test would not be appropriate. Instead, we qwickr package. We want to find out if disease
conduct Fisher’s exact test. state at visit 1 (before treatment) is the same as
that at visit 4 (after treatment) for participants in
With a p-value of 0.235, we fail to reject the group A.
null hypothesis. Therefore, there is no associa-
tion between treatment and improvement in pain. With a p-value of 0.003, we reject a null
hypothesis that disease states before and after
> # Fisher’s Exact test treatment are the same.
> ## Cross-tabulate the data
> pain <- table(catdata$GROUPING, > # McNemar Test
catdata$PAIN_IMPROVED) > ## Create a subset of rmdata contain-
> print(pain) ing visits 1 and 4 for participants in
> group A
0 1 > mndata <- rmdata[rmdata$VISITNUMBER
A 2 18 %in% c(1,4) & rmdata$GROUPING == “A”,]
B 6 14 >
> > ## Create separate vectors for
> DISEASE_STATE variable at visits 1 and
> ## Run Fisher’s Exact test 4
> fisher.test(pain)
8 Analyzing Data from Therapeutic Drug Monitoring, Pharmacokinetics, and Clinical Toxicology Studies 131

> before <- mndata$DISEASE_ regression in R is done using a generalized linear


STATE[mndata$VISITNUMBER == 1] model with a binomial distribution for the error
> after <- mndata$DISEASE_ term.
STATE[mndata$VISITNUMBER == 4]
>
From the output below, the odds of having
> ## Create cross-tabulation of before
improvement in pain in group B was 1.5 times
and after data
lower compared to group A. However, with a
> crosstab <- table(before, after)
p-value of 0.104, we fail to reject the null hypoth-
> print(crosstab)
after
esis. Similarly, for each unit increase in age, the
before 0 1 odds of having pain improvement reduces by
1 0 0.05. However, with a p-value of 0.379, we fail to
1 11 3 reject the null hypothesis regarding age. This is
> confirmed by the 95% confidence intervals (CIs)
> ## Run the McNemar test for study group and age. Both CIs include an
> mcnemar.test(crosstab) odds ratio of 1, suggesting that we are 95% con-
fident that the true population odds of pain
McNemar’s Chi-squared test with conti- improvement could increase or decrease or
nuity correction remain the same with change in study group and
age. In summary, the results from our sample are
not strong enough to reject the null hypothesis.
data: crosstab
McNemar’s chi-squared = 9.0909, df = 1, > # Multiple logistic regression
p-value = 0.002569 > multiplelogistic <- glm(PAIN_IMPROVED
~ GROUPING + AGE,
8.9.1.4 Logistic Regression + data = catdata,
Simple logistic regression is used to assess the + family = “binomial”)
probability of occurrence of an outcome. The >
outcome/dependent variable must be binary, and > ## Display the regression output
> summary(multiplelogistic)
the independent variable can be categorical or
>
continuous. The null hypothesis is that the prob-
Call:
ability of a particular value of the binary outcome
glm(formula = PAIN_IMPROVED ~ GROUPING
variable is not associated with the value of the
+ AGE, family = “binomial”,
independent variable.
data = catdata)
Multiple logistic regression is an extension of
simple logistic regression where there are two or Deviance Residuals:
more independent variables in the model. Ordinal Min 1Q Median 3Q Max
logistic regression is an extension of simple -2.3075 0.3541 0.4578 0.7542 1.0194
logistic regression where the outcome/dependent
variable has more than two levels and is ordered Coefficients:
(ordinal). Estimate Std. Error z value Pr(>|z|)
(Intercept) 5.00185 3.35975 1.489 0.137
Example 8.14 In this multiple logistic regres- GROUPINGB -1.52362 0.93767 -1.625 0.104
sion example, we test the null hypothesis that the AGE -0.04551 0.05173 -0.880 0.379
probability of having pain improvement is asso-
ciated with study treatment (grouping) and age. (Dispersion parameter for binomial fam-
We use catdata from qwickr package. Logistic ily taken to be 1)
132 A. M. Sulley

Null deviance: 40.032 on 39 degrees of >


freedom > ## Shapiro-Wilk test
Residual deviance: 36.673 on 37 degrees > Shapiro.test(mndata$BIOMARKER)
of freedom
AIC: 42.673 Shapiro-Wilk normality test

Number of Fisher Scoring iterations: 5


> ## Generate 95% CI data: mndata$BIOMARKER
> confint(multiplelogistic) W = 0.96155, p-value = 0.7193
Waiting for profiling to be done...
2.5 % 97.5 % >
(Intercept) -1.3888665 12.18110393 > ## t-test
GROUPINGB -3.6594346 0.17225183 > t.test(BIOMARKER ~ 1, mu = 56, data =
AGE -0.1503426 0.05888971 mndata)

One Sample t-test

8.9.2 Continuous Endpoints/


Comparison of Means data: BIOMARKER
t = 12.375, df = 14, p-value =
8.9.2.1 One-Sample T-Test 6.295e-09
The one-sample t-test is used to test the null alternative hypothesis: true mean is
hypothesis that the mean for a normally distrib- not equal to 56
uted variable (interval or ratio) is not statistically 95 percent confidence interval:
significantly different from a prespecified value. 75.62886 83.85931
One key assumption is that the data must be nor- sample estimates:
mally distributed. This can be checked using a mean of x
79.74408
histogram and/or the Shapiro-Wilk test [16]. The
null hypothesis for the Shapiro-Wilk test is that
the data is normally distributed. 8.9.2.2 One-Sample Wilcoxon Test
When the assumption of normality fails to hold,
Example 8.15 In the example below, the continuous data cannot be analyzed using the
Shapiro-Wilk test gives a p-value of 0.719; there- parametric one-sample t-test. Instead, the non-
fore, we fail to reject the null hypothesis that the parametric one-sample median or Wilcoxon or
data is normally distributed. With this, we pro- Mann-Whitney test is used. The null hypothesis
ceed to test the null hypothesis that the mean bio- is that the sample median is equal to a specified
marker level is 57 units. From the output of the number.
one-sample t-test, the mean is 79.7 units, 95%
confidence interval (CI) is 75.6 units to 83.9 units, Example 8.16 In the example below, the
and p-value is <0.001. Therefore, we reject the Shapiro-Wilk test gives a p-value of 0.026; there-
null hypothesis. We conclude that the mean age fore, we reject the null hypothesis that the data is
of our sample is not equal to 60 units. normally distributed. With this, we proceed to
use the one-sample Wilcoxon test for the null
hypothesis that the median age is equal to
> # One-sample t-test 64 years. With a p-value of <0.001, we reject this
> ## Create a subset of rmdata contain- null hypothesis and conclude that the median age
ing visit 1 for participants in group A is not equal to 64 years.
> mndata <- rmdata[rmdata$VISITNUMBER
%in% c(1) & rmdata$GROUPING == “A”,]
8 Analyzing Data from Therapeutic Drug Monitoring, Pharmacokinetics, and Clinical Toxicology Studies 133

> # One-sample Wilcoxon Test Homogeneity of variances can be checked


> ## Shapiro-Wilk test using Levene’s test, whose null hypothesis is that
> Shapiro.test(catdata$AGE)
the variances are equal [18]. Therefore, a statisti-
cally significant test means that the variance of
Shapiro-Wilk normality test
one group is different from the other. Welch’s
t-test does not assume homogeneity of variances,
and this is conducted by default using the t.test
data: catdata$AGE
command in R as illustrated below.
W = 0.93607, p-value = 0.02554

> ## Wilcoxon test Example 8.17 In this example, we create a sub-


> wilcox.test(AGE ~ 1, mu = 64, data = set of rmdata at visit 4 for participants in groups
catdata) A and B. We want to find out if their mean bio-
marker levels are significantly different. As con-
Wilcoxon signed rank test with conti- firmed by the Shapiro-Wilk test, the data are
nuity correction normally distributed; therefore, we proceed with
a two-sample t-test.

data: AGE Although the means of groups A and B are


V = 146.5, p-value = 0.0004034 75.65 units and 78.19 units, respectively, the null
alternative hypothesis: true location hypothesis is supported with a p-value of 0.366.
is not equal to 64 This is confirmed by the 95% CI. We are 95%
confident that the true population difference in
8.9.2.3 Independent T-Test means lies between −8.22 units and 3.14 units,
The independent (or two sample) t-test is used to which includes 0 units.
compare (interval or ratio) data between two
independent groups or samples of participants or > # Independent t-test
observations. The null hypothesis is that there is > ## Create a subset of rmdata contain-
no statistically significant difference in means ing visits 4 for participants in groups
between the two independent groups. In other A and B
words, the difference in means of the two groups > itdata <- rmdata[rmdata$VISITNUMBER
is equal to zero (0). == 4,]
This test is based on four assumptions as fol- > ## Test for normality
> Shapiro.test(itdata$BIOMARKER)
lows: (1) data are normally distributed; (2) vari-
ances (or standard deviations) of the two groups
Shapiro-Wilk normality test
are “equal” or homogeneous; (3) data for the two
groups are independent, i.e., no participant
belongs to both groups; and (4) the two groups
data: itdata$BIOMARKER
are randomly sampled. W = 0.96112, p-value = 0.3308
The independent t-test is robust to violations
of normality, especially when the groups have >
equal sizes and variances are homogeneous [17]. > ## Run the t-test
Furthermore, according to the central theorem, as > t.test(BIOMARKER ~ GROUPING, data =
sample size increases, the distribution of data itdata)
approaches normality. By convention, when
n ≥ 30, normality can be assumed. However, for Welch Two Sample t-test
the purposes of learning, most of the examples in
this chapter involve n < 30 so the test for normal-
ity will be done where necessary. data: BIOMARKER by GROUPING
134 A. M. Sulley

t = -0.92031, df = 25.208, p-value = W = 223.5, p-value = 0.5331


0.3661 alternative hypothesis: true location
alternative hypothesis: true difference shift is not equal to 0
in means between group A and group B is
not equal to 0
8.9.2.5 Paired T-Test
95 percent confidence interval:
The paired t-test is used when the two samples
-8.218544 3.140587
being compared are not independent. This often
sample estimates:
arises when repeated measurements are done for
mean in group A mean in group B
75.65129 78.19027
the same participant, e.g., blood pressure mea-
sured before treatment and after treatment. Here,
participants serve as their own controls. The
8.9.2.4 Wilcoxon Rank Sum (Mann-­ paired t-test assumes that the differences between
Whitney U) Test paired observations are interval and normally
When data from two independent samples are not distributed.
normally distributed, the Wilcoxon Rank Sum
test is a nonparametric alternative for analyzing Example 8.19 In this example, since the bio-
the data. As such the data is ranked (ordinal) and marker is normally distributed (Shapiro-Wilk
the null hypothesis is that the medians of the two p-value = 0.223), we proceed with the paired
samples are equal. t-test. With a p-value of 0.112, the null hypothe-
sis that the mean difference in biomarker levels
Example 8.18 In this example, since age is not between visits 1 and 4 is equal to 0 is supported.
normally distributed, we conduct the Wilcoxon This is confirmed by the 95% CI. We are 95%
Rank Sum (Mann-Whitney U) test. The null confident that the true population mean differ-
hypothesis that the median age for the two groups ence lies between −1.02 and 9.21 units, which
is equal is supported, given a p-value of 0.533. includes 0 units.

> # Wilcoxon Rank Sum (Mann-Whitney U) > # Paired t-test ####


Test #### > ## Create a subset of the data for
> ## Shapiro-Wilk test visits 1 and 4 for group A
> Shapiro.test(catdata$AGE) > mndata <- rmdata[rmdata$VISITNUMBER
%in% c(1,4) & rmdata$GROUPING == “A”,]
Shapiro-Wilk normality test >
> ## Shapiro-Wilk test
> Shapiro.test(mndata$BIOMARKER)
data: catdata$AGE
W = 0.93607, p-value = 0.02554 Shapiro-Wilk normality test

>
> # Wilcoxon test data: mndata$BIOMARKER
> wilcox.test(AGE ~ GROUPING, data = W = 0.95451, p-value = 0.2229
catdata)
>
Wilcoxon rank sum test with continuity > ## Run paired t-test
correction > t.test(BIOMARKER ~ VISITNUMBER, data
= mndata)

data: AGE by GROUPING Welch Two Sample t-test


8 Analyzing Data from Therapeutic Drug Monitoring, Pharmacokinetics, and Clinical Toxicology Studies 135

data: BIOMARKER by VISITNUMBER > wilcox.test(GAIT_LENGTH ~ VISITNUMBER,


t = 1.641, df = 27.086, p-value = 0.1124 data = mndata, paired = T)
alternative hypothesis: true difference
in means between group 1 and group 4 is Wilcoxon signed rank test with conti-
not equal to 0 nuity correction
95 percent confidence interval:
-1.02389 9.20948
sample estimates: data: GAIT_LENGTH by VISITNUMBER
mean in group 1 mean in group 4 V = 0, p-value = 0.0007211
79.74408 75.65129 alternative hypothesis: true location
shift is not equal to 0

8.9.2.6 Wilcoxon Signed Rank Test


The Wilcoxon Signed Rank test is used when the 8.9.2.7 Linear Regression
assumption of interval (or ratio) normally distrib- Simple linear regression is used to test the rela-
uted differences in paired observations fails to tionship between a continuous predictor and a
hold. As such, the data is ranked (ordinal) and the continuous outcome variable. It is useful in pre-
null hypothesis is that the medians of the two dicting what the value of one variable will be
samples are equal. based on the value of the other variable. When
the more than one predictor is used, it is called
Example 8.20 In this example, since the gait multiple regression. Here, the predictors could
length is not normally distributed (Shapiro-Wilk include categorical variables. In this case, each
p-value = <0.001), we proceed with the Wilcoxon categorical variable is recoded to have numeric
Signed Rank test to assess if the medians in this values such that each level of the categorical pre-
paired assessment (before/visit 1 versus after/ dictor is treated as if it was a binary variable by
visit 4) of gait length are equal. itself.
Linear regression is based on four assump-
The null hypothesis that median gait lengths tions. First, the relationship between the depen-
before and after treatment are equal is rejected, dent and independent variable is linear. Figure 8.3
given a p-value of <0.001. is an example showing a simple linear regression
line and associated equation. Equation 8.12 is the
> # Wilcoxon Signed Rank test #### general form of a multiple regression equation.
> ## create a subset of the data for
visits 1 and 4 for group A
yi  a  b1 x1i  b2 x2 i  bz xzi  ei (8.12)
> mndata <- rmdata[rmdata$VISITNUMBER where
%in% c(1,4) & rmdata$GROUPING == “B”,] yi is the outcome/response for the ith participant.
> a is the intercept or the value of y when all predic-
> ## Shapiro-Wilk test tors have a value of 0.
> Shapiro.test(mndata$GAIT_LENGTH)
xi is the value for predictor x for the ith
participant.
Shapiro-Wilk normality test
b is the coefficient for each respective predictor.
It is the slope of the regression line and is
interpreted as the magnitude of change in the
data: mndata$GAIT_LENGTH
response variable for each unit change in the
W = 0.81633, p-value = 0.000132
respective continuous predictor or the differ-
> ence between levels, for a binary predictor.
> ## Run paired t-test This value is adjusted based on the effects of
other predictors.
136 A. M. Sulley

Fig. 8.3 Scatterplot with a regression line

> lmdata <- rmdata[rmdata$VISITNUMBER


ei is the residual or error term, which represents
== 1,]
other effects on y that are not accounted for by
>
the predictors in the model.
>
> ## Create a linear regression model
The second assumption is that (residuals of)
> mod <- lm(GAIT_LENGTH ~ BIOMARKER,
both outcome and continuous predictor variables
data = lmdata)
are normally distributed. Based on the central the- >
orem, if the sample size is about 30 or more, the > ## Test of normality for residuals
residuals can be assumed to be normal. Third, > shapiro.test(resid(mod))
observations are independent of each other. Lastly,
variances of the residual are homogeneous for Shapiro-Wilk normality test
each value of the predictor. When sample sizes for
groups being compared are approximately equal,
homogeneity of variances is not of much concern. data: resid(mod)
W = 0.97715, p-value = 0.7458
Example 8.21 In this simple regression exam-
ple, biomarker is not a statistically significant >
predictor of gait length at visit 1 (p = 0.704). The >
R-squared is a measure of the amount of variance > ## Display linear regression output
in biomarker explained by gait length, and in this > summary(mod)
case, it is only about 0.5%. The adjusted
R-squared excluded variability that is due to Call:
chance, and being negative, it suggests that the lm(formula = GAIT_LENGTH ~ BIOMARKER,
model does not fit the trend in the data. data = lmdata)

Residuals:
Min 1Q Median 3Q Max
> # Linear Regression ####
-6.2170 -2.3407 -0.3289 2.6034 8.3873
> ## create a subset of the data for
visit 1 for groups A and B
8 Analyzing Data from Therapeutic Drug Monitoring, Pharmacokinetics, and Clinical Toxicology Studies 137

Coefficients: Call:
Estimate Std. Error t value Pr(>|t|) lm(formula = GAIT_LENGTH ~ GROUPING +
(Intercept) 71.05733 6.63726 10.706 VISITNUMBER + BIOMARKER,
2.09e-11 *** data = rmdata)
BIOMARKER -0.03153 0.08216 -0.384 0.704
--- Residuals:
Signif. codes: 0 ‘***’ 0.001 ‘**’ 0.01 Min 1Q Median 3Q Max
‘*’ 0.05 ‘.’ 0.1 ‘ ’ 1 -9.982 -2.558 -0.072 2.663 10.235

Residual standard error: 3.702 on 28 Coefficients:


degrees of freedom Estimate Std. Error t value Pr(>|t|)
Multiple R-squared: 0.005233, Adjusted (Intercept) 67.718131 4.182362 16.191 <
R-squared: -0.03029 2e-16 ***
F-statistic: 0.1473 on 1 and 28 DF, GROUPINGB 2.035633 0.725162 2.807
p-value: 0.704 0.00588 **
VISITNUMBER2 0.509900 1.027276 0.496
0.62060
Example 8.22 In this multiple regression exam- VISITNUMBER3 1.410830 1.026279 1.375
ple, for visit 1 (reference visit) gait length 0.17192
increases by 2 cm for group B compared to group VISITNUMBER4 9.124217 1.040061 8.773
A (reference group), keeping biomarker constant 1.97e-14 ***
(p = 0.006). Furthermore, for group A (reference BIOMARKER -0.002646 0.050931 -0.052
group), gait length increases by 9.12 cm at visit 4 0.95866
compared to visit 1, keeping biomarker constant ---
Signif. codes: 0 ‘***’ 0.001 ‘**’ 0.01
(p < 0.001). The regression model explains about
‘*’ 0.05 ‘.’ 0.1 ‘ ’ 1
50% (adjusted to 48%) of the variance in gait
length. An overall p-value of 1.142e-15 for the
Residual standard error: 3.97 on 114
model indicates that the model is better than a
degrees of freedom
model consisting of only the intercept.
Multiple R-squared: 0.4972, Adjusted
R-squared: 0.4752
F-statistic: 22.55 on 5 and 114 DF,
> # Multiple Regression ####
p-value: 1.142e-15
> ## Create a linear regression model
> mmod <- lm(GAIT_LENGTH ~ GROUPING +
VISITNUMBER + BIOMARKER, data = rmdata) 8.9.2.8 Analysis of Variance (ANOVA)
> A one-way analysis of variance (ANOVA) is a
> ## Test of normality for residuals special case of simple linear regression with an
> shapiro.test(resid(mmod)) interval (or ratio) response variable and categori-
cal predictor variable. It is called one-way
Shapiro-Wilk normality test because there is only one predictor.
When the categorical predictor has two levels,
the result is identical to a two-sample indepen-
data: resid(mmod)
dent t-test. When additional continuous predic-
W = 0.99413, p-value = 0.8996
tors are added to the model, these predictors are
called covariates, and the test is called an analysis
>
of covariance (ANCOVA). When additional cat-
> ## Display linear regression output
egorical predictors (factors) are added to an
> summary(mmod)
138 A. M. Sulley

ANOVA, it is called factorial ANOVA. Depending Response: GAIT_LENGTH


on the number of factors included in the model, it Sum Sq Df F value Pr(>F)
GROUPING 124.22 1 15.2003 0.0001659 ***
can also be called a two-way, three-way, etc.
VISITNUMBER 1605.00 3 65.4659 < 2.2e-16
ANOVA or ANCOVA.
***
ANOVA and ANCOVA have similar assump-
BIOMARKER 11.89 1 1.4549 0.2303084
tions as linear regression and are robust against
GROUPING:VISITNUMBER 889.97 3 36.3006 <
minor departures from normality and unequal
2.2e-16 ***
variances of residuals, especially when group
Residuals 907.12 111
sizes are equal and large. ---
ANCOVA is used to assess the effects of a Signif. codes: 0 ‘***’ 0.001 ‘**’ 0.01
variable while controlling for the covariates. ‘*’ 0.05 ‘.’ 0.1 ‘ ’ 1
When the grouping variable has three or more >
groups, the ANOVA/ANCOVA output does not
give information about which pairs of groups are
significantly different from each other. This Given the significant interaction effects we
information is obtained using post-hoc tests. have, we want to know (1) how gait length differs
between groups for each visit, and (2) how gait
Example 8.23 In this two-way ANCOVA length differs between visits for each group. We
example, we want to compare means of gait will answer these questions by performing post-­
length for a combination of factors (group and hoc comparisons.
visit number), while controlling for biomarker. First, we generate the estimated marginal
Therefore, group and visit number are included means from the model, then we run pairwise
in the model as an interaction (denoted by * comparisons. Estimated marginal means are
between them), while biomarker is controlled mean values for the response variable (gait
for (included in the model additively). We use length) for each level of categorical predictor
the Anova command from the car package, (factor) or average continuous predictor (covari-
because it is better suited for models with ate). We use the emmeans package to generate
interactions. the estimated marginal means.

Since Grouping*Visit Number is statisti- 1. How does gait length differ between groups
cally significant, we interpret that result and for each visit?
ignore the main effects of GROUPING and
VISITNUMBER. Where the interaction term is Estimated marginal means for gait length are
not significant, we interpret the main effects. different between groups A and B for each visit.
The ANCOVA output tells us that there is a sig- The largest difference of 7.43 is observed at visit
nificant interaction between the effects of group 4 (p < 0.001). This can be confirmed by finding
and visit number on gait length while control- the difference in emmeans at visit 4. It is impor-
ling for biomarker [F(3, 111) = 36.30, tant to note the direction of these differences. At
p < 0.001]. visits 1 through 3, gait length was smaller in
group A compared to B. Gait length in group A
> # Analysis of variance #### increased gradually over the study period until it
> ## Create a linear regression model became larger than that for group B at visit 4,
> mmod <- lm(GAIT_LENGTH ~ GROUPING * suggesting that the treatment provided for group
VISITNUMBER + BIOMARKER, data = rmdata) A improves gait length.
>
> ## Run ANOVA > ## (1) Estimated marginal means by
> Anova(mmod) visit number
Anova Table (Type II tests) > library(emmeans)
8 Analyzing Data from Therapeutic Drug Monitoring, Pharmacokinetics, and Clinical Toxicology Studies 139

> EM <- emmeans(mmod, ~ GROUPING, A - B 7.43 1.05 111 7.087 <.0001


by=c("VISITNUMBER"))
> EM > ### Another method of getting the
VISITNUMBER = 1: same result
GROUPING emmean SE df lower.CL upper. > ### contrast(EM, "pairwise", adjust =
CL "Tukey" )
A 65.7 0.739 111 64.3 67.2
B 71.2 0.742 111 69.7 72.6
2. How does gait length differ between visits for
VISITNUMBER = 2:
each group?
GROUPING emmean SE df lower.CL upper. For group A, estimated marginal means for
CL gait length are significantly different between
A 66.2 0.745 111 64.7 67.7 visits 1 and 4, 2 and 4, and 3 and 4. This is con-
B 71.8 0.744 111 70.4 73.3 sistent with findings from the first pairwise com-
parison. For group B, the means are only different
VISITNUMBER = 3: between visits 1 and 4.
GROUPING emmean SE df lower.CL upper.
CL > ## (2) Estimated marginal means by
A 67.6 0.738 111 66.2 69.1 grouping
B 72.2 0.738 111 70.7 73.7 > EM2 <- emmeans(mmod, ~ VISITNUMBER,
by=c("GROUPING"))
VISITNUMBER = 4: > EM2
GROUPING emmean SE df lower.CL upper. GROUPING = A:
CL VISITNUMBER emmean SE df lower.CL
A 81.5 0.748 111 80.0 82.9 upper.CL
B 74.0 0.739 111 72.6 75.5 1 65.7 0.739 111 64.3 67.2
2 66.2 0.745 111 64.7 67.7
Confidence level used: 0.95 3 67.6 0.738 111 66.2 69.1
> 4 81.5 0.748 111 80.0 82.9
> ## Pairwise comparisons by visit
number GROUPING = B:
> pairs(EM) VISITNUMBER emmean SE df lower.CL
VISITNUMBER = 1: upper.CL
contrast estimate SE df t.ratio p. 1 71.2 0.742 111 69.7 72.6
value 2 71.8 0.744 111 70.4 73.3
A - B -5.43 1.04 111 -5.197 <.0001 3 72.2 0.738 111 70.7 73.7
4 74.0 0.739 111 72.6 75.5
VISITNUMBER = 2:
contrast estimate SE df t.ratio p. Confidence level used: 0.95
value >
A - B -5.64 1.06 111 -5.312 <.0001 > ## Pairwise comparisons by grouping
> pairs(EM2)
VISITNUMBER = 3: GROUPING = A:
contrast estimate SE df t.ratio p. contrast estimate SE df t.ratio p.
value value
A - B -4.58 1.04 111 -4.386 <.0001 1 - 2 -0.469 1.05 111 -0.448 0.9698
1 - 3 -1.882 1.04 111 -1.803 0.2775
VISITNUMBER = 4: 1 - 4 -15.718 1.06 111 -14.898 <.0001
contrast estimate SE df t.ratio p. 2 - 3 -1.413 1.05 111 -1.350 0.5334
value 2 - 4 -15.249 1.07 111 -14.286 <.0001
140 A. M. Sulley

3 - 4 -13.836 1.05 111 -13.127 <.0001 References


GROUPING = B: 1. R Core Team. R: a language and environment for sta-
contrast estimate SE df t.ratio p. tistical computing [Internet]. Vienna; 2020. Available
value from: https://www.R-­project.org/.
1 - 2 -0.675 1.06 111 -0.638 0.9194 2. RStudio Team. RStudio: integrated development
environment for R [Internet]. Boston; 2020. Available
1 - 3 -1.030 1.05 111 -0.985 0.7582 from: http://www.rstudio.com/.
1 - 4 -2.860 1.05 111 -2.726 0.0369 3. Sulley AM. qwickr: a quick and easy way to analyze
2 - 3 -0.355 1.05 111 -0.339 0.9865 and report phase I–IV clinical trial data including
2 - 4 -2.185 1.05 111 -2.089 0.1629 pharmacokinetics. 2021.
3 - 4 -1.830 1.04 111 -1.751 0.3025 4. Fox J, Weisberg S. An R companion to applied regres-
sion [Internet]. 3rd ed. Thousand Oaks: Sage; 2019.
Available from: https://socialsciences.mcmaster.ca/
P value adjustment: tukey method for jfox/Books/Companion/.
comparing a family of 4 estimates 5. Denney WS, Duvvuri S, Buckeridge C. Simple, auto-
matic noncompartmental analysis: the PKNCA R
package. J Pharmacokinet Pharmacodyn [Internet].
2015;42(1):11–107, S65. Available from: https://
8.9.2.9 Kruskal-Wallis Test github.com/billdenney/pknca.
A Kruskal-Wallis test is a nonparametric test 6. Andri et mult. al. S. DescTools: tools for descriptive
used in place of ANOVA when the assumptions statistics [Internet]. 2021. Available from: https://
are grossly violated. The dependent variable is cran.r-­project.org/package=DescTools.
7. Dionisi M, Cairoli S, Simeoli R, de Gennaro F,
treated as ordinal just like in a Wilcoxon Rank Paganelli V, Carta R, et al. Pharmacokinetic evalua-
Sum test, with the additional benefit of being able tion of Eltrombopag in ITP pediatric patients. Front
to handle categorical independent variables with Pharmacol. 2021;12:772873.
two or more levels. 8. de Rose DU, Cairoli S, Dionisi M, Santisi A, Massenzi
L, Goffredo BM, et al. Therapeutic drug monitoring is
a feasible tool to personalize drug administration in
Example 8.24 In this example, we compare gait neonates using new techniques: an overview on the
length between study groups. With a p-value pharmacokinetics and pharmacodynamics in neonatal
<0.001, we reject the null hypothesis that there is age. Int J Mol Sci. 2020;21(16):5898.
9. Bricheux A, Lenggenhager L, Hughes S, Karmime A,
no difference in gait lengths between study Lescuyer P, Huttner A. Therapeutic drug monitoring
groups. of imipenem and the incidence of toxicity and failure
in hospitalized patients: a retrospective cohort study.
Clin Microbiol Infect. 2019;25(3):383–e1.
> # Kruskal-Wallis test #### 10. Märtson A-G, Sturkenboom MGG, Stojanova J,
Cattaneo D, Hope W, Marriott D, et al. How to
> ## create a subset of the data for design a study to evaluate therapeutic drug monitor-
visit 4 for groups A and B ing in infectious diseases? Clin Microbiol Infect.
> kwdata <- rmdata[rmdata$VISITNUMBER 2020;26(8):1008–16.
== 4,] 11. Wang D, Bakhai A, editors. Clinical trials: a practi-
> ## Run KW test cal guide to design, analysis, and reporting. London:
Remedica; 2006. 480 p.
> kruskal.test(GAIT_LENGTH ~ GROUPING, 12. US Food and Drug Administration. Guidance for
data = kwdata) industry: bioavailability and bioequivalence studies
submitted in NDAs or INDs—general considerations.
Kruskal-Wallis rank sum test Rockville: Food and Drug Administration I; 2014.
p. 2014.
13. Hiemke C, Bergemann N, Clement HW, Conca A,
Deckert J, Domschke K, et al. Consensus guidelines
data: GAIT_LENGTH by GROUPING for therapeutic drug monitoring in neuropsycho-
Kruskal-Wallis chi-squared = 16.021, df pharmacology: update 2017. Pharmacopsychiatry.
= 1, p-value = 6.264e-05 2018;51(01/02):9–62.
8 Analyzing Data from Therapeutic Drug Monitoring, Pharmacokinetics, and Clinical Toxicology Studies 141

14. Rodríguez-Gascón A, Solinís MÁ, Isla A. The 16. Ahad NA, Yin TS, Othman AR, Yaacob CR. Sensitivity
role of PK/PD analysis in the development and of normality tests to non-normal data. Sains Malays.
evaluation of antimicrobials. Pharmaceutics. 2011;40(6):637–41.
2021;13(6):833. 17. Boneau CA. The effects of violations of assumptions
15. de Velde F, Mouton JW, de Winter BCM, van underlying the t test. Psychol Bull. 1960;57(1):49.
Gelder T, Koch BCP. Clinical applications of 18. Levene H. Robust tests for equality of variances.
population pharmacokinetic models of antibiot- In: Contributions to probability and statistics essays
ics: challenges and perspectives. Pharmacol Res. in honor of Harold Hotelling. Palo Alto: Stanford
2018;134:280–8. University Press; 1961. p. 279–92.
Reducing Toxicity in Critically Ill
Patients by Using Therapeutic
9
Drug Monitoring

Zalak Panchal, Khushboo Faldu, and Jigna Shah

Abstract TDM might be useful in the reduction of tox-


icity in critically ill patients with serious
Nowadays, the healthcare system encounters infections.
high morbidity rates which are related to
severe infections in critical illnesses. However, Keywords
by dose individualization, we can minimize
the toxicity in critically ill patients. These Therapeutic drug monitoring · Critically ill
days, the patient and organism complexity patients · Toxicity · Pharmacokinetics ·
demand precision dosing which can be Pharmacodynamics · Antimicrobial ·
achieved through therapeutic drug monitoring Antibiotic · Antifungal · Antiviral ·
(TDM) services. TDM has proven to be an Anticoagulant
effective approach for dose individualization.
Conventionally, TDM of antifungal, antibiot-
ics, antiviral, and antimicrobial classes of Abbreviations
drugs essentially involve quantitative drug
measurement in the plasma of the patients to 5-FC 5-Fluorocytosine
reduce toxicity risks associated with agents of ABCDE Airway, breathing, circulation,
narrow therapeutic indices. But it has been disability, exposure
observed that there is significant institutional ADME Absorption, distribution, metabo-
variation related to TDM-specific criteria like lism, excretion
the selection of patients, concentration moni- AIS Abbreviated Injury Score
toring, time of sampling, assay techniques, APACHE Acute Physiology and Chronic
pharmacokinetic/pharmacodynamic (PK/PD) Health Evaluation
target selection, and dosage optimization pro- ARC Enhanced renal clearance
cedures. The goal of this chapter is to examine AUC Area under the curve
available information on TDM practices for BMD Bone mineral density
different drug classes and to illustrate how CL Drug clearance
CLSI Clinical and Laboratory Standards
Z. Panchal · K. Faldu · J. Shah (*) Institute
Department of Pharmacology, Institute of Pharmacy, CRRT Continuous renal replacement
Nirma University, Ahmedabad, Gujarat, India therapy
e-mail: 21mph212@nirmauni.ac.in; DOACs Direct oral anticoagulants
khushi181@gmail.com; jigna.shah@nirmauni.ac.in

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 143
S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_9
144 Z. Panchal et al.

EC50 Half maximal effective 9.1 Introduction


concentration
ECMO Extracorporeal membrane 9.1.1 Critically Ill Patients
oxygenation
ESBL-PE Extended-spectrum ß-lactamase-­ Critically ill patients are at-risk patients who can
producing Enterobacteriaceae easily acquire infections as a result of multiresis-
EUCAST European Committee on tant organism which could lead to significant
Antimicrobial Susceptibility Testing morbidity or mortality [1–3]. These patients are
FLC Free light chains generally victims of motor vehicle accidents, vio-
HCV Hepatitis C virus lence, burns, drowning, falls, or patients with car-
HIT Heparin-induced thrombocyto- diovascular and cerebrovascular comorbidities
penia [3]. The main clinical indications that define a
HPLC-UV High-performance liquid chroma- critical state in patients are as follows: hypoten-
tography with ultraviolet detection sion, tachycardia, tachypnea, a decreased volume
HSV Herpes simplex virus of urine output, and altered consciousness [4].
IATDMCT International Association of When these observations are analyzed collec-
Therapeutic Drug Monitoring and tively, their sensitivity and specificity for critical
Clinical Toxicology illness are considerably increased [4]. Therefore,
ICU Intensive care unit the management of critically ill patients should
ISS Injury Severity Score be thoughtfully planned. Additionally, the evalu-
IV Intravenous ations of critically ill patients must be carried out
LC-MS Liquid chromatography-mass by a qualified clinician using a standardized
spectrometry ABCDE (airway, breathing, circulation, disabil-
LMWH Low molecular weight heparin ity, and exposure) format [2, 5]. The classifica-
MAP Mean arterial pressure tion of patients into risk groups is based on their
MELD Model for end-stage liver disease severity and identifying discrepancies between
MIC Minimum inhibitory concentration various units or medical centers [6]. In addition
MRSA Methicillin-resistant Staphylo- to the required clinical estimate, score systems
coccus aureus are employed at various stages of in-hospital
NFB Non-fermenter bacilli treatments for acutely or potentially critically ill
PAE Post-antibiotic effect patients [6, 7]. The scoring methods use morpho-
PD Pharmacodynamic logical, physiological, and biochemical factors to
PDDIs Possible drug-drug interactions quantify the severity of critically ill/injured
PK Pharmacokinetic patients and assign them to a risk group [6, 7].
RRT Renal replacement therapy The existing scoring system includes:
SAPS Simplified Acute Physiology Score
SOFA Sepsis-related organ failure 1. Organ-based scoring which is like therapeutic
assessment scoring. It is based on the observation that as
TDM Therapeutic drug monitoring the health of the patient deteriorates, the
TISS Therapeutic Intervention Scoring organs get affected and can malfunction or
System head into failure. One of the assessments is
UFH Unfractionated heparin sepsis-related organ failure assessment
Vd Volume of distribution (SOFA) [6, 7].
VRE Vancomycin-resistant enterococci 2. Simple scale: It is based on clinical judgment
VTE Venous thromboembolism (e.g., survive or die) [7].
9 Reducing Toxicity in Critically Ill Patients by Using Therapeutic Drug Monitoring 145

3. Anatomy-based scoring is based on the TDM is therapeutically relevant and requires a


involvement of anatomical areas and is uti- good correlation between the plasma concentra-
lized mainly in trauma patients. Examples tion of the drug and clinical efficacy [12]. For drug
include Injury Severity Score (ISS) and concentration measures to be effective in treating
Abbreviated Injury Scale (AIS) [7]. patients, they must be strongly related to the drug
4. Physiological assessment is based on the action, toxicity, or both [12]. This allows for the
degree of derangement of routinely measured definition of an effective therapeutic window – the
physiological variables, e.g., Acute concentration range between the minimum effec-
Physiology and Chronic Health Evaluation tive concentration and the concentration at which
(APACHE) and Simplified Acute Physiology noxious effects begin to appear – as well as dose
Score (SAPS) [6, 7]. titration to attain effective concentration within
5. Assessment based on the disease-like model that window [10, 12]. Therapeutic drug monitor-
for end-stage liver disease (MELD), Child-­ ing engages in determining drug concentrations
Pugh for assessment of liver failure, subarach- alongside interpreting the results clinically. This
noid hemorrhage scoring by World Federation necessitates an understanding of pharmacokinet-
of Neurosurgical Societies, and Ranson’s ics, pharmacodynamics, sample time, drug his-
assessment for acute pancreatitis [7]. tory, and the clinical status of the patient [9].
6. Therapeutically weighted scoring like the TDM is essential because pharmacokinetics and
Therapeutic Intervention Scoring System pharmacodynamics (PK/PD) are highly variable
(TISS) utilizes the assumption that critically due to increased patient and organism complex-
ill individuals require multiple complex thera- ity (Fig. 9.1). A highly variable PK/PD is found
peutic interventions and procedures in com- in people with critical illness, obese, and older
parison to comparatively healthy or recovering adults [12]. Precision dosing of drugs through
individuals [7]. TDM is becoming important for these patient
populations to improve outcomes and minimize
toxicities [10, 13]. Currently, TDM is being
9.1.2 Therapeutic Drug Monitoring used on antibiotics, antifungals, antivirals, anti-
microbials, anticoagulants, sedative-analgesic,
Therapeutic drug monitoring (TDM) is an effec- vasopressor-inotropic agents, and neuromuscular
tive approach for dose individualization [8]. In blocking agents [13]. The goal of this chapter is
general, TDM deals with determining drug con- to discuss how TDM can be utilized to reduce
centration in the bloodstream and adjusting doses toxicity in critically ill patients and improve
within a targeted therapeutic window [8, 9]. The patient outcomes from severe infections.
reliability of TDM is predominantly due to the
specificity and sensitivity of the analytical meth-
ods [10]. Moreover, the International Association 9.2 Antimicrobial Agents
of Therapeutic Drug Monitoring and Clinical
Toxicology (IATDMCT) defines TDM as “a mul- 9.2.1 General Pharmacokinetics
tidisciplinary clinical specialty aimed at improv- (PK)/Pharmacodynamics (PD)
ing patient care by individually adjusting the Targets
dose of drugs for which clinical experience or
clinical trials have shown improved outcomes in Pharmacological drug modeling utilizes the char-
the general or special populations” [11]. TDM acteristics, especially pharmacokinetics (PK) and
concedes the evaluation of drug efficacy and pharmacodynamics (PD) parameters of the drug.
safety in a range of clinical contexts by a collec- PK involves studying the action of the body on the
tive understanding of pharmaceutics, pharmaco- drug which involves its absorption (A), distribu-
kinetics, and pharmacodynamics [12]. Thus, tion (D), metabolism (M), and excretion (E). PD
TDM helps to improve the clinical effects of the involves the study of the effect of the drug on the
drug along with patient management. body which involves its interaction with different
146 Z. Panchal et al.

Fig. 9.1 Flowchart for


Diagnosis
reaching dosage
decisions with
therapeutic drug
monitoring Drug selection

The dosage regimen is designed to achieve a specific plasma concentration.

Drug administration

Patient evaluations are Concentration of drugs are


performed determined

Pharmacokinetic modeling is applied and clinical judgement is used

Dosage adjustment (if required)

receptors, enzymes, and signaling pathways to physicochemical properties are utilized for the
produce a desired effect in the body [14]. Many determination of the concentration of drug in body
antimicrobials have shown benefits in terms of fluids and tissues [18].
lower mortality, clinical efficacy, reduced half-life, PD is the relationship between a drug’s phar-
and reduced toxicity. These are related to PK/PD macological influence and its PK which for anti-
target achievement, through modifications in dos- biotics becomes minimum inhibitory
ing [15]. The concentration-­time course of an anti- concentration (MIC). The MIC of antibiotics is
microbial agent is defined by PK parameters, the concentration at which the drug effectively
based on the drug’s ADME profile, adverse drug inhibits the growth of microorganisms or patho-
reactions, and protein binding. Furthermore, these gens [16]. This interlinks PD; PK (blood concen-
parameters are strongly influenced by critical ill- tration); the pharmacological activity of the drug,
ness [16]. The absorption of the drug reflects on its i.e., its inhibitory action on microorganisms or
bioavailability and is influenced by tissue or organ pathogens; and drug toxicity (Fig. 9.2) [15, 18,
characteristics and physicochemical properties. In 19]. Theoretically, MIC should be directly pro-
critical illness, problems regarding absorption portional to PK for achieving the ideal PK/PD
often increase the need for intravenous administra- index [19].
tion, which ensures 100% bioavailability [17]. Sepsis patients not admitted to ICU may
This chapter intends to discuss PK assessment in experience altered exposure to medications [20].
critically ill patients. The area under the curve The conventional antimicrobial dosages can
(AUC), peak plasma concentration, and concen- have altered volume of distribution (Vd) and
tration before the next dosage(Cmin) are some of drug clearance (Cl) which may lead to subthera-
the most important PK parameters which in com- peutic or toxic drug exposure [20]. At the very
bination with strategic dosing as per the drug’s least, the human PK exposure target should be
9 Reducing Toxicity in Critically Ill Patients by Using Therapeutic Drug Monitoring 147

Fig. 9.2 Triangular


Antimicrobial
relationship between
antimicrobial drugs, agents
host, and bacteria during
treatment

Host defense
Host Microorganism

Infection

determined using preclinical PK/PD relation- method, EUCAST, adoption of local antibio-
ships (e.g., fCmax/MIC, free maximal drug con- grams and CLSI breakpoints, and automated
centration to MIC ratio; fAUC/MIC, area under microbiology system (e.g., Phoenix, Vitek 2) can
the free concentration-­time curve to MIC ratio; all be used to define MICs for TDM [20, 22].
fT > MIC, time the free concentration surpasses Clinicians, using TDM to treat serious infections,
the MIC) [21]. especially those involving resistant organisms,
Figure 9.3 lists frequent factors that can affect should be aware of each method’s limitations.
antimicrobial pharmacokinetics in critically ill The tissue distribution of hydrophilic antimi-
patients. The Vd of hydrophilic antimicrobials crobials is restricted to the extracellular space,
like ß-lactams and aminoglycosides, which with the majority of patients relying on renal
approximates the extracellular fluid volume, clearance for survival [23]. However, lipophilic
could be significantly increased by capillary antimicrobials have an intracellular accumulation
leakage, fluid resuscitation, and third space losses and depend on hepatic clearance [21]. As a result,
[20]. Antimicrobials Cl are influenced by the hydrophilic medicines are more affected than
patient organ function, drug clearance systems, lipophilic medications by critical illness PK
and extracorporeal therapies. Antimicrobials Cl changes, particularly in situations of sepsis,
are reduced by renal hypoperfusion, acute kidney which leads to fluctuation in renal function and
injury, and end-organ failure (Fig. 9.3). In criti- increase in Vd due to resuscitation of volume and
cally ill patients, however, enhanced renal clear- leakage of capillaries which indicates whether or
ance (ARC) has been documented, in which not hydrophilic antimicrobials loading and main-
accelerated antibiotic removal leads to subthera- tenance dosages in critically ill patients need to
peutic values [15, 22]. The effects of therapies be adjusted [24]. The blood concentration of AB
like extracorporeal membrane oxygenation is also affected by dose, Vd, and bioavailability.
(ECMO) and renal replacement therapy (RRT) Hydrophilic antimicrobials with an extracellular
on antibiotic pharmacokinetics are multifaceted, distribution have a low Vd, whereas lipophilic
varied, and complex, as previously addressed antimicrobials with a rapid cellular uptake have a
[15, 20] high Vd [23]. Conventionally followed antimi-
All PK/PD attainments are expressed with crobial dosages will be inadequate to meet ­PK/
respect to the pathogen’s MIC, emphasizing that, PD parameters in ICU patients [20]. As a result,
in addition to antimicrobials concentration mea- a tailored approach that takes into account spe-
surements, precise and timely MIC determina- cific MIC and regimens that are very likely to
tion should be considered the base of antibiotic achieve PK/PD objectives can deliver viable
TDM. Disc method, E-test, microdilution broth answers [20, 24].
148 Z. Panchal et al.

Fig. 9.3 Common


factors affected with the 1. Hypoalbuminaemia, leading to
pharmacokinetics of increased unbound drug
antimicrobials in Increased Vd
critically ill patients 2. Capillary leakage

3. Fluid resuscitation

4. Third space loss

5. Renal hypoperfusion
Decreased Cl
6. Acute kidney injury

7. Renal/ hepatic dysfunction

Increased Cl 8. Augmented renal clearance

Variable changes in 9. Extracorporeal interventions (e.g.,


Vd & Cl RRT, ECMO)

9.2.2 Antibacterial Agents/ in the local area must be closely monitored


Antibiotics before empirical administration of antibiotics.
The development of antibiotic-­resistant infec-
Antibiotics are one of the most regularly tions in critically ill patients is the most serious
used medications in ICU patients. Critically adverse effect of the administration of antibac-
ill and ICU patients who are at maximum terial medications [25].
risk of contracting community or hospital- Antibiotics are divided into three PK/PD
acquired infections are administered antibi- groups (concentration-dependent, time-­
otics preemptively [16]. The most common dependent, and concentration-time dependent)
bacterial infections which are acquired in the based on their dose-response relationships [16,
community or hospital settings are caused due 24]. The efficacy of time-dependent antibiotics
to vancomycin-resistant enterococci (VRE), like ß-lactams is determined by the cumulative
methicillin-resistant Staphylococcus aureus time percentage for which the concentration of
(MRSA), extended-­ spectrum ß-lactamase- the free antibiotics is more than MIC (percent
producing Enterobacteriaceae (ESBL-PE), fT > MIC) in the due course of 24 hours. Increased
MDR Pseudomonas aeruginosa, non-fermen- concentrations above MIC do not lead to an
ter bacilli (NFB), and carbapenem-resistant improvement in the killing rate [26]. The effect of
Acinetobacter baumannii [25]. Statistics show concentration-dependent antibiotics, such as
that around 70% pool of critically ill patients aminoglycosides, is determined by the ratio of
in ICU are administered antibiotic treatment peak concentration to MIC [17]. The spectrum
empirically or targeted. Antibiotic resistance and rate of bactericidal activity are directly pro-
9 Reducing Toxicity in Critically Ill Patients by Using Therapeutic Drug Monitoring 149

portional to the concentration of antibiotics [18]. extended interval dosing [29]. Ototoxicity and
Concentration-time-dependent antibiotics like nephrotoxicity have been linked to the elevated
fluoroquinolones and glycopeptides utilize the Cmin and continuous AUC exposures spanning
ratio of AUC0–24 to MIC. across days [33].

9.2.2.1 Aminoglycosides Dose Adjustment Strategies


The commonly used PK/PD targets for amino-
PK/PD Targets glycoside TDM are fT > MIC, AUC/MIC, and
In clinical practice, it has become a standard Cmax/MIC; no other targets have been set specifi-
practice for performing therapeutic drug moni- cally for the critical illness [34, 35]. As TDM data
toring of aminoglycosides like amikacin, genta- from the individual patient is provided, the soft-
micin, and tobramycin to reduce drug toxicity ware can check the expected parameters in an
while maintaining maximum efficacy [27]. The individual patient and hence provide precise dose
toxicity of aminoglycosides rendered it less pop- recommendations that can achieve therapeutic
ular for the treatment of critical illnesses in com- PK/PD targets [30]. Bayesian dose adaptation
parison to other available broad-spectrum software is the best predictor of aminoglycoside
antibiotics [28]. With the growth of multidrug-­ dosing requirements [27, 36]. Although no clini-
resistant (MDR) gram-negative infections, par- cal benefits of software-based dosing methods
ticularly hospital-acquired infections, there is a have been demonstrated, they should be consid-
renewed interest in aminoglycoside therapy [28]. ered only in critically ill patients with severe
Characteristics of aminoglycosides include a infections. TDM by Bayesian dose adaptation
high susceptibility rate for a variety of gram-­ resulted in decreased hospitalization duration and
negative bacteria commonly observed in critical reduced instances of nephrotoxicity in patients
illness [20]. Aminoglycosides have a post-­ receiving gentamicin [36, 37].
antibiotic effect (PAE), in which even after the
sinking of antibiotic concentration below MIC, Bioanalytical Assay
the bacterial growth remains suppressed [29]. Assay of aminoglycoside TDM is well estab-
Aminoglycosides are small, hydrophilic com- lished, with little, if any, disagreement in the
pounds whose volume of distribution (Vd) is scholarly literature [27]. For aminoglycosides,
equivalent to the volume of extracellular fluid current immunoassay approaches like chromato-
and their rate of clearance (Cl) is proportional to graphic assays are still very effective for amika-
the glomerular filtration rate [30]. The studies cin, gentamicin, and tobramycin TDM [29, 30].
have pointed out that fluctuations in Vd and Cl For regular clinical practice, validated, low-cost,
are observed in critically ill patients. Because of and easy-to-use immunoassays are preferred in
hypoalbuminemia, increased capillary permea- comparison to chromatography or capillary zone
bility, or organ dysfunctions, PK (increased vol- electrophoresis [20].
ume of distribution or changed drug clearance) of
critically ill patients is severely affected [31]. 9.2.2.2 ß-Lactams
Aminoglycosides exhibit concentration-­
dependent bactericidal actions, with maximum PK/PD Targets
activity occurring at a peak concentration (Cmax) Critically ill patients are frequently administered
of around eight to ten times the organism’s mini- ß-lactam antibiotics for treatment of severe infec-
mum inhibitory concentration (MIC) ratio (Cmax tions. ß-lactam antibiotics are generally hydro-
is ≥8–10 × MIC) [32]. philic, have a low volume of distribution (Vd),
Current research study suggests ratio of AUC and are mostly eliminated from kidneys [38].
vs time curve during 24 hours period to Protein binding of ß-lactams varies from moder-
MIC(AUC0–24/MIC ratio) is potent indicator of ate (30–70%) to low (30%) [39]. Early stages of
response and can be effectively utilized to achieve infections witness significant alteration in Vd and
target accomplishment in aminoglycoside Cl which can lead to altered PK and inadequate
150 Z. Panchal et al.

concentrations of ß-lactam antibiotics [38]. 9.2.2.3 Fluoroquinolones


Hypoalbuminemia leads to an increase in non-­
protein-­bound drug concentrations of ß-lactams PK/PD Targets
like ceftriaxone, semisynthetic penicillin (flu- Fluoroquinolones are moderately lipophilic, hav-
cloxacillin, temocillin, and oxacillin), and ertape- ing Vd affected by critical sickness, with excre-
nem which are highly protein-bound, while it has tion of one of the fluoroquinolones like
been observed that toward the end of the dosing levofloxacin [44]. The majority of fluoroquino-
interval, they are found in lower concentrations lones bind to proteins in a moderate to low
in unbound form [38]. Piperacillin-tazobactam amount, and some are excreted by kidneys. The
and meropenem are both ß-lactams, and as such, bactericidal efficacy of fluoroquinolones is
they are categorized as time-dependent antibiot- concentration-­dependent and is best predicted by
ics in terms of pharmacodynamics [40]. When the ratio of AUC0–24 to MIC. For optimal bacteri-
the antibiotic’s “free” or unbound plasma con- cidal pursuit, greater Cmax/MIC ratios are neces-
centrations are above the minimum inhibitory sary [45]. Gram-positive species with the
concentration (MIC) during the dosing period AUC0–24/MIC ratios of 25–30 also show efficacy,
(fT > MIC), these medicines have the greatest but gram-negative microorganisms require higher
efficacy [40]. The % fT > (40–70%) MIC is the values [39, 46]. A ratio of >100–200 has also
PK/PD measure linked with excellent ß-lactam been indicated to limit the formation of resis-
action [41]. Despite the fact that myelosuppres- tance against gram-negative microbes in several
sion is required for ß-lactam toxicity, no toxicity investigations [47]. Despite an increase in reports
criteria have been established [29]. of fluoroquinolone-related seizures, the criteria
for the toxicity have not been established, and
Dose Adjustment Strategies still causality is being contested [44, 46].
In most TDM units, generalized but nonspecific
dose modification strategies such as adjusting Dose Adjustment Strategies
dose quantity or frequency, as well as the use of In critical patients, a dosage plan for quinolones
prolonged or continuous infusion, have been should be devised such that AUC0–24/MIC is
used regularly [39, 42]. However, monitoring higher than LD with higher maintenance doses
concentrations for a clinician-selected target [48]. TDM may prove beneficial in patients with
appears to be safe and more appropriate. If con- quinolone resistance or to address inter-­individual
tinual infusions are prescribed, a target of 100% variability in PK. It will be useful in case bacteria
fT > MIC might be a good choice [27]. Cmin sam- exhibit MICs near the susceptibility breakpoint
ples are commonly used at a steady state; how- [16, 49].
ever, dosing software could allow for much
earlier sampling and dose optimization [20, 43]. Bioanalytical Assay
Although Bayesian adaptive feedback software is Literature suggests the use of sensitive tech-
the ideal technique for adjusting doses of niques like LC-MS/MS and HPLC for fluoroqui-
ß-­lactams, its availability at clinical sites may be nolones TDM [29].
limited [30].
9.2.2.4 Vancomycin
Bioanalytical Assay
Liquid chromatography is a widely used assay PK/PD Targets
method for ß-lactam TDM [20]. Several liquid Vancomycin is the first-line therapy for
chromatography-mass spectrometry (LC-MS/ methicillin-­resistant Staphylococcus aureus
MS) and high-performance liquid chromatogra- (MRSA), and it is advised for therapeutic drug
phy with ultraviolet detection (HPLC-UV) assays monitoring (TDM) to avoid nephrotoxicity and
have also been employed for therapeutic drug achieve optimal therapeutic outcomes [50].
monitoring of ß-lactams [20, 27, 29]. While scientific community still debates the
9 Reducing Toxicity in Critically Ill Patients by Using Therapeutic Drug Monitoring 151

efficacy of TDM in reducing toxicity and Bioanalytical Assay


increasing the clinical effectiveness of vanco- Vancomycin TDM immunoassays are widely
mycin. The evidence for a link between high available [29]. It is a homogeneous enzyme assay
serum vancomycin concentrations and nephro- technique for quantitative analysis.
toxicity is mixed [51]. All plasma concentra-
tions were assigned to one of three categories: 9.2.2.5 Linezolid
therapeutic, subtherapeutic, or supratherapeu-
tic. For common infections, a therapeutic van- PK/PD Targets
comycin level was identified as 20–25 mg/L in Linezolid pharmacokinetic variability was for-
adults, 25–35 mg/L in severe infections, and merly thought to be less significant than that of
10–15 mg/L in children and newborns [52]. other antibiotics; therefore, dose modifications
Both CRRT and non-CRRT patients were were avoided in patients with dysfunctional kid-
included [53]. Vancomycin is hydrophilic in neys and liver [56]. The most common serious
nature, has a lower Vd, and is mostly elimi- linezolid ADR is thrombocytopenia, which has
nated by the kidneys. The Vd and Cl of been linked to linezolid plasma concentrations in
Vancomycin are altered by critical illness, numerous investigations [57]. Linezolid is a lipo-
resulting in fluctuating and low drug exposure philic molecule. Its Vd is equivalent to the body’s
[29, 54]. Based on PD of vancomycin, for S. total water, and its elimination is done by nonre-
aureus with MIC less than or equal to 2 mg/L, nal clearance. Varied patient exposure to line-
the ratio of AUC/MIC should be greater than or zolid is due to the factors like intra- and
equal to 400 [51, 54]. inter-patient variability in PK [58]. Renal func-
tion serves as a substantial source of inter-patient
Dose Adjustment Strategies variability in linezolid clearance because 30–40%
Dose adjustments can be accomplished by of the linezolid dose is removed unaltered in the
adjusting the dose according to the ratio between urine (Cl) [59]. Antibiotic efficacy of linezolid
the measured and target concentrations. The tar- can be well characterized by the ratio of area
get concentrations for intermittent (15–20 mg/L) under the curve of plasma concentration vs time
and continuous (20–25 mg/L) administration are graph to MIC (AUC0–24/MIC) and the time period
not the same, requiring a higher continuous infu- for which the antibiotic plasma concentration is
sion target to reach the same AUC as intermittent higher than the minimum inhibitory concentra-
dosing [20]. When treating microorganisms with tion (MIC) for the given pathogen (T > MIC)
MICs >1 mg/L, safely achieving optimum [60]. To exert optimal antimicrobial activity, a
AUC0–24/MIC ratios is extremely difficult [55]. T > MIC of 85 and an AUC0–24/MIC >100 are
Dose individualization methods based on the necessary and are related to better clinical out-
calculation of individual pharmacokinetic comes in seriously ill patients [60].
parameters and PK/PD targets (AUC/MIC) are
available, but they are not widely used in clinical Dose Adjustment Strategies
practice [53, 55]. For dose adaptation, real-time Higher linezolid dosages and/or other adminis-
Bayesian forecasting software combined with tration procedures (e.g., continuous infusion and
TDM is regarded to be the most accurate [52]. front-loaded dosing regimen) may assist criti-
Despite the fact that vancomycin’s current cally ill patients. If TDM is available, these
AUC0–24/MIC attainments have consistently been approaches should be backed up with it [61].
around 400, the index came into existence from Obese patients, patients with acute respiratory
BMD MIC, and its E-test resemblance is 226 distress syndrome and ARC, and patients infected
which is low [29]. with bacteria with MICs of less than 2 mg/L are
152 Z. Panchal et al.

all subsets of patients who should be adminis- 9.2.3 Antifungal Agents


tered higher doses of linezolid [29, 62]. Table 9.1
summarizes the PK/PD index, efficacy, and tox- The antifungal agents have a well-defined dose-­
icity threshold of various antibacterial drugs. response relationship for TDM essentially with
respect to a narrow therapeutic index and substan-
Bioanalytical Assay tial pharmacokinetic variability to impact clear-
Literature suggests the use of immunoassays, ance. Antifungals currently recommended for
HPLC-UV, and LC-MS/MS but is not utilized in therapeutic drug monitoring include flucytosine,
routine clinical TDM practice [27, 29, 63]. fluconazole, itraconazole, and voriconazole.

Table 9.1 PK/PD index, efficacy, and toxicity threshold of antibacterial drugs
Antibacterial Clinical PK/PD threshold for
class PK/PD index Clinical PK/PD threshold for efficacy toxicity
Aminoglycosides
Gentamicin AUC0–24/MIC AUC0–24/MIC greater than or equal to 110 Cmin greater than 1 mg/La
Cmax/MIC greater than or equal to 8–10
Amikacin AUC0–24/MIC Cmax/MIC greater than or equal to 8–10 Cmin greater than 5 mg/La
ß –lactams
Penicillins % fT greater than 50–100% fT greater than MIC Cmin greater than 361 mg/Ld
MIC
Carbapenems % fT greater than 50–100% fT greater than MIC Cmin greater than 44.5 mg/Lb
MIC
Cephalosporins % fT greater than 45–100% fT greater than MIC Cmin greater than 20 mg/Lc
MIC
Daptomycin AUC0–24/MIC AUC0–24/MIC greater than or equal to Cmin greater than 24 mg/Le
666 mg/L
Fluoroquinolones AUC0–24/MIC AUC0–24/MIC greater than or equal to Ambiguous
125–250
Cmax/MIC greater than or equal to 12
Glycopeptides
Linezolid AUC0–24/MIC AUC0–24/MIC: 80–120 greater than or AUC0–24 greater than 700 mg h/
equal to 85% Lf
fT greater than MIC Cmin greater than 20 mg/Lf
Vancomycin AUC0–24/MIC AUC0–24/MIC greater than or equal to 400 AUC0–24 greater than 300g
Cmin greater than 10–20 mg/L Cmin greater than 7g
Teicoplanin AUC0–24/MIC Cmin greater than or equal to 10 mg/L Ambiguous
Polymyxins
Polymyxin B AUC0–24/MIC Data unavailable AUC0–24 greater than 100f
Colistin AUC0–24/MIC Data unavailable Cmin greater than 2.4 mg/Lf
AUC0–24/MIC the ratio of the area under the concentration-time curve during a 24-hour period to minimum inhibitory
concentration, Cmax/MIC the ratio of maximum drug concentration to minimum inhibitory concentration, Cmin trough/
minimum drug concentration, fAUC0–24/MIC the free (unbound drug concentration) ratio of the area under the
concentration-­time curve during a 24-hour period to minimum inhibitory concentration, fT > MIC the duration of time
that the free drug concentration remains above the MIC during a dosing interval, PK/PD pharmacokinetic/
pharmacodynamics
a
Nephrotoxicity or ototoxicity
b
Data only available for meropenem and related to nephrotoxicity or neurotoxicity
c
Data only available for cefepime and related to neurotoxicity
d
Data mostly on piperacillin and related to nephrotoxicity or neurotoxicity
e
Myopathy indicated by creatine phosphokinase elevation
f
Related to nephrotoxicity
g
Related to hematological toxicity
9 Reducing Toxicity in Critically Ill Patients by Using Therapeutic Drug Monitoring 153

9.2.3.1 Fluconazole Candida species as well as Cryptococcus neo-


formans. Because of the rapid selection of
PK/PD Targets resistant isolates when used as monotherapy,
Fluconazole drugs are available in oral and par- clinical usage of 5-FC is mostly limited to
enteral formulations, have a linear PK profile, combination therapy with another antifungal
and are highly absorbed from the gastrointesti- like amphotericin B, for the treatment of cryp-
nal tract [64, 65]. It is somewhat lipophilic with tococcal meningitis [72]. Flucytosine has a Vd
Vd around 1 L/kg and is mostly eliminated by of 0.6–0.9 L/kg and is mostly eliminated by
the kidneys. Substantial inter-individual PK the kidneys [73]. There is a close link between
variableness was observed [66]. The relation- 5-FC serum concentrations and both toxicity
ship between drug dose, exposure, in vitro sus- and efficacy, according to pharmacodynamic
ceptibility, and response to therapy has been investigations [74]. The toxicodynamic link
demonstrated in preclinical and clinical for two relatively common adverse effects,
research, implying that dosing may be crucial bone marrow suppression and hepatic dys-
for outcomes in infections caused by bacteria function, has been the strongest of these cor-
with lower susceptibility [67, 68]. Values of relations [47]. Variable flucytosine
55.2–100 of the ratio of AUC0–24/MIC have been concentrations have been found due to signifi-
defined as having a maximum therapeutic effect cant inter-patient PK variability [75, 76]. In
in patients with candidemia [69]. Since flucon- multiple clinical studies, a Cmax of moreover
azole AUC can be substituted by its dose, it has 100 mg/L is associated with hepatotoxicity
been utilized to calculate the dose/MIC ratio for and myelosuppression. Resistant C. albicans
evaluating its clinical outcomes [70]. mutants may be selectively amplified at con-
Hepatotoxicity and convulsions are possible centrations of less than 25 mg/L [76]. Table 9.2
side effects of higher doses [71]. summarizes the PK/PD attainments and mag-
nitudes associated with antifungal agents.
Dose Adjustment Strategies
This target was associated with a Cmin of approxi- Dose Adjustment Strategies
mately 10e15 mg/L [69, 71]. TDM found that an There is inadequate clinical evidence to recom-
FLC Cmin > 11 mg/L was substantially linked mend changing flucytosine dose in critically ill
with clinical outcomes in adult liver transplant patients for optimal outcomes. Dosing should be
recipients taking FLC for invasive candidiasis based on weight and suited to renal function [67].
[65]. In critically ill patients with normal renal In comparison to toxicity prevention, TDM com-
function, an intravenous low dose of 12 mg/kg manded dosing to optimize flucytosine efficacy
followed by an adjusted intravenous dose of remains little documented. To reduce the toxicity
6–12 mg/kg is recommended to achieve PK/PD and resistance of flucytosine, TDM can be uti-
outcomes – low (AUC0–24/MIC ratio of 25) or lized to achieve Cmax = 100 mg/L and
high (AUC0–24/MIC ratio of 100) [65, 66]. Cmin = 25 mg/L [73, 76].

Bioanalytical Assay Bioanalytical Assay


For fluconazole TDM, several chromatographic For flucytosine TDM, several chromatographic
tests have been documented [29, 64]. assays have been documented [27, 67].

9.2.3.2 Flucytosine
9.2.4 Antiviral Agents
PK/PD Targets
One of the first antifungal chemicals produced Antiviral TDM is useful not only to minimize
was flucytosine (5-FC), a pyrimidine deriva- potential toxicities but also to increase the effec-
tive [27]. The medicine is effective against tiveness of treatment. Acyclovir, ribavirin, fos-
154 Z. Panchal et al.

Table 9.2 PK/PD attainments and magnitudes associated with antifungals


Antifungal PK/PD
drugs index Clinical PK/PD efficacy threshold Clinical PK/PD toxicity threshold
Itraconazole AUC0–24/ Cmin greater than or equal to 0.25–0.5 mg/L Cave greater than or equal to
MIC (Prop) 17.1 mg/Ld
Cmin greater than or equal to 1 mg/L (Tx)
Fluconazole AUC0–24/ AUC0–24/MIC greater than or equal to Ambiguous
MIC 55–100
Flucytosine % fT > MIC Data unavailable Cmax greater than 100 mg/Lb
Posaconazole AUC0–24/ Cmin greater than 0.5 (Prop) Data unavailable
MIC Cmin greater than 1 mg/L (Tx)
Voriconazole AUC0–24/ Cmin greater than or equal to 1–2 mg/L Cmin greater than or equal to
MIC 4.5–6 mg/Lc
Echinocandins AUC0–24/ AUC0–24/MIC greater than 3000a Data unavailable
MIC
Isavuconazole AUC0–24/ Data unavailable Data unavailable
MIC
AUC0–24/MIC the ratio of the area under the concentration-time curve during a 24-hour period to minimum inhibitory
concentration, Cave average drug concentration, Cmin = trough/minimum drug concentration, fAUC0–24/MIC the free
(unbound drug concentration) ratio of the area under the concentration-time curve during a 24-hour period to minimum
inhibitory concentration, fT > MIC the duration of time that the free drug concentration remains above the MIC during
a dosing interval, PK/PD pharmacokinetic/pharmacodynamics, prop prophylaxis, Tx treatment
a
In patients receiving micafungin for invasive candidiasis/candidemia
b
Related to hematological toxicity and hepatotoxicity
c
Mostly related to gastrointestinal toxicity
d
Mostly related to hepatotoxicity and neurotoxicity

carnet, and ganciclovir are few examples of neither favored nor opposed by the panel. A few
antivirals that essentially involve TDM. reports on acyclovir TDM are published, and the
majority of these feature individuals with enceph-
9.2.4.1 Acyclovir/Valacyclovir alitis. If TDM is used, a Cmin of 2–4 mg/L is
advised [79].
PK/PD Targets
Acyclovir and valacyclovir are moderately lipo- Bioanalytical Assay
philic entities with high Vd and are excreted by Multiple techniques like LC-MS/MS and
kidneys [77]. There is a scarcity of information HPLC-UV have been research for TDM of acy-
relating acyclovir exposure to clinical efficacy clovir [27, 81].
and harm [78]. The efficacy of these drugs in the
treatment of the herpes simplex virus (HSV) is 9.2.4.2 Ribavirin
connected to AUC and the time duration for which
it is available above 50% inhibitory concentration PK/PD Targets
(EC50; T > EC50). But these indices need to be Ribavirin has a high Vd (about 18 L/kg) and is
investigated further [79]. Higher doses have been mostly eliminated by the kidneys [82]. This drug
associated with an increased risk of gastrointesti- has been connected to a variety of PK differ-
nal and neurological side effects, especially in ences between individuals, including bioavail-
patients with renal impairment [27, 79]. ability. Literature on the relationship between
drug exposure, efficacy, and toxicity of ribavirin
Dose Adjustment Strategies is conflicting [83]. A conflicting correlation was
In immunocompetent individuals with severe also found between the relationship of Cmin and
viral infections, a typical intravenous dose of AUC to hepatitis C virus (HCV) eradication or
10–15 mg/kg every 8 hours is suggested [80]. long-term virological response [84]. A strong
Routine TDM in critical patients for acyclovir is predictor of sustained virological response is the
9 Reducing Toxicity in Critically Ill Patients by Using Therapeutic Drug Monitoring 155

area under the curve [AUC0–4 (1.76 mg h/L) and ICU patients varies, ranging from 30% in normal
AUC0–12 (3.01 mg h/L)] after initial administra- medical-surgical ICU patients to 70% in ICU
tion. Although a Cmin of >2.3–3.5 has been con- patients with acute ischemic stroke [87]. ICU
nected to hemolytic anemia, some investigations patients’ clinical conditions, such as intubation,
have found no significant association [27, 85]. mechanical ventilation, sedation, delirium, and
Other studies have found that other parameters altered mental status, may mask the common
(such as pegIFN-alpha-2a levels) may have a presentations of VTE, resulting in a silent DVT
higher impact on the outcome than ribavirin lev- in many ICU patients, highlighting the impor-
els [85]. Table 9.3 summarizes the PK/PD attain- tance of prophylactic anticoagulant administra-
ments and magnitudes associated with antiviral tion in complete bed rest patients. However,
agents. many of these people are in danger of serious
bleeding if they are given full-strength anticoagu-
Dose Adjustment Strategies lants [89]. Unfractionated heparin (UFH) and
Hepatitis C is not regarded as a pathogenic entity, low molecular weight heparin (LMWH) are the
and thus ribavirin TDM is not regarded as com- most often used anticoagulants in the ICU. The
pletely essential [83]. Additionally, data linking most serious problem with UFH and, to a lesser
exposure to ribavirin to its efficacy and toxicity is extent, LMWH administration in critically ill
contradictory [85]. While in chronic hepatitis C patients is the development of heparin-induced
genotype 1, TDM dosing of ribavirin has shown thrombocytopenia (HIT), which can be caused by
better virological response in comparison to immunological or non-immune mechanisms and
weight-based administration, and the resulting can result in thrombosis. In this case, calculating
anemia in the TDM group although of severe the “4Ts” score could aid in HIT diagnosis in
grade could be adequately treated with erythro- ICU patients [89, 90]. The 4T score is determined
poietin beta [83]. TDM has recently been pro- by assigning points to each of the following
posed as a technique for guiding ribavirin parameters: thrombocytopenia, platelet count fall
treatment in paramyxovirus-infected lung trans- timing, thrombosis or other sequelae, and other
plant patients [86]. causes of thrombocytopenia [90]. According to a
recent study, the clinical and economic repercus-
Bioanalytical Assay sions of VTE prophylaxis with UFH and LMWH
For ribavirin TDM, the literature suggests the are similar, with no statistically significant differ-
use of LC-MS/MS and HPLC-UV techniques ences in critically ill patients. DOACs, or direct
[27, 86]. oral anticoagulants, are new anticoagulants that
have been used to prevent VTE in critically ill
patients. The simplicity of administration by oral
9.3 Anticoagulant Agents route, shorter half-lives (t1/2), and lack of HIT
adverse effects are the major benefits of DOACs
Prophylactic anticoagulation is required in high-­ [91]. Partial hepatic metabolism and renal elimi-
risk patients because critically sick patients have nation are the most significant drawbacks, both
a higher risk of venous thromboembolism (VTE) of which can lead to possible drug-drug interac-
[87, 88]. VTE may also result in statistically sig- tions (PDDIs) [91]. Clinical trials demonstrated
nificant increases in mortality and morbidity due that rivaroxaban reduced the incidence of VTE
to their reduced cardiopulmonary function. The and the risk of significant bleeding when com-
most common symptom of VTE in ICU patients pared to LMWH, whereas apixaban was not
is deep vein thrombosis (DVT), which can lead to superior to LMWH and was associated with
pulmonary embolism (PE). The rate of DVT in increased bleeding risks [92].
156 Z. Panchal et al.

Table 9.3 PK/PD attainments and magnitudes associated with antivirals


PK/PD Clinical PK/PD efficacy Clinical PK/PD toxicity
Antiviral drugs index threshold threshold
Acyclovir/valacyclovir Ambiguousa Ambiguousa Ambiguous
Ganciclovir/valganciclovir AUC AUC: 40–60 mg h/L (Prop) Ambiguousa
Ribavirin AUC Auc0–4 greater than 1755 mg h/L Cmin greater than 2.3 mg/Lb
AUC0–12 greater than
3014 mg h/L
Cmin greater than or equal to
2 mg/L
Foscarnet Ambiguousa Ambiguousa No dataa
Oseltamivir/oseltamivir Ambiguousa Ambiguousa Ambiguousa
carboxylate
AUC area under the concentration-time curve, AUC0–4 the ratio of the area under the concentration-time curve during
a 4-hour period, AUC0–12 the ratio of the area under the concentration-time curve during a 12-hour period, Cmin trough/
minimum drug concentration, PK/PD pharmacokinetic/pharmacodynamics, prop prophylaxis
a
While in vitro concentrations at which viral replication is inhibited by 50% (i.e., EC50 representing antiviral activity)
have been widely determined, there are no/limited data which correlate these values with in vivo pharmacokinetic
parameters (e.g., AUC) to describe magnitudes for preclinical efficacy
b
Mostly related to anemia

9.4 Sedative and Analgesic 9.5 Vasopressors and Inotropic


Agents Agents

Sedation and analgesia are frequently required Vasopressors are drugs that induce an increase in
in critically ill and mechanically ventilated ICU mean arterial pressure (MAP) by inducing vasocon-
patients due to discomfort, anxiety, or delirium striction. Inotropes improve myocardial contractility
[25]. PK of sedatives and analgesics in critical [96]. Multiple molecules have both the characteris-
patients is altered due to alteration of altered tics of inotropes and vasopressors. The catechol-
protein binding, fluid resuscitation, multi- amine neurotransmitters (dopamine, epinephrine,
organ failure, and unstable hemodynamics. and norepinephrine) induce adrenergic and dopami-
Personalized pharmacotherapy can reduce the nergic receptors and thus can be utilized as moieties
chances of adverse drug reactions and increase for shock control [25]. In hemodynamically unsta-
the chance of achieving optimal efficacy in crit- ble patients, vasopressors improve vascular tone,
ical patients admitted to ICU. Based on the and inotropes improve myocardial contractions
patient’s health parameters, accurate drug and [87]. These drugs are administered in patients who
dose selection is required to achieve optimum are hemorrhagic and cardiogenic, have heart failure,
sedation in critical patients [93]. The use of or are in ­septic shock and also trauma patients and
opioids and/or benzodiazepines in the treat- patients undergoing surgery [96]. Alterations in
ment of ICU delirium has been linked to a lon- metabolism, immune system, receptors, intracellu-
ger delirium episode in these critically ill lar signaling pathways, and renal and hepato-
patients [94]. They give consistent amnesic splanchnic blood flow can later PK in critical
effects while also causing cardiovascular and patients [87, 97]. This necessitates careful medica-
respiratory depression [25]. Midazolam can be tion selection and dosing in critical patients.
effectively utilized in intensive care for pro-
longed sedative effect without the occurrence
of adrenal suppression as it is 94–98% protein-­ 9.6 Neuromuscular Blocking
bound with a shorter half-life, steady-state Vd, Agents
shorter elimination half-life (1.5–5 hours), and
intermediate clearance (Cl) [25]. Delayed ICU Neuromuscular blockers are quaternary ammo-
delirium management has been linked to a nium compounds that are ionized and water-­
higher mortality rate [95]. soluble and can be utilized to facilitate mechanical
9 Reducing Toxicity in Critically Ill Patients by Using Therapeutic Drug Monitoring 157

ventilation and lower respiratory muscle oxygen peutic efficacy as well as decrease the instances
consumption. They are also employed in the of development of serious adverse events and
treatment of tetanus and status epilepticus [25]. drug toxicity.
The trend of administering neuromuscular block-
ers in ICU is reducing with shorter-acting agents
finding prominence [25]. Most commonly pre- References
scribed are nondepolarizing blockers (atracu-
rium, pancuronium, and vecuronium) which are 1. Ron Gilat MD, Haunschild ED, Tracy Tauro BSBA,
Cole BJ, M.D. MB. Since January 2020 Elsevier has
administered as an intravenous bolus or via con- created a COVID-19 resource centre with free infor-
tinuous infusion [87]. Long-term use of neuro- mation in English and Mandarin on the novel corona-
muscular blockers can lead to the buildup of virus COVID-19. Ann Oncol. 2020;(January):19–21.
drugs which can result in long-term weakness in 2. Bennett KA, Robertson LC, Al-Haddad
M. Recognizing the critically ill patient. Anaesth
critically ill patients, and thus twitch monitoring Intensive Care Med. 2016;17(1):1–4.
is practiced [98]. Pancuronium also builds up in 3. Lilian TM, Halima M. Caring critically ill patients
patients with fulminant hepatic failure (FHF), in the general wards in Tanzania: experience of
and thus in the case of patients with renal or nurses and physicians. Int J Crit Care Emerg Med.
2018;4(2):1–7.
hepatic insufficiency, twitch monitoring is neces- 4. Sekulic AD, Trpkovic SV, Pavlovic AP, Marinkovic
sary [99]. OM, Ilic AN. Scoring systems in assessing sur-
vival of critically ill ICU patients. Med Sci Monit.
2015;21:2621–9.
5. National G, Pillars H. No 主観的健康感を中心とし
9.7 Conclusion た在宅高齢者における 健康関連指標に関する共
分散構造分析Title.
TDM is an effective approach for dose individu- 6. Trujillano J, Badia M, Servi L, March J, Rodriguez-­
alization. TDM is becoming increasingly vital for Pozo A. Stratification of the severity of critically
ill patients with classification trees. BMC Med Res
critically ill patients to enhance their outcomes Methodol. 2009;9(1):1–12.
and reduce drug toxicity. Some patients may still 7. Dzaharudin F, Ralib AM, Jamaludin UK, Nor MBM,
receive sub-optimal antimicrobial exposure Tumian A, Har LC, et al. Mortality prediction in criti-
because of the long duration of medication and cally ill patients using machine learning score. IOP
Conf Ser Mater Sci Eng. 2020;788(1):012029.
the PK/PD variability. TDM-guided dose adjust- 8. Dasgupta A. Chapter 1 - Introduction to Therapeutic
ment strategy is safe and effective in assuring that Drug Monitoring: Frequently and Less Frequently
all critically ill patients get therapeutic exposures. Monitored Drugs. In: Therapeutic drug monitor-
TDM of antibiotics and antifungals is highly rec- ing. Elsevier; 2012. p. 1–29. https://doi.org/10.1016/
C2010-­0-­66930-­6
ommended for achieving therapeutic index in 9. Ghiculesco R. Abnormal laboratory results: therapeu-
critically ill and ICU patients. Antivirals will tic drug monitoring: which drugs, why, when and how
require additional clinical investigation to dis- to do it. Aust Prescr. 2008;31(2):42–4.
cover therapeutic targets for patient benefit. TDM 10. Ates HC, Roberts JA, Lipman J, Cass AEG, Urban
GA, Dincer C. On-site therapeutic drug monitoring.
of anticoagulants is restricted to specific clinical Trends Biotechnol. 2020;38(11):1262–77.
conditions and has recently gained importance 11. Swartz HA, Jessica C. Levenson and EFU. 乳鼠
for the treatment of critically ill patients. 心肌提取 HHS Public Access. Physiol Behav.
Understanding the therapeutic indices of seda- 2012;43(2):145–53.
12. Kang JS, Lee MH. Overview of therapeutic drug
tives, analgesics, and vasopressor-inotropic monitoring. Korean J Intern Med. 2009;24(1):1–10.
agents in critically patients will require extensive 13. Oellerich M, Kanzow P, Walson PD. Therapeutic drug
research involving TDM. Few studies of neuro- monitoring – key to personalized pharmacotherapy.
muscular blocking agents employing TDM prac- Clin Biochem. 2017;50(7–8):375–9.
14. Ahmad I, Huang L, Hao H, Sanders P, Yuan
tices have shown beneficial effects in critical Z. Corrigendum to “Application of PK/PD modeling
illness. From the clinical studies, it is recom- in veterinary field: dose optimization and drug resis-
mended that TDM should be introduced as bed- tance prediction”. Biomed Res Int. 2017;2017(Vich
side practice for optimizing the therapeutic doses 2003):1408737.
15. Mabilat C, Gros MF, Nicolau D, Mouton JW, Textoris
in critically ill patients so as to maintain thera- J, Roberts JA, et al. Diagnostic and medical needs for
158 Z. Panchal et al.

therapeutic drug monitoring of antibiotics. Eur J Clin 32. Cristea S, Smits A, Kulo A, Knibbe CAJ, van
Microbiol Infect Dis. 2020;39(5):791–7. Weissenbruch M, Krekels EHJ, Allegaert K. Amikacin
16. Póvoa P, Moniz P, Pereira JG, Coelho L. Optimizing pharmacokinetics to optimize dosing in neonates
antimicrobial drug dosing in critically ill patients. with perinatal asphyxia treated with hypothermia.
Microorganisms. 2021;9(7):1–27. Antimicrob Agents Chemother. 2017;61(12):1–9.
17. Phe K, Heil EL, Tam VH. Optimizing 33. Hurkacz M, Nowakowska JM, Paluszyńska D,
pharmacokinetics-­ pharmacodynamics of antimicro- Królak-Olejnik B. Individualization of treatment
bial management in patients with sepsis: a review. J with gentamicin in neonates based on drug con-
Infect Dis. 2021;222(Suppl 2):S132–41. centration in the blood serum. Dev Period Med.
18. Craig WA. Pharmacokinetic/pharmacodynamic 2019;23(1):21–7.
parameters: rationale for antibacterial dosing of mice 34. Hartman SJF, Brüggemann RJ, Orriëns L, Dia N,
and men. Clin Infect Dis. 1998;26(1):1–12. Schreuder MF, De Wildt SN. Pharmacokinetics
19. Moniz P, Coelho L, Póvoa P. Antimicrobial steward- and target attainment of antibiotics in critically ill
ship in the intensive care unit: the role of biomarkers, children: a systematic review of current literature.
pharmacokinetics, and pharmacodynamics. Adv Ther. Clin Pharmacokinet. 2020;59:173–205. https://doi.
2021;38(1):164–79. org/10.1007/s40262-­019-­00813-­w.
20. Wong G, Sime FB, Lipman J, Roberts JA. 1471-2334-­ 35. Nicolau DP, Freeman CD, Belliveau PP, Nightingale
14-288.Pdf. 2014:1–11. CH, Ross JW, Quintiliani R. Experience with a
21. Märtson AG, Sturkenboom MGG, Stojanova J, once-daily aminoglycoside program administered to
Cattaneo D, Hope W, Marriott D, et al. How to 2,184 adult patients. Antimicrob Agents Chemother.
design a study to evaluate therapeutic drug monitor- 1995;39(3):650–5.
ing in infectious diseases? Clin Microbiol Infect. 36. Begg EJ, Barclay ML, Duffull SB. A suggested
2020;26(8):1008–16. approach to once-daily aminoglycoside dosing. Br J
22. Carrié C, Chadefaux G, Sauvage N, De Courson H, Clin Pharmacol. 1995;39:605–9.
Petit L, Nouette-Gaulain K, et al. Increased β-Lactams 37. Roberts JA, Field J, Visser A, Whitbread R, Tallot M,
dosing regimens improve clinical outcome in criti- Lipman J, et al. Using population pharmacokinet-
cally ill patients with augmented renal clearance ics to determine gentamicin dosing during extended
treated for a first episode of hospital or ventilator-­ daily diafiltration in critically ill patients with acute
acquired pneumonia: a before and after study. Crit kidney injury. Antimicrob Agents Chemother.
Care. 2019;23(1):1–9. 2010;54(9):3635–40.
23. Pea F, Viale P, Furlanut M. Antimicrobial ther- 38. Guilhaumou R, Benaboud S, Bennis Y, Dahyot-­
apy in critically ill patients. Clin Pharmacokinet. Fizelier C, Dailly E, Gandia P, et al. Optimization of
2005;44(10):1009–34. the treatment with beta-lactam antibiotics in critically
24. 123.pdf. ill patients. Crit Care. 2019;23(1):104.
25. Power BM, Forbes AM, Heerden PV, Van Ilett 39. Pistolesi V, Morabito S, Di MF, Regolisti G, Cantarelli
KF. Pharmacokinetics of drugs used in critically ill C, Fiaccadori E. A guide to understanding antimi-
adults. Clin Pharmacokinet. 1998;34(1):25–56. crobial drug dosing in critically ill patients on renal
26. Mouton JW, Punt N, Vinks AA. Concentration-effect replacement therapy. Antimicrob Agents Chemother.
relationship of ceftazidime explains why the time 2019;63(8):e00583–19.
above the MIC is 40 percent for a static effect in vivo. 40. McDonald C, Cotta MOC, Little PJ, McWhinney B,
Antimicrob Agents Chemother. 2007;51(9):3449–51. Ungerer JPJ, Lipman J, et al. Is high-dose β-lactam
27. Balakrishnan I, Shorten RJ. Therapeutic drug therapy associated with excessive drug toxic-
monitoring of antimicrobials. Ann Clin Biochem. ity in critically ill patients? Minerva Anestesiol.
2016;53(3):333–46. 2016;82(9):957–65.
28. De Waele JJ, De Neve N. Aminoglycosides for life-­ 41. Veiga RP, Paiva J-A. Pharmacokinetics-­
threatening infections: a plea for an individualized pharmacodynamics issues relevant for the clinical use
approach using intensive therapeutic drug monitoring. of beta-lactam antibiotics in critically ill patients 11
Minerva Anestesiol. 2014;80(10):1135–42. Medical and Health Sciences 1103 Clinical Sciences.
29. Abdul-Aziz MH, Alffenaar JWC, Bassetti M, Bracht Crit Care. 2018;22:1–34.
H, Dimopoulos G, Marriott D, et al. Antimicrobial 42. Sime FB, Roberts MS, Peake SL, Lipman J, Roberts
therapeutic drug monitoring in critically ill adult JA. Does beta-lactam pharmacokinetic variability in
patients: a Position Paper#. Intensive Care Med. critically ill patients justify therapeutic drug moni-
2020;46(6):1127–53. toring? A systematic review. Ann Intensive Care.
30. Roberts JA, Norris R, Paterson DL, Martin 2012;2(1):1.
JH. Therapeutic drug monitoring of antimicrobials. 43. Tängdén T, Ramos Martín V, Felton TW, Nielsen
Br J Clin Pharmacol. 2012;73(1):27–36. EI, Marchand S, Brüggemann RJ, et al. The role of
31. Roger C, Wallis SC, Muller L, Saissi G, Lipman infection models and PK/PD modelling for optimis-
J, Lefrant J, et al. Influence of renal replacement ing care of critically ill patients with severe infections.
modalities on amikacin population pharmacoki- Intensive Care Med. 2017;43(7):1021–32.
netics in critically ill patients on continuous renal 44. Dalhoff A. Resistance surveillance studies: a mul-
replacement therapy. Antimicrob Agents Chemother. tifaceted problem — the fluoroquinolone example.
2016;60(8):4901–9. Infection. 2012;40:239–62.
9 Reducing Toxicity in Critically Ill Patients by Using Therapeutic Drug Monitoring 159

45. Abdulla A, Ewoldt TMJ, Hunfeld NGM, Muller AE, 60. Villa G, Di Maggio P, De Gaudio AR, Novelli A,
Rietdijk WJR, Polinder S, et al. The effect of therapeu- Antoniotti R, Fiaccadori E, et al. Effects of continu-
tic drug monitoring of beta-lactam and fluoroquino- ous renal replacement therapy on linezolid pharmaco-
lones on clinical outcome in critically ill patients: the kinetic/pharmacodynamics: a systematic review. Crit
DOLPHIN trial protocol of a multi-centre randomised Care. 2016;20:1–11.
controlled trial. BMC Infect Dis. 2020;20(1):1–9. 61. Kawasuji H, Tsuji Y, Ogami C, Kimoto K, Ueno
46. Mckenzie C. Antibiotic dosing in critical illness. J A, Miyajima Y, et al. Proposal of initial and main-
Antimicrob Chemother. 2011;66(Suppl 2):25–31. tenance dosing regimens with linezolid for renal
47. Aminimanizani A, Beringer P, Jelliffe R. Comparative impairment patients. BMC Pharmacol Toxicol.
pharmacokinetics and pharmacodynamics of 2021;22(1):13.
the newer fluoroquinolone antibacterials. Clin 62. Soraluce A, Barrasa H, As E, Jose ÁS, Maynar J, Isla
Pharmacokinet. 2001;40(3):169–87. A, et al. Novel population pharmacokinetic model
48. Bilbao-Meseguer I, Rodríguez-Gascón A, Barrasa for linezolid in critically ill patients and evaluation of
H, Isla A, Solinís MÁ. Augmented renal clearance the adequacy of the current dosing recommendation.
in critically Ill patients: a systematic review. Clin Pharmaceutics. 2020;12(1):54.
Pharmacokinet. 2018;57(9):1107–21. 63. Ashbee HR, Barnes RA, Johnson EM, Richardson
49. Kumta N, et al. A systematic review of studies MD, Gorton R, Hope WW. Therapeutic drug monitor-
reporting antibiotic pharmacokinetic data in the ing (TDM) of antifungal agents: guidelines from the
cerebrospinal fluid of critically ill patients with unin- British Society for Medical Mycology. J Antimicrob
flamed meninges. Antimicrob Agents Chemother. Chemother. 2014;69:1162–76.
2021;65(1):1–19. 64. Andes D, Pascual A, Marchetti O. Antifungal thera-
50. Ye Z, Li C, Zhai S. Guidelines for therapeutic drug peutic drug monitoring: established and emerg-
monitoring of vancomycin: a systematic review. PLoS ing indications. Antimicrob Agents Chemother.
One. 2014;9(6):e99044. 2009;53(1):24–34.
51. Ye Z, Tang H, Zhai S. Benefits of therapeutic drug 65. Sandaradura I, Wojciechowski J, Marriott DJE, Day
monitoring of vancomycin: a systematic review and RO, et al. Model-optimized fluconazole dose selec-
meta-analysis. PLoS One. 2013;8(10):e77169. tion for critically ill patients improves early phar-
52. Zamoner W, Prado IRS, Balbi AL, Ponce macodynamic target attainment without the need
D. Vancomycin dosing, monitoring and toxicity: criti- for therapeutic drug monitoring. Antimicrob Agents
cal review of the clinical practice. Clin Exp Pharmacol Chemother. 2021;65(3):1–10.
Physiol. 2019;46(4):292–301. 66. Muilwijk EW, de Lange DW, Schouten JA, Wasmann
53. Bakke V, Sporsem H, Von Der LE, Nordøy I, Lao Y, RE, Ter Heine R, Burger DM, Colbers A, Haas PJ,
Nyrerød HC, et al. Vancomycin levels are frequently Verweij PE, Pickkers P, Brüggemann RJ. Suboptimal
subtherapeutic in critically ill patients: a prospec- dosing of fluconazole in critically ill patients: time
tive observational study. Acta Anaesthesiol Scand. to rethink dosing. Antimicrob Agents Chemother.
2017;61:627–35. 2020;64(10):e00984–20.
54. He N, Su S, Ye Z, Du G, He B, Li D, Liu Y, et al. 67. Smith J, Andes D. Therapeutic drug monitoring of
Evidence-based Guideline for Therapeutic Drug antifungals: pharmacokinetic and pharmacodynamic
Monitoring of Vancomycin: 2020 Update by considerations. Ther Drug Monit. 2008;30(2):1–6.
the Division of Therapeutic Drug Monitoring, 68. Märtson A-G, Boonstra JM, Sandaradura I, Kosterink
Chinese Pharmacological Society. Clin Infect Dis. JGW, van der Werf TS, Marriott DJE, Zijlstra JG,
2020;71(4):S363–S371. Touw DJ, Alffenaar JWC. Reply to Van Daele et al.,
55. Marella P, Roberts J, Hay K, Shekar K. Effectiveness “Fluconazole underexposure in critically ill patients: a
of vancomycin dosing guided by therapeutic drug matter of using the right targets?”. Antimicrob Agents
monitoring in adult patients receiving extracorpo- Chemother. 2021;65(6):e00465–21.
real membrane oxygenation. Antimicrob Agents 69. Boonstra JM, Märtson AG, Sandaradura I, Kosterink
Chemother. 2020;64(9):1–7. JGW, Van Der Werf TS, Marriott DJE, et al.
56. Pea F, Furlanut M, Cojutti P, Cristini F, Zamparini Optimization of fluconazole dosing for the preven-
E, Franceschi L, et al. Therapeutic drug moni- tion and treatment of invasive candidiasis based
toring of linezolid: a retrospective monocen- on the pharmacokinetics of fluconazole in criti-
tric analysis. Antimicrob Agents Chemother. cally ill patients. Antimicrob Agents Chemother.
2010;54(11):4605–10. 2021;65(3):1–11.
57. Cheng C, Wu C, Kuo C, Jhang R, Lin S. ScienceDirect 70. Vena A, Muñoz P, Mateos M, Guinea J, Galar A, Pea
Impact of high plasma concentrations of linezolid in F, Alvarez-Uria A, Escribano P, Bouza E. Therapeutic
Taiwanese adult patients – therapeutic drug monitor- drug monitoring of antifungal drugs: another tool
ing in improving adverse drug reactions. J Formos to improve patient outcome ? Infect Dis Ther.
Med Assoc. 2021;120(1):466–75. 2020;9:137–49.
58. Therapeutic S, Monitoring D. Crossm systematic 71. Gómez-López A. Antifungal therapeutic drug moni-
therapeutic drug monitoring. toring: focus on drugs without a clear recommenda-
59. Crass RL, Cojutti PG, Pai MP, Pea F. Reappraisal of tion. Clin Microbiol Infect. 2020;26:1481–7.
linezolid dosing in renal impairment to improve safety. 72. Bellmann R, Smuszkiewicz P. Pharmacokinetics
Antimicrob Agents Chemother. 2019;63(8):6–8. of antifungal drugs: practical implications for
160 Z. Panchal et al.

optimized treatment of patients. Infection. plasma by capillary electrophoresis. Electrophoresis.


2017;45(6):737–79. 2004;25(10–11):1615–22.
73. Pasqualotto, A. Aspergillosis: from diag- 86. Müller DM, Rentsch KM. Therapeutic drug monitor-
nosis to prevention. 2010. https://doi. ing by LC-MS-MS with special focus on anti-infective
org/10.1007/978-­90-­481-­2408-­4 drugs. Anal Bioanal Chem. 2010;398(6):2573–94.
74. Myers E, Ashley ED. Antifungal drug therapeutic 87. Ghasemiyeh P, Mohammadi-Samani S, Firouzabadi
monitoring: what are the issues? Curr Clin Microbiol N, Vazin A, Zand F. A brief ICU residents’ guide:
Rep. 2015;2:55–66. Pharmacotherapy, pharmacokinetic aspects and dose
75. Kunka ME, Cady EA, Woo HC, Bastin MLT. Case adjustments in critically ill adult patients admitted to
report flucytosine pharmacokinetics in a critically ill ICU. Trends Anaesth Crit Care. 2021;41:11–31.
patient receiving continuous renal replacement ther- 88. Boonyawat K, Crowther MA. Venous thromboem-
apy. Case Rep Crit Care. 2015;2015:5–10. bolism prophylaxis in critically ill patients. Semin
76. Vermes A, Guchelaar HJ, Dankert J. Flucytosine: Thromb Hemost. 2015;41(1):68–74.
a review of its pharmacology, clinical indications, 89. Sakr Y. Heparin-induced thrombocytopenia in the
pharmacokinetics, toxicity and drug interactions. J ICU: an overview. Crit Care. 2011;15(2):211.
Antimicrob Chemother. 2000;46:171–9. 90. Lo GK, Juhl D, Warkentin TE, Sigouin CS, Eichler P,
77. Back DJ, Khoo SH, Gibbons SE, Merry C. The role Greinacher A. Evaluation of pretest clinical score (4
of therapeutic drug monitoring in treatment of HIV T’s) for the diagnosis of heparin-induced thrombocy-
infection. Br J Clin Pharmacol. 2001;51(4):301–8. topenia in two clinical settings. J Thromb Haemost.
78. Svensson JO, Barkholt L, Säwe 2006;4(4):759–65.
J. Determination of acyclovir and its metabolite 91. Bertoletti L, Murgier M, Stelfox HT. Direct oral anti-
9-­carboxymethoxymethylguanine in serum and urine coagulants for venous thromboembolism prophylaxis
using solid-phase extraction and high-performance in critically ill patients: where do we go from here ?
liquid chromatography. J Chromatogr B Biomed Sci Intensive Care Med. 2019;45(4):549–51.
Appl. 1997;690(1–2):363–6. 92. Samuel Z, Goldhaber AL, Kakkar AK, Haas SK,
79. Watson WA, Rhodes NJ, Echenique IA, Angarone Merli G, Knabb RM, Weitz JI, ADOPT Trial
MP, Scheetz MH. Resolution of acyclovir-associated Investigators. Apixaban versus enoxaparin for throm-
neurotoxicity with the aid of improved clearance boprophylaxis in medically ill patients. N Engl J Med.
estimates using a Bayesian approach: a case report 2011;365(23):2167–77.
and review of the literature. J Clin Pharm Ther. 93. Berghaus G, Sticht G, Grellner W. Meta-analysis of
2017;42(3):350–5. empirical studies concerning the effects of medicines
80. Peyrière H, Jeziorsky E, Jalabert A, Cociglio M, and illegal drugs including pharmacokinetics on safe
Benketira A, Blayac JP, et al. Neurotoxicity related driving meta-analysis of empirical studies concerning
to valganciclovir in a child with impaired function: the effects of medicines and illegal drugs including
usefulness of therapeutic drug monitoring. Ann pharmacokinetics on safe. 2011.
Pharmacother. 2006;40(1):143–6. 94. Pharm D, Van Ness PH, Inouye SK. NIH Public
81. Ritchie BM, Barreto JN, Barreto EF, Crow SA, Access. 2010;37(1):177–83.
Dierkhising RA, Jannetto PJ, Tosh PK, Razonable RR. 95. Heymann A, Radtke F, Schiemann A, Lütz A,
Relationship of ganciclovir therapeutic drug monitor- MacGuill M, Wernecke KD, Spies C. Delayed treat-
ing with clinical efficacy and patient safety. Antimicrob ment of delirium increases mortality rate in intensive
Agents Chemother. 2019;63(3):e01855–18. care unit patients. J Int Med Res. 2010;38:1584–95.
82. Conti M, Matulli Cavedagna T, Ramazzotti E, Mancini 96. Bangash MN, Kong M-L, Pearse RM. Use of inotro-
R, Calza L, Rinaldi M, et al. Multiplexed therapeutic pes and vasopressor agents in critically ill patients. Br
drug monitoring (TDM) of antiviral drugs by LC– J Pharmacol. 2012;165(7):2015–33.
MS/MS. Clin Mass Spectrom. 2018;7(December 97. De Backer D, Biston P, Devriendt J, Madl C, Chochrad
2017):6–17. D, Aldecoa C, Brasseur A, Defrance P, Gottignies P,
83. Aouri M, Moradpour D, Cavassini M, Mercier Jean-Louis V, SOAP II Investigators. Comparison
T, Buclin T, Csajka C, et al. Multiplex liquid of dopamine and norepinephrine in the treatment of
chromatography-­tandem mass spectrometry assay for shock. N Engl J Med. 2010;362(9):779–89.
simultaneous therapeutic drug monitoring of ribavi- 98. Khuenl-Brady KS, Reitstätter B, Schlager A,
rin, boceprevir, and telaprevir. Antimicrob Agents Schreithofer D, Luger T, Seyr M, Mutz N, Agoston
Chemother. 2013;57(7):3147–58. S. Long-term administration of pancuronium and
84. Chan AHW, Partovi N, Ensom MHH. The utility of pipecuronium in the intensive care unit. Anesth
therapeutic drug monitoring for ribavirin in patients Analg. 1994;78(6):1082–6.
with chronic hepatitis C – a critical review. Ann 99. Murray MJ, Deblock H, Erstad B, Gray A, Jacobi J,
Pharmacother. 2009;43(12):2044–63. Jordan C, et al. Clinical practice guidelines for sus-
85. Breadmore MC, Theurillat R, Thormann tained neuromuscular blockade in the adult critically
W. Determination of ribavirin in human serum and ill patient. Crit Care Med. 2016;44:2079–103.
Quality Assurance of Samples
for Therapeutic Drug Monitoring
10
and Clinical Toxicology

Samuel O. Bekoe, Samuel Asare-Nkansah,


and Kwabena F. M. Opuni

Abstract emphasis that the assurance of the quality of


samples meant for TDM and CT assessments
The integrity of samples employed for thera- correlates strongly with the quality of data
peutic drug monitoring (TDM) and clinical obtained for clinical interpretation and imple-
toxicology (CT) has a significant impact mentation of optimized care. This chapter thus
on the quality of data obtained for clinical addresses major concerns, regarding the qual-
decision-making. Samples usually employed ity assurance of samples for TDM and CT.
for TDM and/or CT are obtained either using
invasive or non-invasive approaches, and these Keywords
include blood, plasma, or serum (invasive
approaches), urine, and saliva (non-invasive Bioanalysis · Biological samples · Clinical
approaches). Lack of standard operating pro- toxicology · Quality assurance · Therapeutic
cedures for samples in the pre-analytical stage drug monitoring
could lead to variabilities in analysis results
because of factors such as haemolysis (blood
samples), inappropriate cooling and storage 10.1 Introduction
(urine samples), and desiccation of saliva.
Therefore, assuring the quality and integrity Measurement uncertainties and variabilities are
of samples obtained for TDM and CT studies usually associated with TDM and CT studies.
is a requirement for reliable data suitable for These challenges could result in the generation of
informed clinical decisions for patients as well unreliable data, which could affect the clinical
as medical emergencies. It is an irrefutable decision-making required for optimal patient
therapy ultimately. Several factors could account
for such variabilities, but key amongst them is the
S. O. Bekoe · S. Asare-Nkansah (*) integrity of the sample meant for the TDM and
Department of Pharmaceutical Chemistry, Faculty of CT studies. The nature, integrity, and handling of
Pharmacy and Pharmaceutical Sciences, Kwame
Nkrumah University of Science and Technology, TDM and CT samples strongly influence the
Kumasi, Ghana quality of analytical data that can be generated
e-mail: sankansah.pharm@knust.edu.gh from such samples for the purpose of critical
K. F. M. Opuni (*) healthcare decisions. Thus, certain key variables
Department of Pharmaceutical Chemistry, School of such as sample acquisition and handling (before,
Pharmacy, University of Ghana, Legon, Ghana during, and after analysis) and sample prepara-
e-mail: kfopuni@ug.edu.gh

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 161
S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_10
162 S. O. Bekoe et al.

tion and storage are to be considered carefully serum, or plasma, while capillary blood speci-
before any TDM and CT studies are carried out mens are collected for babies. In the collection
[1–6]. Other important factors include calibration of blood samples, drug levels are allowed to
of equipment, temperature monitoring of freez- reach steady state, which can be predicted by at
ers, and data management. In a typical workflow, least five half-lives of the specific pharmacody-
the factors that impact the quality of samples for namic agent. However, in collecting samples
TDM and CT are as indicated in Table 10.1 which for drugs with long half-­ lives (e.g. amioda-
will be respectively discussed in the ensuing rone), samples are taken before steady state is
sections. achieved in order to prevent toxicity in patients
suffering from renal and metabolic impair-
ments. It must also be noted that drugs such as
10.2 Sample Acquisition, cyclosporin A require whole blood to be sam-
Handling, and Storage pled because the drug partitions itself between
the red blood cells and plasma [7]. Therefore,
Given the importance of the pre-analytical phase the pharmacokinetic and pharmacodynamic
(sample acquisition, handling, and storage) of properties of a drug are important to consider
TDM and CT projects, a well-developed and for a standard sample acquisition.
tested plan/protocols, and standard operating In essence, standard operating procedures
procedures, that encompass sampling, handling, for the collection of samples (blood, serum,
and storage of samples must be followed (opti- plasma, urine, and saliva) for TDM or CT stud-
mized and standardized best practices). This ulti- ies should require the availability of appropri-
mately ensures that the integrity of biological ate storage facilities, equipment, and qualified
specimens is maintained in order to secure qual- personnel, amongst others. Samples, be it
ity analytical data needed for optimum clinical blood, serum, plasma, urine, or saliva (in cer-
decision for patients. Usually, laboratories tain instances), should be obtained using well-
responsible for the analytical phase of TDM or established and approved techniques in
CT undergo proficiency testing and ultimately appropriate containers. For instance, it is not
get certified for their purpose. acceptable to use lithium heparin as an antico-
In TDM, for example, drug levels in adults agulant for blood samples meant for lithium
are usually profiled in whole venous blood, analysis. Again, in the monitoring of certain
drugs such as phenytoin, some types of gel sep-
arator tubes are not suitable [8] because of the
Table 10.1 Workflow for therapeutic drug monitoring potential diffusion of drugs into the gel from
and clinical toxicology cycle
serum. Guided by best practices, samples for
General sequence of technical activities for TDM or CT should be collected to avoid
therapeutic drug monitoring and clinical toxicology
unwanted contamination and losses which
1 Study design
2 Informed consent
could affect the quality of analytical data gener-
3 Sample collection protocola ated. It is also important to have protocols and
4 Sample collectiona standard operating procedures (SOPs) fol-
5 Sample annotationa lowed, supervised, and approved by qualified
6 Sample labellinga personnel to ensure the quality of the various
7 Sample storagea stages involved in specimen acquisition. In sit-
8 Data recordsa
uations where specimens must be transported to
9 Sample transportationa
10 Sample analysis (using analytical methods)a
another site for laboratory testing, appropriate
11 Data analysis and validated sample transport and handling
12 Reporting systems must be ensured. Samples that are not
a
Activities that impact the quality assurance of samples analysed immediately must be stored in appro-
for therapeutic drug monitoring and clinical toxicology priate containers and environments (e.g. tem-
10 Quality Assurance of Samples for Therapeutic Drug Monitoring and Clinical Toxicology 163

perature and light) to ensure sample integrity is ment should be regularly checked to avoid any
maintained [1, 9–14]. form of instrumental effects that could result in
measurement uncertainties and significant vari-
abilities in data [3, 11, 13].
10.3 Sample Preparation
and Validation Techniques
10.5 Automation of Testing
The stability of sample matrices and drug ana- Laboratory
lytes is essential to be considered in TDM or
CT, as these candidates can sometimes be sus- The use and application of advanced technology
ceptible to degradation. Sample degradation involving appropriate statistical tools will be an
may be observed either at the pre-analytical essential tool required for the assurance of qual-
stage or during the analytical phase through ity in TDM and CT. Such tools help to provide
visual inspection or instrumental analysis. valuable information and also monitor the reli-
Therefore, comprehensive and validated proce- ability and reproducibility of analytical data. The
dures should be developed and be made acces- replicate determinations on samples are a key
sible for easy identification of bio/chemical requirement that should be considered for TDM
degradation of samples. Thus, separation meth- and CT, and having such automation could pro-
ods or procedures should be designed to ensure vide the needed data. Moreover, batch-to-batch
that possible interferences from the matrix being in-process controls are ensured to establish sound
studied or degraded or metabolic products from scientific patterns amongst samples analysed.
the analyte of interest are significantly avoided Such tools also do help in preventing analysts or
or minimized. For this purpose, the quality of laboratory/clinical scientists from making any
solvents and reagents should be routinely form of alteration to data obtained [1, 3, 11, 13].
checked, paying particular attention to storage
conditions and expiry dates. The maintenance,
calibration, and cleaning of equipment such as 10.6 Conclusion and Outlook
centrifuges should be routinely done, notwith-
standing the availability of appropriate glass- The integrity of samples for TDM and CT are
ware, plasticware, and consumables. Where assured when appropriate sample quality man-
necessary, the use and application of preserva- agement approaches are used [15]. More impor-
tives should be well documented since com- tantly, invasive approaches to sampling of
pounds employed as preservatives could blood, plasma, or serum can be minimized using
negatively influence analytical data generated. microsampling techniques which are less inva-
Internal standards could also be considered to sive. Emerging microsampling techniques such
help minimize random errors associated with as dried blood spot, plasma microsampling, vol-
both extraction procedures, sample dilution, and umetric absorptive microsampling, and
instrumental effects [3, 10–14]. microneedle have been implemented success-
fully for sample collection [16]. Also, dried
blood and serum spots have proven an important
10.4 Sample Testing or Analysis technique for sample storage and transportation
especially in resource constraint countries [17].
It must be ensured that appropriate sensitive ana- Although the suggested strategies have some
lytical techniques that have been well validated challenges, optimization and improvement in
are used for the testing of samples. The calibra- these techniques can improve sample integrity
tion of equipment used for the method develop- prior to analysis.
164 S. O. Bekoe et al.

References 9. Delanghe J, Speeckaert M. Preanalytical require-


ments of urinalysis. Biochem Med (Zagreb).
2014;24(1):89–104.
1. Guidelines for Forensic Science Laboratories. http://
10. Lippi G, von Meyer A, Cadamuro J, Simundic
www.sadcmet.org/SADCWaterLab/Archived_
AM. Blood sample quality. Diagnosis (Berl).
Reports/2006%20Reports%20and%20Docs/Ilac-­g19.
2019;6(1):25–31.
pdf. Accessed on 14 Apr 2022.
11. Moffat AC, Osselton MD, Widdop B, Watts
2. Summary of the responses received for the forensic
J. Clarke’s analysis of drugs and poisons. London:
Science regulator’s consultation paper on “A review
Pharmaceutical Press; 2011.
of the options for the accreditation of forensic practi-
12. Peplies J, Fraterman A, Scott R, Russo P, Bammann
tioners”. https://assets.publishing.service.gov.uk/gov-
K. Quality management for the collection of biologi-
ernment/uploads/system/uploads/attachment_data/
cal samples in multicentre studies. Eur J Epidemiol.
file/117558/practitioner-­reg-­summary.pdf. Accessed
2010;25(9):607–17.
on 14 Apr 2022.
13. ISO. ISO/IEC 17025: 1999: general requirements
3. Bush DM. The U.S. mandatory guidelines for fed-
for the competence of testing and calibration labo-
eral workplace drug testing programs: current sta-
ratories. Geneva: International Organization for
tus and future considerations. Forensic Sci Int.
Standardization; 1999.
2008;174(2–3):111–9.
14. Vargas RJ, Cobar OM. The urgent need for manage-
4. Ellison SL, Williams A. Quantifying uncertainty in
ment of biological samples and data accessibility in
analytical measurement. London: EURACHEM;
Latin America. Front Pharmacol. 2021;12:620043.
2012.
15. Redrup MJ, Igarashi H, Schaefgen J, et al. Sample
5. Ellison SLR. Implementing measurement uncer-
management: recommendation for best practices
tainty for analytical chemistry: the Eurachem
and harmonization from the Global Bioanalysis
Guide for measurement uncertainty. Metrologia.
Consortium Harmonization Team. AAPS J.
2014;51(4):S199–205.
2016;18(2):290–3.
6. Fraser J, Williams R. Handbook of forensic science.
16. Lei BUW, Prow TW. A review of microsampling tech-
New York: Routledge; 2013.
niques and their social impact. Biomed Microdevices.
7. Gross AS. Best practice in therapeutic drug monitor-
2019;21(4):81.
ing. Br J Clin Pharmacol. 1998;46(2):95–9.
17. Mercatali L, Serra P, Miserocchi G, et al. Dried
8. Quattrocchi F, Karnes HT, Robinson JD, Hendeles
blood and serum spots as a useful tool for sample
L. Effect of serum separator blood collection
storage to evaluate cancer biomarkers. J Vis Exp.
tubes on drug concentrations. Ther Drug Monit.
2018;(136):57113.
1983;5(3):359–62.
Therapeutic Drug Monitoring
and Toxicology of Anticancer
11
Drugs

Seema Kohli and Lavakesh Kumar Omray

Abstract commonly employed for chemotherapeutic


agents. However, a number of antineoplastic
The successful management of disease is agents may have narrow therapeutic indices.
completely dependent on drug dosage regi- As such, it is very difficult to provide right
men strategies. Appropriate dosage regimen individualized treatment for cancer chemo-
helps to achieve optimum level of a drug at the therapy. Despite several limitations of TDM
receptor site in order to obtain optimum thera- for cytotoxic drugs, several clinical trials of
peutic response with minimal adverse reac- anticancer drugs have shown the benefits of
tions/events. But there always occurs maximum chemotherapy using TDM proto-
inter-individual variation toward drug cols. The chapter details TDM applications for
response, which can be attributed to individu- the therapeutic efficacy and toxicological
al’s pharmacokinetics/pharmacodynamics implications of various anticancer drugs.
(PK/PD) characteristics. Therefore, there is a
need for clinical evaluation and therapeutic Keywords
monitoring of drug. Drugs with narrow thera-
peutic indices such as digoxin, aminoglyco- Therapeutic index · Individualized drug
sides, antiarrhythmics, anticonvulsants, and therapy · Drug monitoring · Anticancer drugs
anti-asthmatic agents need strict individual-
ization of drug regimen. The dose of these
drugs is carefully individualized to avoid fluc- 11.1 Introduction
tuations in plasma drug concentration. Often,
the individualization of dose and adjustment The successful management of disease is com-
depends on the toxicity of the drug and the pletely dependent on the drug dosage regimen
patient’s ability to tolerate the drug. strategies. Additionally, appropriate dosage regi-
Therapeutic drug monitoring (TDM) is not men helps to achieve optimum level of drug at the
receptor site in order to obtain optimum thera-
peutic response with minimal adverse reactions/
S. Kohli (*)
events. There always occurs inter-individual vari-
Department of Pharmaceutical Sciences, Kalaniketan
Polytechnic College, Jabalpur, Madhya Pradesh, India ation toward drug response, which can be attrib-
uted to individual’s pharmacokinetics/
L. K. Omray
Radharaman Radharaman Institute of Pharmaceutical pharmacodynamics (PK/PD). Therefore, there is
Sciences, Bhopal, Madhya Pradesh, India a substantial need for clinical evaluation and

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 165
S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_11
166 S. Kohli and L. K. Omray

therapeutic monitoring of drugs. It is important to dose and adjustment depends on the toxicity of
remember some basic aspects of treating patients the drug and the patient’s ability to tolerate the
and using medicines before identifying drugs that drug.
require monitoring. For most medicines, the ben-
efits could be easily observed and measured, so it
is possible to know if the administered dose is 11.2 Therapeutic Drug
appropriate for that patient or if some dose adjust- Monitoring (TDM)
ments are required either to avoid toxic effects or
to obtain the desired benefit. The course of measurement of the amount of
For most medicines, routine monitoring is not drug or its metabolism product in blood at a
recommended. Only clinically relevant testing defined period is expressed as therapeutic drug
should be carried out. Due to the fact that drug monitoring. Some of the drugs are being evalu-
assays are expensive, it’s important to think about ated to have a low “therapeutic index.”
why you’re monitoring and what new informa- Therapeutic index is described as the proportion
tion you’ll get (if any). As a result, in a clinically of the toxic (lethal) and therapeutic (effective)
stable patient, routine monitoring of many medi- doses of medication.
cines is not required [1].
LD50
Therapeutic Index =
TD50
11.1.1 Individualization of Drug In order to maintain the drug’s effective concen-
Therapy tration in the body, a patient needs to be adminis-
tered a dose of drug at consistent interludes.
In the past, drugs were used in similar doses for Maintaining a constant state for some medica-
almost every patient. This theory is known as flat tions is more difficult than just administering a
dosing. But the pharmacokinetic/pharmacody- normal dose. Drugs are absorbed, metabolized,
namic parameters and individual’s variation, viz., utilized, and eliminated at varied rates depending
body weight and surface area, result in variations in on the person’s age, general health, and genetic
drug responses. Individualizing of dosage regimen composition. The concentration in circulation
is not required for all drugs. Drugs having wide eventually yields a pharmacological response
margin of safety with wide therapeutic window do (Fig. 11.1).
not need strict individualization of therapy. The TDM approach involves monitoring the
Drugs with narrow therapeutic window such incidence and intensity of both the desired
as digoxin, aminoglycosides, antiarrhythmics, therapeutic and adverse drug effects by mea-
anticonvulsants, and anti-asthmatics need strict suring the average plasma drug concentration.
individualization of drug regimen. The objec- The therapeutic range of a drug is a probability
tive of dosage design for the abovementioned concept, and care should be taken toward pre-
category of drugs is to produce a maximum scribing/administering drugs to a patient in
therapeutic response with minimal adversities that range. While administering drugs to the
or both. The dose of these drugs is carefully patient, the drug plasma level must be main-
individualized to avoid fluctuations in plasma tained within a narrow range of the therapeutic
drug concentration. Pharmacokinetic and phar- window. The dose of a drug is frequently
macodynamic monitoring is advocated for drugs accustomed as per the body weight/body sur-
having fluctuations in plasma concentration lev- face of the individual and the PK/PD of the
els. In concern therapy, the individualization of drug [2].
11 Therapeutic Drug Monitoring and Toxicology of Anticancer Drugs 167

Fig. 11.1 Drug dose and response

11.2.1 Criteria for TDM • Drugs taken as a preventative measure to avert


convulsions, arrhythmias related to the heart,
• Drugs having low therapeutic index. CNS depression, events of mania, asthma
• Direct correlation between drug and its metab- reversions, or refusal of organs
olite’s plasma level and the pharmacological • Avoidance of drug’s major toxic effects. For
or toxic effects. example, aminoglycoside antibiotics, which
• Individual variability in drug plasma concen- have a narrow therapeutic range as compared
tration profile on single dosing. to other antibiotics
• Assessment of therapeutic effect cannot be
made clinically. The apposite suggestions for TDM are as fol-
• Suitable analytical techniques not available lows [1]:
for estimation of drug and its metabolite.
• Toxicity: When the clinical pattern is not sim-
ple to identify, it’s difficult to diagnose toxic-
11.2.2 Features of Drug for Apt ity as the case of inexplicable nausea in a
Candidature for TDM person on digoxin therapy.
• Dosing: Adjustment of dose after attaining
• Drugs with significant pharmacokinetic steady levels; evaluation of loading dose, e.g.,
variations. on commencement of phenytoin therapy;
• Drugs having dose-associated healing and prognosis of drug dosing to estimate patient
adverse effects. dose requirement, e.g., aminoglycosides.
• Drugs with low or narrow therapeutic index. • Monitoring: Evaluating acquiescence; diag-
• Drugs having distinct targeted therapeutic nosing under treatment; and futile treatment.
range.
• Drug monitoring is problematic. The fact that TDM is the measurement of
medication concentrations in plasma or blood to
If the clinical efficacy of a drug can be assessed aid patient management is an important compo-
by simply as the heart rate/ blood pressure, the nent of TDM that should be addressed. This
dose may be adjusted based on response. If this is means that the laboratory-assisted medication
not feasible, then therapeutic drug monitoring is concentration levels are evaluated to personalize
used in the following situation [3]: and optimize dose schedule of the patient and
168 S. Kohli and L. K. Omray

Fig. 11.2 Process of TDM

treatment results using drug dose in a precise help in individualizing the drug dosing. The eval-
therapeutic window [4]. Figure 11.2 shows the uation of liver and kidney functions, as well as
process of dosage regimen adjustment using the monitoring of therapeutic drugs, may assist in
TDM approach. determination of body’s ability to metabolize and
eliminate therapeutic drugs. Several categories of
drugs which need monitoring are shown in
11.3 Therapeutic Drug Table 11.1.
Monitoring for Various
Drugs
11.4 Importance of Clinical
It is imperative to express that various drugs Pharmacokinetic Parameters
which need intense observations during treat- in Context to Anticancer
ment have been used chronically. These monitor- Agents
ing must be well maintained at steady
concentrations even for years. Pregnancies, The investigation of the correlation between drug
infections, transient diseases, physical and men- dosage regimens and drug concentration-time
tal stressors, accidents, and surgeries are just a profiles is known as clinical pharmacokinetics
few examples of life events that can change a per- that is the study of absorption, distribution,
son’s therapeutic level. Over time, the patient metabolism, and elimination (ADME) status of
may develop other chronic disorders that neces- drugs. The parameters usually investigated are as
sitate lifelong medicine, which could interfere follows:
with the monitoring of drugs. Kidney disorder,
heart problems, liver disease, thyroid disorders, • First-order elimination rate constant (ke)
and HIV are just a few examples. These fluctua- • Clearance (C)
tions are traced by therapeutic drug monitoring • Volume of distribution (Vd)
and allow them to be accommodated. This would • Elimination half-life (t1/2)
11 Therapeutic Drug Monitoring and Toxicology of Anticancer Drugs 169

Table 11.1 Details of drugs that need monitoring


Drug category Drugs Treatment used
Immunosuppressant Tacrolimus, cyclosporine, azathioprine, sirolimus, Inhibit refusal of transplanted organs
mycophenolate mofetil and autoimmune ailments
Anticancer drug Methotrexate and all cytotoxic agents Psoriasis, rheumatoid arthritis,
various cancers, non-Hodgkin
lymphoma, osteosarcoma
Psychiatric drugs Lithium, valproic acid, some antidepressants Bipolar disorder (manic depression),
(imipramine, amitriptyline, nortriptyline, doxepin, depression
desipramine)
Cardiac drugs Digoxin, digitoxin, amiodarone, lidocaine, quinidine, Congestive heart failure, angina,
procainamide, N-acetylprocainamide (a metabolite of arrhythmias
procainamide)
Antibiotics Aminoglycosides (gentamicin, tobramycin, amikacin), Resistant cases of bacterial infections
vancomycin, chloramphenicol
Antiepileptics Phenobarbital, phenytoin, valproic acid, Protection from epilepsy and seizures
carbamazepine, ethosuximide, and other antiepileptic and also as stabilizer of moods
drugs
Bronchodilators Theophylline, caffeine Asthma, chronic obstructive
pulmonary disease (COPD), neonatal
apnea

Precision dosing or personalized dosing in a The utility of clinical pharmacokinetics must


patient is made easier using these criteria. be assessed in terms of patient results, such as
Anticancer drug dosing has traditionally been whether or not to include dose-related toxicity,
based on body surface area (BSA), with the and how long it takes to reach a conclusion, tak-
assumption that there is a link between BSA and ing into consideration the most essential vari-
clearance (CL) or volume of distribution (Vd). ables and deciding on the most favorable
However, in many case, this relationship is not concentration-effect connection [5].
accurate and does not present factual reasons for
variation in drug responses in population [4, 5].
This happens when there is drug-to-drug interac- 11.5 Methods
tion, hepatic/renal impairment and in the case of for Individualization of Drug
geriatric/pediatric patients. These factors lead to Therapy
a wide range of therapeutic practices, making it
difficult for doctors to make decisions based on The methods employed for individualization of
their prior experience. Patients with several drug therapy are as follows:
comorbidities/co-medications may receive insuf-
ficient pharmacotherapy which could result in • A priori method: A priori approaches deter-
undesirable intensities of toxicity or effective- mine the dose required to achieve a desired
ness. There are several sources for inter-­ exposure in a given patient based on morpho-
individual/intra-individual variations in PKs of logical, biological, and physiological factors
anticancer drugs, viz.: such as body weight, age, gender, serum cre-
atinine level, and glomerular filtration rate
• Age and age-related alterations (GFR).
• Obesity • A test dose method: This method comprises
• Concomitant medications following:
• Pharmacogenomics –– In the first step, specific pharmacokinetic
• Renal/hepatic impairment characteristics are assessed after a bolus
• Alcohol consumption injection of a moderate dose is adminis-
• Racial groups tered using numerous blood samples.
170 S. Kohli and L. K. Omray

–– This step comprises the use of PK parame- tory concentrations of drug for their target sites
ters to estimate the complete dose required only. It is useful for treatment-limiting side
to accomplish the target revelation. effects also [8].
• A posteriori method: In this nomogram, mul- The therapeutic index of cytotoxic anticancer
tiple linear regression (MLR) and maximum a medicines is limited, and they have a highly toxic
posteriori (MAP) Bayesian method are applied property and a significant level of interpatient
[6]. pharmacokinetic changeability. These are the
most important characteristics that support
TDM’s usefulness [9]. However, these features
11.6 Therapeutic Drug are often not sufficient in some case where TDM
Monitoring Approach has been employed in the treatment of cancer
of Anticancer Drug patients in normal practice. Thus, currently the
role and importance of TDM for the patient care
High intersubject variance, a narrow therapeutic in cancer treatment are limited [10].
index, minimal inter-occasion variation, and a
robust relationship between concentrations of
drug in plasma and clinical effects of the treat- 11.6.2 Limitations of TDM
ment are all characteristics of successful thera- for Anticancer Drugs
peutic drug monitoring. Process, methodological,
funding, logistical, social, and religion barriers Therapeutic drug monitoring approach is not
also contribute in the challenges for the success- commonly employed for the administration of
ful implementation of right methods to individu- chemotherapeutic drugs. Antineoplastic agents
alized drug treatment. have narrow therapeutic indices. As such, it is
very difficult to provide precise individualized
treatment for cancer chemotherapy. The clinical
11.6.1 Importance of TDM value of TDM for chemotherapeutic agents is
in the Management of Cancer presently constrained by several factors [11–13].
Limitations for the use of TDM are given below:
The international association of TDM and clini-
cal toxicology defines that TDM is a multidisci- 1. An incomplete knowledge of the pharmaco-
plinary approach in the treatment of cancer, kinetics and pharmacodynamics of most
which is aimed at improving patient treatment antineoplastic therapeutic agents.
and care. On the basis of clinical experience, the 2. Blood concentration of an anticancer drug is
dose of the medicine is adjusted individually. an incidental measurement of the amount of
Clinical studies have proven that treatments drug present in the objected tissue, as the site
based on clinical trials improved outcomes in the of action of the drug may be distant from
target populations suffering from cancer disease. intravascular spaces. Therefore, it is difficult
It can be based on a priori, posteriori and/or bio- to correlate and quantify the drug at the site
markers effective measurement and measurement of action.
of concentrations of drugs in plasma [7]. 3. In cancer chemotherapy, there is a large lag
Targeting of anticancer drugs is intended for period between the assessment of drug quan-
action on explicit recognized tumor substrate in tity in biological tissues and variation in
cells in a biopsy from an individual patient which efficacy.
is generally protein. These proteins are tremen- 4. Cancer is summarized as a group of hetero-
dous substances for TDM. The anticancer drugs geneous complex disease. It is having inher-
are administered by patient through suitable ent features between the concentration and
route, i.e., generally parenteral or oral adminis- effect relationship for antineoplastic drugs.
tration. This approach offers appropriate inhibi- This affects the activity of drugs. Anticancer
11 Therapeutic Drug Monitoring and Toxicology of Anticancer Drugs 171

medicines frequently exhibit variability in 10. TDM is costly in comparison with conven-
blood supply and cellular characteristics, tional therapy, is time consuming, and
resulting in varying levels of susceptibility, required technical field force which is diffi-
tolerance, and resistance [14]. In this condi- cult to manage all the time.
tion, only few antineoplastic drugs fulfill the
entire criterion required for the implementa-
tion of TDM, since there aren’t any well-­ 11.7 Beneficial Aspects of TDM
defined correlations between therapeutic for Cancer Therapy
efficacy and systemic exposure. Such rela-
tionships have been demonstrated by few Many drugs need special attention during the
drugs in particular cancer disease only. For therapy due to its narrow therapeutic window.
example, methotrexate is used as a TDM Among these, the anticancer drugs have major
approach in acute lymphoblastic leukemia concerns for successful treatment [15]. TDM
(B lineage), and 5-fluorouracil (5-FU) is refers to the need to treat cancer patients with the
used as the right TDM approach in metasta- smallest dose possible in order to achieve the
sis colorectal [12]. required concentrations in the upper end of the
5. Solid tumors present in any part of the body therapeutic window’s nontoxic range, increasing
may have their own distinctive source of the likelihood of drug response.
blood and affect the concentration of drugs. Many anticancer drug agents satisfy the cri-
This results in poor relationship between teria for the selection of TDM in the treatment
plasma and tumor concentration of the drug. of cancer. Variability of pharmacokinetics and
6. There is a natural holdup period between the narrow therapeutic indices of antineoplastic
measurement of drug concentration in agents makes it a suitable candidate for TDM.
plasma and the evaluation of the drug’s final In addition to this, while treating patients of
pharmacodynamic effect, and hence it is dif- cancer disease, seeking the highest efficacy of
ficult to establish a correlation between drug is an important principle because of the
them. magnitude of the consequences for cancer
7. If the outcome variable in TDM therapy is an patients. Physician cannot take the risk of sub-
increase in cancer cure rates, then at least optimal therapy. It can greatly reduce the prob-
5 years of therapy follow-up is usually ability of treatment for curable cancer diseases.
required to accurately analyze the outcome On the other hand, side effects, such as myelo-
and establish the correct link. As a result, suppression, can be life-­threatening and may be
these trials take longer to complete and have related to higher than optimal therapy. It can
a more complicated treatment approach. also be addressed using TDM. Despite the vari-
This appears to be in contrast to research ous limitations facing the TDM approach for
conducted on medicines with faster effects, the treatment of antineoplastic agents, the pos-
such as antibiotics exemplified by clarithro- sibilities for development of cancer treatment
mycin, erythromycin, etc. are also available, provided TDM is properly
8. There is a difficulty in establishing a rela- accomplished. Some important benefits of anti-
tionship between concentration and effect cancer TDM are outlined below:
because anticancer drugs are given in combi-
nation. Therefore, it is very difficult and 1. TDM is useful in identifying the efficacy of
problematic to precisely define the pharma- anticancer agents that are being given either in
cokinetics and pharmacodynamics of each overdose or underdose in any given
individual drug for TDM approach. condition.
9. With combination therapy, it is also difficult 2. TDM in the treatment of cancer chemotherapy
to determine pharmacodynamics of drug provides added advantages apart from indi-
toxicity [15]. vidualizing drug therapy to improve patient
172 S. Kohli and L. K. Omray

compliance and effectiveness and avoid toxic- cancer for 5 years. The clearance (CL) of carbo-
ity of drugs. platin quantity in plasma is diligently interrelated
3. It is easy to correlate and treat the efficacy and with kidney functioning. It is employed to moni-
toxicity of combination antineoplastic drug tor the carboplatin dose to accomplish an antici-
therapy. pated target of AUC by using Calvert’s equation
4. TDM provides the possibility of dose modifi- as given below [19].
cation in patients with liver and kidney impair-
Calvertsequation : Dose  mg  =Area under the curve
ment [14].
of drug  AUC   mg / ml / min 
5. There are chances of minimization of patient
  Glomerular filtration rate  GFR   25 ml / min 
to patient pharmacokinetic variability [14].
6. TDM is useful in the detection of drug inter- The constant of 25 ml/min is used to account for
actions and drug-food interaction [16]. non-renal clearance. TDM of carboplatin is
7. TDM enhances patient compliance. mainly proposed for high-dose protocols [20].
8. Subsequently, therapeutic range of drugs in
the plasma expresses efficacious as well as
toxicity concentrations having massive clini- 11.7.2 Methotrexate
cal usefulness.
Methotrexate is known as antimetabolites
TDM approach for some anticancer drugs is dis- immune suppressant and used as an anticancer
cussed underneath. drug. It is employed in the management of certain
cancerous diseases, adult and childhood acute
lymphoblastic leukemia, severe psoriasis, and
11.7.1 Carboplatin autoimmune diseases like rheumatoid arthritis
[11–13]. Methotrexate exhibits its anticancer
Carboplatin, an antimetabolite, is mainly used in effect by competitively suppressing dihydrofo-
ovarian cancer. It is also used in pediatric cancers late reductase enzyme present inside the cell.
and other cancers including neuroblastoma and This type of enzyme is accountable for conver-
retinoblastoma [17]. Carboplatin shows dose sion of folates to tetrahydrofolate in the cell. The
dependent response up to a certain level, and reduced folate helps in the transfer of carbon
after that an increase in dose, the toxic effect of units.
the drug starts. In other words, it has a clear rela- Purine synthesis and the methylation of uracil
tion between plasma concentration and the phar- to thymine in DNA synthesis both utilize these
macological effect of drugs on the patient [18]. carbon units [21]. Leucovorin is an antidote (a
Mechanism of action and TDM approach of car- folate analogue) and used to avoid excessive
boplatin and other anticancer drugs are given in destruction of host normal cell. Leucovorin is a
Table 11.2. Small increments of drugs in AUC man-made material for dihydrofolate reductase,
result in increased antineoplastic activity of and it allows the renewal of thymine develop-
drugs. Due to narrow therapeutic index, further ment and the re-initiation of DNA production.
augmentations in carboplatin dose result in aug- Mechanism of action of methotrexate is shown in
mented toxic effect of drug. The foremost toxic Fig. 11.3.
effect of carboplatin in small increased dose is In effective doses, methotrexate has the
myelosuppression. Successful TDM concentra- potential for significant toxic effects. Patients
tion of carboplatin AUC is about 4–7 mg/min/ml being treated with methotrexate should be con-
recommended in cancer of the ovary. As a result, tinuously observed for their pharmacokinetic
even minor modifications in carboplatin dose and and ­ pharmacodynamic effects so that toxic
exposure can have a significant clinical impact. effects are detected promptly to give antidote or
Similarly, a reduction in the dose of carboplatin rescue therapy. Methotrexate is the only anti-
up to 10%, results in double the reversion rate of cancer drug whose dose and concentration are
11 Therapeutic Drug Monitoring and Toxicology of Anticancer Drugs 173

Table 11.2 List of anticancer drugs and their TDM approach


Drug Indication Mechanism of action TDM approach References
Carboplatin Ovarian cancer, pediatric Carboplatin activates and Concentration of carboplatin [18, 19]
cancers, neuroblastoma, generates reactive AUC kept between 4 and
and retinoblastoma complexes inside the cell 7 mg/min/ml recommended
that cause intra- and in ovarian cancer for
inter-strand cross-linking of optimum effect
DNA molecules
Methotrexate Childhood acute Competitively inhibiting High-dose methotrexate and [22, 23]
lymphoblastic leukemia, dihydrofolate reductase initiation of treatment with
non-Hodgkin lymphoma, fixed time interval and
and osteosarcoma tailored dose of folinic acid
rescue approach
13-cis-retinoic Neuroblastoma, skin and It inhibits ornithine Dose intensity, duration, and [26, 28]
acid ovarian cancer by decarboxylase, thereby plasma drug concentration
(Isotretinoin) increasing cell apoptosis decreasing polyamine
and cell growth inhibition synthesis
Busulfan Bone marrow stem cell Bifunctional alkylating The target concentration [29]
transplantation and agent of DNA. Alkylation range for optimum effect of
treatment of hematologic leads to breaks in the DNA busulfan AUC between 78
malignancies such as molecule as well as and 101 mg∙h/L
acute and chronic myeloid cross-linking of the DNA
leukemia, etc. strands
5-Fluorouracil Used for the treatment of 5-FU acts in several ways Administered as a continuous [30]
colorectal cancer, as an anticancer drug but infusion via an implanted
esophageal cancer, principally acts as a port system using a body
stomach cancer, thymidylate synthase surface area (BSA)-based
pancreatic cancer, breast inhibitor dose calculation
cancer, cervical cancer,
etc.
Mitotane Adrenocortical carcinoma Interfere with the Concentration-dependent [31]
peripheral metabolism of effect and achievement of
steroids and suppresses the steady-state concentration
adrenal cortex
Tamoxifen Used to treat breast Selective estrogen receptor Monitoring endoxifen plasma [32]
cancer modulator concentration
Imatinib Philadelphia Specific tyrosine kinase Establishing relationship [34]
chromosome-positive receptor inhibitor between dose and plasma
chronic myelogenous concentration
leukemia and
gastrointestinal stromal
tumors
Pazopanib Inhibition of spread of Kinase inhibitor inhibits Exposure-response [35]
cancer cells the vascular endothelial correlation specifies that
growth factor receptor, pazopanib would benefit
platelet-derived growth from pharmacokinetically
factor receptor, etc. guided dosing

regularly individualized based on TDM. Plasma function of cell restored by using leucovorin
concentration of methotrexate can be used to treatment. The timing of leucovorin introduc-
monitor the toxic effect. Higher concentration tion and dose is guided by the routine use of
of methotrexate therapy can be neutralized with TDM for high-dose methotrexate therapy.
leucovorin or folinic acid as rescue treatment. Leukemia, osteogenic sarcoma, non-Hodgkin
Therefore, in the treatment of methotrexate, ini- lymphoma, and breast and lung cancer have all
tially higher concentration is given and normal benefited by leucovorin rescue. After adminis-
174 S. Kohli and L. K. Omray

tration of methotrexate infusion, the patients 11.7.3 13-Cis-Retinoic Acid


having plasma concentrations above 10 μM
after 24 h, 1 μM after 48 h, and 0.1 μM after Among the retinoids, mainly 13-cis-retinoic acid
72 h are known to have threat of bone marrow exhibits pronounced potential in numerous can-
and gastric organs [22, 23]. cer diseases; however, severe skin toxicity may
Methotrexate is metabolized via two path- occur. Whether a cancer or a tumor is caused by
ways, 7-hydroxylation and polyglutamation as chemical, physical, or viral source, 13-cis-­
given in Fig. 11.3. Methotrexate and its metabo- retinoic acid is an effective anti-promoter. Anti-­
lites are active. Aldehyde oxidase catalyzed proliferative activity of 13-cis-retinoic acid has
7-hydroxylation and produced a less effective been validated in in vitro investigation. The
inhibitor of dihydrofolate reductase. This metab- mechanism of action of retinoic acid is presented
olite is responsible for nephrotoxicity which in Fig. 11.4. The results of various preneoplastic
occurs during high dose of methotrexate treat- and neoplastic lesions of tissue histology have
ment. Methotrexate and its metabolite 7-hydroxy been demonstrated and are being explored fur-
methotrexate convert to intracellular polygluta- ther for cancer prevention. The ADME of 13-cis-­
mate formation. These polyglutamate metabo- retinoic acid has been explored in the number of
lites are as potent as methotrexate and cancer patients. 13-cis-retinoic acid is employed
dihydrofolate reductase inhibitors. In both meth- for the management of cancer in children with
otrexate metabolic pathways, restoration of DNA diseases such as neuroblastoma having great risk
synthesis is required [24]. factor and increases a 3-year survival benefit up

Fig. 11.3 Mechanism Source of Folic Acid from Diet or Intestinal Flora
of action of
methotrexate and
leucovorin rescue. FH2,
dihydrofolate; FH4, Outside of Cell Folate Methotrexate
tetrahydrofolate; dTMP,
deoxythymidine
monophosphate; dUMP, Target Cell
deoxyuridine Folate Active Transport
Process
monophosphate

Dihydrofolate Methotrexate
Reductase
dTMP FH2 FH4

Dihydrofolate
Reductase

dUMP N5, N10–Methylene-FH4

Adenine
Gaunine
Leucovorin Thymidine
Rescue Methinone
Serine
11 Therapeutic Drug Monitoring and Toxicology of Anticancer Drugs 175

Fig. 11.4 Mechanism


of action of retinoic acid

to 46% improvement as compared to 29% in imum Cmax of 2 μM was attained in 90% of


other conditions [25]. The standard dosing proto- patients in the given protocol. The pharmacoki-
col of drug about 160 mg/m2/day for 2 weeks netic variability in patient was also considerably
treatment study, observed to have up to 20 times- lowered particularly in the pediatric patients suf-
variability in Cmax and AUC. The lack of clinical fering from neuroblastoma (<12 kg) who were
benefit with an alternate drug regimen of low and given dose of 5.33 mg/kg. The TDM protocol of
continuous dosing suggests that dose intensity 13-cis-retinoic acid is currently suggested in the
and plasma drug concentration are critical for high-risk neuroblastoma of European patients.
pharmacological therapeutic efficacy. Dose-­ However, further intensive studies for proper
dependent toxicity is also related with the amount implementation of TDM are required to get con-
of drug in plasma having more than 10 μM quan- firmed clinical benefit of the drug [28].
tity in the blood [26].
By stimulating cell differentiation, retinoic
acid as a medication aids in the transformation of 11.7.4 Busulfan
cell types from proliferative to maturation phage.
Retinol converts to retinal in the presence of reti- Busulfan is an anticancer drug having bifunc-
nol dehydrogenase (RDH), which changes to tional alkylating property of DNA in the target
retinoic acid in the presence of retinaldehyde cell. Alkylation leads to breakdown of the DNA
dehydrogenase (RALDH). The cellular retinoic molecule along with cross-linking of the strands
acid-binding protein (CRABP) site binds retinoic of DNA. It consequently leads to interloping of
acid in the cytoplasm, and in the cell nucleus, the DNA replication and RNA transcription. It is
receptors retinoic acid receptor (RAR) and reti- used in high doses as preparative chemothera-
noid X receptor (RXR) are involved in its bind- peutic cures in patients experiencing hemato-
ing. Retinoic acid comes in a variety of poietic stem cell transplantation (HSCT) for
biochemically active forms, each of which isom- patients with numerous cancerous conditions.
erizes under different physiological conditions. The oral dose of busulfan in cancer patients is
These isomeric forms of retinoic acid act on about 1 mg/kg body weight given four times
diverse receptors present in the cell for the daily or 3.2 mg/kg IV in continuous mode once
expression of gene response. In the nucleus, it is every day. Pharmacokinetic profile of busulfan
having mainly three steps, i.e., transcription, his- follows single compartment model which is dif-
tone modification, and DNA methylation through ficult to c­ orrelate concentration of drug in the
retinoic acid response element (RARE) [27]. target site. Oral absorption of busulfan is rapid
In a controlled dosing study of 13-cis-retinoic and maximum concentration achieved within
acid for TDM, it was found that the target of min- 1 h of administration of the drug. Oral bioavail-
176 S. Kohli and L. K. Omray

ability of busulfan is approximately 70–90% BSA-based dosing. However, results of BSA do


due to erratic absorption of the drug. This not link with any PK parameters in adult patients.
absorption is variable and associated with very A strong correlation has been established among
wide inter- and intra-individual unpredictable concentration, efficacy, and toxicity. The dose
intestinal absorption. These problems are modification of 5-FU is a viable and suggestively
resolved by developing an IV formulation of important technique for positive clinical results
busulfan, which has a more predictable pharma- by reducing toxicities and boosting efficacy of
cokinetic profile with less interpatient variabil- TDM therapy [30].
ity. TDM of busulfan is based on the right dose
selection which involves the administration and
gathering of some [4–7] blood samples at known 11.7.6 Mitotane
predetermined period. These are later employed
to estimate busulfan clearance of an individual Mitotane is used as an anticancer drug in some
and study pharmacodynamic effects. The effec- cases. It serves as a standard of drug to care
tive management of busulfan pharmacokinetic for the patient after completion of surgical
dose-based targeting reveals reduced rejection, resection of adrenocortical carcinoma. It is
relapse, discontinuity of drug, and death in useful in the treatment of cancer of the adre-
patients of HSCT receivers. However, pharma- nal gland that cannot be treated with surgical
cokinetic sampling of drugs has been a barrier procedure. It reduces the growth or size of the
to common acceptance of busulfan based on adrenal tumor. Mitotane interferes with the
dose targeting [29]. steroidal metabolism of peripheral parts and
suppresses adrenal cortex activity. It reduces
17-­hydroxycorticosteroids concentration in the
11.7.5 5-Fluorouracil (5-FU) absence of decreased corticosteroid level and
increases formation of 6β-hydroxycortisol. The
5-Fluorouracil is an antimetabolite given as a FDA recommends a starting dose of mitotane
parenteral dosage form to treat breast, colon, rec- from 2 to 6 mg daily in divided doses, with a
tum, stomach, and pancreatic cancer and as a gradual increase in dose from 9 to 10 grams per
cream to treat actinic keratosis (it is a skin condi- day. This increment is dependent on the patient
tion that may convert into cancer) and certain tolerance. The long half-life of mitotane occurs
other types of basal cell skin cancer. 5-FU inter- after the achievement of steady-state concen-
face with the combination of fluorouridine tri- trations which is possible after some weeks
phosphate (FUTP) into RNA strand, combination of treatment. The recommended therapeutic
of fluorodeoxyuridine triphosphate (FdUTP) into dose of mitotane is from 14 to 20 mg/L. This is
DNA strand and inhibition of thymidylate syn- based on the recommendation of the European
thase (TS) enzyme by fluorodeoxyuridine mono- Medicines Agency in the treatment of adrenal
phosphate (FdUMP), leads to consequential cancer [31].
DNA damage which finally results in apoptosis.
Figure 11.5 shows mechanism of action of
5-fluorouracil. 11.7.7 Tamoxifen
Therapy is based on measurement of body
surface area (BSA) and calculated administration Tamoxifen is an antiestrogen medication that
of a dose of 5-FU. This produces extensive devia- works by blocking estrogen’s effects in breast
tion of 5-FU systemic exposure in different cells and tissues. It is the most commonly pre-
patients that can be linked with the efficacy of the scribed selective estrogen receptor modulator
drug. The TDM of 5-FU administration is per- (SERM) for breast cancer treatment, and it has
sonalized for particular patients to the extent of been approved by the US Food and Drug
11 Therapeutic Drug Monitoring and Toxicology of Anticancer Drugs 177

Fig. 11.5 Mechanism


of action of 5-FU
5-fluorouracil

FdUMP FTUP

Inhibit thymidylate Mistakenly


Synthetase incorporate to RNA

dTMP Depletion Alteration of RNA processing


(required for DNA synthesis) and protein synthesis

Cell Apoptosis

Administration. It can help with estrogen hypereosinophilic syndrome, and other tumor con-
receptor-­positive breast cancer and its complica- ditions. The above conditions are noticeable by an
tions. Another agent mediates the activity of the unusual, constitutively articulated tyrosine kinase,
selective estrogen receptor modulator (tamoxi- and it results in nonregulated cell growth. Imatinib
fen); it is primarily influenced by significant and displays inter-individual variability in their phar-
more potent metabolites such as endoxifen. macokinetic. Greater body weight and increased
Endoxifen is largely processed by the hepatic apparent clearance of imatinib have both been
microsomal enzyme CYP2D6, which is an aber- linked to chronic myeloid leukemia. Decreased
rant enzyme. This is the problem of common apparent clearance is associated with renal impair-
genetic polymorphism in cancer patient. ment and patients on concomitant medications
Enzyme CYP2D6 is an indirect measure to with potent inhibition of cytochrome P450 3A4
predict the endoxifen plasma concentration. enzyme. Patients with Philadelphia chromosome-
Therefore, TDM of tamoxifen is based on the positive chronic myelogenous leukemia and gas-
monitoring of endoxifen plasma concentrations, trointestinal stromal tumors are treated for a long
and it has been advocated for individualizing period of time. As per available data, TDM for
tamoxifen therapy. The plasma concentration of imatinib may give more information on efficacy,
the metabolite endoxifen can be increased by compliance, and safety than clinical evaluation
raising the tamoxifen dose without causing sub- alone [33]. The substantial variability in the rela-
stantial adverse effects such as hot flushes or tionship between medication dose and concentra-
severe gastrointestinal discomfort and regardless tion is due to variations in CYP3A4 enzyme
of the patient’s CYP2D6 genetic problem [32]. activity, which mediates the major metabolic route
and controls both efflux from the gut wall and
active secretion in the bile [34].
11.7.8 Imatinib

Imatinib is a monoclonal antibody. It serves as a 11.7.9 Pazopanib


specific tyrosine kinase receptor inhibitor
employed in the treatment of different types of leu- Pazopanib is another monoclonal antibody anti-
kemia, myelodysplastic disease, systemic masto- neoplastic agent; it is a kinase inhibitor type of
cytosis, dermatofibrosarcoma protuberans, anticancer drug. Pazopanib is used for stopping or
178 S. Kohli and L. K. Omray

slowing the spread of cancer cells in the affected References


organ. Pazopanib is used in the treatment of
advanced renal cell carcinoma (RCC, a type of 1. Ghiculescu RA. Therapeutic drug monitoring: which
drugs, why, when and how to do it. Aust Prescr.
cancer that begins in the cells of the kidneys) in 2008;31:42–1.
adults’ cancer patient. The vascular endothelial 2. Darwich AS, Ogungbenro K, Hatley OJ, et al. Role
growth factor receptor (VEGFR) is inhibited by of pharmacokinetic modeling and simulation in preci-
pazopanib. It also inhibits other receptors like sion dosing of anticancer drugs. Transl Cancer Res.
2017;6(Suppl 10):S1512–29.
platelet-derived growth factor (PDGF), fibroblast 3. Birkett DJ. Therapeutic drug monitoring. Aust Prescr.
growth factor (FGF), and stem cell (c-Kit). 1997;20:9–11.
Pazopanib similar to other tyrosine kinase inhibi- 4. Kang JS, Lee M-H. Overview of therapeutic drug
tors also has substantiation of interpatient variabil- monitoring. Korean J Intern Med. 2009;24:1–1.
5. Salman B, Al-Khaboori M. Applications and chal-
ity in pharmacokinetic profile resulting in lenges in therapeutic drug monitoring of cancer treat-
increased risk of suboptimal dosing when given in ment: a review. J Oncol Pharm Pract. 2020;0(0):1–9.
empirical fixed dose schedules of drugs in conven- 6. Alnaim L. Therapeutic drug monitoring of cancer
tional form. chemotherapy. J Oncol Pharm Pract. 2007;13:207–21.
7. IATDMCT. Definitions of TDM and CT. 2013.
A study assessing the efficacy of pazopanib in Available online: https://www.iatdmct.org/about-­us/
renal cell carcinoma found a clear link between about-­association/about-­definitions-­tdm-­ct.html.
plasma concentration and medication outcome, 8. Fujita K, Ishida H, Kubota Y, et al. Toxicities of recep-
with patients who achieved a steady-state trough tor tyrosine kinase inhibitors in cancer pharmacother-
apy: management with clinical pharmacology. Curr
concentration of pazopanib above 20.5 mg/L hav- Drug Metab. 2017;18:186–98.
ing a longer response and better tumor reduction. 9. Decosterd LA, Widmer N, Zaman K, et al. Therapeutic
However, greater pazopanib plasma concentra- drug monitoring of targeted anticancer therapy.
tions were linked to several side events, including Biomark Med. 2015;9:887–93.
10. Bardin C, Veal G, Paci A, et al. Therapeutic drug
diarrhea, hypertension, an increase in hepatic monitoring in cancer- are we missing a trick? Eur J
enzymes, and stomatitis. These issues can be Cancer. 2014;50:2005–9.
managed if supportive medicine is administered 11. Milano G, Etienne MC, Renee N, et al. Relationship
concurrently. Pazopanib would benefit from a between fluorouracil systemic exposure and
tumor response and patient survival. J Clin Oncol.
pharmacokinetically guided dosage approach due 1994;15:1291–5.
to its low safety margin, variable PK, and indica- 12. Joel S, O’Byrne Penson K, Papamichael R, et al.
tions of an exposure-response link [35, 36]. A randomized concentration-controlled compari-
son of Standard (5-day) vs. 260 A. Rousseau and
P. Marquet prolonged (15-day) infusions of etopo-
side phosphate in small cell lung cancer. Ann Oncol.
11.8 Final Remark 1998;11:1205–11.
13. Santini J, Milano G, Thyss A, et al. 5-FU thera-
Cancer is a major concern worldwide. The use of peutic monitoring with dose adjustment leads to an
improved therapeutic index in head and neck cancer.
various techniques of TDM in oncology is a Br J Cancer. 1989;59:287–90.
debatable issue as evident from the above discus- 14. Pignon T, Lacarelle B, Duffaud F, et al.
sions. The use of anticancer drugs is associated Pharmacokinetics of high-dose methotrexate in adult
with severe side effects (mainly because some osteogenic sarcoma. Cancer Chemother Pharmacol.
1994;33:420–4.
have narrow therapeutic indices), and often 15. Chatelut E, Pivot X, Otto J, et al. A limited sampling
depending upon the patient’s condition and state strategy for determining carboplatin AUC and moni-
of cancer, drugs are given in combination. toring drug dosage. Eur J Cancer. 2000;36:264–9.
Consequently, the application of TDM may be 16. Reynolds DJM, Aronson JK. Making the most of
plasma drug concentration measurements. Br Med J.
required to assess the efficacy of anticancer 1993;306:48–51.
drugs. However, the estimation of drug levels in 17. Veal GJ, Errington J, Hayden J, et al. Carboplatin
cancer is a challenge in TDM. Finally, the TDM therapeutic monitoring in preterm and full-term neo-
approach in cancer needs more research and stud- nates. Eur J Cancer. 2015;51:2022–30.
18. Jodrell DI, Egorin MJ, Canetta RM, et al. Relationships
ies for effective implementation of individualized between carboplatin exposure and tumour response
therapy.
11 Therapeutic Drug Monitoring and Toxicology of Anticancer Drugs 179

and toxicity in patients with ovarian cancer. J Clin 28. Veal GJ, Errington J, Rowbotham SE, et al. Adaptive
Oncol. 1992;10:520–8. dosing approaches to the individualization of 13-cis-­
19. Calvert AH, Newell DR, Gumbrell LA, et al. retinoic acid (isotretinoin) treatment for children
Carboplatin dosage: prospective evaluation of a sim- with high-risk neuroblastoma. Clin Cancer Res.
ple formula based on renal function. J Clin Oncol. 2013;19:469–79.
1989;7:1748–56. 29. Salman B, Al-Za’abi M, Al-Huneini M, et al.
20. Casal MA, Nolin TD, Beumer JH. Estimation of kid- Therapeutic drug monitoring-guided dosing of busul-
ney function in oncology: implications for anticancer fan differs from weight-based dosing in hematopoi-
drug selection and dosing. Clin J Am Soc Nephrol. etic stem cell transplant patients. Hematol Oncol
2019;14:587–95. Stem Cell Ther. 2017;10:70–8.
21. Leveque D, Santucci R, Gourieux B, et al. 30. Codacci-Pisanelli G, Pinedo HM, Lankelma J,
Pharmacokinetic drug-drug interactions with metho- et al. Pharmacokinetics of bolus 5-fluorouracil:
trexate in oncology. Expert Rev Clin Pharmacol. relationship between dose, plasma concentrations,
2011;4:743–50. area-under-the curve and toxicity. J Chemother.
22. Saeter G, et al. Treatment of osteosarcoma of the 2005;17:315–20.
extremities with the T-10 protocol, with emphasis 31. Terzolo M, Angeli A, Fassnacht M, et al. Adjuvant
on the effects of preoperative chemotherapy with mitotane treatment for adrenocortical carcinoma. N
single-agent high-dose methotrexate: a Scandinavian Engl J Med. 2007;356:2372–80.
Sarcoma Group Study. J Clin Oncol. 1991;9:1766–75. 32. Desta Z, Ward BA, Soukhova NV, et al. Comprehensive
23. Abromowitch M, et al. High-dose methotrexate evaluation of tamoxifen sequential biotransformation
improves clinical outcome in children with acute lym- by the human cytochrome P450 system in vitro: prom-
phoblastic leukemia: St. Jude Total Therapy Study inent roles for CYP3A and CYP2D6. J Pharmacol
X. Med Pediatr Oncol. 1988;16:297–303. Exp Ther. 2004;310:1062–75.
24. Hann IM, et al. ‘MACHO’ chemotherapy for stage IV 33. Teng JFT, Mabasa VH, Ensom MHH. The role of
B cell lymphoma and B cell acute lymphoblastic leu- therapeutic drug monitoring of imatinib in patient
kaemia of childhood. Br J Haematol. 1990;76:359–64. with chronic myeloid leukemia and metastatic or
25. Matthay KK, Villablanca JG, Seeger RC, et al. unresectable gastrointestinal stromal tumor. Ther
Treatment of high-risk neuroblastoma with intensive Drug Monit. 2012;34(1):85–97.
chemotherapy, radiotherapy, autologous bone marrow 34. Hannah YK, Jennifer HM, Andrew JM, et al.
transplantation, and 13-cis-retinoic acid. Children’s Precision dosing of targeted anticancer drugs-
Cancer Group. N Engl J Med. 1999;341:1165–73. challenges in the real world. Transl Cancer Res.
26. Villablanca JG, Khan AA, Avramis VI, et al. Phase I 2017;6(Suppl 10):S1500–11.
trial of 13-cis-retinoic acid in children with neuroblas- 35. Suttle AB, Ball HA, Molimard M, et al. Relationships
toma following bone marrow transplantation. J Clin between pazopanib exposure and clinical safety and
Oncol. 1995;13:894–901. efficacy in patients with advanced renal cell carci-
27. Hunsu VO, Facey COB, Fields JZ, Boman noma. Br J Cancer. 2014;111:1909–16.
BM. Retinoid as chemo-preventive and molecular-­ 36. Joel SP, Ellis P, O’Byrne K, et al. Therapeutic moni-
targeted anti-cancer therapies. Int J Mol Sci. toring of continuous infusion etoposide in small-cell
2021;22(14):7731. Published online. lung cancer. J Clin Oncol. 1996;14:1903–12.
Therapeutic Drug Monitoring
and Toxicology
12
of Immunosuppressant

Anshul Shakya, Rajdeep Sarma, Neha Ghimire,


Surajit Kumar Ghosh, Hans Raj Bhat,
and Obaidur Rahman

Abstract cated inter- or intra-patient variability, getting


the desired outcome isn’t always as simple as
Immunosuppressive medications have reached it appears. Monitoring drug levels is critical,
a great milestone in the treatment of numerous to ensure that immunosuppressants are main-
autoimmune diseases and the maintenance of tained within their therapeutic ranges in the
immune response after organ transplantation. blood, thus minimizing the risk of rejection or
Antibodies, glucocorticoids, calcineurin toxicity. Some immunosuppressant drugs that
inhibitors, and antiproliferative are the four have been shown to be effective (azathioprine,
main immunosuppressive drug classes now mycophenolic acid, tacrolimus, cyclosporine,
being utilized successfully in the current situ- sirolimus, everolimus, etc.) are briefly
ation. It has been suggested for the treatment reviewed in this chapter, with a summary of
of severe autoimmune disorders and acute their mechanisms of action, adverse effects,
immunological rejection of organ transplants, and toxicities followed by a general discus-
but these medicines necessitate long-term sion of the monitoring of these immunosup-
usage and non-specifically inhibit all the pressive agents. Finally, a brief discussion of
immune system, resulting in patients suffering future trends in immunosuppression therapy is
from higher risks of unwanted effects. Since, provided, as well as information on monitor-
the pharmacokinetics of immunosuppressive ing individual immunosuppressive
medications is complicated and unpredictable; medications.
drug parameters such as therapeutic index,
absorption, distribution, and elimination are Keywords
unique and changeable to each individual.
Every physician’s goal is to personalize a Immunosuppressants · Therapeutic drug
patient’s drug treatment to achieve the best monitoring · Immunosuppressants toxicity ·
feasible balance between therapeutic efficacy Drug interactions · Cyclosporine
and the risk of side effects. Due to compli-

12.1 Introduction
A. Shakya (*) · R. Sarma · N. Ghimire · S. K. Ghosh
· H. R. Bhat · O. Rahman
Department of Pharmaceutical Sciences, Faculty of Immunosuppressants, also known as antirejec-
Science and Engineering, Dibrugarh University, tion drugs, are those medicines that suppress the
Dibrugarh, Assam, India body’s immune system and also lower the ability
e-mail: anshulshakya@dibru.ac.in

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 181
S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_12
182 A. Shakya et al.

to reject a transplanted organ. For a long time, tic drug monitoring measures certain medications
organ failure resulted in the patient’s immediate at predetermined intervals. Medical professionals
death. Human organ transplantation has rescued use therapeutic drug monitoring to keep track of a
a large portion of the population, during the last patient’s blood levels of immunosuppressant and
three decades. Unfortunately, there aren’t enough adjust their dosage accordingly. Therapeutic drug
organs to go around to save every patient’s life. monitoring is only used to monitor drugs with
The available organs must be used as efficiently limited therapeutic ranges, drugs with substantial
as possible, and the function must be maintained pharmacokinetic variability, medications with dif-
continuously, and for that organ rejection must be ficult target concentrations to monitor, and drugs
minimized. To avoid rejection of the transplanted with known therapeutic toxicity and adverse
organ, immunosuppressive medication must be effects. Due to their limited therapeutic index and
administered after transplantation. considerable inter-individual variability in blood
Immunosuppressive medicines or immunosup- levels, immunosuppressants necessitate therapeu-
pressants play a key role in preserving trans- tic drug monitoring. There are many factors that
planted organ function and immunogenicity of play a key role in this blood level variability, viz.,
the receiver’s physiological system [1]. Earlier, sex, age, drug-nutrient interactions, inflammation,
allograft survival rates for organ transplants are liver mass, infection, polymorphism, drug-­disease
very less because of their powerful cellular and interactions, and renal clearance impairments [4].
humoral immunological responses. The imple- Some of the most frequently used immunosup-
mentation of novel immunosuppressive drugs has pressants in several organ transplants are everoli-
improved allogeneic transplantation survival mus, cyclosporine A, sirolimus, and tacrolimus.
rates dramatically over the past quarter century. In which, lymphocyte proliferation and cytokine
Now, allogeneic bone marrow transplants are productions are inhibited by cyclosporine A and
being performed more than 100,000 times a year tacrolimus, which act as calcineurin inhibitors,
throughout the world. In contrast to the early and by preventing the synthesis of interleukin-2.
immunosuppressive drugs, which are mostly glu- Sirolimus and everolimus impede the progression
cocorticoids and antimetabolites, newer immu- of the T-cell cycle. Serious adverse effects may
nosuppressive drugs have been launched, appear if plasma blood level concentration of
including belatacept (Nulojix, a CTLA4-Ig immunosuppressants is too high (including neu-
fusion protein), the first biologic drug, which dis- rological side effects, nephrotoxicity, cardiotoxic-
rupts a vital stage in the beginning of T-cell-­ ity, and increased risk of infections) [5].
mediated immune responses [2].
The therapeutic range of the immunosuppres-
sive drugs currently in use is extremely low. As a 12.2 Therapeutic Drug
result, for the patients who are in immunosup- Monitoring
pressant therapy, their blood plasma concentra- of Immunosuppressants
tion levels for immunosuppressant must be
monitored continuously [3]. In immunosuppres- Numerous studies have indicated that therapeutic
sant therapy, it is hard to predict an individual’s drug monitoring of immunosuppressants
reaction to a certain dose of immunosuppressive improves the clinical outcomes of transplanted
drugs, as immunosuppressants have notable phar- patients. A summary of classification of the vari-
macokinetic variations between peoples; due to ous immunosuppressants is shown in Fig. 12.1.
this, administration of an immunosuppressive These medications need constant monitoring due
drug is difficult. Therapeutic drug monitoring is to the limited therapeutic index as well as the
an assay that determines a drug’s concentration in various drug-dietary interactions and drug-­
blood plasma. So, the dosage of medication that disease interactions that they have. When used in
has been taken is both safe and effective. In order combination therapy, immunosuppressive
to optimize individual dosing regimens, therapeu- ­medications have an additive effect because of
12 Therapeutic Drug Monitoring and Toxicology of Immunosuppressant 183

Everolimus

Cyclosporine Tacrolimus

Sirolimus IMMUNOSUPPRESSANT Mycophenolate


DRUGS mofetil

Glucocorticoids Corticosteroids
Azathioprin

Fig. 12.1 Immunosuppressant drugs

their complementary modes of action. In thera- when it is caused by either BK virus nephrop-
peutic practice, they are commonly combined in athy or calcineurin inhibitors (cyclosporine
order to minimize negative effects. This combi- and tacrolimus) which occurs during the
nation therapy of medications necessitates a impairment of kidney graft function [10].
simultaneous determination of immunosuppres- 4. In immunosuppressants therapy, the dose/
sive medicines. Drug monitoring is common for exposure relationship is highly varied between
drugs like cyclosporine, tacrolimus, sirolimus, intra- and inter-patient. When it comes to
and mycophenolic acid [6, 7]. adjusting the dosage and target ranges for
For several reasons, immunosuppressant drug immunosuppressive drugs, it is all patient-­
treatment needs constant drug concentration specific. The dose/concentration relationship
monitoring. depends on a number of factors, such as
genetic polymorphisms and drug-drug inter-
1. Immunosuppressive medicines have narrow actions, as well as interactions with food, and
therapeutic ranges. Immunosuppressants such the environment might complicate [11].
as cyclosporine, tacrolimus, sirolimus, and 5. Immunosuppressant drugs treatment cause
mycophenolate have a narrow therapeutic drug dependency which can lead to critical
index and are critical-dose medicaments that conditions, especially in adolescents and
show the intended therapeutic potential with young patients, and requires regular monitor-
acceptable tolerability only within a narrow ing [12].
blood concentration range [8].
2. Immunosuppressive medicines have severe
consequences if the required therapeutic range 12.3 Classification
is not met during therapy. There is an increased of Immunosuppressant
risk of infection and malignancies when over-­ Drugs
immunosuppression or drug toxicity has
occurred, and also there is a possibility of 12.3.1 Calcineurin Inhibitors:
graft dysfunction and graft loss [9]. (Cyclosporine and Tacrolimus)
3. In between clinical disease stage, toxicody-
namic effects of immunosuppressants can be Cyclosporine Cyclosporine is a fungal cyclic
difficult to diagnose, for example, distinguish- polypeptide which contains of 11 amino acids,
ing the source of nephrotoxicity is difficult which has drastically altered the field of organ
184 A. Shakya et al.

transplant in the past 30 years. It acts as an immu- eases [16]. The presence of clinical findings
nosuppressive agent by disrupting the initiation along with fever, decrease in urine output, rise in
and escalation of the cytotoxic T cells [13]. blood pressure, weight gain, and increase in the
Cyclosporine acts by blocking the production of level of serum creatinine leads to the determina-
a T-lymphocyte lymphokine, i.e., interleukin-2. tion of acute renal transplant refusal. Percutaneous
Lymphokines are responsible for the regulation renal transplant biopsy confirms the suspected
of the immune response to a transplanted organ. cases of rejection (acute) before induction of
Cytochrome P450 is the enzyme that breaks antirejection treatment [20]. Tacrolimus inhibits
down cyclosporine and oxidized to at least 12 T-cell activation by attaching to the FK-binding
metabolites in the liver [14]. In kidney transplant protein. The resultant complex binds to calcineu-
patients, poorly absorbed cyclosporine may lead rins and hinders dephosphorylation of the tran-
to graft rejection. The ethno-pharmacokinetics of scription factor [21], and signal transduction
racial groups haven’t been noted, but it has been pathways in T cells are interrupted by the
observed that the black patients absorb less than tacrolimus-­FKBP12 complex [22]. Blockade of
that of the whites [15]. interleukin-2 gene transcription leads to failure
of T-cell clonal expansion and differentiation of
Cyclosporine is used as a combination therapy precursor to mature cytotoxic T cells [21]. The
with corticosteroids, azathioprine, and mycophe- oral dose of tacrolimus is bid 0.15–0.30 mg/kg/
nolic acid for kidney, liver, heart, skin, and bone day and by i.v. route is 0.05–0.20 mg/kg/day [22].
marrow transplantation. Cyclosporine is admin- Tacrolimus has a low oral bioavailability, with
istered orally at a dose of 7–9 mg/kg/day [16]. considerable intra- and inter-individual variabil-
The inhibitors of CYP3A4 (viz., erythromycin, ity ranging from 4% to 89% [21]. The plasma
fluvoxamine, nefazodone, losartan, grapefruit concentrations are reached after about 0.5–1 h.
juice) increase the level of cyclosporine by inhib- Further, tacrolimus is eliminated by hepatic
iting CYP3A4. The inducers of CYP3A4 or metabolism in the bile [22]. Inhibitor of CYP3A4,
P-glycoproteins (like phenobarbital, phenytoin, which enhances the tacrolimus activity, includes
rifampin, modafinil, St. John’s wort) decrease the diltiazem, fluconazole, erythromycin, clarithro-
level of cyclosporine [13]. Higher than normal mycin, itraconazole, and indinavir. Rifampin,
levels of cyclosporine may lead to neurotoxicity phenobarbital, and phenytoin are the inducers of
or nephrotoxicity but below the therapeutic range CYP3A4 which decreases the level of tacroli-
of drugs may lead to transplant rejection [13]. mus. The adverse effect includes, hyperkalemia,
The pharmacokinetic study of cyclosporine hypomagnesemia, tremors, insomnia, paresthe-
has a low oral bioavailability of about 30% [17]. sias, and irritability [23].
Distribution of cyclosporine in the blood is
41–58% approximately in erythrocytes, 5–12%
in granulocytes, 33–47% in plasma, and 4–9% in 12.3.2 Antiproliferative Agents:
lymphocytes [18]. It is metabolized by the cyto- (Mycophenolate Mofetil,
chrome P450 3A (CYP3A). 90% of the drug is Azathioprine, Methotrexate,
excreted via bile [19]. Absorption of cyclospo- Cyclophosphamide,
rine is affected by P-glycoproteins after oral and Chlorambucil)
administration, as it clears the drug from the cells
to the intestinal lumen and is metabolized by 3A4 Mycophenolate Mofetil Mycophenolate
intestinal activity [13]. mofetil is a prodrug of the mycophenolic acid
that inhibits the proliferation of T and B lympho-
Tacrolimus Tacrolimus is procured from cytes through reversible and non-competitive
Streptomyces tsukubensis and is used for the pre- inhibition of the inosine monophosphate dehy-
ventive treatment of liver and kidney transplant drogenase. Mycophenolate mofetil is basically
exclusion and T-cell-mediated autoimmune dis- administered at a fixed dose. The recommended
12 Therapeutic Drug Monitoring and Toxicology of Immunosuppressant 185

starting dose is 720 mg/kg/day [24]. It may be nance drug protocol, azathioprine is used in com-
active against both acute and chronic refusal [25]. bination with cyclosporine/tacrolimus and
The mechanism of action of mycophenolic acid corticosteroids [36]. Azathioprine and allopuri-
is based on blocking of purine synthesis [26] and nol when given in combined form cause leukope-
mesangial proliferation [27]. Purine synthesis nia. The adverse effect of azathioprine is
occurs by two major pathways: the salvage path- gastrointestinal disturbances, hepatic toxicity,
way and the de novo pathway [28]. In the de novo and bone marrow suppression [37].
pathway, ribose 5-phosphate which is a product
of the pentose phosphate pathway is phosphory-
lated to 5-phosphoribosyl-l-pyrophosphate by 12.3.3 mTOR Inhibitors: (Sirolimus
adenosine triphosphate. The glycosylation and and Everolimus)
expression of adhesion molecules, as well as
lymphocyte and monocyte migration to inflam- Sirolimus Sirolimus obtained from
matory sites, are all inhibited by mycophenolic Streptomyces hygroscopicus consists of a macro-
acid [29]. Mycophenolic acid reversibly inhibits cyclic lactone, which is characterized by its
inosine 50-monophosphate dehydrogenase, potent immunosuppressant activity. Interestingly,
which is a key enzyme in the immune system. sirolimus is used in organ transplantation because
The de novo synthesis of guanosine nucleotides of its distinctive mechanism of action, low side
is blocked by the inhibition of inosine effect, and ability to synergize with other immu-
50-­monophosphate dehydrogenase, which are nosuppressants. At the later stage of the cell
necessary substrates for RNA and DNA synthesis cycle, sirolimus reduces the activation of T lym-
[30]. Oral administration of mycophenolate phocyte by preventing the post-interleukin-2
mofetil is rapid and quickly gets converted to its receptor mTOR signal transduction pathway.
active metabolite, i.e., mycophenolic acid [31]. Cytochrome P450 and 3A4 isoenzyme metabo-
The glucuronide metabolites are eliminated lize the drug [38]. The mechanism of sirolimus is
through the kidney, and 95% of a given dose of that it operates in the cytosol by binding to the
mycophenolate mofetil is excreted in the urine as immunophilin FK-binding protein-12. This com-
glucuronide metabolites [32]. The adverse effects plex prevents the activation of the mammalian
related to mycophenolate mofetil are abdominal target of sirolimus (mTOR), which is a key regu-
pain, nausea, diarrhea, decreased appetite accom- latory kinase. Hence, the inhibition will suppress
panying weight loss, vomiting, hematological the cytokine-mediated T-cell proliferation inhib-
problems such as leukopenia, and symptomless iting the progression from the G1 to the S phase
anemia due to bone marrow toxicity [33]. of the cell cycle. Thus, at the later stage, siroli-
mus will inhibit the progression of the cell cycle.
The oral dose of sirolimus is 3–5 mg/kg [39].
Azathioprine Azathioprine is considered the Sirolimus is absorbed rapidly; the mean peak
first immunosuppressive agent that was accept- whole-blood sirolimus concentrations (Cmax)
able for successful renal transplantation in occur 1 h after administration of a single dose
humans [34]. The initial dose of azathioprine is and 2 h after multiple doses in renal transplant
3–5 mg/kg orally or i.v. once a day. The medical patients. The systemic availability of sirolimus is
efficacy of azathioprine is reliant on its metabo- 14% [40]. In human whole blood, sirolimus is
lism to 6-thioguanine nucleotides and the amal- distributed among red blood cells (94.5%),
gamation of 6-thioguanine into cellular DNA plasma (3.1%), lymphocytes (1.01%), and granu-
[35]. Azathioprine interferes with the synthesis locytes (1.0%) [41]. Sirolimus is primarily
of RNA and DNA and exerts its action by pre- metabolized not only by CYP3A4 but also by
venting the differentiation and escalation of B CYP3A5 and CYP2C8 [42]. The area under the
and T lymphocytes. Azathioprine is considered curve of sirolimus is increased by diltiazem and
an inhibitor of nucleotide synthesis. In mainte- ketoconazole by 60% and 90%; however, rifamy-
186 A. Shakya et al.

cin decreases the area under the curve by 82% Glucocorticoids (Prednisolone) Immunosuppr
[41]. The concentration of sirolimus is increased essive medications such as glucocorticoids were
by diltiazem, ketoconazole, and cyclosporine, first introduced to the market in the 1960s and
whereas rifampin reduces sirolimus exposure played a key role in making organ transplantation
[42]. Inhibitors like diltiazem, verapamil, itra- possible. Peripheral blood lymphocyte counts are
conazole, ketoconazole, voriconazole, flucon- reduced rapidly due to the lysis and redistribution
azole, erythromycin, and clarithromycin increase of lymphocytes induced by glucocorticoids. In
the concentration of sirolimus. Enzyme inducers, the long run, receptors, glucocorticoid-induced
viz., phenytoin, carbamazepine, and rifampin, proteins, or other interacting proteins regulate the
decrease the sirolimus exposure [43]. The adverse transcription of numerous genes in response to
effects related to sirolimus are bone marrow sup- hormones [51]. IkB expression is enhanced by
pression, hyperlipidemia [44], anemia, post-­ glucocorticoid receptor complexes, which sup-
transplant diabetes mellitus, proteinuria, edema, presses NF-kB activation and induces apoptosis
mouth ulcer, and joint pain [45]. in activated cells. The downregulation of key pro-­
inflammatory cytokines including IL-1 and IL-6
is critical. There is a block on the production and
Everolimus The hydroxyethyl derivative of siro- proliferation of IL-2 by T cells. Cytotoxic
limus is everolimus [46]. A comparable immuno- T-lymphocyte activation is suppressed.
suppressive mechanism to that of sirolimus, Chemotaxis is poor in neutrophils and mono-
namely, the suppression of the mammalian target cytes, and also lysosomal enzyme release is
for rapamycin, is found in everolimus [47]. It also reduced. As a result, glucocorticoids exert anti-­
lowers angiogenesis while reducing cytokine-­ inflammatory effects on a variety of immune sys-
mediated T-cell proliferation. In comparison with tem components [52]. An oral dose of
sirolimus, the half-life of everolimus is shorter, prednisolone is 5–60 mg/kg/day. Diltiazem and
and it quickly achieves steady-state trough con- ketoconazole both being the inhibitors of
centrations. It does, however, have a small thera- CYP3A4 increase the levels of prednisolone,
peutic window of 3–8 mg/mL [48]. When given in thus enhancing the toxic activity of the drug. The
doses of 2.5 mg/kg, everolimus is rapidly absorbed inducers of CYP3A4 such as phenytoin, rifampin,
following oral administration, with an average C- and phenobarbital increase the metabolism of
max of 45 mg/L. After 24–78 minutes, the maxi- glucocorticoids and thereby lower the level of
mal concentration of the drug is reached. prednisolone. Side effects related to glucocorti-
Additionally, CYP3A4, 3A5, and 2C8 are coids are osteoporosis, predisposition to infec-
involved in the metabolism of everolimus, and it tion, hyperglycemia, weight gain, hypertension,
is a substrate for P-glycoprotein. Everolimus used glucose intolerance, adrenal suppression, psy-
as 1 mg/kh of body weight twice a day for renal chosis, and depression [53].
transplants and 0.75 mg/kg of body weight for
liver transtplants recipients [49].
12.4 Adverse Effects and Toxicity
of Newer
Corticosteroids Corticosteroids largely affect Immunosuppressant Drugs
T-cell activation by decreasing the synthesis of
T-cell cytokines including IL-2, IL-6, and inter- Leflunomide and Malononitriloamides The
feron gamma, which are necessary for lympho- newest class of immunosuppressant medicines,
cytes and macrophages to respond to allograft leflunomide and the malononitriloamides, are
antigens. They also promote T-cell migration currently being studied for use in transplantation.
from the intravascular compartment to lymphoid Leflunomide’s anti-inflammatory and immuno-
tissue by inhibiting antibody and complement modulating capabilities were discovered in 1985,
binding [50]. which set it apart from other anti-inflammatory
12 Therapeutic Drug Monitoring and Toxicology of Immunosuppressant 187

and immunosuppressive medications. developed for clinical use as an antibiotic [62].


Leflunomide’s immunosuppressive effects have Preclinical investigations and Phase I and II stud-
been studied extensively in animal transplanta- ies in stable kidney recipients have largely pre-
tion models. Leflunomide’s clinical development dicted the current profile of adverse events in
has been limited to usage in patients with autoim- humans. Short-term sirolimus administration has
mune illnesses such as rheumatoid arthritis due been linked to headache, nausea, dizziness,
to its long half-life (11–16 days) in humans [54, changes in blood glucose levels, epistaxis, infec-
55]. The most important side effect of lefluno- tion, and a decrease in platelets and white blood
mide and the malononitriloamides in cynomol- cells [63, 64]. The long-term usage of rapamycin
gus monkeys was anemia. GI problems, rashes, has been linked to an increased risk of hypertri-
allergic reactions, weight loss, and reversible glyceridemia. In multiple trials in rats and pigs,
baldness were among the side events documented sirolimus was found to be free of the nephrotox-
in the leflunomide research. Infections were not icity seen with tacrolimus and cyclosporine A,
more common in the leflunomide group, but probably due to the lack of calcineurin inhibition.
hematocrit and hemoglobin levels were lower in However, in normal rats receiving sirolimus,
all groups [56]. hypomagnesemia and tubular damage were
observed, and in spontaneously hypertensive rats,
the progression of kidney failure has been docu-
Mycophenolic Acid Penicillium spp. cultures mented. In rats, excessive doses of sirolimus have
were used by Gosio in 1896 to produce mycophe- caused heart and retinal infractions [65–67].
nolic acid, which was refined by Alsberg and
Black in 1913. In the 1940s, antibacterial and
antifungal properties were discovered. Tacrolimus Tacrolimus is a metabolite of
Mycophenolic acid was researched for psoriasis Streptomyces tsukubaensis, an actinomycete that
but did not acquire clinical use when its antitu- was initially shown to be immunologically effec-
mor action was discovered in 1968 [57]. Its tive in rat heart allograft recipients in 1987 [68].
immunosuppressive characteristics have been In various experimental models, it was quickly
demonstrated by Mitsui and Suzuki [58]. discovered to be a strong alternative to cyclospo-
Mycophenolic acid was approved by the FDA in rine A. Patients using tacrolimus have experi-
1995 for the treatment of acute renal allograft enced significant nephrotoxicity and
rejection. For heart transplant recipients, it was neurotoxicity. The suppression of calcineurin
approved for usage in 1998 [59]. Diarrhea, vom- phosphatase is one plausible mechanism for neu-
iting, opportunistic infections, and leukopenia rotoxicity, but the etiology of its renal vasculo-
are the most prevalent side effects of mycopheno- pathic consequences is unknown. Increased
lic acid in humans. Mycophenolic acid’s myelo- thromboxane A2, endothelin production, or
toxic mechanism is still a mystery. Only enhanced intrarenal renin production are all
proliferating lymphocytes will be affected by linked to decreased renal glomerular and cortical
mycophenolic acid’s specific suppression of de blood flow and increased renal vascular resis-
novo purine production. Patients with psoriasis tance [69, 70]. Cardiomyopathy, anemia, persis-
treated with mycophenolic acid rarely experience tent diarrhea, diabetes, and allergies have all been
leukopenia, unlike transplant recipients [60]. documented in tacrolimus patients.
Hypercholesterolemia and hypertension are less
common when compared with cyclosporine A,
Sirolimus Sirolimus was discovered initially as while gingival hyperplasia and hirsutism are
an antifungal agent in the mid-1970, which is a almost non-existent in patients using chronic
microbial product derived from the actinomycete tacrolimus [71]. Tacrolimus-based immunosup-
Streptomyces hygroscopicus [61]. Because of its pressive treatments have been linked to lymphop-
immunosuppressive effects, it was not further roliferative illness and infections [7].
188 A. Shakya et al.

12.5 Immunosuppressant Drug et al. studied renal transplant recipients taking the
Interactions immunosuppressive drugs such as tacrolimus,
mycophenolate mofetil, methylprednisolone, and
Immunosuppressants can have a number of inter- basiliximab were showed substantial gender-­
actions, some of which are enumerated in related differences after the first oral dose [76].
Fig. 12.2.

Drug-Nutrient Interactions The importance of Polymorphism Polymorphism has demon-


drug-nutrient interactions is increasingly becom- strated functional consequences of many drug-­
ing recognized. Depending on the patient’s diet, metabolizing enzymes. CYP3A4/5 and
the metabolism of some medicines might be P-glycoprotein are recognized substrates for
affected. Many medication’s metabolisms are cyclosporine A. One of the most important
altered when eaten in large quantities with grape- CYP3A enzymes, CYP3A4/5, has a polymorphic
fruit juice, for example. Studies have shown that expression pattern and is ethnically dependent.
the CYP3A-mediated metabolism of cyclospo- Cytochrome P450 (CYP)3A enzymes in the
rine is inhibited by grapefruit juice and enhanc- intestines and liver are primarily responsible for
ing medication absorption by blocking the metabolism of tacrolimus. It also serves as a
P-glycoprotein efflux transporters. As a result, if substrate for P-glycoprotein, which carries dif-
a patient is taking cyclosporine and grapefruit fused tacrolimus back into the gut lumen after it
juice at the same time, the medication concentra- has left intestinal cells. Tacrolimus is predicted to
tion should be checked [72, 73]. have an effect on the elderly because of age-­
related variations in CYP3A and P-glycoprotein
expression as well as changes in liver mass and
Drug-Disease Interactions Drug-disease inter- body composition [19, 77].
actions may possibly have a role in the diversity
of plasma immunosuppressive concentrations
across individuals. For example, due to a decrease 12.6 Toxicity Related
in protein binding, renal insufficiency can cause a to Immunosuppressant
change in the free fraction of mycophenolic acid. Agents
Furthermore, a decrease in cytochrome P450
enzyme activity is commonly related with inflam- As new immunosuppressive drugs with novel
mation and infection. Cytochrome P450 enzyme mechanisms of action, new formulations, and
enhances the amount of cyclosporine A metabo- improved means for routine therapeutic drug
lism [74]. monitoring have become available to transplant
recipients, the efficacy and tolerance of therapy
have improved. Life expectancy and quality of
Gender Gender also has an impact on drug con- life for organ transplant recipients have improved
centration. Variances in drug responses between dramatically in recent years. Neurotoxicity,
men and women can be caused by differences in nephrotoxicity, hepatotoxicity, and bone marrow
their biology. Both pharmacokinetic and pharma- depression (Fig. 12.3) are all the serious side
codynamic parameters vary in men and women, effects that come along with the use of most
but more evidences have been reported on vari- immunosuppressive drugs that have not yet been
ance of pharmacokinetic parameters. For exam- adequately studied. Hematological toxicity, such
ple, women may have slightly better as leukopenia, thrombocytopenia, and anemia
bioavailability following oral medication admin- due to bone marrow suppression or hemolysis,
istration than men, particularly in CYP3A sub- may occur as a result of immunosuppressive drug
strates. It is known that mycophenolic acid is delivery. Allogeneic bone marrow suppression is
primarily metabolized in the liver [75]. Velickovic more common with the use of azathioprine and
12 Therapeutic Drug Monitoring and Toxicology of Immunosuppressant 189

Fig. 12.2 Immunosuppressant drug interactions

mycophenolate mofetil, but hemolytic uremic creatinine levels. On the other hand, cyclospo-
syndrome is more common with the use of cyclo- rine prescribed in lower doses or when the
sporine, tacrolimus, or muromonab. Long-term patient is prescribed with other immunosup-
usage of immunosuppressant drugs such as pressants such as azathioprine leads to a
cyclosporine can lead to side effects like hirsut- decrease in serum creatinine levels [14].
ism and gingival hyperplasia as well as alopecia
or cushingoid facies and an increase in blood cre-
atinine [45]. Hepatotoxicity Hepatotoxicity indicates
chemical-­driven liver damage. It is the leading
cause of liver failure. Cyclosporine and azathio-
12.7 The Toxicities Related prine have been reported to cause infrequent but
to Various significant hepatotoxicity. Conjugated hyperbili-
Immunosuppressants Are rubinemia is a common symptom of cholestasis
as Follows caused by cyclosporine. Cyclosporine-induced
cholestasis appears to be a result of bile acid
Nephrotoxicity Nephrotoxicity is defined as transport impairment. In several studies, cyclo-
the functional abnormalities of the kidney, sporine has been associated with the develop-
which are often caused by drugs. Nephrotoxicity ment of bile duct calculi. Hyperbilirubinemia
is basically dose-dependent and related to the may occur in patients receiving large oral doses
patient’s bioavailability. Increased renal vascu- of cyclosporine to an extent of 30–50% [78].
lar resistance leads to acute nephrotoxicity. High doses of glucocorticoids may cause hepato-
Acute nephrotoxicity is categorized by the megaly, and macrovesicular steatosis may result
decrease in glomerular filtration rate and reduc- from a low dose of prednisolone. Azathioprine
tion of renal blood flow with an increase of fil- has been related to a wide range of hepatotoxic
tration fraction. However, nephrotoxicity is responses, all of which are believed to be caused
quite difficult to distinguish from long-term by azathioprine-induced destruction to the endo-
renal transplant rejection. Though nephrotoxic- thelial cells that line the hepatic terminal venules
ity has been reported in both cyclosporine and and sinusoids. Azathioprine elevates liver dam-
tacrolimus, it has also been reported that cyclo- age when used for a longer period of time. Some
sporine is more nephrotoxic than tacrolimus research suggests that liver transplant recipients
[46]. Cyclosporine, when prescribed for a lon- who have viral hepatitis or are experiencing con-
ger period of time, may cause a rise in serum tinuous rejection are more susceptible to devel-
oping these hepatotoxic responses [79].
190 A. Shakya et al.

Fig. 12.3 Toxicity related to immunosuppressant agents

Neurotoxicity The frequency of neurotoxicity by the third month in over half of 402 kidney
varies according to the organ transplanted. Fine transplant recipients. The greatest number of
tremor, generalized seizures, paresthesia, and people who were affected were children.
encephalopathy have been related to cyclosporine Cyclosporine may cause hypertrichosis by rais-
therapy [80]. The symptoms of mild neurotoxicity ing the activity of 5-reductase in peripheral tis-
using tacrolimus include tremor, headache, night- sues, but the exact mechanism is unknown.
mares, vertigo, insomnia, photophobia, mood dis- Tacrolimus stimulated hair growth in animal
turbance, or dysesthesia, and the symptoms of experiments [14].
severe neurotoxicity include akinetic mutism,
focal deficits, cortical blindness, seizures, psycho-
sis, or encephalopathy [81]. Gingival Hyperplasia Dose-related gingival
hyperplasia is more common in children, and it
can be severe enough to necessitate gingivec-
Hirsutism Hirsutism is a well-known, dose-­ tomy. Another well-known adverse effect of
dependent pharmacological effect of cyclospo- cyclosporine is gingival hyperplasia. McGaw
rine therapy, occurring in approximately 40% of et al. [84] found fibrous hyperplasia of the gums
the patients. The combination of minoxidil and in up to 33% of patients, and King et al. [85]
cyclosporine leads to dramatic hair growth [82]. documented it in 22% of kidney transplant
In renal transplantation allograft patients, the use recipients. Gingival hyperplasia is especially
of cyclosporine for immunosuppression has been dangerous and stressful for pediatric transplant
associated to hypertrichosis. Patients who are recipients since it might cause teeth to erupt
switched to tacrolimus may have an improve- later and speech development to be hampered.
ment in these effects with little risk of rejection or Recently, a link between gingival overgrowth
allograft failure [83]. Eighty percent of individu- and alterations in renal function in juvenile
als assessed had hirsutism, alopecia, acne, or renal transplant r­ ecipients has been discovered.
cushingoid facies as a result of immunosuppres- This shows a link between the fibrosis seen in
sion. In cyclosporine-treated patients, 94% had gingival overgrowth and cyclosporine-induced
hirsutism, compared to non-cyclosporine-treated nephrotoxicity, and it’s tempting to think that
patients. Hypertrichosis, with hair growth con- transforming growth factor beta is involved in
centrating on the upper body and face, appeared the mechanism [86].
12 Therapeutic Drug Monitoring and Toxicology of Immunosuppressant 191

Hematologic Toxicity Immunosuppressants can Diabetogenic Toxicity Some immunosuppres-


cause hematologic damage, such as bone marrow sants have been found to be diabetogenic, mean-
suppression or hemolysis, thrombocytopenia, and ing they can cause diabetes in people who aren’t
leukopenia, when taken for a long time. Hemolytic diabetic. This makes pre-transplant and post-­
uremic syndrome can be caused by the use of transplant care even more difficult. Furthermore,
tacrolimus, cyclosporine, or muromonab. The those with diabetes mellitus after a transplant are
most prevalent and dangerous side effect of aza- more prone to get serious infections and have
thioprine therapy is myelosuppression, particu- lower patient and graft survival rates. The devel-
larly leukopenia [87]. In addition, platelet and opment of glucose intolerance and post-­transplant
leucocyte counts were reduced, resulting in a diabetes mellitus has long been regarded to be
myelosuppression. Immunosuppressive medica- one of the main side effects of high-dose steroid
tions may be reduced or discontinued, blood regimens [93]. In a recent study, glucose intoler-
transfusions may be required, or cytokines, such ance was evaluated in 173 kidney transplant
as erythropoietin and colony-stimulating factors, patients utilizing oral glucose tolerance tests or
may be used to treat bone marrow failure [88]. diabetes mellitus diagnosis 10 weeks after dona-
tion. It has been found that 0.01 mg/kg/day of
prednisolone increases the likelihood of develop-
Bone Toxicity Risk factors for bone disease, ing diabetes mellitus following a transplant by
such as limited mobility, menopause, cirrhosis, 5%. Higher steroid dosage and older age are both
diabetes, hyperparathyroidism, and renal osteo- connected to the development of post-transplant
dystrophy, are common among transplant patients. glucose intolerance. Steroids’ diabetogenic side
Heart, kidney, liver, lung, and bone marrow trans- effects are likely to be more severe in black peo-
plantation have all been linked to the development ple, who have been proven to metabolize steroid
of post-transplantation osteopenia. Kidney trans- dosages more slowly, resulting in increased drug
plant recipients, on the other hand, appear to be exposure. Steroids are still implicated in the
less vulnerable than heart and liver transplant development of post-transplant diabetes mellitus,
recipients [89]. Immunosuppressant-­ induced or according to new research [94].
immunosuppressant-exacerbated bone loss can
occur after transplantation and is usually identi-
fied as a decrease in bone mineral density. The Dyslipidemic Toxicity One of the most com-
immunosuppressants most strongly linked to mon side effects of immunosuppressant therapy
osteopenia and osteoporosis are steroids [90]. is dyslipidemia, which affects 80% of heart trans-
More than half of people who had long-­term glu- plant recipients, 60–70% of kidney transplant
cocorticoid medication developed osteoporosis, recipients, and 45% of liver transplant recipients.
according to a retrospective evaluation of 160 Allograft survival is reduced when cholesterol
research published since 1970. Many studies have and triglyceride levels are increased [94]. It was
linked steroids to post-­transplantation osteopenia shown that cyclosporine has a detrimental influ-
and osteoporosis in the context of transplantation. ence on the serum lipid levels of people with
The incidence of demineralization is linked to amyotrophic lateral sclerosis (ALS). These
both cumulative and daily steroid dosage levels. patients exhibited elevated levels of serum low-­
Experiments on rats have demonstrated that density lipoprotein cholesterol. In a study of liver
cyclosporine and tacrolimus alter the equilibrium transplant recipients, tacrolimus and cyclospo-
between bone formation and bone resorption, rine were found to dramatically raise serum lev-
resulting in considerable bone loss [91, 92]. els of cholesterol and triglycerides [95].
192

Table 12.1 Classification of the immunosuppressive agents with their possible mechanism of action, indication(s), toxicity, and dose
Sl
No. Classification Sub class Mechanism of action Indication(s) Toxicity Dose Citation
1. Calcineurin Cyclosporine Cyclosporine suppresses the Immunosuppressive agents used in Nephrotoxicity, hirsutism and 7–9 mg/kg [14]
inhibitors generation of the T-cell organ transplantation gingival hyperplasia,
lymphokines y-interferon, hepatotoxicity, neurotoxicity,
macrophage inhibitory factor, lymphoproliferative neoplasms
and macrophage chemotactic
factor, which decrease
monocyte activity indirectly
2. Calcineurin Tacrolimus Tacrolimus works by attaching Prevent the rejection of liver and Nephrotoxic, diabetogenic, i.v. dose is [96]
inhibitors to FKBP-12, an intracellular kidney transplants. According to neurological, and cardiovascular 0.05–0.20 mg/
protein that inhibits the pediatric liver transplantation effects kg/day and
T-lymphocyte activation studies registry, it is the most oral dose bid
commonly utilized maintenance 0.15–0.30 mg/
therapy for pediatric transplant kg/day 12 h
recipients apart
3. Antiproliferative Mycophenolate Guanosine monophosphate Add on drug to glucocorticoid and Gastrointestinal toxicity, 720 mg/kg/day [32]
agents mofetil nucleotide synthesis is cyclosporine in renal occasional leukopenia
inhibited, and purine synthesis transplantation
is blocked, preventing T- and
B-cell proliferation
4. Antiproliferative Azathioprine It inhibits purine synthesis, Azathioprine has a long history of Bone marrow depression 3–5 mg/kg [97, 98]
agents affects T cells, and inhibits use in the treatment of hepatotoxicity, post-transplant orally
cytolytic lymphocytes inflammatory bowel disease lymphoproliferative disorder
(PTLD)
5. m-TOR Sirolimus Target of rapamycin and For therapy and prophylaxis of Edema, diabetes, elevation in 3–5 mg/kg [44, 99]
inhibitors interleukin-2 inhibited by the graft rejection reaction lipids, anemia, thrombocytopenia, daily
complex which is formed by proteinuria, impaired wound
binding to FKBP12 healing, interstitial lung disease,
lymphoma, and various cancers
are only a few examples
6. IL 2 receptor Daclizumab and Activated T cells are depleted, It prevents renal and other 5 mg/mL; [100]
antagonist basiliximab and interleukin-2-induced transplant rejection reaction 150 mg/mL
T-cell activation is inhibited
by binding and blocking the
interleukin-2 receptor A chain
(CD25 antigen)
A. Shakya et al.
12 Therapeutic Drug Monitoring and Toxicology of Immunosuppressant 193

12.8 Conclusion and Future pharmacokinetic variability equivalent to already


Perspective used immunosuppressants, which may be useful
in monitoring therapy. Along with the develop-
Based on the above information, it can be con- ment of new immunosuppressive medications,
cluded that the therapeutic drug monitoring of generic formulations of existing potent immuno-
immunosuppressants has undergone an evolu- suppressant drugs, such as cyclosporine, will be
tionary change to minimize drug toxicity. available in the coming age, and the possibility of
Therapeutic drug monitoring is still evolving rap- xenotransplantation will be a future challenge
idly in the field of organ transplantation with con- that will require a significant amount of therapeu-
tinuous practices and currently has become a tic drug monitoring. It is clear from the book
significant standard for most of the immunosup- chapter that therapeutic drug monitoring has sig-
pressive drugs (compiled in the Table 12.1) such nificantly improved treatment outcomes, but
as tacrolimus, sirolimus, cyclosporine, mycophe- understanding the limitations of therapeutic drug
nolic acid, etc. It is necessary to perform thera- monitoring is critical for the appropriate develop-
peutic drug monitoring for all of these drugs, ment of immunosuppressive drug therapy in the
despite the fact that their pharmacokinetics are organ transplantation.
complex and variable, because of the small thera-
peutic ranges, large inter-individual variability in
blood concentrations, gender differences in drug References
metabolism, and interactions between drugs,
nutrients, and other drugs. Nearly two decades, 1. Zhang Y, Zhang R. Recent advances in analyti-
cal methods for the therapeutic drug monitoring
cyclosporine has been the most widely investi- of immunosuppressive drugs. Drug Test Anal.
gated immunosuppressant medication in trans- 2018;10(1):81–94.
plantation, leading to its advanced role in 2. Adams DH, Sanchez-Fueyo A, Samuel D. From
combination therapy that has brought a major immunosuppression to tolerance. J Hepatol.
2015;62(1):S170–85.
improvement in clinical outcomes in transplant 3. Mohammadpour N, et al. A review on therapeutic
recipients. Therefore, combination therapy is drug monitoring of immunosuppressant drugs. Iran
becoming more common. Physicians strive to J Basic Med Sci. 2011;14(6):485.
personalize a patient’s drug therapy in order to 4. Oellerich M, Armstrong VW. The role of therapeutic
drug monitoring in individualizing immunosuppres-
establish the best possible balance between thera- sive drug therapy: recent developments. Ther Drug
peutic efficacy and the risk of adverse effects, but Monit. 2006;28(6):719–25.
because of complicated inter- or intra-patient 5. Krnáč D, Reiffová K, Rolinski B. A new HPLC-MS/
variability, getting the desired outcome isn’t easy, MS method for simultaneous determination
of Cyclosporine A, Tacrolimus, Sirolimus and
which concludes that understanding the pharma- Everolimus for routine therapeutic drug monitoring.
cokinetic precepts of immunosuppressant drugs J Chromatogr B. 2019;1128:121772.
is critical for transplant success. 6. Tiwari P. Therapeutic drug monitoring of immu-
Most given immunosuppressants attach to red nosuppressants: an overview. Indian J Pharmacol.
2007;39(2):66.
blood cells and plasma proteins; the unbound 7. Johnston A, Holt DW. Immunosuppressant drugs–
drug is the only one that can interact with the tar- the role of therapeutic drug monitoring. Br J Clin
gets. As a result, determining the free drug con- Pharmacol. 2001;52(Suppl 1):61S.
centration rather than the total drug concentration 8. Kang J-S, Lee M-H. Overview of therapeutic drug
monitoring. Korean J Intern Med. 2009;24(1):1.
in whole blood may offer more information on 9. Mohammadpour N, Elyasi S, Vahdati N,
immunosuppressive effects. The adaptation of Mohammadpour AH, Shamsara J. A review on
appropriate extraction techniques and the accom- therapeutic drug monitoring of immunosuppressant
plishment of extremely low quantification limits drugs. Iran J Basic Med Sci. 2011;14(6):485–98.
10. Taddeo A, Prim D, Bojescu ED, Segura JM, Pfeifer
are two of the key drawbacks of this methodol- ME. Point-of-care therapeutic drug monitoring for
ogy. New immunosuppressants, such as sotrastau- precision dosing of immunosuppressive drugs. J
rin, are now being investigated and show Appl Lab Med. NLM (Medline). 2020;5:738–61.
194 A. Shakya et al.

11. Jorga A, Holt D, Johnston A. Therapeutic drug mon- 32. Jeong H, Kaplan B. Therapeutic monitoring of
itoring of cyclosporine. In: Transplantation proceed- mycophenolate mofetil. Clin J Am Soc Nephrol.
ings. Elsevier; 2004 Mar;36(2 Suppl):396S–403S. 2007;2(1):184–91.
https://doi.org/10.1016/j.transproceed.2004.01.013. 33. Manzia, T. M., et al. Use of mycophenolate
12. Kahan B. Therapeutic drug monitoring of cyclo- mofetil in liver transplantation: a literature
sporine: 20 years of progress. In: Transplantation review. In: Transplantation proceedings. Elsevier;
proceedings. Elsevier; 2004 Mar;36(2 2005;37(6):2616–2617. https://doi.org/10.1016/j.
Suppl):378S–391S. https://doi.org/10.1016/j. transproceed.2005.06.073.
transproceed.2004.01.091. 34. Stockfleth E, Ulrich C. Skin cancer after organ trans-
13. Fireman M, et al. Immunosuppressants. plantation, vol. 146. Springer Science & Business
Psychosomatics. 2004;45(4):354–60. Media; 2009.
14. Bennett WM, Norman DJ. Action and toxicity of 35. Perrett C, et al. Azathioprine treatment photosen-
cyclosporine. Annu Rev Med. 1986;37(1):215–24. sitizes human skin to ultraviolet A radiation. Br J
15. Lindholm A, Säwe J. Pharmacokinetics and thera- Dermatol. 2008;159(1):198–204.
peutic drug monitoring of immunosuppressants. 36. Suthanthiran M, Morris RE, Strom
Ther Drug Monit. 1995;17(6):570–3. TB. Immunosuppressants: cellular and molecu-
16. Wong SH. Therapeutic drug monitoring lar mechanisms of action. Am J Kidney Dis.
for immunosuppressants. Clin Chim Acta. 1996;28(2):159–72.
2001;313(1–2):241–53. 37. Casetta I, Iuliano G, Filippini G. Azathioprine for
17. Freeman D. Pharmacology and pharmacokinetics of multiple sclerosis. Cochrane Database Syst Rev.
cyclosporine. Clin Biochem. 1991;24(1):9–14. 2007;(4):CD003982.
18. Han K, Pillai VC, Venkataramanan R. Population 38. Cattaneo D, et al. Therapeutic drug monitoring of
pharmacokinetics of cyclosporine in transplant sirolimus: effect of concomitant immunosuppres-
recipients. AAPS J. 2013;15(4):901–12. sive therapy and optimization of drug dosing. Am J
19. Milone MC. Overview of the pharmacology and tox- Transplant. 2004;4(8):1345–51.
icology of immunosuppressant agents that require 39. Oellerich M, et al. Immunosuppressive drug moni-
therapeutic drug monitoring. In: Personalized immu- toring of sirolimus and cyclosporine in pediatric
nosuppression in transplantation. Elsevier; 2016. patients. Clin Biochem. 2004;37(6):424–8.
p. 1–27. 40. MacDonald A, et al. Clinical pharmacokinetics and
20. Bloom RD, et al. Association of hepatitis C with therapeutic drug monitoring of sirolimus. Clin Ther.
posttransplant diabetes in renal transplant patients on 2000;22:B101–21.
tacrolimus. J Am Soc Nephrol. 2002;13(5):1374–80. 41. Mahalati K, Kahan BD. Clinical pharmacokinetics of
21. Shrestha BM. Two decades of tacrolimus in renal sirolimus. Clin Pharmacokinet. 2001;40(8):573–85.
transplant: basic science and clinical evidences. Exp 42. Moes DJA, Guchelaar H-J, de Fijter JW. Sirolimus
Clin Transplant. 2017;15(1):1–9. and everolimus in kidney transplantation. Drug
22. Spencer CM, Goa KL, Gillis JC. Tacrolimus. Drugs. Discov Today. 2015;20(10):1243–9.
1997;54(6):925–75. 43. Augustine JJ, Bodziak KA, Hricik DE. Use of
23. Hooks MA. Tacrolimus, a new immunosuppres- sirolimus in solid organ transplantation. Drugs.
sant—a review of the literature. Ann Pharmacother. 2007;67(3):369–91.
1994;28(4):501–11. 44. Stenton SB, Partovi N, Ensom MH. Sirolimus. Clin
24. Van Gelder T, et al. Therapeutic drug monitoring Pharmacokinet. 2005;44(8):769–86.
of mycophenolate mofetil in transplantation. Ther 45. Ponticelli C, Glassock RJ. Prevention of complica-
Drug Monit. 2006;28(2):145–54. tions from use of conventional immunosuppressants:
25. Sievers TM, et al. Mycophenolate mofetil. a critical review. J Nephrol. 2019;32(6):851–70.
Pharmacotherapy. 1997;17(6):1178–97. 46. De Mattos AM, Olyaei AJ, Bennett
26. Johnston A, Holt DW. Therapeutic drug monitoring WM. Nephrotoxicity of immunosuppressive drugs:
of immunosuppressant drugs. Br J Clin Pharmacol. long-term consequences and challenges for the
1999;47(4):339. future. Am J Kidney Dis. 2000;35(2):333–46.
27. Sepe V, et al. Mycophenolate mofetil in primary glo- 47. Shipkova M, et al. Therapeutic drug monitoring of
merulopathies. Kidney Int. 2008;73(2):154–62. everolimus: a consensus report. Ther Drug Monit.
28. Mele TS, Halloran PF. The use of myco- 2016;38(2):143–69.
phenolate mofetil in transplant recipients. 48. Di Maira T, Little EC, Berenguer
Immunopharmacology. 2000;47(2–3):215–45. M. Immunosuppression in liver transplant. Best
29. Allison A. Mechanisms of action of mycophenolate Pract Res Clin Gastroenterol. 2020;46–47:101681.
mofetil. Lupus. 2005;14(3_suppl):2–8. 49. Kirchner GI, Meier-Wiedenbach I, Manns
30. Fulton B, Markham A. Mycophenolate mofetil. MP. Clinical pharmacokinetics of everolimus. Clin
Drugs. 1996;51(2):278–98. Pharmacokinet. 2004;43(2):83–95.
31. Allison AC, Eugui EM. Mechanisms of action 50. Kaufman DB, et al. Immunosuppression: practice
of mycophenolate mofetil in preventing acute and trends. Am J Transplant. 2004;4:38–53.
and chronic allograft rejection. Transplantation. 51. Coutinho AE, Chapman KE. The anti-inflammatory
2005;80(2S):S181–90. and immunosuppressive effects of glucocorticoids,
12 Therapeutic Drug Monitoring and Toxicology of Immunosuppressant 195

recent developments and mechanistic insights. Mol Pt 2):1284–1286 Publisher name: New York, N.Y.:
Cell Endocrinol. 2011;335(1):2–13. Elsevier Science Inc.
52. Franchimont D. Overview of the actions of gluco- 69. Pham SM, et al. A prospective trial of tacroli-
corticoids on the immune response: a good model mus (FK 506) in clinical heart transplantation:
to characterize new pathways of immunosuppres- intermediate-­term results. J Thorac Cardiovasc Surg.
sion for new treatment strategies. Ann N Y Acad Sci. 1996;111(4):764–72.
2004;1024(1):124–37. 70. Abu-Elmagd K, et al. The effect of graft function on
53. Meier CA. Mechanisms of immunosuppression by FK506 plasma levels, dosages, and renal function,
glucocorticoids. Eur J Endocrinol. 1996;134(1):50. with particular reference to the liver. Transplantation.
54. Lucien J, et al. Blood distribution and single-dose 1991;52(1):71.
pharmacokinetics of leflunomide. Ther Drug Monit. 71. Atkison P, et al. Hypertrophic cardiomyopathy
1995;17(5):454–9. associated with tacrolimus in paediatric transplant
55. Mladenovic V, et al. Safety and effectiveness patients. Lancet. 1995;345(8954):894–6.
of leflunomide in the treatment of patients with 72. Edwards DJ, et al. 6′, 7′-Dihydroxybergamottin
active rheumatoid arthritis. Arthritis Rheumatol. in grapefruit juice and Seville orange juice:
1995;38(11):1595–603. effects on cyclosporine disposition, enterocyte
56. Morris, R. E., et al. Leflunomide (HWA 486) and its CYP3A4, and P-glycoprotein. Clin Pharm Therap.
analog suppress T- and B-cell proliferation in vitro, 1999;65(3):237–44.
acute rejection, ongoing rejection, and antidonor 73. Ducharme MP, et al. Trough concentrations of cyclo-
antibody synthesis in mouse, rat, and cynomol- sporine in blood following administration with grape-
gus monkey transplant recipients as well as arte- fruit juice. Br J Clin Pharmacol. 1993;36(5):457–9.
rial intimal thickening after balloon catheter injury. 74. Tamási V, et al. Some aspects of interindividual vari-
Transplant Proc. 1995;27(1):445-447. Publisher ations in the metabolism of xenobiotics. Inflamm
name: New York, N.Y. : Elsevier Science Inc. Res. 2003;52(8):322–33.
57. Williams RH, et al. Mygophenolig acid: antiviral and 75. Schwartz JB. The influence of sex on pharmacoki-
antitumor properties. J Antibiot. 1968;21(7):463–4. netics. Clin Pharmacokinet. 2003;42(2):107–21.
58. Mitsui A, Suzuki S. Immunosuppressive effect of 76. Kovarik JM, Koelle EU. Cyclosporin pharmacokinet-
mycophenolic acid. J Antibiot. 1969;22(8):358–63. ics in the elderly. Drugs Aging. 1999;15(3):197–205.
59. Klupp J, et al. Mycophenolate mofetil added to 77. Staatz CE, Tett SE. Clinical pharmacokinet-
immunosuppression after liver transplantation–first ics and pharmacodynamics of tacrolimus in
results. Transpl Int. 1997;10(3):223–8. solid organ transplantation. Clin Pharmacokinet.
60. Shaw LM, et al. Current opinions on therapeutic 2004;43(10):623–53.
drug monitoring of immunosuppressive drugs. Clin 78. Reuben A. Hepatotoxicity of immunosuppressive
Ther. 1999;21(10):1632–52. drugs. In: Drug-induced liver disease. Elsevier;
61. Morris RE. Rapamycins: antifungal, antitumor, anti- 2013. p. 569–91.
proliferative, and immunosuppressive macrolides. 79. Wijdicks EF. Neurotoxicity of immunosuppressive
Transplant Rev. 1992;6(1):39–87. drugs. Liver Transpl. 2001;7(11):937–42.
62. Kahan BD, et al. Therapeutic drug monitoring of 80. Bechstein WO. Neurotoxicity of calcineurin inhibi-
immunosuppressant drugs in clinical practice. Clin tors: impact and clinical management. Transpl Int.
Ther. 2002;24(3):330–50. 2000;13(5):313–26.
63. Almond PS, et al. Rapamycin: immunosuppres- 81. Anghel D, et al. Neurotoxicity of immunosuppres-
sion, hyporesponsiveness, and side effects in a sive therapies in organ transplantation. Maedica.
porcine renal allograft model. Transplantation. 2013;8(2):170.
1993;56(2):275–81. 82. Scott J, Higenbottam T. Adverse reactions and inter-
64. Yocum DE. Cyclosporine, FK-506, rapamycin, actions of cyclosporin. Med Toxicol Adverse Drug
and other immunomodulators. Rheum Dis Clin. Exp. 1988;3(2):107–27.
1996;22(1):133–54. 83. Kahan B, et al. Complications of cyclosporine-­
65. Whiting P, et al. Toxicity of rapamycin--a com- prednisone immunosuppression in 402 renal
parative and combination study with cyclospo- allograft recipients exclusively followed at a single
rine at immunotherapeutic dosage in the rat. center for from one to five years. Transplantation.
Transplantation. 1991;52(2):203–8. 1987;43(2):197–204.
66. DiJoseph JF, Mihatsch MJ, Sehgal SN. Renal effects 84. McGaw T, Lam S, Coates J. Cyclosporin-induced
of rapamycin in the spontaneously hypertensive rat. gingival overgrowth: correlation with dental plaque
Transpl Int. 1994;7(2):83–8. scores, gingivitis scores, and cyclosporin levels in
67. Chan C-C, et al. Side effects of rapamycin in the rat. serum and saliva. Oral Surg Oral Med Oral Pathol.
J Ocul Pharmacol Ther. 1995;11(2):177–81. 1987;64(3):293–7.
68. Ochiai T., et al. Effect of a new immunosuppressive 85. King GN, et al. Prevalence and risk factors associ-
agent, FK 506, on heterotopic cardiac allotrans- ated with leukoplakia, hairy leukoplakia, erythema-
plantation in the rat. Transplant Proc. 1987;19(1 tous candidiasis, and gingival hyperplasia in renal
196 A. Shakya et al.

transplant recipients. Oral Surg Oral Med Oral of posttransplant diabetes in renal allograft recipi-
Pathol. 1994;78(6):718–26. ents. Transplantation. 1987;44(3):376–81.
86. Kilpatrick N, et al. Gingival overgrowth in pediatric 94. Tornatore KM, et al. Methylprednisolone pharma-
heart and heart-lung transplant recipients. J Heart cokinetics, cortisol response, and adverse effects
Lung Transplant. 1997;16(12):1231–7. in black and white renal transplant recipients.
87. Danesi R, Del Tacca M. Hematologic toxicity of Transplantation. 1995;59(5):729–36.
immunosuppressive treatment. In: Transplantation 95. Kirk JK, Dupuis RE. Approaches to the treatment of
proceedings. Elsevier; 2004;36(3):703–704. https:// hyperlipidemia in the solid organ transplant recipi-
doi.org/10.1016/j.transproceed.2004.03.016. ent. Ann Pharmacother. 1995;29(9):879–91.
88. Lowenthal RM, Eaton K. Toxicity of chemotherapy. 96. Fung JJ. Tacrolimus and transplantation: a decade in
Hematol Oncol Clin. 1996;10(4):967–90. review. Transplantation. 2004;77(9):S41–3.
89. Connell W, et al. Bone marrow toxicity caused by 97. Elion GB. The pharmacology of azathioprine. Ann
azathioprine in inflammatory bowel disease: 27 N Y Acad Sci. 1993;685(1):401–7.
years of experience. Gut. 1993;34(8):1081–5. 98. Matell G. Immunosuppressive drugs: azathioprine in
90. Bagni B, et al. Continuing loss of vertebral mineral the treatment of myasthenia gravis. Ann N Y Acad
density in renal transplant recipients. Eur J Nucl Sci. 1987;505:589–94.
Med. 1994;21(2):108–12. 99. Sehgal SN. Sirolimus: its discovery, biological prop-
91. Rodino MA, Shane E. Osteoporosis after organ erties, and mechanism of action. In: Transplantation
transplantation. Am J Med. 1998;104(5):459–69. proceedings. Elsevier; 2003;35(3 Suppl):7S–14S.
92. Meys E, et al. Bone loss after orthotopic liver trans- https://doi.org/10.1016/s0041-­1345(03)00211-­2.
plantation. Am J Med. 1994;97(5):445–50. 100. Pham K, et al. Limited-dose daclizumab vs. basilix-
93. Boudreaux JP, et al. The impact of cyclosporine and imab: a comparison of cost and efficacy in preventing
combination immunosuppression on the incidence acute rejection. Transplantation. 2004;78(2):277–8.
Therapeutic Drug Monitoring
and Toxicology: Relevance
13
of Measuring Metabolites

James Akingbasote, Sandra Szlapinski,


Elora Hilmas, Patrik Miller, and Natalie Rine

Abstract and mechanisms involved in metabolite-­


induced drug toxicity are discussed, drawing
Therapeutic drug monitoring (TDM) is a valu- from several examples of drugs that are used
able tool that healthcare providers have in the clinical setting. This chapter empha-
employed to optimize therapeutic efficacy sizes the utility of TDM of metabolites, dem-
of drugs while also maintaining drug concen- onstrating how select drugs can be converted
trations below toxic levels. Although TDM to reactive metabolites with toxicity potential,
has predominantly focused on determining the and these should be considered when evaluat-
level of the parent drug compound in order to ing the efficacy and toxicity of a drug. The
guide the dosing regimen, it is important to impact of pharmacogenetics in drug metabo-
consider that some drugs are inactive in their lism and the potential impact on efficacy and
parental forms and only their metabolites have safety of drugs subject to TDM are also dis-
therapeutic applications. Moreover, some cussed. Finally, a cost-benefit analysis is made
drug metabolites possess distinct toxicity pro- to support the inclusion of metabolite determi-
files from the parental compounds which are nation in routine TDM practice toward optimi-
important to consider when evaluating the zation of drug therapy.
toxicity of a drug as a whole. This chapter
investigates the role of metabolites in the Keywords
safety assessment of drugs that are subject to
TDM, such as those with narrow therapeutic Bioactivation · Cytochrome P450 ·
windows. Examples of TDM of metabolites Metabolites · Polymorphism · Pro-drugs ·
Uridine diphosphate glucuronosyltransferase

J. Akingbasote (*) · S. Szlapinski


Regulatory Toxicologist, London, ON, Canada
13.1 Brief Overview
E. Hilmas · P. Miller of Therapeutic Drug
Nationwide Children’s Hospital, Columbus, OH,
USA Monitoring (TDM)
e-mail: Elora.Hilmas@nationwidechildrens.org;
Patrik.Miller@nationwidechildrens.org As described in previous chapters in this book,
N. Rine TDM involves the determination of drug concen-
Central Ohio Poison Center, Nationwide Children’s trations in the plasma, serum, or blood during
Hospital, Columbus, OH, USA treatment or for diagnostic purposes. In the clini-
e-mail: Natalie.Rine@nationwidechildrens.org

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 197
S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_13
198 J. Akingbasote et al.

cal setting, TDM is aimed at guiding drug dosing essential to ensure that clinically meaningful
regimens to ensure that drug concentrations drug concentrations are attained since it relies on
remain within their target range in the blood [1]. a rapid and accurate determination of drug levels
This is especially important to guide dosing for [1, 3]. The accurate interpretation of analytical
drugs with variable or unpredictable pharmacoki- readouts of drug concentrations by the multidis-
netics and for drugs with narrow therapeutic indi- ciplinary team requires information on the time
ces, to guide withdrawal of drug therapy, to of blood sample collection, dose administered,
monitor and detect drug interactions, to monitor dosage regimen, patient demographics, comedi-
efficacy failure, to monitor unexpected treatment cations, indication for monitoring and pharmaco-
failures or adverse effects, and to monitor drug kinetics, and therapeutic range of the drug [8],
compliance particularly for drugs with non-­ with an ultimate goal of ensuring accurate drug
readily observable therapeutic responses and for dosing to bring about an optimal therapeutic
drugs with long-term therapy (e.g., anticonvul- effect while reducing toxicity.
sants) and life-threatening situations (e.g., epi-
lepsy or sepsis) [2–6]. While TDM may help
account for pharmacokinetic differences among 13.1.1 Brief History of TDM
drugs, pharmacokinetics of these drugs can also
be further complicated by intra- or inter-­ While TDM presents as a powerful tool for ensur-
individual variability due to body size and com- ing an optimal benefit in drug dosing, its clinical
position, age, organ function, disease states and benefits have been underappreciated in the
differences in expression and activity of drug-­ present-­day clinical setting [9]. Nonetheless, the
metabolizing enzymes (e.g., cytochrome P450 critical influence of dosage in a drug regimen has
(CYP450) and uridine diphosphate glucuronos- long been acknowledged in toxicology and can
yltransferase (UGT), among others), enzyme be attributed to the Renaissance physician
induction/inhibition, and drug-drug interaction Paracelsus (1493–1541) credited to be the “Father
[4]. These differences are especially important of Toxicology,” who famously stated:
for drugs that have a narrow target range or those What is there that is not poison? All things are poi-
with concentration-dependent pharmacokinetics son and nothing is without poison. Solely the dose
[1]. Faced with these factors, TDM is a very determines that a thing is not a poison [10].
important tool in the hand of healthcare providers
in order to optimize therapeutic efficacy while Therefore, the scientific community has known
also maintaining drug concentrations below toxic for centuries that the dosage of a drug is impor-
levels. tant for optimal patient outcomes. However, it
In TDM, drug concentrations are quantified was not until 1932 that the Swedish Scientist,
using techniques such as radioimmunoassay, Erik Widmark, demonstrated the ability to moni-
high-performance liquid chromatography tor the blood concentration of a drug after he
(HPLC), fluorescence polarization immunoassay developed two formulae commonly used today
(FPIA), enzyme-multiplied immunoassay tech- for the determination of blood alcohol levels
nique (EMIT), enzyme-linked immunosorbent [11]. A classic historical example that demon-
assay (ELISA), and chemiluminescence immu- strates the importance of TDM is lithium – origi-
noassay (CLIA), among others. The method cho- nally used as used as a sodium-free table salt for
sen for TDM must be sensitive, must be specific individuals with hypertension in the 1940s before
for the detection of the compound of interest, being withdrawn due to reports of severe toxicity
must be feasible for use as a routine assay, and and death [2]. However, there were also concur-
must be able to distinguish the drug of interest rent reports of improvement in symptoms in
from other co-administered drugs [7]. hypertensive patients with manic-depressive dis-
For robust TDM, a multidisciplinary team of orders who used lithium. Despite its potential
scientists, clinicians, nurses, and pharmacists is therapeutic benefits, the inability to accurately
13 Therapeutic Drug Monitoring and Toxicology: Relevance of Measuring Metabolites 199

monitor plasma levels of lithium so as to ensure large dataset generated by the Human Genome
that drug concentrations were not in the toxic Project between 1990 and 2001 [20]. Considering
range, led to its withdrawal [2], and it would be a that there are numerous reported genetic poly-
few more years before TDM was finally intro- morphisms that affect the disposition of drugs,
duced, thereby enabling the establishment of the TDM can be combined with pharmacogenetics in
therapeutic dose range of lithium [12, 13]. order to optimize pharmacotherapy for patients
By the 1960s, landmark papers were pub- [2]. In these situations, a patient receives a dose
lished that introduced TDM as a new aspect of of the drug based on a pharmacogenetic assess-
clinical practice based on a publication of phar- ment, after which conventional TDM can be used
macokinetic studies that associated mathematical to ensure optimal patient outcomes [2]. Scientific
theories to patient outcomes; there was also a advances of TDM were also supported by the
review published outlining the importance of introduction of noninvasive and minimally inva-
drug monitoring [14, 15]. In the late 1960s to sive methods to be used for monitoring, including
early 1970s, a new discipline known as “clinical wearable sensors. For example, the first wearable
pharmacokinetics” emerged, which enabled sci- sensor that consists of an ingestible sensor for
entists to study adverse drug reactions and estab- TDM was introduced in 2013 for monitoring
lish therapeutic ranges for drugs so as to reduce adherence to anti-tubercular therapy [21]. The
the incidence of their toxicity [3]. system was able to correctly identify the ingest-
Improvements in analytical technology and ible sensors with high accuracy and confirm med-
high-throughput computerization subsequently ication compliance while also posing a low risk
resulted in significant advancements in the field to users. On the other hand, another minimally
of clinical pharmacokinetic monitoring [16]. For invasive technique was introduced in 2016 which
instance, the development of chromatographic relied on painless hollow microneedles for drug
techniques such as gas chromatography, high-­ quantification rather than requiring a blood draw
performance liquid chromatography (HPLC), [22]. The needles were suitable to extract small
and mass spectrometry enabled the monitoring of volumes of interstitial fluid and drug analytes
the concentration of drugs, while the use of which were rapidly quantified with high sensitiv-
immunoassays for saliva samples facilitated the ity using appropriate detection techniques. A fast
ease of performing TDM [9, 17]. In 1978, the and versatile multianalyte (up to eight enzyme-­
applicability of TDM was further demonstrated linked assays) single-use biosensor assay was
as simultaneous quantification of drugs was also introduced using human plasma for simulta-
reported, demonstrating the powerful potential of neous detection of antibiotics in less than 15 min-
TDM on patient outcomes in a clinical setting utes [23]. In 2019, the clinical relevance of TDM
where patients often co-administered drugs [18]. was further demonstrated as the World Health
The clinical utility of TDM was demonstrated as Organization included TDM on the list of essen-
early as 1981, wherein TDM of the beta-lactam tial in vitro diagnostic tests for a list of drugs to
antibiotic, cefoperazone, was reported in patients be monitored on a priority basis [24].
[19]. Cefoperazone displays differing pharmaco- Despite the plethora of advancements in
kinetics relative to other first-generation cephalo- TDM, the first in-human study for continuous
sporins (e.g., cephalothin, cephapirin), as it is not drug monitoring in healthy volunteers was only
readily metabolized and has a much longer serum published in 2019. In this study, TDM was used
half-life, necessitating the use of TDM in patients for monitoring phenoxymethylpenicillin using a
taking the drug. minimally invasive microneedle-based beta-­
From the 1990s onward, the advancement of lactam biosensor [25]. While chromatographic
much more sophisticated techniques further techniques have largely been used in the field,
advanced the field of TDM as pharmacogenetics they are limited due to the lack of standardiza-
and pharmacogenomics research found its appli- tion, high turnaround time and instrumentation
cation in TDM; this was made possible by the cost, and laborious sample preparation process
200 J. Akingbasote et al.

[9]. Furthermore, sample preparations requiring for determining blood levels of the parent com-
blood draws pose a hindrance as a potentially pound. In clinical practice, the use of drug trough
painful process to many individuals. As such, the levels (drug level that is drawn at the point where
scientific advancements in recent years using the amount of drug in the body would be at its
noninvasive or minimally invasive methods for lowest amount when the drug level is at a steady
TDM will be revolutionary for the feasibility and state in the blood) is commonplace [30]. An
applicability in the field. example of the use of drug troughs is the moni-
toring of vancomycin. If the vancomycin is given
every 8 hours, a trough is usually drawn after
13.1.2 Importance of TDM in Clinical three doses, and the blood is obtained right
Settings before the 4th dose is given [31]. Since a few
doses have been given, it would be expected that
As shown above, therapeutic drug monitoring a measurable amount of the drug be detected in
(TDM) is an important clinical tool that ensures the body when a trough is drawn due to drug
maximum therapeutic effectiveness and avoids accumulation [31]. Troughs can be used in
toxicity for medications with narrow therapeutic ensuring that the medications do not accumulate;
indices [3]. While most drugs in clinical practice achieving a certain trough has been shown to
have a wide therapeutic index [3], there are sev- correlate with treatment success [30]. On the
eral agents with narrow therapeutic windows for other end of the spectrum of the parameters mea-
which TDM is required to determine dosing and sured in TDM are drug peaks, which correlate
dosing frequency and to prevent toxicity. with the highest detectable drug concentration.
Medications are often developed to provide a Drug peaks are important in ensuring that target
therapeutic effect at a standard dose; however, ranges are not exceeded, in mitigating unwanted
due to patient variability, there can be quite a toxicities, and in ensuring therapeutic success
wide range of drug exposure leading to differ- [3]. The importance of drug peak measurement
ences in efficacy and toxicity that results from is illustrated in the case of amikacin where the
the manufacturer-recommended standard dose use of an extended interval dosing strategy
[26]. Use of TDM aligns with precision medi- helped achieve a peak drug concentration, which
cine as it allows individualized dosing to assist directly correlated with a successful antimicro-
clinicians in determining the right dose to give to bial effect [32]. Since a higher plasma concen-
a specific patient [26]. In the age of tremendous tration of amikacin is required for the
advancements in pharmacogenomics, where the management of sepsis [33], the measurement of
genotyping of an individual patient’s drug- its peak concentration is an important step to
metabolizing enzymes or transporters is becom- ensuring efficacy [34]. Finally, another approach
ing more commonplace, it may be assumed that in TDM is the measurement of several sequential
the practice of TDM may eventually be retired. levels such as through the estimation of area
However, due to the fact that TDM can achieve under the curve (AUC). In this approach, thera-
target drug concentrations in patients more accu- peutic success is determined by the exposure of
rately than pharmacogenomics, perhaps the bet- the drug in the body over time, such as in the
ter approach is to use TDM and cases of vancomycin and mycophenolate [30,
pharmacogenomics together to achieve the high- 34, 35]. Beyond measuring blood levels of the
est level of precision medicine [26]. Examples of parent compound is the determination of the
drug classes that may require TDM include anti- level of surrogate efficacy markers which are
biotics, antiepileptics, and antineoplastics [27]. measurable indicators of the intended clinical
While TDM has primarily focused on measuring outcome [36]. In the course of therapy with the
levels of the parent compound, metabolite moni- anticoagulant drug, enoxaparin, the surrogate
toring is more of an academic exercise [28, 29]. marker of efficacy is a measurement of the inhi-
A wide variety of approaches have been taken bition of clotting factor Xa [37]. In the case of
13 Therapeutic Drug Monitoring and Toxicology: Relevance of Measuring Metabolites 201

Table 13.1 List of drug classes requiring TDM


Drug class Example medications References
Antineoplastics Methotrexate, mercaptopurine, carboplatin, busulfan, thioguanine, azathioprine [1–5]
Immunomodulators Tacrolimus, sirolimus, mycophenolate, cyclosporine [6–9]
Antiepileptics Valproic acid, phenytoin, phenobarbital, levetiracetam, carbamazepine [2, 10–12]
Anticoagulants Enoxaparin, heparin, bivalirudin, argatroban, fondaparinux [13]
Antifungals Posaconazole, voriconazole, itraconazole [14, 15]
Antibiotics Amikacin, tobramycin, gentamicin, vancomycin [2, 16, 17]
Antivirals Valganciclovir [18]
Antipsychotic Lithium [19]
Antiarrhythmics Digoxin, lidocaine, procainamide, quinidine [19, 20]
Respiratory agents Theophylline, caffeine [19]

warfarin, prothrombin time serves as the surro- of gene and gene expression on the response of
gate efficacy marker which is employed in the individual patients to drugs to which they are
calculation of international normalized ratio exposed [46].
(INR), a ­measurement used to determine effi- The impact of pharmacogenomics on TDM is
cacy and safety of medications that affect clot- aptly illustrated in the case of tacrolimus, a com-
ting time [38, 39]. monly used immunosuppressant with a narrow
In clinical practice there are a wide variety of therapeutic window [47]. Tacrolimus, in complex
drug classes where dose individualization based with immunophilin, binds to calcineurin and
on TDM has been a part of medical management inhibits calcineurin phosphatase activity [48]
[3, 27, 34, 37, 40, 41]. In Table 13.1, a listing of which results in an inhibition of calcium-­
drug classes where TDM is commonly used dependent activities including nitric oxide syn-
along with specific examples is shown. thase activation, cell degranulation, and cytokine
production (proinflammatory IL-2, which results
in suppressed T-cell proliferation) [45]. Hence,
13.1.3 Current Clinical Practice Gaps tacrolimus is effective in preventing xenograft
Associated with Measuring attack [49]. TDM is an important part of tacroli-
Parent Compounds mus therapy, and regular blood draws of whole
blood tacrolimus levels are critical to guide thera-
Current clinical practice often focuses on the peutic management. Elevated tacrolimus levels
measurement of blood levels of parent compound are associated with increased risk of nephrotoxic-
administered. However, drugs are often biotrans- ity, hypertension, and hyperglycemia [50], while
formed within the biological system with the a subtherapeutic blood level of tacrolimus results
active entities being the metabolites thereof as is in rejection of the implanted organ thereby caus-
the case with pro-drugs. A pro-drug is a com- ing organ dysfunction or failure [47]. One key
pound which after having been administered is factor involved in this varied response to tacroli-
metabolized within a body compartment into a mus therapy that is observed among various
pharmacologically active drug [42, 43]. Some patient populations is the polymorphism in the
examples of pro-drugs include codeine, trama- expression and activity of the enzymes princi-
dol, clopidogrel, and mercaptopurine [41, 44, pally responsible for its metabolism – cyto-
45]. However, because not all patients can bio- chromes P450 (CYP) 3A4 and 3A5 [51]. In 2015,
transform the parent compound into the active guidelines were published by the Clinical
compound to the same extent, a field of research Pharmacogenetics Implementation Consortium
known as pharmacogenetics (or pharmacoge- (CPIC) to provide initial dosing guidance based
nomics) has been developed to study the effects on an individual patient’s genotype [51]. They
202 J. Akingbasote et al.

suggested that extensive and intermediate metab- distinct therapeutic and toxicity profiles that dif-
olizers tend to have lower tacrolimus trough con- fer from the parental compound. In such a case,
centrations and a decreased chance of achieving there is need for the determination of the concen-
target tacrolimus concentrations. Therefore, their tration of the parental compound and its metabo-
initial doses were recommended to be 1.5–2 lite in biological fluids in order to show the
times the recommended starting dose of normal relationship between the amount detected and
metabolizers. Although poor metabolizers may their pharmacological effects. While this may not
have higher trough concentrations, the recom- be part of what is typically done on a day-to-day
mendation for this population is to start with basis in clinical practice, it is key to recognize the
standard recommended doses [51, 52]. In most overall impact of measurement of metabolites in
cases, therapeutic drug monitoring is recom- such a scenario. As an example,
mended to guide any dose adjustments after ini- N-acetylprocainamide (acecainide), a metabolite
tiation [45]. Although tacrolimus is not a pro-drug of procainamide, has been shown to possess anti-
and may not have an active metabolite, this arrhythmic potency, similar to the parental com-
example demonstrates the impact of metabolism pound [53–56]. Given the fact that this metabolite
and its variability within a patient population. of procainamide possesses similar (and possibly
The remainder of this chapter discusses the equal) pharmacological activity as the parental
importance of metabolites in TDM and drug compound, a knowledge of the metabolite plasma
safety, the mechanism of metabolite-induced concentration would be key in determining dos-
drug toxicity, and the impact of pharmacogenet- ing so as to modulate efficacy and reduce any
ics in drug metabolism and the potential impact potential toxicity. Other drugs with active metab-
on efficacy and safety of drugs subject to TDM. olites include losartan [57], whose metabolite
E-3174 has 10- to 40-fold the potency of the par-
ent compound [58] and is implicated in “on-­
13.2 Metabolites and Drug target” effects which are directly related to the
Monitoring primary mechanism of action of the parent com-
pound. Alternatively, there are drugs, metabolites
Although TDM is principally concerned with the of which are involved in secondary effects or
measurement of parent compounds with direct those characterized as “off-target.” In these cases,
clinical use, its use can also be applied to the the metabolites result in pharmacological effects
measurement of metabolites of the parental com- different from those of the parent compound.
pounds. Metabolite measurement is important in Such is the case with fenfluramine, an anorectic
TDM because some drugs including those with drug [59] whose metabolite(s) (+)- and (−)-nor-
narrow therapeutic windows are converted to fenfluramine have diverse pharmacological
reactive metabolites and these need to be ana- effects including substrate activity at norepineph-
lyzed. Furthermore, some drugs are inactive in rine transporter (NET) to release norepinephrine
their parental forms, and only their metabolites (NE) and direct agonist activation of
have therapeutic applications. Since some of 5-­hydroxytryptamine, 5-HT2B and 5-HT2C,
these metabolites have a narrow therapeutic win- receptor subtypes [60]. Carbamazepine, a drug
dow, they require the application of TDM. with a narrow therapeutic window, is also metab-
olized to carbamazepine-10,11-epoxide, which is
an active metabolite of this anti-convulsant drug
13.2.1 Importance of Metabolite [61]. It is imperative to measure levels of this
Measurements in TDM metabolite as carbamazepine overdose has been
reported not only with the parent compound but
As discussed in a review by Kang and Lee [3], in association with this metabolite as well [62]. A
one of the difficulties inherent in TDM is the pos- case was reported by Russell et al. [63] of an
sible presence of metabolites which may have overdose of carbamazepine wherein the level of
13 Therapeutic Drug Monitoring and Toxicology: Relevance of Measuring Metabolites 203

carbamazepine-10,11-epoxide was shown to be the Pharmaceutical Research and Manufacturers


450% higher than the parent compound. The Association (PhRMA) which highlighted the
authors suggested that this metabolite played a need for more rigorous studies to investigate the
role in the carbamazepine toxicity. Doxorubicin role of metabolites in the safety assessment of
is an anthracycline antineoplastic drug with a low drugs with narrow therapeutic windows, such as
therapeutic index that induces cardiovascular those which are primarily measured in TDM
toxicity [64–66]. This toxicity is attributed to its [76]. They also determined that if metabolites
metabolite, doxorubicinol [67], which upon constitute a minimum of 25% of the circulating
accumulation following long-term use causes drug-related material in the study system, such a
myocardial toxicity in humans [65]. Since car- metabolite should be considered as being of a
diotoxicity results from the accumulation of safety concern. However, in the review by
doxorubicin and doxorubicinol in cardiovascular Robison and Jacobs [74], there are other cases in
tissues, monitoring plasma levels of this drug and which further safety study of metabolites might
its metabolite in breast cancer patients is key to not be required such as in the case of metabolites
minimizing toxicity [68]. with no structural alerts for reactive products.
Other drugs like acetaminophen, when taken The nature of the metabolite is also an important
at a high dose, form the highly reactive metabo- factor to consider. Therefore, while oxidative
lite, N-acetyl-p-benzoquinone imine (NAPQI), metabolites (commonly called phase I metabo-
with hepatotoxic potential [69]. Further, other lites) are readily linked to reactive species with
drugs such as troglitazone [70], felbamate [71], potential for toxicity, conjugative metabolites
and diclofenac [72] also generate metabolites (commonly called phase II metabolites) are
which induce idiosyncratic drug toxicity. In the mostly water soluble and easily excreted. The
example of procainamide above, it has been general assumption is that most oxidative metab-
shown that the procainamide-induced agranulo- olites would require further safety evaluation
cytosis was due to the formation of protein free provided other criteria are met, while conjugative
radicals [73]. Since there is potential of forma- metabolites do not require further evaluation
tion of pharmacologically active metabolites and [77]. An exception to this assumption lies in the
metabolites with toxicity potentials, metabolite fact that other products of conjugation like acyl
measurement can help optimize therapy and glucuronides and acyl-CoA thioesters are unsta-
abrogate possible toxicity in the clinical setting. ble and have been shown to interact covalently
The importance of metabolite measurement with cellular macromolecules to cause organ-­
has been highlighted in FDA’s Center for Drug system toxicity [78]. This is particularly true for
Evaluation and Research Guidance for Industry drug molecules that contain a carboxylic acid
on Safety Testing of Metabolites and the moiety, 14% of which were withdrawn from the
International Conference on Harmonization global pharmaceutical market in the past century
(ICH) which provided guidance on the amount of due to toxicity [79]. This would be a concern for
metabolite relative to the parent compound which carboxylic acid-containing drugs such as levo-
is of safety concern. The threshold of human thyroxine, valproic acid, argatroban, mycopheno-
metabolites which can raise a safety concern can lic acid, methotrexate, and vincristine which have
be as little as 10% of the parent drug concentra- been shown to have a narrow therapeutic window
tion during systemic exposure at steady state [80–86]. Although the metabolism of these drugs
[74], and the recommendation is that a series of may not be primarily through the glucuronidation
tests be conducted to evaluate the safety of pathway, the mere presence of the carboxylic
metabolites in a manner similar to the parent acid group is a potential structural alert for the
compound [75]. A similar stance was adopted by generation of a reactive metabolite [78, 87]. The
204 J. Akingbasote et al.

relationship between drug metabolism and toxic- therapy [96]. Another layer of complexity associ-
ity will be discussed in Sect. 13.2.3. ated with carbamazepine therapy lies in the fact
that it is a potent inducer of CYP3A4, other oxi-
dative enzymes involved in its own metabolism
13.2.2 Metabolite Measurement as well as the conjugative enzyme family of gluc-
in TDM uronosyltransferases [97]. CYP3A4 is polymor-
phic [98] as are other enzymes involved in the
In the context of TDM and metabolite determina- metabolism of carbamazepine, making for a wide
tion following oral administration, highly reac- variation in the metabolism of carbamazepine
tive metabolites may form other unstable with a potential for toxicity in the case extensive
metabolites that are difficult to detect in biologi- metabolizers via the oxidative pathway [99]
cal fluids like plasma due to their relatively short (Fig. 13.2).
half-lives [74]. However, the potential they have The examples cited above – valproic acid,
to form stable conjugates with endogenous mol- phenytoin, carbamazepine, and mycophenolic
ecules like glutathione and cysteine which can be acid – are all drugs in clinical use and which have
readily measured can provide insight into their a narrow therapeutic window. While the mainstay
formation and presence in the plasma [88]. This of clinical practice is to determine the levels of
has been reported in the case of acetaminophen the administered drug compound, the fact that
(APAP) which is primarily metabolized by some of the metabolites of these drugs have tox-
CYP2E1 to form N-acetyl-p-benzoquinone imine icity potential is a reason that metabolite mea-
(NAPQI) [89], and NAPQI readily reacts with surement is important in TDM. This will help
hepatic glutathione (GSH) to form the detoxified mitigate the overall risk posed by exposure to the
product APAP-GSH [89]. This concept is further parent compound and the active substance, in
discussed below. Other examples of drugs that addition to possibly toxic metabolites [3]. As a
are metabolized to toxic metabolites include val- consequence, metabolite measurement is war-
proic acid which is metabolized to 4-ene-valproic ranted in TDM for:
acid [90] (Fig. 13.1), mycophenolic acid which is
glucuronidated to an acyl glucuronide [91], phe- 1. Drugs with metabolites possessing pharmaco-
nytoin which is metabolized to logical actions, e.g., procainamide and carba-
5-(4′-hydroxyphenyl)-5-phenylhydantoin which mazepine [100, 101]
is produced via the formation of a reactive arene 2. Pro-drugs, e.g., tenofovir disoproxil [102]
oxide intermediate [92], and carbamazepine 3. Drugs which produce metabolites with estab-
which is converted to carbamazepine-10,11-­ lished toxicity, e.g., doxorubicin, procain-
epoxide [62]. Although carbamazepine-10,11-­ amide, daunorubicin, and valproic acid [103]
epoxide is an active metabolite with a similar 4. Drugs with high variability in their pharmaco-
pharmacology to the parent compound, the fact kinetics (e.g., absorption, distribution, metab-
that it is an epoxide poses a structural alert for olism, and elimination), e.g., carbamazepine,
toxicity. Epoxides are highly reactive electro- thiopurines, clopidogrel, and codeine
philic moieties that have been shown to induce [103–105]
genetic toxicity [93, 94]. Little wonder authors 5. Drugs known to demonstrate a nonlinear rela-
have suggested that the toxicity seen in carbam- tionship between the administered dose and
azepine might be associated with the formation blood/serum/plasma concentrations, e.g., phe-
of its epoxide metabolite [63, 95], and other nytoin and theophylline [104, 105]
authors have shown a positive correlation
between the blood level of carbamazepine-­10,11-­ Because some drugs with narrow therapeutic win-
epoxide and adverse effects in carbamazepine dows have been shown to produce reactive metab-
13 Therapeutic Drug Monitoring and Toxicology: Relevance of Measuring Metabolites 205

Fig. 13.1 Valproic acid metabolism and bioactivation. 3-oxo-VPA, CoA-SH, and propionyl-CoA and pentanoyl-­
There are three routes of VPA metabolism in humans, CoA. Bioactivation of VPA involves the entry of 4-ene-­
including glucuronidation (major route, 50%), beta-­ VPA into the mitochondria, followed by conversion to
oxidation in the mitochondria (major route, 40%), and 4-ene-VPA-CoA ester via 2-methyl-branched chain acyl-­
cytochrome P450 (CYP)-mediated oxidation (minor CoA dehydrogenase. Subsequently, beta-oxidation forms
route, 10%). It has been shown that CYP2C9 and CYP2A6 the reactive 2,4-diene-VOA-CoA ester. This putative cyto-
are the key enzymes involved in CYP-mediated oxidation toxic metabolite gets further conjugated with glutathione
of VPA, while CYP2B6 is a less predominant route. Also, and together with 2,4-diene-VPA-CoA ester can deplete
glucuronidation of VPA is mediated by several UGTs mitochondrial glutathione pools and form conjugates with
including UGT1A4, UGT1A10, UGT1A3, UGT1A9, CoA, ultimately inhibiting enzymes involved in the beta-­
UGT1A8, UGT1A6, UGT2B7, and UGT2B15. In the oxidation pathway. (Figure created with BioRender.com)
mitochondria, mitochondrial beta-oxidation generates

olites which need to be measured in biological toxic and have been shown to result in severe
fluids, the following section gives an overview of organ/system damage, as was observed with
the mechanisms by which these metabolites acetaminophen (APAP) [106]. In the example
induce toxicity in the biological system. below, the drug acetaminophen is not a drug
requiring TDM. It is, however, a classical
example of a drug that produces toxic metabo-
13.2.3 Drug Metabolism and Toxicity lites which may need to be measured and moni-
tored as a determinant of its safety in cases of
As described in the sections above, in the overdose.
majority of situations, the parent drug is the Reviewing the metabolism of acetaminophen
active compound, and alteration in its metabo- in an overdose scenario provides some insight on
lism can greatly impact its efficacy, as was the impact of metabolism on drug toxicity.
observed with tacrolimus [47]. However, there Although acetaminophen is one of the most com-
are cases in which the metabolites of a drug are monly used analgesic and antipyretic medica-
206 J. Akingbasote et al.

Fig. 13.2 Metabolism and reactive metabolites of carba- tion of proinflammatory cytokines and chemokines,
mazepine. Carbamazepine undergoes hepatic metabolism resulting in liver inflammation. In excess, the active
mediated by several CYP enzymes. Select metabolites of metabolite carbamazepine 10,11 epoxide has also been
carbamazepine, including 2-OH carbamazepine and 3-OH associated with adverse events including toxicity, over-
carbamazepine, have been shown to induce the production dose, and genetic toxicity. Similarly, excessive intake of
of reactive oxygen species in macrophages. The macro- carbamazepine itself can lead to toxicity and overdose.
phages subsequently release signals that lead to the secre- (Figure created with BioRender.com)

tions, it is one of the principal causes of acute aminophen adducts can be more accurate than
liver failure in industrialized nations [107]. acetaminophen levels because the adducts persist
Acetaminophen is metabolized by CYP2E1 to much longer in serum with a half-life of 1–2 days
yield N-acetyl-p-benzoquinone imine (NAPQI), compared to the half-life of acetaminophen
a highly reactive compound that forms adducts which is typically around 5 hours [111]. A high
with cellular macromolecules, including glutathi- proportion of ingested acetaminophen is metabo-
one. Conjugation of NAPQI with the cysteine lized via glucuronidation and catalyzed by uri-
residue of glutathione molecule results in its dine 5′-diphospho-glucuronosyltransferase to
depletion and removal from the body [108]. yield APAP-glucuronide or by sulfotransferases
Additionally, the interaction between NAPQI and thereby enhancing the renal elimination of the
glutathione has been shown to be involved in the drug [112, 113]. Another reaction that occurs is
inactivation of NAPQI leading to APAP-CYS glutathionylation, but this usually occurs after
which can be measured in the plasma as a bio- phase I metabolism [114].
marker of impending liver toxicity [109]. Studies Although APAP is not a drug that typically
in the past decades have demonstrated novel requires TDM, a knowledge of the levels of cir-
methods of efficiently quantifying the formation culating metabolites can better inform clinical
of toxic metabolites via the measurement of decision-making for specific patients and creates
acetaminophen-­protein adduct levels in human an opportunity to implement precision medicine
serum [106, 110, 111]. Measurement of acet- [115, 116].
13 Therapeutic Drug Monitoring and Toxicology: Relevance of Measuring Metabolites 207

13.2.3.1 An Overview of Mechanisms the liver [123–125]. In regard to conjugative


Involved in the Bioactivation metabolism of xenobiotics (also called phase II
of Drugs to Toxic Metabolites metabolism), reactions may involve a series of
Metabolism plays a key role in the conversion of enzymes including uridine 5′-diphospho-­
drugs, which are foreign substances to the body, glucuronosyltransferase (UDP-­
to various forms including less toxic, water solu- glucuronosyltransferase, UGT) which is a
ble, and excretable forms [117, 118]. While in microsomal enzyme belonging to the glycosyl-
many cases, metabolism is a detoxification pro- transferase family; they are involved in the trans-
cess, it can produce pharmacologically active fer of the glucuronic acid component of
forms of the compounds with therapeutic appli- UDP-glucuronic acid to a small hydrophobic
cations, as is observed with pro-drugs [119] (fur- molecule [126]. Other conjugative reactions
ther discussed in Sect. 13.4). In other cases, drugs include sulfation, acetylation, glutathionylation,
are metabolized to forms that are reactive with and methylation [89].
the potential to induce organ-system damage. The cytochrome P450 system catalyzes a
This organ-system damage can arise from the series of reactions that can bioactivate drugs and
interaction between these metabolites – many of other xenobiotics into toxic forms. Located pre-
which are electrophiles in nature – and cellular dominantly in liver microsomes, this enzyme
macromolecules like proteins, nucleic acids, or system catalyzes oxidative reactions such as C-,
unsaturated lipids, ultimately causing degrada- S-, and N-oxidations; O-, S-, and
tion of these macromolecules, cellular dysfunc- N-dealkylations; dehalogenation; and deamina-
tion, and cell death [120–122]. tion [127]. Other enzyme systems involved in
This section gives an overview of the mecha- drug metabolism include FAD-containing
nisms of toxicity induced by metabolites using monooxygenase which oxidizes nucleophilic
specific examples of, wherever applicable, drugs sulfur, nitrogen, and organophosphorus com-
with narrow therapeutic windows. Briefly, drug pounds [128]; xanthine oxidases which oxidize
metabolism can take place in different ways purine derivatives like theophylline and doxoru-
depending on the enzymes involved and the bicin, among others [129]; and alcohol dehydro-
chemical structure of the compound in question. genase and aldehyde dehydrogenase which
In what has been typically called a phase I oxidize a vast array of alcohol and aldehydes
reaction,1 drugs undergo different reactions into aldehyde and acids, respectively [130].
including oxidation, reduction, substitution, There are also peroxidases which catalyze the
hydrolysis, and elimination under the action of oxidation of hydrogen peroxide and organic
different enzyme systems including the cyto- peroxides [131]. Some of the reactions above
chrome P450 system, most of which is resident in are known to generate reactive metabolites in
drugs as in the case of chloramphenicol.
1
While it is common to refer to this as phase one reaction, Chloramphenicol is metabolized by the CYP
this is not an accurate description of the reactions that take monooxygenases to chloramphenicol oxamyl
place. Referring to this as phase I only suggests that they
take place first and other reactions follow. However, it has
chloride, which is produced via oxidative
been shown that the other kind of reaction in the liver, dechlorination of the dichloromethyl moiety of
which is generally referred to as phase II, may actually the compound and then the elimination of the
take place before “phase I,” simultaneously with “phase hydrochloric acid portion of the ensuing product
I,” or may even be the only major reaction that the xenobi-
otic undergoes. Therefore, phase I reactions are more
[132]. Chloramphenicol oxamyl chloride is a
accurately and broadly referred to as oxidative metabo- reactive metabolite that reacts with the epsilon-­
lism because a vast majority of reactions that take place amino group of the lysine portion of the CYP
via this route are oxidative in nature, while phase II reac- enzyme, which subsequently results in the inhi-
tions are more accurately referred to as conjugative
metabolism as the other chemical moieties are added
bition or inactivation of the enzyme [133, 134].
(conjugated) to the parent compound or its oxidative CYP enzymes have also been shown to catalyze
metabolite to create a new form. oxidation of unsaturated bonds into epoxides
208 J. Akingbasote et al.

which are reactive metabolites involved in the facilitating their excretion, some of these deriva-
toxicity of the unsaturated compounds. This is tives have been implicated in the toxicity of these
the case with aflatoxin B1 which is oxidized by compounds. UGT-mediated conjugative metabo-
the CYP enzyme system to aflatoxin B1 epoxide. lism is involved in the formation of acyl glucuro-
Aflatoxin B1 epoxide subsequently adducts with nides of carboxylic acid containing-drugs like
the guanine portion of DNA, thereby resulting diclofenac [148, 149]. The aromatic amines in
in hepatic carcinogenicity [135, 136]. A clini- these drugs can also be converted to hydroxyl-
cally relevant epoxide metabolite of a drug with amine o-glucuronide which after being broken
a narrow therapeutic window is that of carbam- down in the bladder forms a carcinogenic proxi-
azepine, as was discussed previously. mate hydroxylamine [150].
Carbamazepine is oxidized by CYP 3A4 to Since drugs can be bioactivated to toxic
carbamazepine-­10,11-epoxide which, although metabolites, an essential aspect of TDM should
possesses a similar efficacy profile to the parent include the measurement of metabolites espe-
compound [137], has been shown to induce cially in cases where metabolites have been
adverse drug reactions and has been implicated implicated in the toxicity of drugs with a narrow
in carbamazepine toxicity [63]. In other cases, therapeutic window. The next section examines
unsaturated compounds with aromatic rings the effect of genetic polymorphisms in drug
have also been shown to be oxidized to benzo- metabolism and how this can impact drugs sub-
quinone and hydroquinone which are highly ject to TDM.
reactive compounds [138] as is the case with
acetaminophen. Acetaminophen is converted by
CYP2E1 to N-acetyl-p-­ benzoquinone imine 13.3 Polymorphism in Drug
(NAPQI); this metabolic product is involved in Metabolism
its hepatotoxicity as discussed in details above
[139]. Other oxidation reactions involve Since TDM involves monitoring for an optimal
N-oxidation of primary and secondary amines to plasma concentration of drugs to achieve the
hydroxylamines which have been shown to be desired therapeutic effect at the right dose while
hepatotoxic and mutagenic as is the case with also avoiding the toxic effects, the plasma con-
acetamino-2-fluorene [140, 141]. Examples of centration of the substance of interest can be
CYP-mediated toxicity metabolism giving rise affected by their metabolism, resulting in an
to reactive metabolites include the oxidation of alteration of the effective plasma concentration,
heteroatoms like sulfur or nitrogen and the or generation of toxic metabolites (as shown in
reduction of polyhalogenated, nitro-, keto-, and the last section) with potential organ-system
azo derivatives in such compounds as daunoru- dysfunction. Genetic polymorphism in the
bicin. The oxidation of heteroatoms occurs in expression and activity of drug-metabolizing
the metabolism of thiophene, which becomes enzymes has been shown to have a significant
oxidized to sulfoxide, thiophene S-oxide, or effect on the efficacy and safety of drugs, includ-
epoxides and subsequently adducts with gluta- ing those with narrow therapeutic windows [151,
thione or proteins [142–145]. The reduction of 152]. Genetic polymorphism describes the
polyhalogenated, nitro-, keto-, and azo deriva- simultaneous occurrence of two or more discon-
tives occurs in compounds such as daunorubi- tinuous alleles or genotypes in a population and
cin, which forms a hydroxyl radical causing results in an alteration in gene expression; an
oxidative stress and cardiac toxicity; it also alteration in gene expression could also result in
forms another free radical derivative, the a change in the structure and function of the
7-deoxydaunorubicin radical, which alkylates resulting gene products [153, 154]. Cytochrome
DNA resulting in genotoxicity [146, 147]. P450 enzymes (CYPs) are prone to polymor-
While conjugative metabolism mainly results phism, which may account for inter-individual
in the formation of water-soluble derivatives, differences in drug pharmacokinetics and phar-
13 Therapeutic Drug Monitoring and Toxicology: Relevance of Measuring Metabolites 209

macodynamics [155, 156]. Several factors may most drugs used clinically [171]. We will focus
play a role in polymorphism, including tran- our discussion on members of these three
scriptional regulation, transcription errors, sin- families.
gle-nucleotide polymorphisms (SNPs), and copy
number variants [157–160]. Polymorphisms 13.3.1.1 CYP1A1
which result in a gain of function may lead to CYP1A1 is thought to be expressed in the liver
either decreased response to a therapeutic medi- [172, 173], but with a significant variability
cation or increased metabolism and toxic metab- [174]. It is also expressed in human extrahepatic
olite formation in some cases [161]. On the other tissues including intestine [175]. While CYP1A1
hand, polymorphisms which lead to loss of is involved in the metabolism of several common
function could result in overexposure to a medi- drugs, including caffeine [176], amiodarone
cation and increased undesirable off-target and [162, 177], R-warfarin [178, 179], ondansetron
on-­target effects [162]. [180], haloperidol [181], cyclobenzaprine [182],
and propranolol [183], there have been no studies
to date showing clinically relevant polymor-
13.3.1 Polymorphism in Expression phisms in the expression of CYP1A1 enzymes.
of CYPs To date, 15 allelic variants and subvariants (*2A
to *13) have been described in the CYP1A1 gene
The cytochrome P450 enzymes (CYPs) are pri- [184].
marily expressed in hepatic tissue where they
metabolize both endogenous compounds and 13.3.1.2 CYP1A2
xenobiotics [163]. CYPs are also expressed in the CYP1A2 accounts for about 10% of the total
small intestinal mucosa, lung, kidney, brain, pla- CYP content in human liver [185]. Several clini-
centa, olfactory mucosa, and skin, with the intes- cally relevant drugs are metabolized by CYP1A2
tinal mucosa being the main extrahepatic site of including caffeine [186], clozapine [187], ropiva-
drug biotransformation [164, 165]. Inter-­ caine [188], olanzapine [189], lidocaine [190],
individual variation in the expression of CYPs imipramine [191], propranolol [183], verapamil
may be partially explained by differential tran- [192], propafenone [193], and bortezomib [194].
scriptional regulation via nuclear receptors [166]. There are notable inter-individual differences
Thus, the extent to which each person metabo- (40- to 130-fold) in CYP1A2 expression and
lizes a certain drug is a combination of gene activity [195]. CYP1A2 is also influenced by
expression which is influenced by environmental environmental factors such as smoking and food
factors and individual variations in amino acid components [196]. There have been at least 40
sequences as a result of transcription errors [167]. variant alleles and a series of subvariants (*1B to
CYP polymorphisms are denoted as * (star) *21) identified [197], with CYP1A2*1A being
alleles, each corresponding to particular sequence the wild type.
variations within the coding sequence for the CYP1A2 accounts for about 30% of the
enzymes [168]. The subsection below explores metabolism of clozapine, an atypical antipsy-
the differences in metabolism as a result of CYP chotic, which is subject to TDM [198, 199]. In
enzyme polymorphisms. In humans, there are 57 smokers with the CYP1A2*1F genotype, a vari-
genes considered to be functional and 58 pseudo- ant that results in increased enzyme activity, low
genes [169]. Based on sequence similarity, these plasma levels have been reported to result in
genes are divided into 18 families and 44 sub- resistance to therapy [200]. Likewise, patients in
families. Most of the CYP’s roles relate to metab- whom two CYP1A2 variants with decreased
olizing endogenous substances (bile acids, enzyme activity (CYP1A2*1C and *1D) are
eicosanoids, and steroids) [170]. Of the 18 fami- present have been found to have higher clozapine
lies, CYP1, CYP2, and CYP3 are the main CYPs levels [201], resulting in hyperthermia, altera-
that contribute to the oxidative metabolism of tions in consciousness, seizures, cardiac arrhyth-
210 J. Akingbasote et al.

mias, excessive mucus production in bronchi, Clopidogrel is an antiplatelet agent used to


hepatitis, and cardiac arrhythmias [202, 203]. treat several different indications including sec-
ondary prevention in recent stroke or myocardial
13.3.1.3 CYP2C9 infarction and primary prevention of thromboem-
CYP2C8, 2C9, 2C18, and 2C19 make up the bolism atrial fibrillation, to name a few [217].
CYP2C subfamily and are primarily located in Clopidogrel is an irreversible inhibitor of platelet
the liver; these enzymes account for approxi- P2Y12 adenosine diphosphate receptor which
mately 26% of total CYP contents [185]. leads to decreased platelet aggregation. It is a
Together, the CYP2C subfamily is responsible pro-drug which is activated in a two-step path-
for the metabolism of about 20% of clinically way that involves CYP2C19. Several studies
relevant drugs [204]. In particular, CYP2C9 is have shown that PMs do not activate clopidogrel
among the most abundant CYP enzymes in the to the same extent as normal metabolizers (NMs)
liver (∼24% of total CYP content), where it [218]. The CYP2C19 genotype has also been
metabolizes approximately 15% of clinically linked with clinical outcomes for several indica-
used drugs, including S-warfarin and phenytoin – tions [219–223]. For example, the CYP2C19*17
drugs with narrow therapeutic indices [205, 206]. allele leads to increased function and has been
There are more than 60 variants and subvari- associated with an increased risk of bleeding
ants of CYP2C9 [207]. Two of the most common [224]. Because of the strength of evidence, a
allelic polymorphisms in the CYP2C9 gene, *2 guideline published by CPIC suggests alternative
and *3, result in the “poor metabolizer” (PM) antiplatelet therapy for CYP2C19 intermediate
phenotype and thus have a marked effect on metabolizers (IMs) and PMs [218].
S-warfarin metabolism [208]. CYP2C9 PMs
clear S-warfarin up to 85% less efficiently than 13.3.1.5 CYP2D6
normal metabolizers, which leads to longer war- Though it is one of the less abundant CYPs in the
farin half-life, longer time to achieve goal INR, liver (~5%) [185], CYP2D6 plays a role in the
and increased bleeding risk [209]. Likewise, metabolism of about 25% of drugs used clini-
CYP2C9 PMs are at risk for phenytoin toxicity as cally [225]. Drugs that are extensively metabo-
they require 30–50% dose reduction compared to lized by CYP2D6 include tricyclic
normal metabolizers [210]. antidepressants, SSRIs, other nontricyclic antide-
pressants, beta-blockers, opioids, antiemetics,
13.3.1.4 CYP2C19 and antihistamines. CYP2D6 is highly polymor-
Located on chromosome 10, CYP2C19 is found phic and leads to significant inter-individual vari-
primarily in hepatic tissue, but the intestinal wall ation in enzyme expression and activity. In fact, it
also contains a significant amount of the enzyme. has been reported that there are nearly 150 vari-
CYP2C19 is responsible for the metabolism of ants of CYP2D6 (*1B to *149) [226]. CYP2D6
approximately 10% of commonly used drugs also exhibits gene duplication, which results in
[211, 212], including proton pump inhibitors multiple functional copies of the enzyme and
(PPIs), tricyclic antidepressants (TCAs), selec- increased activity. Hence, as a result of the vari-
tive serotonin reuptake inhibitors (SSRIs), ben- ability in the CYP2D6 phenotype, individuals
zodiazepines, phenobarbital, phenytoin, can be classified as poor, intermediate, normal, or
bortezomib, and voriconazole [213, 214]. ultrarapid metabolizers [227], and these can vary
CYP2C19 is also involved in the activation of by racial backgrounds [228]. The example of
clopidogrel, along with several other CYP codeine aptly illustrates the variability in the
enzymes [215]. Thus far, over 40 variants and expression and catalytic activity of CYP2D6
subvariants of CYP2C19 (*1B to *39) have been [229]. Codeine is inactive but is bioactivated to
described, and the *1A allele has been reported morphine by CYP2D6, the extent of which
to be the wild type [216]. depends on the variant of CYP2D6 present in the
13 Therapeutic Drug Monitoring and Toxicology: Relevance of Measuring Metabolites 211

patient. While poor or intermediate metabolizers molecule unto the substrate to enhance its water
show decreased conversion to morphine and a solubility thereby enhancing its renal excretion
reduced analgesic effect, ultrarapid metabolizers [237]. Like the CYP enzymes, UGTs are primar-
may show life-threatening adverse effects after ily located in the liver [238–240] but can be found
codeine administration [229, 230]. This example in a number of other tissues including the kid-
is further discussed in Sect. 13.4.1. neys, gastrointestinal tract, lungs, epithelium,
ovaries, testis, mammary glands, and prostate
13.3.1.6 CYP3A4/5 [238, 240–242]. While functional genetic varia-
CYP3A4 and CYP3A5 are expressed in the liver, tions have been found in UGTs, including those
and these enzymes are responsible for metabo- which are important in drug metabolism [243–
lizing more than 50% of medications used clini- 254], less is known about which variants lead to
cally [231, 232]. While CYP3A4 activity is altered enzyme function.
highly variable, genetic differences do not
account for all of the variability [233]. CYP3A5
is structurally similar to 3A4 and thus results in 13.3.3 Other Polymorphisms in Drug
substrate overlap. Tacrolimus, a CYP3A5 sub- Metabolism
strate, is subject to therapeutic drug monitoring
in clinical practice; however, evidence is scant 13.3.3.1 TPMT and NUDT15
when assessing clinical outcomes associated Thiopurine methyltransferase (TPMT) and nudix
with genotyping. There are no genotype-based (nucleoside diphosphate-linked moiety X)-type
guidelines available for CYP3A4 or CYP3A5, motif 15 (NUDT15) play an important role in the
but the CYP3A5 genotype has been associated metabolism of thiopurines (azathioprine, thio-
with varying tacrolimus concentrations [51]. guanine, and mercaptopurine) [255]. TPMT inac-
CYP3A4 and CYP3A5 also play a role in carba- tivates mercaptopurine via catabolism, and
mazepine (CBZ) metabolism [234]. Particular individuals who inherit two nonfunctional TPMT
variants such as CYP3A5*3, the most common alleles are at greatly increased risk of severe
nonfunctional variant, results in higher levels of myelosuppression with standard doses of mer-
CBZ plasma concentration [235]. Patients who captopurine or azathioprine [256]. A guideline
harbor this variant may require lower doses of developed and published by CPIC suggests dose
CBZ, though the clinical impact of this is dis- adjustments for TPMT phenotypes based on gen-
puted. Furthermore, CYP3A4*22 results in a otype [257].
lower CBZ-diol/CBZ epoxide ratio, the clinical NUDT15 catalyzes the conversion of thiogua-
significance of which has yet to be fully eluci- nine triphosphate metabolites to thioguanine
dated [99]. monophosphates which are less toxic [257].
Patients with defective or less active NUDT15
can show severe myelosuppression [258]. Several
13.3.2 Uridine Diphosphate-­ variants have been assigned star alleles, and a
Glucuronosyltransferases guideline exists in tandem with TPMT to assign
(UGTs) metabolizer status to individuals based on geno-
type and accordingly dose-adjust the thiopurines
Mostly complementary to oxidative metabolism, to minimize the risk of toxicity [257]. Medications
mediated by the CYP enzyme family, uridine in the thiopurine class (i.e., azathioprine, mercap-
diphosphate-glucuronosyltransferases (UGTs) topurine, and thioguanine) are often monitored
are a superfamily of enzymes and probably the by drug levels in clinical practice as they are one
most important conjugative enzyme [236]. UGTs of the oldest examples of drugs where metabolic
catalyze the conjugation of a glucuronic acid differences lead to a variety of responses in
212 J. Akingbasote et al.

patients [45]. Thiopurines are antineoplastic have TPMT and NUDT15 genetic testing per-
agents commonly used for a variety of condi- formed [41, 45]. If a patient has one nonfunc-
tions, including acute lymphoblastic leukemia tional TPMT or NUDT15 allele, it is
(ALL) and inflammatory bowel disease [259]. recommended to start with reduced doses (by
These agents are considered pro-drugs with three 30–70% with mercaptopurine and azathioprine).
main metabolic pathways that include phosphor- If the patient has two nonfunctional alleles, it is
ylation, methylation, and catabolism [41]. The recommended to reduce the thrice-weekly dose
metabolism is illustrated in Fig. 13.3. by 90% or to consider an alternative agent [259].
It is thought that the phosphorylation pathway Once a patient has started a thiopurine, ongoing
wherein inosine monophosphate dehydrogenase dosing should be adjusted based on the degree of
creates 6-thioguanine nucleotides (TGNs) is key myelosuppression and advised through TDM
for resulting in a therapeutic effect because the [45].
triphosphate can be incorporated into DNA and TDM of thiopurines is an excellent example
RNA as a false purine analog causing cell death of the measuring of metabolites versus the usual
[41]. These pro-drugs are also converted into drug level monitoring of the parent compound
inactive metabolites by xanthine oxidase (XO), [41, 265]. Laboratories measure metabolite con-
TPMPT, and NUDT15 enzymes, leading to 6- centrations in red blood cells, by a quantification
thiouric acid (6-TUA), 6-­methylmercaptopurine of the hydrolysis products from several metabo-
(6- MMP), 6-thioguanosine triphosphate lites [41]. With the use of high-performance liq-
(6-TGMP), and 6-thioguanine diphosphate uid chromatography (HPLC), levels of TGN
(6-TGDP) [45]. The metabolite 6-MMP has levels and methylated thioinosine derivatives are
been linked to thiopurine-induced liver toxicity determined [266]. While higher 6-TGN levels are
[260]. In non-malignant conditions, thiopurine associated with improved clinical response and
use is associated with potential toxicities such as remission in inflammatory bowel disease patients,
gastrointestinal effects and myelosuppression, levels of 6-MMP are monitored to avoid toxicity
and over 20% of patients discontinue their thio- [45, 267]. When applying TDM to patients who
purine medication based on drug-related toxici- continue to have disease progression, patients
ties [259]. TPMT and NUDT15 deficiency is with normal 6-TGN levels are unlikely to respond
­relatively rare but affects all thiopurine drugs; it to treatment with thiopurines [268]. Patients with
is responsible for approximately 30% of all low concentrations of both 6-TGN and 6-MMP
thiopurine-­ associated neutropenia cases [259]. could be showing noncompliance or a subopti-
NUDT15 deficiency is considered another risk mal dosing strategy, and those with low 6-TGN
factor for thiopurine-induced neutropenia. levels and high 6-MMP levels are suggested to be
Patients with two normal function variants, con- metabolically shunting away from 6-TGN pro-
sidered TPMT or NUDT15 normal metabolizers, duction and creating more of the toxic 6-MMP
are expected to tolerate mercaptopurine at stan- [269]. Thus, the metabolism of thiopurines and
dard doses, while patients with a low or deficient the polymorphism in their metabolizing enzymes
activity variant and one normal function variant illustrate the importance of measurement of
are classified as intermediate metabolizers [259, metabolites as this could make a significant dif-
261]. Finally, patients with two nonfunctional ference in achieving optimal therapeutic efficacy
variants are considered poor metabolizers [262]. while avoiding toxicities.
TPMT deficiency is observed more in Caucasian
and African populations, whereas NUDT15 defi- 13.3.3.2 Vitamin K Epoxide
ciency is relatively less frequent [263]. In con- Reductase Complex Subunit
trast, NUDT15 deficiency is seen more in Asian 1 (VKORC1)
and Hispanic populations, whereas TPMT defi- VKORC1 encodes the target enzyme of warfarin,
ciency is relatively less frequent [264]. Before vitamin K epoxide reductase [270]. It catalyzes
starting this medication, it is recommended to vitamin K formation from vitamin K epoxide,
13 Therapeutic Drug Monitoring and Toxicology: Relevance of Measuring Metabolites 213

Fig. 13.3 Metabolism of thiopurine. Azathioprine is con- incorporated into DNA and RNA and cause apoptosis
verted to 6-mercaptopurine by GST. Subsequently, through the mismatch repair system. As such, higher
6-­mercaptopurine can be converted to either 6-thiouric 6-TGN levels have been associated with improved clinical
acid by XO or 6-methylmercaptopurine by TPMPT. The response. The thiopurine drug, azathioprine, is an immu-
toxic metabolite 6-methylmercaptopurine has been linked nosuppressive drug used in individuals with diseases such
to liver toxicity, and neutropenia can occur in cases of as rheumatoid arthritis, Crohn’s disease, and systemic
TPMT deficiency. Following the conversion of lupus, among others. Abbreviations: GST glutathione
6-­mercaptopurine to 6-thioinosine monophosphate by S-transferase, HPRT hypoxanthine-guanine phosphoribo-
HPRT, the metabolite is converted to 6-thioguanine nucle- syltransferase, IMPDH inosine-5′-monophosphate dehy-
otide (6-TGN). 6-TGN is responsible for the therapeutic drogenase, TPMT thiopurine S-methyltransferase, XO
effects of thiopurine drugs, since the triphosphate can be xanthine oxidase. (Figure created with BioRender.com)

which is the rate-limiting step in vitamin K recy- types [273]. These and other types of
cling [271]. Upstream of VKORC1, one variant polymorphism in VKORC1 expression play a
(c.-1639G>A, rs9923231) is associated with key role in dose selection for warfarin [274].
increased warfarin sensitivity. Individuals with In summary, there are many known variants
one or two -1639A require lower warfarin doses of drug-metabolizing enzymes found for medi-
than patients homozygous for -1639G, and a cations with narrow therapeutic index. Several
CPIC guideline exists to guide dosing when com- guidelines exist for starting doses of certain
bined with other patient factors including medications based on a patient’s pharmacoge-
CYP2C9 status [272]. There is a race-dependent nomic profile; however, there are limited rec-
variation in the expression of VKORC1 with ommendations which guide therapeutic drug
Asian-Americans having a higher proportion of monitoring of metabolites. TDM of parent
group A haplotypes and African-Americans dis- compounds remains the standard for monitor-
playing a higher proportion of group B haplo- ing efficacy and toxicity of drugs with a narrow
214 J. Akingbasote et al.

Table 13.2 Effect of polymorphism in drug-metabolizing enzymes on efficacy and safety of drugs with narrow thera-
peutic window
Effects in
Enzyme class involved and Effect in poor extensive
Drug polymorphic variants metabolizers metabolizers References
Tacrolimus CYP3A4 and CYP3A5 May have higher Subtherapeutic [6]
troughs but no troughs so initial
dosing changes dose is
recommended recommended to
start higher
Thiopurines (i.e., Thiopurine S-methyltransferase, Dose reduction Myelotoxicity due [1]
azathioprine, glutathione S-transferase, would be warranted to elevated
mercaptopurine, hypoxanthine-guanine based on level of 6-thioguanine
and thioguanine phosphoribosyltransferase, inosine-5′- enzyme deficiency nucleotide
monophosphate dehydrogenase,
thiopurine S-methyltransferase,
xanthine oxidase, nudix hydrolase 15
Clozapine CYP1A2 Increased serum Decreased plasma [23, 24]
clozapine level level and
therapeutic
resistance
Warfarin CYP2C9, VKORC1 Increased half-life, [25]
longer time to
achieve goal INR,
increased bleeding
risk (2C9)
Phenytoin CYP2C9 Increased risk for [26]
toxicity, require
30–50% dose
reduction
Clopidogrel CYP2C19 Decreased Increased risk of [27, 28]
antiplatelet activity, bleeding
alternative therapy
recommended
Codeine CYP2D6 Decreased May result in [29, 30]
conversion to life-threatening
morphine and adverse effects
decreased analgesic
effect
Carbamazepine CYP3A5, UGT2B7 Higher Lower [31]
carbamazepine carbamazepine
serum concentration, concentration and
patients may require escalating dose
lower doses, though requirements
this is disputed (UGT2B7)
(CYP3A5)

therapeutic index in a clinical setting. The con- 13.4 Pro-drugs, TDM,


tribution of pharmacogenomics to drug metab- and Metabolite
olism and inherently the safety of these drugs Measurement
suggest the need to monitor metabolite in TDM
of drugs with narrow therapeutic window Pro-drugs are inactive substances that are metab-
(Table 13.2). olized in the body to their active metabolites
[275]. Pro-drugs often contain functional groups
13 Therapeutic Drug Monitoring and Toxicology: Relevance of Measuring Metabolites 215

of esters, amides, phosphates, carbonates, or car- diate substance, followed by spontaneous loss of
bamates that are cleaved either enzymatically or CO2 and formaldehyde, producing the monoester
chemically once in the body [276]. Upon bio- intermediate [276]. Phosphodiesterases were also
transformation within the body, the active con- suggested to play a key role in the final hydroly-
stituent is released, and the active metabolite can sis step of the monoester intermediate into the
subsequently confer the intended pharmacologi- active drug “tenofovir” [276].
cal benefit of the pro-drug substance. While some As was described in the previous sections of
examples of pro-drugs in clinical use are not this book chapter, TDM of metabolites is impor-
associated with any clinical benefits and were tant for many drugs in order to ensure that the
only found to be pro-drugs retrospectively, other drug remains within a safe therapeutic range.
pro-drugs confer biological benefits [277]. For Similarly, TDM of the metabolites of pro-drugs
example, the cytotoxic drug cyclophosphamide is is also performed as the outcomes are associated
a pro-drug that only becomes active after under- with comparable clinical utility. For example,
going hepatic metabolism [277]. Another clinical therapeutic drug monitoring of the active compo-
benefit of pro-drugs is to mask the polar or ioniz- nent of the pro-drug fosphenytoin (phenytoin) is
able functional groups of the active molecules, performed to guide anticonvulsant therapy [278].
ultimately improving oral bioavailability of the Fosphenytoin is a phosphate ester pro-drug of
active substance [276]. For instance, tenofovir is phenytoin that is rapidly hydrolyzed (half-life of
a nucleotide inhibitor of reverse transcriptase 5–15 min) and is associated with superior solu-
involved in human immunodeficiency virus 1 bility and tolerance upon administration [278].
(HIV-1) infections. Tenofovir was once a drug Due to its narrow therapeutic index, saturable
with limited clinical utility due to the high hydro- elimination kinetics, and its concentration-­
philicity of the phosphonic acid group, resulting dependent side effects, it is recommended that
in very low bioavailability in humans (<5%) drug plasma concentrations be monitored in
[276]. However, the pro-drug “tenofovir diso- order to guide dose therapy individualization
proxil” had better oral bioavailability in humans [278]. The first step in the enzymatic hydrolysis
(39%). The conversion of this pro-drug was dem- of the fosphenytoin sodium salt substance by
onstrated to occur by the chemical or enzymatic phosphatase results in the formation of
hydrolysis of tenofovir disoproxil to an interme- 3-hydroxymethylphenytoin (Fig. 13.4).

Fig. 13.4 Metabolic bioactivation of the pro-drug fos- an unstable intermediate, 3-hydroxymethylphenytoin,
phenytoin. Fosphenytoin sodium salt contains a phos- which spontaneously converts to phenytoin. This is a good
phate ester group (red circle) linked to an acidic amine of example of the need to monitor the blood levels of a com-
the antiepileptic agent, phenytoin, through an oxymeth- pound which doubles as the active metabolite and one
ylene spacer. The first step in the enzymatic hydrolysis of which merits TDM due to its narrow therapeutic window.
fosphenytoin by phosphatase results in the formation of (Figure created with BioRender.com)
216 J. Akingbasote et al.

This unstable intermediate subsequently such medications [44, 45]. On the other hand,
spontaneously converts to phenytoin [279]. The patients who highly express these enzymes (rapid
conversion of the fosphenytoin to the active drug or ultrarapid metabolizers) produce a very high
metabolite, phenytoin, occurs rapidly in the level of the active metabolite such that there may
blood. Another example of the clinical utility of be safety concerns [289]. A clinical example of a
TDM of pro-drug metabolites can be demon- direct relationship between metabolism and tox-
strated with the use of mycophenolate mofetil icity is in codeine, a pro-drug narcotic analgesic
(MMF). MMF is a pro-drug that is converted to [44]. As a parent drug, codeine has no therapeutic
the active metabolite, mycophenolic acid (MPA), effect [290]. However, metabolism mediated by
by esterase enzymes in the intestine, liver, and hepatic CYP2D6 forms morphine, its active
plasma [280]. MPA is associated with immuno- metabolite. Codeine may also undergo glucuron-
suppressant activity. As such, MMF is often used idation to produce codeine-6-glucuronide [291].
after solid organ transplantation, including lung,Morphine and codeine-6-glucuronide are two
heart, and renal transplantation [281]. Many active metabolites with well-pronounced thera-
studies have suggested that TDM of MPA in peutic effects for pain relief (Fig. 13.5).
patients receiving these organ transplants may In patients who undergo ultrarapid metabo-
improve clinical outcomes and allow for dose lism by CYP2D6, there is a dramatic shift of
individualization of MMF [282–286]. Effective metabolism of codeine toward morphine. An
dose individualization by using TDM of MPA increase in the levels of morphine leads to an
ultimately increases the safety of the drug by increased risk of oversedation, respiratory
potentially minimizing toxicity but also results depression, and death, even with a low dose of
in a minimal risk of organ transplant rejection, codeine. Alternatively, for patients who are poor
thus improving efficacy [282]. These findings CYP2D6 metabolizers, they often experience
also extend to pediatric transplant patients and poor pain control from codeine therapy [292]. In
children with pediatric lupus nephritis, wherein 2013, the US FDA issued a warning to restrict the
TDM of MPA was suggested to improve the effi- use of codeine by adding a contraindication to the
cacy of MMF [287, 288]. Altogether, these drug label of codeine alerting that codeine should
examples demonstrate the clinical utility of not be used to treat pain or cough in children
TDM of the active drug MPA as it can be used to younger than 12 years [293]. A similar example
guide clinical management and MMF dosing is tramadol, an opioid analgesic pro-drug which
[282]. is transformed into O-desmethyltramadol, the
active metabolite, via metabolic pathways that
include CYP2D6 [44, 294]. Excessive levels of
13.4.1 Toxicity of Metabolites this metabolite lead to the same adverse effects as
of Pro-drugs a morphine overdose which includes overseda-
tion, respiratory depression, and death [44]. The
As shown above, the wide variation in the expres- FDA has also issued a similar warning to restrict
sion of drug-metabolizing enzymes among tramadol use in children older than 12 years of
patient populations can impact the efficacy and/ age for pain management after adenotonsillec-
or toxicity of drugs used in clinical practice. tomy and in children younger than 18 years of
Hence, in the case of pro-drugs which require age [293]. The examples above illustrate the
prior metabolism before conversion to a thera- importance of measuring the active metabolite
peutically active molecule, individuals who do which would be of greater clinical value to deter-
not express the relevant enzymes (slow or non-­ mine drug toxicity in comparison with measuring
metabolizers) would not benefit from the use of the parent compound.
13 Therapeutic Drug Monitoring and Toxicology: Relevance of Measuring Metabolites 217

Fig. 13.5 Metabolism of codeine. Codeine undergoes exhibit rapid metabolism of codeine by CYP2D6 display
hepatic metabolism into morphine (10%), norcodeine a shift of metabolism toward morphine, resulting in
(5–10%), and codeine-6-glucuronide (70–80%). While adverse effects such as oversedation, respiratory depres-
codeine has no direct therapeutic effect, the active metab- sion, and possibly death. On the other hand, poor CYP2D6
olites (morphine, codeine-6-glucoronide) have analgesic metabolizers experience a lack of efficacy from codeine
effects, exploited in pain management. Patients who therapy. (Figure created with BioRender.com)

these assays on a routine basis. The first step is


13.5 Conclusions and Future the screening of the entity involved in the toxicity
Directions of the drug of interest, to see if the toxicity of
concern is a result of the parent compound or the
While the main focus of TDM over the years has metabolites. This will require establishing an
been the determination of the level of the parent association between the dose ingested, blood
compound in order to achieve an optimal thera- level of the parent compounds and/or metabo-
peutic benefit while also avoiding toxicity, this lites, and emergence of toxicity. Since detection
chapter has highlighted the importance of mea- of metabolites at levels up to 25% of circulating
suring metabolites of drugs subject to TDM. drug levels can be a safety concern [74–76], such
Through the various examples that have been metabolites should be further investigated as to
provided in this chapter, where clinical decision-­ their possible role in the toxicity of the drug, par-
making based on the blood level of the adminis- ticularly in cases of the adverse reactions. These
tered medication leads to an incomplete picture results can then be followed with further investi-
of efficacy or toxicity, we hope to provide further gations in a larger patient population to d­ etermine
justification for the value and importance of tak- the association between the emergence of the
ing into consideration the impact of metabolic adverse effect and metabolite detection. In devel-
pathways. If this approach is adopted, the ability oping an assay for the detection of the metabo-
to provide a higher level of precision medicine lites, the same standard processes and
for our patients will be optimized. considerations can be implemented as those used
One major setback in the measurement of for measuring parent compounds. Important con-
metabolites is the practicality and cost of running siderations include the timing of sample collec-
218 J. Akingbasote et al.

tion, sample processing (including choice of life, lower incidence of adverse reactions, and
anticoagulant to use for blood collection), and increased longevity may outweigh these costs
sample storage conditions [3]. These factors [301]. As reviewed by Kang and Lee [3], the
depend on the compound of interest and the example of antiepileptic therapy was used to
metabolite of interest, and method development exemplify the benefit of monitoring metabolites
and validation are key in this endeavor. Key fac- in TDM. This review reported improved thera-
tors affecting the robustness of the chosen method peutic outcomes including effective seizure con-
include its accuracy, precision, limit of quantifi- trol, a reduced incidence of adverse reactions,
cation/detection/identification, linear dynamic cost savings from reduced hospitalization per sei-
range, reproducibility, and repeatability [295]. To zure, and greater chances of remission in patients
a large extent, techniques including radioimmu- who underwent TDM. Further, these factors
noassay, high-performance liquid chromatogra- enhanced the quality of life of the patients, result-
phy (HPLC), immunobinding assays, ing in better earning capacity, with reduced eco-
fluorescence polarization immunoassay (FPIA), nomic losses due to hospital stay [301]. A similar
enzyme-multiplied immunoassay technique outcome has been reported for other therapeutic
(EMIT), and enzyme-linked immunosorbent interventions in which TDM has been applied,
assay (ELISA) have been used in the determina- including aminoglycoside drugs [302, 303],
tion of the blood level of parent compounds immunosuppressants [304], and other indications
[296–298] and can be used in metabolite mea- [305]. Of particular interest in these cases was the
surement. Another concern in metabolite mea- fact that dose optimization and individualization,
surement is in the fact that cross-reactivity has especially upon consideration of pharmacogenet-
been reported between the metabolites and cel- ics, resulted in cost-effectiveness. A review by
lular macromolecules such as proteins and even McNeill and Barclay [306] showed that TDM has
antibodies, thereby resulting in an alteration in proven to be cost-effective overall in the manage-
the measurable concentration of the metabolite in ment of inflammatory bowel disease when drugs
question. Therefore, cross-reactivity may ulti- such as thiopurine were considered. Additional
mately interfere with the result of the assay [299, modeling data to predict the cost-effectiveness
300]. For this reason, other specific methods and beneficial outcomes of metabolite measure-
would need to be developed to accurately mea- ment in the TDM of thiopurine was reported in
sure the concentration of metabolites. another study [307]. In addition to enhanced out-
Furthermore, since reactive metabolites are comes, it was predicted that metabolite measure-
mostly unstable, a delay between sample collec- ment would result in approximately 18%
tion and analysis can result in an undetectable reduction in the cost of treatment. Since the stud-
amount of the metabolite. This characteristic ies described above were based on a modeling
highlights the importance of developing a robust data, further studies may be required to further
method for quantification of reactive demonstrate the cost-effectiveness of metabolite
metabolites. measurement in TDM with a particular focus on
Finally, another key consideration in the metabolites.
implementation of metabolite measurement in Although TDM focuses primarily on the mea-
TDM is the cost of running such analyses vis-à-­ surement of parent compounds administered to
vis the benefit to the patients. The pharmacoeco- patients in clinical practice, this chapter has high-
nomic impact of TDM has been studied to lighted the importance of including metabolite
compare the advantage that accrues to the patients measurement in TDM as these measurements
in comparison with the cost involved in running further enhance the quality of healthcare, assist to
such analyses. While it is expected that adding monitor and predict the emergence of drug-­
metabolite measurement to standard TDM may induced organ-system toxicity, and help optimize
be associated with considerable cost, the overall therapy. Not all drugs requiring TDM would ben-
advantage to the patients in terms of quality of efit from metabolite measurement. However,
13 Therapeutic Drug Monitoring and Toxicology: Relevance of Measuring Metabolites 219

those which are known to be metabolized to bio- 8. Gross AS. Best practice in therapeutic drug moni-
toring. Br J Clin Pharmacol [Internet]. 2001 [cited
active or toxic metabolites need to be monitored, 2021 Dec 30];52(Suppl 1):5S–10S. Available
as they can have significant adverse impacts on from: https://www.ncbi.nlm.nih.gov/pmc/articles/
patient outcomes. In order for an all-­encompassing PMC2014621/.
adoption of metabolite determination in TDM to 9. Ates HC, Roberts JA, Lipman J, Cass AEG, Urban
GA, Dincer C. On-site therapeutic drug monitor-
be implemented, there is a need for a well-defined ing. Trends Biotechnol [Internet]. 2020 [cited 2021
strategy for sample collection, analytical method Dec 29];38(11):1262–77. Available from: https://
development, and cost-benefit analysis. This www.cell.com/trends/biotechnology/abstract/
would ultimately facilitate the robust implemen- S0167-­7799(20)30061-­5.
10. Grandjean P. Paracelsus revisited: the dose concept
tation of TDM, resulting in a reduced incidence in a complex world. Basic Clin Pharmacol Toxicol
of drug toxicity and optimal utilization of [Internet]. 2016 [cited 2021 Dec 29];119(2):126–32.
therapeutics. Available from: https://www.ncbi.nlm.nih.gov/pmc/
articles/PMC4942381/.
11. Widmark E. Die theoretischen Grundlagen und
die praktische Verwendbarkeit der gerichtlich-­
References medizinischen Alkoholbestimmung. J Am Med
Assoc. 1932;98:1834.
1. Ghiculescu RA. Therapeutic drug monitoring: 12. Sharma S, Joshi S, Chadda RK. Therapeutic drug
which drugs, why, when and how to do it. 2008 monitoring of lithium in patients with bipolar affec-
[cited 2021 Dec 19]. Available from: https://www. tive disorder: experiences from a tertiary care hospi-
nps.org.au/australian-­prescriber/articles/therapeutic-­ tal in India. Am J Ther. 2009;16(5):393–7.
drug-­monitoring-­which-­drugs-­why-­when-­and-­how-­ 13. Shorter E. The history of lithium therapy. Bipolar
to-­do-­it. Disord. 2009;11(Suppl 2):4–9.
2. Advani M, Seetharaman R, Pawar S, Mali S, 14. Finney DJ. The design and logic of a monitor of drug
Lokhande J. Past, present and future perspectives of use. J Chronic Dis. 1965;18:77–98.
therapeutic drug monitoring in India. Int J Clin Pract 15. Nelson E. Kinetics of drug absorption, distribu-
[Internet]. 2021 [cited 2021 Dec 30];75(8):e14189. tion, metabolism, and excretion. J Pharm Sci
Available from: http://onlinelibrary.wiley.com/doi/ [Internet]. 1961 [cited 2021 Dec 29];50(3):181–
abs/10.1111/ijcp.14189. 92. Available from: https://jpharmsci.org/article/
3. Kang J-S, Lee M-H. Overview of therapeutic S0022-­3549(15)33106-­3/abstract.
drug monitoring. Korean J Intern Med [Internet]. 16. Ensom MH, Davis GA, Cropp CD, Ensom
2009 [cited 2021 Dec 28];24(1):1–10. Available RJ. Clinical pharmacokinetics in the 21st century.
from: https://www.ncbi.nlm.nih.gov/pmc/articles/ Does the evidence support definitive outcomes? Clin
PMC2687654/. Pharmacokinet. 1998 Apr;34(4):265–79.
4. Kidd JM, Asempa TE, Abdelraouf K. Chapter 13 – 17. Horning MG, Brown L, Nowlin J, Lertratanangkoon
Therapeutic drug monitoring. In: Adejare A, editor. K, Kellaway P, Zion TE. Use of saliva in therapeu-
Remington. 23rd ed [Internet]. Academic Press; tic drug monitoring. Clin Chem. 1977;23(2 PT.
2021 [cited 2021 Dec 19]. p. 243–62. Available 1):157–64.
from: https://www.sciencedirect.com/science/ 18. Lagerström P, Persson B. Liquid chromatography in
article/pii/B9780128200070000131. the monitoring of plasma levels of antiarrhythmic
5. Petre M, Strah A. Therapeutic drug monitoring of drugs. J Chromatogr A. 1978;149:331–40.
anti-epileptic drugs. Farm Vestn. 2015;66:35–41. 19. Neu HC. A review and summary of the phar-
6. Raj Panday D, Panday KR, Basnet M, Kafle S, Shah macokinetics of cefoperazone: a new, extended-­
B, Rauniar G. therapeutic drug monitoring of carba- spectrum beta-lactam antibiotic. Ther Drug Monit.
mazepine. Int J Neurorehabilitation [Internet]. 2017 1981;3(2):121–8.
[cited 2021 Dec 30];04(01). Available from: https:// 20. Ganguly NK, Bano R, Seth SD. Human genome
www.omicsgroup.org/journals/therapeutic-­d rug-­ project: pharmacogenomics and drug development.
monitoring-­of-­carbamazepine-­2376-­0281-­1000245. Indian J Exp Biol. 2001;39(10):955–61.
php?aid=85963. 21. Belknap R, Weis S, Brookens A, Au-Yeung KY,
7. Saleem MA, Basharat R, Rana NA, Khattak Moon G, DiCarlo L, et al. Feasibility of an ingestible
SAK. Role of clinician in therapeutic drug moni- sensor-based system for monitoring adherence to
toring practice. Clin Pract [Internet]. 2020 Jan 20 tuberculosis therapy. PLoS One. 2013;8(1):e53373.
[cited 2021 Dec 30];17(1):1429–35. Available from: 22. Ranamukhaarachchi SA, Padeste C, Dübner M,
https://www.openaccessjournals.com/abstract/ Häfeli UO, Stoeber B, Cadarso VJ. Integrated hollow
role-­of-­clinician-­in-­therapeutic-­drug-­monitoring-­ microneedle-optofluidic biosensor for therapeutic
practice-­13152.html. drug monitoring in sub-nanoliter volumes. Sci Rep
220 J. Akingbasote et al.

[Internet]. 2016 [cited 2021 Dec 29];6(1):29075. area under the curve calculations in pediatric solid
Available from: https://www.nature.com/articles/ organ transplant recipients. Pediatr Transplant.
srep29075. 2013;17(1):80–5.
23. Kling A, Chatelle C, Armbrecht L, Qelibari E, 36. Katz R. Biomarkers and surrogate markers: An FDA
Kieninger J, Dincer C, et al. Multianalyte antibiotic perspective. NeuroRx [Internet]. 2004 [cited 2022
detection on an electrochemical microfluidic plat- Feb 21];1(2):189–95. Available from: https://www.
form. Anal Chem. 2016;88(20):10036–43. ncbi.nlm.nih.gov/pmc/articles/PMC534924/.
24. WHO. Review of the evidence to include TDM in 37. Monagle P, Chan A, Massicotte P, Chalmers E,
the Essential in vitro diagnostics list and prioritiza- Michelson AD. Antithrombotic therapy in chil-
tion of medicines to be monitored. 2019. dren: the seventh ACCP conference on antithrom-
25. Rawson TM, Gowers SAN, Freeman DME, Wilson botic and thrombolytic therapy. Chest. 2004;126(3
RC, Sharma S, Gilchrist M, et al. Microneedle bio- Suppl):645S–87S.
sensors for real-time, minimally invasive drug moni- 38. Costa-Lima C, Fiusa MML, Annichino-Bizzacchi
toring of phenoxymethylpenicillin: a first-in-human JM, de Paula EV. Prothrombin complex concen-
evaluation in healthy volunteers. Lancet Digit trates in warfarin anticoagulation reversal. Rev Bras
Health. 2019;1(7):e335–43. Hematol E Hemoter [Internet]. 2012 [cited 2022 Feb
26. Jang SH, Yan Z, Lazor JA. Therapeutic drug moni- 21];34(4):302–4. Available from: https://www.ncbi.
toring: a patient management tool for precision nlm.nih.gov/pmc/articles/PMC3460405/.
medicine. Clin Pharmacol Ther. 2016;99(2):148–50. 39. Shikdar S, Vashisht R, Bhattacharya PT. International
27. Marshall WJ, Bangert SK. Clinical chemistry. Normalized Ratio (INR). In: StatPearls [Internet].
Edinburgh. New York: Mosby; 2004. Treasure Island: StatPearls Publishing; 2022 [cited
28. Gex-Fabry M, Balant-Gorgia AE, Balant 2022 Feb 21]. Available from: http://www.ncbi.nlm.
LP. Therapeutic drug monitoring databases for post- nih.gov/books/NBK507707/.
marketing surveillance of drug-drug interactions. 40. Paci A, Veal G, Bardin C, Levêque D, Widmer
Drug Saf. 2001;24(13):947–59. N, Beijnen J, et al. Review of therapeutic drug
29. Schoretsanitis G, Paulzen M, Unterecker S, Schwarz monitoring of anticancer drugs part 1 – cytotox-
M, Conca A, Zernig G, et al. TDM in psychiatry and ics. Eur J Cancer [Internet]. 2014 [cited 2022
neurology: A comprehensive summary of the con- Feb 21];50(12):2010–9. Available from: https://
sensus guidelines for therapeutic drug monitoring in www.sciencedirect.com/science/article/pii/
neuropsychopharmacology, update 2017; a tool for S0959804914005991.
clinicians. World J Biol Psychiatry [Internet]. 2018 41. Vikingsson S, Carlsson B, Almer SHC, Peterson
[cited 2022 Jan 16];19(3):162–74. Available from: C. Monitoring of thiopurine metabolites in patients
https://doi.org/10.1080/15622975.2018.1439595. with inflammatory bowel disease-what is actually
30. Burns AN, Goldman JL. A moving target-­ measured? Ther Drug Monit. 2009;31(3):345–50.
vancomycin therapeutic monitoring. J Pediatr Infect 42. Hacker M, Messer WS, Bachmann
Dis Soc. 2020 Sep 17;9(4):474–8. KA. Pharmacology: principles and practice.
31. Rybak M, Lomaestro B, Rotschafer JC, Moellering Academic Press; 2009. 607 p.
R, Craig W, Billeter M, et al. Therapeutic monitor- 43. Rautio J, Meanwell NA, Di L, Hageman MJ. The
ing of vancomycin in adult patients: a consensus expanding role of prodrugs in contemporary drug
review of the American Society of Health-System design and development. Nat Rev Drug Discov
Pharmacists, the Infectious Diseases Society of [Internet]. 2018 [cited 2022 Jan 19];17(8):559–87.
America, and the Society of Infectious Diseases Available from: https://www.nature.com/articles/
Pharmacists. Am J Health-Syst Pharm: AJHP. nrd.2018.46.
2009;66(1):82–98. 44. Fortenberry M, Crowder J, So T-Y. The use of
32. Jenkins A, Thomson AH, Brown NM, Semple Y, codeine and tramadol in the pediatric population-­
Sluman C, MacGowan A, et al. Amikacin use and what is the verdict now? J Pediatr Health Care.
therapeutic drug monitoring in adults: do dose regi- 2019;33(1):117–23.
mens and drug exposures affect either outcome or 45. Berends SE, Strik AS, Löwenberg M, D’Haens GR,
adverse events? A systematic review. J Antimicrob Mathôt RAA. Clinical pharmacokinetic and pharma-
Chemother. 2016;71(10):2754–9. codynamic considerations in the treatment of ulcer-
33. Duszynska W, Taccone FS, Hurkacz M, Kowalska-­ ative colitis. Clin Pharmacokinet [Internet]. 2019
Krochmal B, Wiela-Hojeńska A, Kübler [cited 2022 Feb 28];58(1):15–37. Available from:
A. Therapeutic drug monitoring of amikacin in sep- https://doi.org/10.1007/s40262-­018-­0676-­z.
tic patients. Crit Care Lond Engl. 2013;17(4):R165. 46. McInnes G, Lavertu A, Sangkuhl K, Klein TE,
34. Filler G. Abbreviated mycophenolic acid AUC from Whirl-Carrillo M, Altman RB. Pharmacogenetics
C0, C1, C2, and C4 is preferable in children after at scale: an analysis of the UK Biobank. Clin
renal transplantation on mycophenolate mofetil and Pharmacol Ther. 2021;109(6):1528–37.
tacrolimus therapy. Transpl Int. 2004;17(3):120–5. 47. Hendijani F, Azarpira N, Kaviani M. Effect of
35. Villeneuve D, Brothers A, Harvey E, Kemna M, CYP3A5*1 expression on tacrolimus required dose
Law Y, Nemeth T, et al. Valganciclovir dosing using
13 Therapeutic Drug Monitoring and Toxicology: Relevance of Measuring Metabolites 221

for transplant pediatrics: A systematic review and Available from: https://www.ncbi.nlm.nih.gov/pmc/


meta-analysis. Pediatr Transplant. 2018;19:e13248. articles/PMC2695569/.
48. Thomson AW, Bonham CA, Zeevi A. Mode of 61. Potter JM, Donnelly A. Carbamazepine-10,11-­
action of tacrolimus (FK506): molecular and cellular epoxide in therapeutic drug monitoring. Ther Drug
mechanisms. Ther Drug Monit. 1995;17(6):584–91. Monit. 1998;20(6):652–7.
49. Morgan RA. Human tumor xenografts: the good, the 62. Yacobi A, Zlotnick S, Colaizzi JL, Moros D, Masson
bad, and the ugly. Mol Ther [Internet]. 2012 [cited E, Abolfathi Z, et al. A multiple-dose safety and
2022 Jan 19];20(5):882–4. Available from: https:// bioequivalence study of a narrow therapeutic index
www.ncbi.nlm.nih.gov/pmc/articles/PMC3345993/. drug: a case for carbamazepine. Clin Pharmacol
50. Golshayan D, Pascual M. Minimization of calci- Ther. 1999;65(4):389–94.
neurin inhibitors to improve long-term outcomes 63. Russell JL, Spiller HA, Baker DD. Markedly elevated
in kidney transplantation. Transpl Immunol. carbamazepine-10,11-epoxide/carbamazepine ratio
2008;20(1–2):21–8. in a fatal carbamazepine ingestion. Case Rep Med
51. Birdwell KA, Decker B, Barbarino JM, Peterson JF, [Internet]. 2015 [cited 2022 Jan 23];2015:369707.
Stein CM, Sadee W, et al. Clinical Pharmacogenetics Available from: https://www.ncbi.nlm.nih.gov/pmc/
Implementation Consortium (CPIC) guidelines for articles/PMC4621337/.
CYP3A5 genotype and tacrolimus dosing. Clin 64. Fan Y, Lin N, Luo L, Fang L, Huang Z, Yu H,
Pharmacol Ther. 2015;98(1):19–24. et al. Pharmacodynamic and pharmacokinetic
52. Willuweit K, Frey A, Hörster A, Saner F, Herzer study of pegylated liposomal doxorubicin com-
K. Real-world administration of once-daily bination (CCOP) chemotherapy in patients with
MeltDose® prolonged-release tacrolimus (LCPT) peripheral T-cell lymphomas. Acta Pharmacol Sin.
allows for dose reduction of tacrolimus and stabilizes 2011;32(3):408–14.
graft function following liver transplantation. J Clin 65. Licata S, Saponiero A, Mordente A, Minotti
Med [Internet]. 2020 [cited 2022 Feb 28];10(1):124. G. Doxorubicin metabolism and toxicity in human
Available from: https://www.ncbi.nlm.nih.gov/pmc/ myocardium: role of cytoplasmic deglycosida-
articles/PMC7795274/. tion and carbonyl reduction. Chem Res Toxicol.
53. Giardina EG. Procainamide: clinical pharmacology 2000;13(5):414–20.
and efficacy against ventricular arrhythmias. Ann N 66. Rebollo J, Valenzuela B, Duart-Duart M, Escudero-­
Y Acad Sci. 1984;432:177–88. Ortiz V, Gonzalez MS, Brugarolas A. Use of thera-
54. Harron DW, Brogden RN. Acecainide peutic drug monitoring of cancer chemotherapy
(N-acetylprocainamide). A review of its pharma- to modify initial per-protocol doses. J Clin Oncol.
codynamic and pharmacokinetic properties, and 2010;28(15_suppl):e13015.
therapeutic potential in cardiac arrhythmias. Drugs. 67. Zeng X, Cai H, Yang J, Qiu H, Cheng Y, Liu
1990;39(5):720–40. M. Pharmacokinetics and cardiotoxicity of doxoru-
55. Kluger J, Drayer D, Reidenberg M, Ellis G, Lloyd V, bicin and its secondary alcohol metabolite in rats.
Tyberg T, et al. The clinical pharmacology and antiar- Biomed Pharmacother. 2019;116:108964.
rhythmic efficacy of acetylprocainamide in patients 68. Harahap Y, Ardiningsih P, Corintias Winarti A,
with arrhythmias. Am J Cardiol. 1980;45(6):1250–7. Purwanto DJ. Analysis of the doxorubicin and doxo-
56. Winkle RA, Jaillon P, Kates RE, Peters rubicinol in the plasma of breast cancer patients for
F. Clinical pharmacology and antiarrhythmic effi- monitoring the toxicity of doxorubicin. Drug Des
cacy of N-acetylprocainamide. Am J Cardiol. Devel Ther. 2020;14:3469–75.
1981;47(1):123–30. 69. Manyike PT, Kharasch ED, Kalhorn TF, Slattery
57. Prasaja B, Sasongko L, Harahap Y, Hardiyanti, JT. Contribution of CYP2E1 and CYP3A to acet-
Lusthom W, Grigg M. Simultaneous quantification aminophen reactive metabolite formation. Clin
of losartan and active metabolite in human plasma by Pharmacol Ther. 2000;67(3):275–82.
liquid chromatography–tandem mass spectrometry 70. Masubuchi Y. Metabolic and non-metabolic factors
using irbesartan as internal standard. J Pharm Biomed determining troglitazone hepatotoxicity: a review.
Anal [Internet]. 2009 [cited 2022 Jan 9];49(3):862– Drug Metab Pharmacokinet. 2006;21(5):347–56.
7. Available from: https://www.sciencedirect.com/ 71. Thompson CD, Barthen MT, Hopper DW, Miller
science/article/pii/S0731708509000326. TA, Quigg M, Hudspeth C, et al. Quantification in
58. Sica DA, Gehr TWB, Ghosh S. Clinical phar- patient urine samples of felbamate and three metab-
macokinetics of losartan. Clin Pharmacokinet. olites: acid carbamate and two mercapturic acids.
2005;44(8):797–814. Epilepsia. 1999 Jun;40(6):769–76.
59. Pinder RM, Brogden RN, Sawyer PR, Speight TM, 72. Grillo MP, Knutson CG, Sanders PE, Waldon DJ,
Avery GS. Fenfluramine: a review of its pharmaco- Hua F, Ware JA. Studies on the chemical reactiv-
logical properties and therapeutic efficacy in obesity. ity of diclofenac acyl glucuronide with glutathi-
Drugs. 1975;10(4):241–323. one: identification of diclofenac-S-acyl-glutathione
60. Rothman RB, Baumann MH. Serotonergic drugs in rat bile. Drug Metab Dispos Biol Fate Chem.
and valvular heart disease. Expert Opin Drug Saf 2003;31(11):1327–36.
[Internet]. 2009 [cited 2022 Jan 9];8(3):317–29.
222 J. Akingbasote et al.

73. Siraki AG, Deterding LJ, Bonini MG, Jiang J, Available from: https://doi.org/10.1517/17425255.2
Ehrenshaft M, Tomer KB, et al. Procainamide, but 014.885016.
not N-acetylprocainamide, induces protein free 84. Stamp LK, Barclay M. Therapeutic drug monitoring
radical formation on myeloperoxidase: a potential in rheumatic diseases: utile or futile? Rheumatology
mechanism of agranulocytosis. Chem Res Toxicol [Internet]. 2014 [cited 2022 Jan 23];53(6):988–
[Internet]. 2008 [cited 2022 Jan 3];21(5):1143–53. 97. Available from: https://doi.org/10.1093/
Available from: https://doi.org/10.1021/tx700415b. rheumatology/ket355.
74. Robison TW, Jacobs A. Metabolites in safety testing. 85. Venkataramanan R, Shaw LM. Therapeutic monitor-
Bioanalysis. 2009;1(7):1193–200. ing of mycophenolic acid in liver transplant patients.
75. FDA. Safety testing of drug metabolites Liver Transpl [Internet]. 2004 [cited 2022 Jan
[Internet]. U.S. Food and Drug Administration. 23];10(4):503–5. Available from: https://onlineli-
FDA; 2020 [cited 2022 Jan 9]. Available brary.wiley.com/doi/abs/10.1002/lt.20125.
from: https://www.fda.gov/regulatory-­­ 86. Wartofsky L. Levothyroxine: therapeutic use and
information/search-­f da-­g uidance-­d ocuments/ regulatory issues related to bioequivalence. Expert
safety-­testing-­drug-­metabolites. Opin Pharmacother. 2002;3(6):727–32.
76. Baillie TA, Cayen MN, Fouda H, Gerson RJ, Green 87. Akingbasote JA. Utility of the HRN™ (hepatic cyp
JD, Grossman SJ, et al. Drug metabolites in safety reductase null) mice for investigating mechanisms
testing. Toxicol Appl Pharmacol [Internet]. 2002 of liver toxicity of carboxylic-acid-containing drugs
[cited 2022 Jan 9];182(3):188–96. Available from: [Internet] [m_rs]. University of Birmingham; 2012
https://www.sciencedirect.com/science/article/pii/ [cited 2017 Oct 20]. Available from: http://etheses.
S0041008X02994408. bham.ac.uk/3717/.
77. Garg U, Frazee C. Chapter 7 – Therapeutic drug 88. Quadri SS, Stratford RE, Boué SM, Cole
monitoring in infants and children. In: Clarke W, RB. Identification of glyceollin metabolites
Dasgupta A, editors. Clinical challenges in therapeu- derived from conjugation with glutathione and
tic drug monitoring [Internet]. San Diego: Elsevier; glucuronic acid in male ZDSD rats by online liq-
2016 [cited 2022 Mar 5]. p. 165–84. Available from: uid chromatography-­ electrospray ionization tan-
https://www.sciencedirect.com/science/article/pii/ dem mass spectrometry. J Agric Food Chem.
B9780128020258000076. 2014;62(12):2692–700.
78. Skonberg C, Olsen J, Madsen KG, Hansen SH, Grillo 89. Macherey A-C, Dansette PM. Chapter 25 –
MP. Metabolic activation of carboxylic acids. Expert Biotransformations leading to toxic metabolites:
Opin Drug Metab Toxicol. 2008;4(4):425–38. chemical aspects. In: Wermuth CG, Aldous D,
79. Fung M, Thornton A, Mybeck K, Wu JH, Raboisson P, Rognan D, editors. The practice of
Hornbuckle K, Muniz E. Evaluation of the charac- medicinal chemistry. 4th ed [Internet]. San Diego:
teristics of safety withdrawal of prescription drugs Academic Press; 2015 [cited 2022 Jan 23]. p. 585–
from worldwide pharmaceutical markets-1960 to 614. Available from: https://www.sciencedirect.com/
1999. Drug Inf J [Internet]. 2001 [cited 2022 Jan science/article/pii/B9780124172050000250.
22];35(1):293–317. Available from: https://doi. 90. Ghodke-Puranik Y, Thorn CF, Lamba JK, Leeder
org/10.1177/009286150103500134. JS, Song W, Birnbaum AK, et al. Valproic acid path-
80. Chang JB, Quinnies KM, Realubit R, Karan C, Rand way: pharmacokinetics and pharmacodynamics.
JH, Tatonetti NP. A novel, rapid method to compare Pharmacogenet Genomics [Internet]. 2013 [cited
the therapeutic windows of oral anticoagulants using 2022 Jan 23];23(4):236–41. Available from: https://
the Hill coefficient. Sci Rep [Internet]. 2016 [cited www.ncbi.nlm.nih.gov/pmc/articles/PMC3696515/.
2022 Jan 23];6(1):29387. Available from: https:// 91. Shipkova M, Strassburg CP, Braun F, Streit F,
www.nature.com/articles/srep29387. Gröne H-J, Armstrong VW, et al. Glucuronide
81. Gosselin RC, Dager WE, King JH, Janatpour K, and glucoside conjugation of mycophenolic acid
Mahackian K, Larkin EC, et al. Effect of direct by human liver, kidney and intestinal micro-
thrombin inhibitors, bivalirudin, lepirudin, and arg- somes. Br J Pharmacol [Internet]. 2001 [cited
atroban, on prothrombin time and INR values. Am J 2022 Jan 23];132(5):1027–34. Available from:
Clin Pathol. 2004;121(4):593–9. https://onlinelibrary.wiley.com/doi/abs/10.1038/
82. Raebel P, Nikki M. Carroll MS, Susan E. Andrade sj.bjp.0703898.
S, Elizabeth A. Chester P, Jennifer Elston Lafata P, 92. Claesen M, Moustafa MA, Adline J, Vandervorst D,
Adrianne Feldstein MD, et al. Monitoring of drugs Poupaert JH. Evidence for an arene oxide-NIH shift
with a narrow therapeutic range in ambulatory care. pathway in the metabolic conversion of phenytoin to
2006 [cited 2022 Jan 23]; Available from: https:// 5-(4-hydroxyphenyl)-5-phenylhydantoin in the rat
www.ajmc.com/view/may06-­2302p268-­274. and in man. Drug Metab Dispos Biol Fate Chem.
83. Said R, Tsimberidou AM. Pharmacokinetic evalu- 1982;10(6):667–71.
ation of vincristine for the treatment of lymphoid 93. Manson MM. Epoxides--is there a human health
malignancies. Expert Opin Drug Metab Toxicol problem? Br J Ind Med. 1980;37(4):317–36.
[Internet]. 2014 [cited 2022 Jan 23];10(3):483–94. 94. Niederer C, Behra R, Harder A, Schwarzenbach
RP, Escher BI. Mechanistic approaches for evalu-
13 Therapeutic Drug Monitoring and Toxicology: Relevance of Measuring Metabolites 223

ating the toxicity of reactive organochlorines and 2013 [cited 2022 Mar 21];22(3):198–202.
epoxides in green algae. Environ Toxicol Chem Available from: https://doi.org/10.1177/20101
[Internet]. 2004 [cited 2022 Jan 23];23(3):697–704. 0581302200307.
Available from: https://onlinelibrary.wiley.com/doi/ 106. McGill MR, Jaeschke H. Metabolism and disposi-
abs/10.1897/03-­83. tion of acetaminophen: recent advances in rela-
95. Simper GS, Hò G-GT, Celik AA, Huyton T, Kuhn tion to hepatotoxicity and diagnosis. Pharm Res.
J, Kunze-Schumacher H, et al. Carbamazepine-­ 2013;30(9):2174–87.
mediated adverse drug reactions: CBZ-10,11-­ 107. Larson AM, Polson J, Fontana RJ, Davern TJ,
epoxide but not carbamazepine induces the Lalani E, Hynan LS, et al. Acetaminophen-induced
alteration of peptides presented by HLA-B∗15:02. acute liver failure: results of a United States mul-
J Immunol Res [Internet]. 2018 [cited 2022 Feb ticenter, prospective study. Hepatol Baltim MD.
6];2018:5086503. Available from: https://www.ncbi. 2005;42(6):1364–72.
nlm.nih.gov/pmc/articles/PMC6158965/. 108. Coles B, Wilson I, Wardman P, Hinson JA, Nelson
96. Yoshimura R, Nakamura J, Eto S, Ueda N. Possible SD, Ketterer B. The spontaneous and enzymatic
relationships between plasma carbamazepine-­10,11-­ reaction of N-acetyl-p-benzoquinonimine with
epoxide levels and antimanic efficacy and side glutathione: a stopped-flow kinetic study. Arch
effects in patients with schizoaffective disorde. Hum Biochem Biophys. 1988;264(1):253–60.
Psychopharmacol. 2000;15(4):237–40. 109. Koenderink JB, van den Heuvel JJMW, Bilos
97. Spina E, Pisani F, Perucca E. Clinically signifi- A, Vredenburg G, Vermeulen NPE, Russel
cant pharmacokinetic drug interactions with car- FGM. Human multidrug resistance protein 4 (MRP4)
bamazepine. An update. Clin Pharmacokinet. is a cellular efflux transporter for paracetamol glu-
1996;31(3):198–214. tathione and cysteine conjugates. Arch Toxicol.
98. Guttman Y, Nudel A, Kerem Z. Polymorphism in 2020;94(9):3027–32.
cytochrome P450 3A4 is ethnicity related. Front 110. Davern TJ, James LP, Hinson JA, Polson J, Larson
Genet [Internet]. 2019 [cited 2022 Jan 23];10:224. AM, Fontana RJ, et al. Measurement of serum
Available from: https://www.ncbi.nlm.nih.gov/pmc/ acetaminophen-­protein adducts in patients with acute
articles/PMC6433705/. liver failure. Gastroenterology. 2006;130(3):687–94.
99. Iannaccone T, Sellitto C, Manzo V, Colucci F, 111. James LP, Letzig L, Simpson PM, Capparelli E,
Giudice V, Stefanelli B, et al. Pharmacogenetics of Roberts DW, Hinson JA, et al. Pharmacokinetics
carbamazepine and valproate: focus on polymor- of Acetaminophen-Protein Adducts in Adults with
phisms of drug metabolizing enzymes and transport- Acetaminophen Overdose and Acute Liver Failure.
ers. Pharm Basel Switz. 2021;14(3):204. Drug Metab Dispos [Internet]. 2009 [cited 2022 Mar
100. Burianová I, Bořecká K. Routine therapeutic 6];37(8):1779–84. Available from: https://www.
monitoring of the active metabolite of carbam- ncbi.nlm.nih.gov/pmc/articles/PMC2712440/.
azepine: is it really necessary? Clin Biochem. 112. Cohen IV, Cirulli ET, Mitchell MW, Jonsson TJ, Yu
2015;48(13–14):866–9. J, Shah N, et al. Acetaminophen (paracetamol) use
101. Zevin S. Chapter 42 – Pharmacologic interactions in modifies the sulfation of sex hormones. eBioMedi-
the CICU. In: Jeremias A, Brown DL, editors. Cardiac cine [Internet]. 2018 [cited 2022 Mar 12];28:316–
intensive care. 2nd ed [Internet]. Philadelphia: 23. Available from: https://www.thelancet.com/
W.B. Saunders; 2010 [cited 2022 Mar 20]. p. 516– article/S2352-­3964(18)30037-­9/fulltext.
31. Available from: https://www.sciencedirect.com/ 113. Kane RE, Li AP, Kaminski DR. Sulfation and gluc-
science/article/pii/B9781416037736100424 uronidation of acetaminophen by human hepatocytes
102. Vivithanaporn P, Kongratanapasert T, Suriyapakorn cultured on Matrigel and type 1 collagen reproduces
B, Songkunlertchai P, Mongkonariyawong P, conjugation in vivo. Drug Metab Dispos Biol Fate
Limpikirati PK, et al. Potential drug-drug interac- Chem. 1995;23(3):303–7.
tions of antiretrovirals and antimicrobials detected 114. Xiong Y, Uys JD, Tew KD, Townsend
by three databases. Sci Rep [Internet]. 2021 [cited DM. S-glutathionylation: from molecular mecha-
2022 Mar 20];11(1):6089. Available from: https:// nisms to health outcomes. Antioxid Redox Signal
www.nature.com/articles/s41598-­021-­85586-­8. [Internet]. 2011 [cited 2022 Jan 21];15(1):233.
103. Ludden TM. Nonlinear pharmacokinetics: Available from: https://www.ncbi.nlm.nih.gov/pmc/
clinical Implications. Clin Pharmacokinet. articles/PMC3110090/.
1991;20(6):429–46. 115. Schmidt JC, Dougherty BV, Beger RD, Jones
104. Ogilvie RI. Monitoring plasma theophylline con- DP, Schmidt MA, Mattes WB. Metabolomics as a
centrations. Ther Drug Monit [Internet]. 1980 truly translational tool for precision medicine. Int J
[cited 2022 Mar 21];2(2):111–8. Available from: Toxicol. 2021;40(5):413–26.
https://journals.lww.com/drug-­­m onitoring/ 116. Wilson ID. Drugs, bugs, and personalized medicine:
Abstract/1980/04000/Monitoring_Plasma_ pharmacometabonomics enters the ring. Proc Natl
Theophylline_Concentrations.1.aspx. Acad Sci U S A. 2009;106(34):14187–8.
105. Wu MF, Lim WH. Phenytoin: a guide to therapeu- 117. Niemann C, Gauthier J-C, Richert L, Ivanov M-A,
tic drug monitoring. Proc Singap Healthc [Internet]. Melcion C, Cordier A. Rat adult hepatocytes in pri-
224 J. Akingbasote et al.

mary pure and mixed monolayer culture: compari- 131. Everse J. Heme proteins. In: Lennarz WJ, Lane
son of the maintenance of mixed function oxidase MD, editors. Encyclopedia of biological chemistry
and conjugation pathways of drug metabolism. [Internet]. New York: Elsevier; 2004 [cited 2022 Feb
Biochem Pharmacol. 1991;42(2):373–9. 5]. p. 354–61. Available from: https://www.science-
118. Sipes IG. Biotransformation of toxicants. direct.com/science/article/pii/B0124437109003045.
Toxicology. 1991:88–126. 132. Pohl LR, Nelson SD, Krishna G. Investigation of
119. Choi-Sledeski YM, Wermuth CG. Chapter the mechanism of the metabolic activation of chlor-
28 – Designing prodrugs and bioprecursors. In: amphenicol by rat liver microsomes. Identification
Wermuth CG, Aldous D, Raboisson P, Rognan D, of a new metabolite. Biochem Pharmacol.
editors. The practice of medicinal chemistry. 4th 1978;27(4):491–6.
ed [Internet]. San Diego: Academic Press; 2015 133. Fontana E, Dansette PM, Poli SM. Cytochrome
[cited 2022 Jan 30]. p. 657–96. Available from: p450 enzymes mechanism based inhibitors: com-
https://www.sciencedirect.com/science/article/pii/ mon sub-structures and reactivity. Curr Drug Metab.
B9780124172050000286. 2005;6(5):413–54.
120. Attia SM. Deleterious effects of reactive metabolites. 134. Halpert JR, Miller NE, Gorsky LD. On the mecha-
Oxidative Med Cell Longev. 2010;3(4):238–53. nism of the inactivation of the major phenobarbital-­
121. Hughes TB, Dang NL, Miller GP, Swamidass inducible isozyme of rat liver cytochrome P-450 by
SJ. Modeling reactivity to biological macromol- chloramphenicol. J Biol Chem [Internet]. 1985 [cited
ecules with a deep multitask network. ACS Cent Sci. 2022 Feb 6];260(14):8397–403. Available from:
2016;2(8):529–37. https://www.sciencedirect.com/science/article/pii/
122. Juan CA, Pérez de la Lastra JM, Plou FJ, Pérez-­ S0021925817394875.
Lebeña E. The chemistry of reactive oxygen species 135. Benasutti M, Ejadi S, Whitlow MD, Loechler
(ROS) revisited: outlining their role in biological EL. Mapping the binding site of aflatoxin B1 in
macromolecules (DNA, lipids and proteins) and DNA: systematic analysis of the reactivity of afla-
induced pathologies. Int J Mol Sci. 2021;22(9):4642. toxin B1 with guanines in different DNA sequences.
123. Guengerich FP. Cytochrome P450 oxidations in Biochemistry [Internet]. 1988 [cited 2022 Feb
the generation of reactive electrophiles: epoxida- 6];27(1):472–81. Available from: https://doi.
tion and related reactions. Arch Biochem Biophys. org/10.1021/bi00401a068.
2003;409(1):59–71. 136. Hamid AS, Tesfamariam IG, Zhang Y, Zhang
124. Guengerich FP. Common and uncommon cyto- ZG. Aflatoxin B1-induced hepatocellular carcinoma
chrome P450 reactions related to metabo- in developing countries: Geographical distribution,
lism and chemical toxicity. Chem Res Toxicol. mechanism of action and prevention. Oncol Lett
2001;14(6):611–50. [Internet]. 2013 [cited 2022 Feb 6];5(4):1087–92.
125. Isin EM, Guengerich FP. Complex reac- Available from: https://www.ncbi.nlm.nih.gov/pmc/
tions catalyzed by cytochrome P450 enzymes. articles/PMC3629261/.
Biochim Biophys Acta BBA – Gen Subj. 137. Tomson T, Bertilsson L. Potent therapeutic effect of
2007;1770(3):314–29. carbamazepine-10,11-epoxide in trigeminal neural-
126. King CD, Rios GR, Green MD, Tephly TR. UDP-­ gia. Arch Neurol. 1984;41(6):598–601.
glucuronosyltransferases. Curr Drug Metab 138. Rietjens IM, den Besten C, Hanzlik RP, van
[Internet]. 2000 [cited 2022 Feb 5];1(2):143–61. Bladeren PJ. Cytochrome P450-catalyzed oxida-
Available from: https://www.eurekaselect.com/ tion of halobenzene derivatives. Chem Res Toxicol.
article/10514. 1997;10(6):629–35.
127. Timbrell JA. Principles of biochemical toxicology. 139. Aldred EM, Buck C, Vall K. Chapter 7 – Free
4th ed. Boca Raton: CRC Press; 2013. 464 p. radicals. In: Aldred EM, Buck C, Vall K, editors.
128. Damani LA, Houdi AA. Cytochrome P-450 and FAD-­ Pharmacology [Internet]. Edinburgh: Churchill
monooxygenase mediated S- and N-oxygenations. Livingstone; 2009 [cited 2022 Feb 6]. p. 41–52.
Drug Metabol Drug Interact [Internet]. 1988 Dec 1 Available from: https://www.sciencedirect.com/
[cited 2022 Feb 5];6(3–4):235–44. Available from: science/article/pii/B9780443068980000074.
https://www.degruyter.com/document/doi/10.1515/ 140. Dybing E, Søderlund E, Haug LT,
DMDI.1988.6.3-­4.235/html. Thorgeirsson SS. Metabolism and activation of
129. Battelli MG, Polito L, Bortolotti M, Bolognesi 2-­acetylaminofluorene in isolated rat hepatocytes.
A. Xanthine oxidoreductase in drug metabolism: Cancer Res. 1979 Aug;39(8):3268–75.
beyond a role as a detoxifying enzyme. Curr Med 141. Wilson RH, DeEds F, Cox A. The carcinogenic
Chem. 2016;23(35):4027–36. activity of 2-acetaminofluorene. IV. Action of
130. Puente RA, Watkins L, Correa J. Specificity of related compounds. undefined [Internet]. 1947
alcohol dehydrogenases among various sub- [cited 2022 Feb 15]; Available from: https://www.
strates. FASEB J [Internet]. 2013 [cited 2022 Feb semanticscholar.org/paper/The-­C arcinogenic-­
5];27(S1):lb221. Available from: https://onlineli- Activity-­o f-­2 -­A cetaminofluorene.-­Wilson-­D eE
brary.wiley.com/doi/abs/10.1096/fasebj.27.1_sup- ds/3e78c8681dc19a1e7179f5b9b1fb17633af53
plement.lb221. 0fa.
13 Therapeutic Drug Monitoring and Toxicology: Relevance of Measuring Metabolites 225

142. Dansette PM, Bertho G, Mansuy D. First evi- 152. Iohom G, Fitzgerald D, Cunningham AJ. Principles
dence that cytochrome P450 may catalyze both of pharmacogenetics--implications for the anaesthe-
S-oxidation and epoxidation of thiophene deriva- tist. Br J Anaesth. 2004;93(3):440–50.
tives. Biochem Biophys Res Commun [Internet]. 153. Gupta A. Chapter 8 – Etiopathogenesis of insulin
2005 [cited 2022 Feb 16];338(1):450–5. Available resistance. In: Gupta A, editor. Understanding insu-
from: https://www.sciencedirect.com/science/ lin and insulin resistance [Internet]. Elsevier; 2022
article/pii/S0006291X05018036. [cited 2022 Mar 13]. p. 231–73. Available from:
143. Dansette PM, Thang DC, Mansuy HEAD. Evidence https://www.sciencedirect.com/science/article/pii/
for thiophene-s-oxide as a primary reactive metabo- B978012820234000010X.
lite of thiophene in vivo: Formation of a dihydro- 154. Wang Y, He W. Chapter 21 – Endogenous mitochon-
thiophene sulfoxide mercapturic acid. Biochem drial aldehyde dehydrogenase-2 as an antioxidant in
Biophys Res Commun [Internet]. 1992 [cited liver. In: Patel VB, Rajendram R, Preedy VR, editors.
2022 Feb 16];186(3):1624–30. Available from: The liver [Internet]. Boston: Academic Press; 2018
https://www.sciencedirect.com/science/article/pii/ [cited 2022 Mar 12]. p. 247–59. Available from:
S0006291X05815943. https://www.sciencedirect.com/science/article/pii/
144. Treiber A, Dansette PM, Mansuy D. Mechanism B9780128039519000215.
of the aromatic hydroxylation of thiophene by 155. Preissner SC, Hoffmann MF, Preissner R, Dunkel M,
acid-catalyzed peracid oxidation. J Org Chem. Gewiess A, Preissner S. Polymorphic cytochrome
2002;67(21):7261–6. P450 enzymes (CYPs) and their role in personal-
145. Treiber A, Dansette PM, El Amri H, Girault J-P, ized therapy. PLoS One [Internet]. 2013 [cited 2022
Ginderow D, Mornon J-P, et al. Chemical and bio- Mar 12];8(12):e82562. Available from: https://www.
logical oxidation of thiophene: preparation and com- ncbi.nlm.nih.gov/pmc/articles/PMC3858335/.
plete characterization of thiophene S-oxide dimers 156. Zhou S-F, Liu J-P, Chowbay B. Polymorphism
and evidence for thiophene S-oxide as an interme- of human cytochrome P450 enzymes and its
diate in thiophene metabolism in vivo and in vitro. clinical impact. Drug Metab Rev [Internet].
J Am Chem Soc [Internet]. 1997 [cited 2022 Feb 2009 [cited 2022 Feb 6];41(2):89–295.
16];119(7):1565–71. Available from: https://doi. Available from: http://www.tandfonline.com/doi/
org/10.1021/ja962466g. full/10.1080/03602530902843483.
146. Garner AP, Paine MJI, Rodriguez-Crespo I, Chinje 157. Chorley BN, Wang X, Campbell MR, Pittman GS,
EC, De Montellano PO, Stratford IJ, et al. Nitric Noureddine MA, Bell DA. Discovery and verifica-
oxide synthases catalyze the activation of redox tion of functional single nucleotide polymorphisms
cycling and bioreductive anticancer agents. Cancer in regulatory genomic regions: current and devel-
Res. 1999;59(8):1929–34. oping technologies. Mutat Res [Internet]. 2008
147. Gaudiano G, Koch TH. Redox chemistry of anthra- [cited 2022 Mar 13];659(1–2):147–57. Available
cycline antitumor drugs and use of captodative from: https://www.ncbi.nlm.nih.gov/pmc/articles/
radicals as tools for its elucidation and control. PMC2676583/.
Chem Res Toxicol [Internet]. 1991 [cited 2022 158. Johnson AD, Zhang Y, Papp AC, Pinsonneault JK,
Feb 16];4(1):2–16. Available from: https://doi. Lim J-E, Saffen D, et al. Polymorphisms affecting
org/10.1021/tx00019a001. gene transcription and mRNA processing in phar-
148. Dickinson RG. Iso-glucuronides. Curr Drug Metab. macogenetic candidate genes: detection through
2011;12(3):222–8. allelic expression imbalance in human target tis-
149. Stachulski AV, Meng X. Glucuronides from metabo- sues. Pharmacogenet Genomics [Internet]. 2008
lites to medicines: a survey of the in vivo generation, [cited 2022 Mar 13];18(9):781–91. Available
chemical synthesis and properties of glucuronides. from: https://www.ncbi.nlm.nih.gov/pmc/articles/
Nat Prod Rep [Internet]. 2013 [cited 2022 Feb PMC2779843/.
16];30(6):806–48. Available from: https://pubs.rsc. 159. Schrider DR, Hahn MW. Gene copy-number poly-
org/en/content/articlelanding/2013/np/c3np70003h. morphism in nature. Proc R Soc B Biol Sci [Internet].
150. IARC. General discussion of common mecha- 2010 [cited 2022 Mar 13];277(1698):3213–21.
nisms for aromatic amines [Internet]. Some aro- Available from: https://www.ncbi.nlm.nih.gov/pmc/
matic amines, organic dyes, and related exposures. articles/PMC2981937/.
International Agency for Research on Cancer; 2010 160. Sugatani J. Function, genetic polymorphism,
[cited 2022 Feb 16]. Available from: https://www. and transcriptional regulation of human UDP-­
ncbi.nlm.nih.gov/books/NBK385423/. glucuronosyltransferase (UGT) 1A1. Drug Metab
151. Ahmed S, Zhou Z, Zhou J, Chen Pharmacokinet. 2013;28(2):83–92.
S-Q. Pharmacogenomics of drug metabolizing 161. Shenfield GM. Genetic polymorphisms, drug metab-
enzymes and transporters: relevance to precision olism and drug concentrations. Clin Biochem Rev
medicine. Genomics Proteomics Bioinformatics [Internet]. 2004 [cited 2022 Mar 13];25(4):203–6.
[Internet]. 2016 [cited 2022 Mar 12];14(5):298–313. Available from: https://www.ncbi.nlm.nih.gov/pmc/
Available from: https://www.ncbi.nlm.nih.gov/pmc/ articles/PMC1934960/.
articles/PMC5093856/.
226 J. Akingbasote et al.

162. Ohyama K, Nakajima M, Nakamura S, Shimada 175. Paine MF, Schmiedlin-Ren P, Watkins
N, Yamazaki H, Yokoi T. A significant role of PB. Cytochrome P-450 1A1 expression in human
human cytochrome P450 2C8 in amiodarone small bowel: interindividual variation and inhibi-
N-deethylation: an approach to predict the contribu- tion by ketoconazole. Drug Metab Dispos Biol Fate
tion with relative activity factor. Drug Metab Dispos Chem. 1999;27(3):360–4.
Biol Fate Chem. 2000;28(11):1303–10. 176. Eugster HP, Probst M, Würgler FE, Sengstag
163. Zanger UM, Turpeinen M, Klein K, Schwab C. Caffeine, estradiol, and progesterone interact
M. Functional pharmacogenetics/genomics of human with human CYP1A1 and CYP1A2. Evidence
cytochromes P450 involved in drug biotransforma- from cDNA-directed expression in Saccharomyces
tion. Anal Bioanal Chem. 2008;392(6):1093–108. cerevisiae. Drug Metab Dispos Biol Fate Chem.
164. Lin JH, Lu AY. Interindividual variability in inhibi- 1993;21(1):43–9.
tion and induction of cytochrome P450 enzymes. 177. Soyama A, Hanioka N, Saito Y, Murayama N, Ando
Annu Rev Pharmacol Toxicol. 2001;41:535–67. M, Ozawa S, et al. Amiodarone N-deethylation by
165. Paine MF, Hart HL, Ludington SS, Haining RL, CYP2C8 and its variants, CYP2C8*3 and CYP2C8
Rettie AE, Zeldin DC. The human intestinal cyto- P404A. Pharmacol Toxicol. 2002;91(4):174–8.
chrome P450 “pie”. Drug Metab Dispos Biol Fate 178. Kaminsky LS, Zhang ZY. Human P450 metabolism
Chem. 2006;34(5):880–6. of warfarin. Pharmacol Ther. 1997;73(1):67–74.
166. Tracy TS, Chaudhry AS, Prasad B, Thummel KE, 179. Zhang Z, Fasco MJ, Huang Z, Guengerich FP,
Schuetz EG, Zhong X, et al. Interindividual variabil- Kaminsky LS. Human cytochromes P4501A1
ity in cytochrome P450–mediated drug metabolism. and P4501A2: R-warfarin metabolism as a
Drug Metab Dispos [Internet]. 2016 [cited 2022 Mar probe. Drug Metab Dispos Biol Fate Chem.
13];44(3):343–51. Available from: https://www. 1995;23(12):1339–46.
ncbi.nlm.nih.gov/pmc/articles/PMC4767386/. 180. Dixon CM, Colthup PV, Serabjit-Singh CJ, Kerr
167. Yang X, Zhang B, Molony C, Chudin E, Hao K, Zhu BM, Boehlert CC, Park GR, et al. Multiple forms
J, et al. Systematic genetic and genomic analysis of of cytochrome P450 are involved in the metabolism
cytochrome P450 enzyme activities in human liver. of ondansetron in humans. Drug Metab Dispos Biol
Genome Res. 2010;20(8):1020–36. Fate Chem. 1995;23(11):1225–30.
168. Botton MR, Whirl-Carrillo M, Del Tredici 181. Fang J, McKay G, Song J, Remillrd A, Li X, Midha
AL, Sangkuhl K, Cavallari LH, Agúndez JAG, K. In vitro characterization of the metabolism of
et al. PharmVar GeneFocus: CYP2C19. Clin haloperidol using recombinant cytochrome p450
Pharmacol Ther [Internet]. 2021 [cited 2022 Mar enzymes and human liver microsomes. Drug Metab
13];109(2):352–66. Available from: https://onlineli- Dispos Biol Fate Chem. 2001;29(12):1638–43.
brary.wiley.com/doi/abs/10.1002/cpt.1973. 182. Wang RW, Liu L, Cheng H. Identification of human
169. Nelson DR, Zeldin DC, Hoffman SMG, Maltais LJ, liver cytochrome P450 isoforms involved in the
Wain HM, Nebert DW. Comparison of cytochrome in vitro metabolism of cyclobenzaprine. Drug Metab
P450 (CYP) genes from the mouse and human Dispos Biol Fate Chem. 1996;24(7):786–91.
genomes, including nomenclature recommendations 183. Masubuchi Y, Hosokawa S, Horie T, Suzuki T,
for genes, pseudogenes and alternative-splice vari- Ohmori S, Kitada M, et al. Cytochrome P450
ants. Pharmacogenetics. 2004;14(1):1–18. isozymes involved in propranolol metabo-
170. Nebert DW, Russell DW. Clinical importance lism in human liver microsomes. The role of
of the cytochromes P450. Lancet Lond Engl. CYP2D6 as ring-­ hydroxylase and CYP1A2 as
2002;360(9340):1155–62. N-desisopropylase. Drug Metab Dispos Biol Fate
171. Nebert DW, Wikvall K, Miller WL. Human cyto- Chem. 1994;22(6):909–15.
chromes P450 in health and disease. Philos Trans 184. Zhou S-F, Yang L-P, Zhou Z-W, Liu Y-H, Chan
R Soc B Biol Sci [Internet]. 2013 [cited 2022 Mar E. Insights into the substrate specificity, inhibitors,
13];368(1612):20120431. Available from: https:// regulation, and polymorphisms and the clinical
www.ncbi.nlm.nih.gov/pmc/articles/PMC3538421/. impact of human cytochrome P450 1A2. AAPS J
172. Drahushuk AT, McGarrigle BP, Larsen KE, [Internet]. 2009 [cited 2022 Mar 13];11(3):481–94.
Stegeman JJ, Olson JR. Detection of CYP1A1 Available from: https://www.ncbi.nlm.nih.gov/pmc/
protein in human liver and induction by TCDD in articles/PMC2758120/.
precision-cut liver slices incubated in dynamic organ 185. Zhang H-F, Wang H-H, Gao N, Wei J-Y, Tian X,
culture. Carcinogenesis. 1998;19(8):1361–8. Zhao Y, et al. Physiological content and intrinsic
173. Schweikl H, Taylor JA, Kitareewan S, Linko activities of 10 cytochrome P450 isoforms in human
P, Nagorney D, Goldstein JA. Expression of normal liver microsomes. J Pharmacol Exp Ther
CYP1A1 and CYP1A2 genes in human liver. [Internet]. 2016 [cited 2022 Feb 6];358(1):83–93.
Pharmacogenetics. 1993;3(5):239–49. Available from: http://jpet.aspetjournals.org/cgi/
174. Lang D, Radtke M, Bairlein M. Highly variable doi/10.1124/jpet.116.233635.
expression of CYP1A1 in human liver and impact 186. Fuhr U, Rost KL, Engelhardt R, Sachs M, Liermann
on pharmacokinetics of riociguat and granisetron in D, Belloc C, et al. Evaluation of caffeine as a test
humans. Chem Res Toxicol. 2019;32(6):1115–22. drug for CYP1A2, NAT2 and CYP2E1 phenotyp-
13 Therapeutic Drug Monitoring and Toxicology: Relevance of Measuring Metabolites 227

ing in man by in vivo versus in vitro correlations. Available from: https://www.ncbi.nlm.nih.gov/pmc/


Pharmacogenetics. 1996;6(2):159–76. articles/PMC8401128/.
187. Bertilsson L, Carrillo JA, Dahl ML, Llerena A, Alm 198. Olesen OV, Linnet K. Contributions of five human
C, Bondesson U, et al. Clozapine disposition cova- cytochrome P450 isoforms to the N-demethylation
ries with CYP1A2 activity determined by a caffeine of clozapine in vitro at low and high concentrations.
test. Br J Clin Pharmacol. 1994;38(5):471–3. J Clin Pharmacol. 2001;41(8):823–32.
188. Oda Y, Furuichi K, Tanaka K, Hiroi T, Imaoka S, 199. Ulrich S, Baumann B, Wolf R, Lehmann D, Peters
Asada A, et al. Metabolism of a new local anesthetic, B, Bogerts B, et al. Therapeutic drug monitoring
ropivacaine, by human hepatic cytochrome P450. of clozapine and relapse--a retrospective study of
Anesthesiology. 1995;82(1):214–20. routine clinical data. Int J Clin Pharmacol Ther.
189. Ring BJ, Catlow J, Lindsay TJ, Gillespie T, Roskos 2003;41(1):3–13.
LK, Cerimele BJ, et al. Identification of the human 200. Eap CB, Bender S, Jaquenoud Sirot E, Cucchia G,
cytochromes P450 responsible for the in vitro forma- Jonzier-Perey M, Baumann P, et al. Nonresponse
tion of the major oxidative metabolites of the anti- to clozapine and ultrarapid CYP1A2 activity:
psychotic agent olanzapine. J Pharmacol Exp Ther. clinical data and analysis of CYP1A2 gene. J Clin
1996;276(2):658–66. Psychopharmacol. 2004;24(2):214–9.
190. Orlando R, Piccoli P, De Martin S, Padrini R, 201. Melkersson KI, Scordo MG, Gunes A, Dahl
Floreani M, Palatini P. Cytochrome P450 1A2 is a M-L. Impact of CYP1A2 and CYP2D6 polymor-
major determinant of lidocaine metabolism in vivo: phisms on drug metabolism and on insulin and lipid
effects of liver function. Clin Pharmacol Ther. elevations and insulin resistance in clozapine-treated
2004;75(1):80–8. patients. J Clin Psychiatry. 2007;68(5):697–704.
191. Koyama E, Chiba K, Tani M, Ishizaki T. Reappraisal 202. Kamış GZ, Ayhan Y, Basar K, Özer S, Yağcıoğlu
of human CYP isoforms involved in imipramine AEA. A case of clozapine intoxication pre-
N-demethylation and 2-hydroxylation: a study using senting with atypical NMS symptoms. Int J
microsomes obtained from putative extensive and Neuropsychopharmacol [Internet]. 2014 [cited 2022
poor metabolizers of S-mephenytoin and eleven Mar 13];17(5):819–21. Available from: https://doi.
recombinant human CYPs. J Pharmacol Exp Ther. org/10.1017/S1461145713001624.
1997;281(3):1199–210. 203. Sartorius A, Hewer W, Zink M, Henn FA. High-dose
192. Kroemer HK, Gautier JC, Beaune P, Henderson C, clozapine intoxication. J Clin Psychopharmacol.
Wolf CR, Eichelbaum M. Identification of P450 2002;22(1):91–2.
enzymes involved in metabolism of verapamil in 204. Totah RA, Rettie AE. Cytochrome P450 2C8: sub-
humans. Naunyn Schmiedeberg's Arch Pharmacol. strates, inhibitors, pharmacogenetics, and clinical
1993;348(3):332–7. relevance. Clin Pharmacol Ther. 2005;77(5):341–52.
193. Botsch S, Gautier JC, Beaune P, Eichelbaum M, 205. Daly AK, Rettie AE, Fowler DM, Miners
Kroemer HK. Identification and characteriza- JO. Pharmacogenomics of CYP2C9: functional and
tion of the cytochrome P450 enzymes involved in clinical considerations. J Pers Med. 2017;8(1):E1.
N-dealkylation of propafenone: molecular base for 206. Kirchheiner J, Brockmöller J. Clinical consequences
interaction potential and variable disposition of active of cytochrome P450 2C9 polymorphisms. Clin
metabolites. Mol Pharmacol. 1993;43(1):120–6. Pharmacol Ther. 2005;77(1):1–16.
194. Uttamsingh V, Lu C, Miwa G, Gan L-S. Relative 207. Shao Z, Kyriakopoulou LG, Ito S. Chapter
contributions of the five major human cytochromes 14 – Pharmacogenomics. In: Hempel G, editor.
P450, 1A2, 2C9, 2C19, 2D6, and 3A4, to the Handbook of analytical separations [Internet].
hepatic metabolism of the proteasome inhibitor Elsevier Science B.V.; 2020 [cited 2022 Mar 13].
bortezomib. Drug Metab Dispos Biol Fate Chem. p. 321–53. (Methods of therapeutic drug monitoring
2005;33(11):1723–8. including pharmacogenetics; vol. 7). Available from:
195. Gunes A, Dahl M-L. Variation in CYP1A2 activ- https://www.sciencedirect.com/science/article/pii/
ity and its clinical implications: influence of envi- B9780444640666000149.
ronmental factors and genetic polymorphisms. 208. Gage BF, Lesko LJ. Pharmacogenetics of warfarin:
Pharmacogenomics. 2008;9(5):625–37. regulatory, scientific, and clinical issues. J Thromb
196. Coffee and Caffeine Genetics Consortium, Cornelis Thrombolysis. 2008;25(1):45–51.
MC, Byrne EM, Esko T, Nalls MA, Ganna A, et al. 209. Jorgensen AL, FitzGerald RJ, Oyee J, Pirmohamed
Genome-wide meta-analysis identifies six novel loci M, Williamson PR. Influence of CYP2C9 and
associated with habitual coffee consumption. Mol VKORC1 on patient response to warfarin: a sys-
Psychiatry. 2015;20(5):647–56. tematic review and meta-analysis. PLoS One.
197. Kumondai M, Gutiérrez Rico EM, Hishinuma E, 2012;7(8):e44064.
Nakanishi Y, Yamazaki S, Ueda A, et al. Functional 210. Caudle KE, Rettie AE, Whirl-Carrillo M, Smith LH,
characterization of 21 rare allelic cyp1a2 variants Mintzer S, Lee MTM, et al. Clinical pharmacoge-
identified in a population of 4773 Japanese individu- netics implementation consortium guidelines for
als by assessing phenacetin O-deethylation. J Pers CYP2C9 and HLA-B genotypes and phenytoin dos-
Med [Internet]. 2021 [cited 2022 Mar 13];11(8):690. ing. Clin Pharmacol Ther. 2014;96(5):542–8.
228 J. Akingbasote et al.

211. Desta Z, Zhao X, Shin J-G, Flockhart DA. Clinical 222. Sun W, Li Y, Li J, Zhang Z, Zhu W, Liu W, et al.
significance of the cytochrome P450 2C19 Variant recurrent risk among stroke patients with dif-
genetic polymorphism. Clin Pharmacokinet. ferent CYP2C19 phenotypes and treated with clopi-
2002;41(12):913–58. dogrel. Platelets. 2015;26(6):558–62.
212. Gardiner SJ, Begg EJ. Pharmacogenetics, drug-­ 223. Tornio A, Backman JT. Cytochrome P450 in phar-
metabolizing enzymes, and clinical practice. macogenetics: an update. In: Advances in pharma-
Pharmacol Rev. 2006;58(3):521–90. cology [Internet]. Elsevier; 2018 [cited 2022 Feb 6].
213. Dean L. Diazepam therapy and CYP2C19 genotype. p. 3–32. Available from: https://linkinghub.elsevier.
In: Pratt VM, Scott SA, Pirmohamed M, Esquivel com/retrieve/pii/S1054358918300267.
B, Kane MS, Kattman BL, et al., editors. Medical 224. Sibbing D, Koch W, Gebhard D, Schuster T, Braun
genetics summaries [Internet]. Bethesda: National S, Stegherr J, et al. Cytochrome 2C19*17 allelic
Center for Biotechnology Information (US); 2012 variant, platelet aggregation, bleeding events, and
[cited 2022 Mar 13]. Available from: http://www. stent thrombosis in clopidogrel-treated patients
ncbi.nlm.nih.gov/books/NBK379740/. with coronary stent placement. Circulation.
214. Purkins L, Wood N, Ghahramani P, Love ER, Eve 2010;121(4):512–8.
MD, Fielding A. Coadministration of voriconazole 225. He Z-X, Chen X-W, Zhou Z-W, Zhou S-F. Impact
and phenytoin: pharmacokinetic interaction, safety, of physiological, pathological and environmental
and toleration. Br J Clin Pharmacol [Internet]. 2003 factors on the expression and activity of human
[cited 2022 Mar 13];56(Suppl 1):37–44. Available cytochrome P450 2D6 and implications in precision
from: https://www.ncbi.nlm.nih.gov/pmc/articles/ medicine. Drug Metab Rev. 2015;47(4):470–519.
PMC1884312/. 226. Dalton R, Lee S, Claw KG, Prasad B, Phillips BR,
215. Kazui M, Nishiya Y, Ishizuka T, Hagihara K, Shen DD, et al. Interrogation of CYP2D6 structural
Farid NA, Okazaki O, et al. Identification of variant alleles improves the correlation between
the human cytochrome P450 enzymes involved CYP2D6 genotype and CYP2D6-mediated meta-
in the two oxidative steps in the bioactivation bolic activity. Clin Transl Sci [Internet]. 2020 [cited
of clopidogrel to its pharmacologically active 2022 Mar 13];13(1):147–56. Available from: https://
metabolite. Drug Metab Dispos Biol Fate Chem. www.ncbi.nlm.nih.gov/pmc/articles/PMC6951848/.
2010;38(1):92–9. 227. Gaedigk A, Sangkuhl K, Whirl-Carrillo M, Klein T,
216. Zhong Z, Hou J, Li B, Zhang Q, Liu S, Li C, et al. Leeder JS. Prediction of CYP2D6 phenotype from
Analysis of CYP2C19 genetic polymorphism in a genotype across world populations. Genet Med.
large ethnic Hakka population in Southern China. 2017;19(1):69–76.
Med Sci Monit Int Med J Exp Clin Res [Internet]. 228. Darney K, Lautz LS, Béchaux C, Wiecek W, Testai
2017 [cited 2022 Mar 13];23:6186–92. Available E, Amzal B, et al. Human variability in polymor-
from: https://www.ncbi.nlm.nih.gov/pmc/articles/ phic CYP2D6 metabolism: Implications for the
PMC5757864/. risk assessment of chemicals in food and emerging
217. Beavers CJ, Naqvi IA. Clopidogrel. In: StatPearls designer drugs. Environ Int. 2021;156:106760.
[Internet]. Treasure Island: StatPearls Publishing; 229. Crews KR, Gaedigk A, Dunnenberger HM,
2022 [cited 2022 Mar 13]. Available from: http:// Leeder JS, Klein TE, Caudle KE, et al. Clinical
www.ncbi.nlm.nih.gov/books/NBK470539/. Pharmacogenetics Implementation Consortium
218. Scott SA, Sangkuhl K, Stein CM, Hulot J-S, Mega guidelines for cytochrome P450 2D6 genotype and
JL, Roden DM, et al. Clinical Pharmacogenetics codeine therapy: 2014 update. Clin Pharmacol Ther.
Implementation Consortium guidelines for 2014;95(4):376–82.
CYP2C19 genotype and clopidogrel therapy: 2013 230. Somogyi AA, Coller JK, Barratt
update. Clin Pharmacol Ther. 2013;94(3):317–23. DT. Pharmacogenetics of opioid response. Clin
219. Mao L, Jian C, Changzhi L, Dan H, Suihua H, Wenyi Pharmacol Ther. 2015;97(2):125–7.
T, et al. Cytochrome CYP2C19 polymorphism and 231. Shimada T, Yamazaki H, Mimura M, Inui Y,
risk of adverse clinical events in clopidogrel-treated Guengerich FP. Interindividual variations in
patients: a meta-analysis based on 23,035 subjects. human liver cytochrome P-450 enzymes involved
Arch Cardiovasc Dis. 2013;106(10):517–27. in the oxidation of drugs, carcinogens and toxic
220. Mega JL, Simon T, Collet J-P, Anderson JL, chemicals: studies with liver microsomes of 30
Antman EM, Bliden K, et al. Reduced-function Japanese and 30 Caucasians. J Pharmacol Exp Ther.
CYP2C19 genotype and risk of adverse clinical 1994;270(1):414–23.
outcomes among patients treated with clopidogrel 232. Zhou S-F. Drugs behave as substrates, inhibitors
predominantly for PCI: a meta-analysis. JAMA. and inducers of human cytochrome P450 3A4. Curr
2010;304(16):1821–30. Drug Metab. 2008;9(4):310–22.
221. Pan Y, Chen W, Xu Y, Yi X, Han Y, Yang Q, et al. 233. Werk AN, Cascorbi I. Functional gene variants of
Genetic polymorphisms and clopidogrel efficacy for CYP3A4. Clin Pharmacol Ther. 2014;96(3):340–8.
acute ischemic stroke or transient ischemic attack: 234. Kerr BM, Thummel KE, Wurden CJ, Klein SM,
a systematic review and meta-analysis. Circulation. Kroetz DL, Gonzalez FJ, et al. Human liver car-
2017;135(1):21–33. bamazepine metabolism. Role of CYP3A4 and
13 Therapeutic Drug Monitoring and Toxicology: Relevance of Measuring Metabolites 229

CYP2C8 in 10,11-epoxide formation. Biochem diphosphate-glucuronosyltransferase 2B7 promoter


Pharmacol. 1994;47(11):1969–79. with significant impact on promoter activity. Clin
235. Ganesapandian M, Ramasamy K, Adithan S, Pharmacol Ther. 2004;75(3):223–33.
Narayan SK. Influence of cytochrome P450 3A5 246. Ehmer U, Vogel A, Schütte JK, Krone B, Manns MP,
(CYP3A5) genetic polymorphism on dose-adjusted Strassburg CP. Variation of hepatic glucuronida-
plasma levels of carbamazepine in epileptic patients tion: novel functional polymorphisms of the UDP-­
in South Indian population. Indian J Pharmacol glucuronosyltransferase UGT1A4. Hepatology
[Internet]. 2019 [cited 2022 Mar 13];51(6):384–8. Baltim MD. 2004;39(4):970–7.
Available from: https://www.ncbi.nlm.nih.gov/pmc/ 247. Iwai M, Maruo Y, Ito M, Yamamoto K, Sato H,
articles/PMC6984020/. Takeuchi Y. Six novel UDP-glucuronosyltransferase
236. Guillemette C, Lévesque É, Rouleau (UGT1A3) polymorphisms with varying activity. J
M. Pharmacogenomics of human uridine diphospho-­ Hum Genet. 2004;49(3):123–8.
glucuronosyltransferases and clinical implications. 248. Jinno H, Saeki M, Saito Y, Tanaka-Kagawa T,
Clin Pharmacol Ther. 2014;96(3):324–39. Hanioka N, Sai K, et al. Functional characterization
237. Kondo T, Ikenaka Y, Nakayama SMM, Kawai YK, of human UDP-glucuronosyltransferase 1A9 vari-
Mizukawa H, Mitani Y, et al. Uridine diphosphate-­ ant, D256N, found in Japanese cancer patients. J
glucuronosyltransferase (UGT) 2B subfamily Pharmacol Exp Ther. 2003;306(2):688–93.
interspecies differences in carnivores. Toxicol Sci 249. Korprasertthaworn P, Rowland A, Lewis BC,
[Internet]. 2017 [cited 2022 Mar 13];158(1):90–100. Mackenzie PI, Yoovathaworn K, Miners JO. Effects
Available from: https://doi.org/10.1093/toxsci/ of amino acid substitutions at positions 33 and 37 on
kfx072. UDP-glucuronosyltransferase 1A9 (UGT1A9) activ-
238. Court MH, Zhang X, Ding X, Yee KK, Hesse LM, ity and substrate selectivity. Biochem Pharmacol.
Finel M. Quantitative distribution of mRNAs encod- 2012;84(11):1511–21.
ing the 19 human UDP-glucuronosyltransferase 250. Krishnaswamy S, Hao Q, Al-Rohaimi A, Hesse
enzymes in 26 adult and 3 fetal tissues. Xenobiotica LM, von Moltke LL, Greenblatt DJ, et al. UDP
Fate Foreign Compd Biol Syst. 2012;42(3):266–77. glucuronosyltransferase (UGT) 1A6 pharmacoge-
239. Izukawa T, Nakajima M, Fujiwara R, Yamanaka H, netics: II. Functional impact of the three most com-
Fukami T, Takamiya M, et al. Quantitative analy- mon nonsynonymous UGT1A6 polymorphisms
sis of UDP-glucuronosyltransferase (UGT) 1A and (S7A, T181A, and R184S). J Pharmacol Exp Ther.
UGT2B expression levels in human livers. Drug 2005;313(3):1340–6.
Metab Dispos Biol Fate Chem. 2009;37(8):1759–68. 251. Mackenzie PI, Miners JO, McKinnon
240. Ohno S, Nakajin S. Determination of mRNA expres- RA. Polymorphisms in UDP glucuronosyltransfer-
sion of human UDP-glucuronosyltransferases and ase genes: functional consequences and clinical rel-
application for localization in various human tis- evance. Clin Chem Lab Med. 2000;38(9):889–92.
sues by real-time reverse transcriptase-polymerase 252. Miners JO, McKinnon RA, Mackenzie PI. Genetic
chain reaction. Drug Metab Dispos Biol Fate Chem. polymorphisms of UDP-glucuronosyltransferases
2009;37(1):32–40. and their functional significance. Toxicology.
241. Gaganis P, Miners JO, Brennan JS, Thomas A, 2002;181–182:453–6.
Knights KM. Human renal cortical and medullary 253. Villeneuve L, Girard H, Fortier L-C, Gagné J-F,
UDP-glucuronosyltransferases (UGTs): immu- Guillemette C. Novel functional polymorphisms
nohistochemical localization of UGT2B7 and in the UGT1A7 and UGT1A9 glucuronidating
UGT1A enzymes and kinetic characterization of enzymes in Caucasian and African-American sub-
S-naproxen glucuronidation. J Pharmacol Exp Ther. jects and their impact on the metabolism of 7-ethyl-­
2007;323(2):422–30. 10-hydroxycamptothecin and flavopiridol anticancer
242. Tukey RH, Strassburg CP. Human UDP-­ drugs. J Pharmacol Exp Ther. 2003;307(1):117–28.
glucuronosyltransferases: metabolism, expres- 254. Wilson W, Pardo-Manuel de Villena F, Lyn-Cook
sion, and disease. Annu Rev Pharmacol Toxicol. BD, Chatterjee PK, Bell TA, Detwiler DA, et al.
2000;40:581–616. Characterization of a common deletion polymor-
243. Bernard O, Guillemette C. The main role of phism of the UGT2B17 gene linked to UGT2B15.
UGT1A9 in the hepatic metabolism of mycophe- Genomics. 2004;84(4):707–14.
nolic acid and the effects of naturally occurring 255. Shah SAV, Paradkar M, Desai D, Ashavaid
variants. Drug Metab Dispos Biol Fate Chem. TF. Nucleoside diphosphate-linked moiety X-type
2004;32(8):775–8. motif 15 C415T variant as a predictor for thiopurine-­
244. Bosma PJ, Chowdhury JR, Bakker C, Gantla S, induced toxicity in Indian patients. J Gastroenterol
de Boer A, Oostra BA, et al. The genetic basis Hepatol. 2017;32(3):620–4.
of the reduced expression of bilirubin UDP-­ 256. Nerenz RD. Chapter 32 – Pharmacogenomics and
glucuronosyltransferase 1 in Gilbert’s syndrome. N personalized medicine in the treatment of human
Engl J Med. 1995;333(18):1171–5. diseases. In: Coleman WB, Tsongalis GJ, editors.
245. Duguay Y, Báár C, Skorpen F, Guillemette C. A Molecular pathology. 2nd ed [Internet]. Academic
novel functional polymorphism in the uridine Press; 2018 [cited 2022 Mar 13]. p. 731–43.
230 J. Akingbasote et al.

Available from: https://www.sciencedirect.com/ 266. Lennard L, Singleton HJ. High-performance liquid


science/article/pii/B9780128027615000328. chromatographic assay of the methyl and nucleotide
257. Relling MV, Schwab M, Whirl-Carrillo M, Suarez- metabolites of 6-mercaptopurine: quantitation of red
Kurtz G, Pui C-H, Stein CM, et al. Clinical blood cell 6-thioguanine nucleotide, 6-thioinosinic
Pharmacogenetics Implementation Consortium acid and 6-methylmercaptopurine metabolites in a
guideline for thiopurine dosing based on TPMT and single sample. J Chromatogr. 1992;583(1):83–90.
NUDT15 genotypes: 2018 update. Clin Pharmacol 267. Gilissen LPL, Derijks LJJ, Bos LP, Bus PJ,
Ther. 2019;105(5):1095–105. Hooymans PM, Engels LGJB. Therapeutic drug
258. Devasia AJ, Illangeswaran RSS, Raj IX, George monitoring in patients with inflammatory bowel dis-
B, Balasubramanian P. NUDT15 polymorphism ease and established azathioprine therapy. Clin Drug
explains serious toxicity to azathioprine in Indian Investig. 2004;24(8):479–86.
patients with chronic immune thrombocytopenia and 268. Yarur AJ, Abreu MT, Deshpande AR, Kerman
autoimmune hemolytic anemia: a case series. Drug DH, Sussman DA. Therapeutic drug monitoring in
Metab Pers Ther [Internet]. 2020 [cited 2022 Mar patients with inflammatory bowel disease. World J
13];35(4). Available from: https://www.degruyter. Gastroenterol WJG [Internet]. 2014 [cited 2022 Mar
com/document/doi/10.1515/dmpt-­2 020-­0 128/ 14];20(13):3475–84. Available from: https://www.
html?lang=en. ncbi.nlm.nih.gov/pmc/articles/PMC3974514/.
259. Weitzel KW, Smith DM, Elsey AR, Duong BQ, 269. Cuffari C. A physician’s guide to azathioprine
Burkley B, Clare-Salzler M, et al. Implementation of metabolite testing. Gastroenterol Hepatol [Internet].
standardized clinical processes for TPMT testing in a 2006 [cited 2022 Mar 14];2(1):58–63. Available
diverse multidisciplinary population: challenges and from: https://www.ncbi.nlm.nih.gov/pmc/articles/
lessons learned. Clin Transl Sci. 2018;11(2):175–81. PMC5307263/.
260. Dubinsky MC, Yang H, Hassard PV, Seidman EG, 270. Rost S, Fregin A, Ivaskevicius V, Conzelmann
Kam LY, Abreu MT, et al. 6-MP metabolite profiles E, Hörtnagel K, Pelz H-J, et al. Mutations in
provide a biochemical explanation for 6-MP resis- VKORC1 cause warfarin resistance and mul-
tance in patients with inflammatory bowel disease. tiple coagulation factor deficiency type 2. Nature.
Gastroenterology. 2002;122(4):904–15. 2004;427(6974):537–41.
261. Tanaka Y, Saito Y. Importance of NUDT15 poly- 271. Limdi NA, Wadelius M, Cavallari L, Eriksson N,
morphisms in thiopurine treatments. J Pers Med Crawford DC, Lee M-TM, et al. Warfarin phar-
[Internet]. 2021 [cited 2022 Mar 14];11(8):778. macogenetics: a single VKORC1 polymorphism
Available from: https://www.ncbi.nlm.nih.gov/pmc/ is predictive of dose across 3 racial groups. Blood.
articles/PMC8399029/. 2010;115(18):3827–34.
262. Yu C-H, Chang Y-H, Wang D-S, Jou S-T, Lin C-Y, 272. Johnson JA, Caudle KE, Gong L, Whirl-Carrillo M,
Lin K-H, et al. Determination of NUDT15 variants Stein CM, Scott SA, et al. Clinical Pharmacogenetics
by targeted sequencing can identify compound het- Implementation Consortium (CPIC) guideline for
erozygosity in pediatric acute lymphoblastic leuke- pharmacogenetics-guided warfarin dosing: 2017
mia patients. Sci Rep [Internet]. 2020 [cited 2022 update. Clin Pharmacol Ther. 2017;102(3):397–404.
Mar 14];10(1):14400. Available from: https://www. 273. Rieder MJ, Reiner AP, Gage BF, Nickerson DA, Eby
nature.com/articles/s41598-­020-­71468-­y. CS, McLeod HL, et al. Effect of VKORC1 haplo-
263. Evans WE, Hon YY, Bomgaars L, Coutre S, types on transcriptional regulation and warfarin
Holdsworth M, Janco R, et al. Preponderance of dose. N Engl J Med. 2005;352(22):2285–93.
thiopurine S-methyltransferase deficiency and het- 274. Yan X, Yang F, Zhou H, Zhang H, Liu J, Ma K, et al.
erozygosity among patients intolerant to mercap- Effects of VKORC1 genetic polymorphisms on war-
topurine or azathioprine. J Clin Oncol [Internet]. farin maintenance dose requirement in a Chinese
2016 [cited 2022 Mar 14]; Available from: https:// Han population. Med Sci Monit Int Med J Exp Clin
ascopubs.org/doi/pdf/10.1200/JCO.2001.19.8.2293. Res [Internet]. 2015 [cited 2022 Mar 14];21:3577–
264. Fan X, Yin D, Men R, Xu H, Yang L. NUDT15 84. Available from: https://www.ncbi.nlm.nih.gov/
polymorphism confer increased susceptibil- pmc/articles/PMC4657763/.
ity to thiopurine-induced leukopenia in patients 275. Cho S, Yoon Y-R. Understanding the pharmacokinet-
with autoimmune hepatitis and related cirrhosis. ics of prodrug and metabolite. Transl Clin Pharmacol
Front Pharmacol [Internet]. 2019 [cited 2022 Mar [Internet]. 2018 [cited 2022 Jan 23];26(1):1–5.
14];10. Available from: https://www.frontiersin.org/ Available from: https://www.ncbi.nlm.nih.gov/pmc/
article/10.3389/fphar.2019.00346. articles/PMC6989223/.
265. Wusk B, Kullak-Ublick GA, Rammert C, von 276. Zhang Z, Tang W. Drug metabolism in drug discov-
Eckardstein A, Fried M, Rentsch KM. Therapeutic ery and development. Acta Pharm Sin B [Internet].
drug monitoring of thiopurine drugs in patients 2018 [cited 2022 Jan 23];8(5):721–32. Available
with inflammatory bowel disease or autoim- from: https://www.ncbi.nlm.nih.gov/pmc/articles/
mune hepatitis. Eur J Gastroenterol Hepatol. PMC6146880/.
2004;16(12):1407–13. 277. Rang H, Dale M, Ritter J, Flower R. Absorption and
distribution of drugs. 2007.
13 Therapeutic Drug Monitoring and Toxicology: Relevance of Measuring Metabolites 231

278. Oellerich M, Armstrong VW. Prodrug metabo- apy and CYP2D6 genotype. In: Medical genetics
lites: implications for therapeutic drug monitor- summaries [Internet]. Bethesda: National Center for
ing. Clin Chem [Internet]. 2001 [cited 2022 Jan Biotechnology Information (US); 2012 [cited 2022
27];47(5):805–6. Available from: https://doi. Mar 5]. Available from: http://www.ncbi.nlm.nih.
org/10.1093/clinchem/47.5.805. gov/books/NBK100662/.
279. Huttunen KM, Raunio H, Rautio J. Prodrugs--from 292. Kirchheiner J, Schmidt H, Tzvetkov M, Keulen
serendipity to rational design. Pharmacol Rev. J-T, Lötsch J, Roots I, et al. Pharmacokinetics
2011;63(3):750–71. of codeine and its metabolite morphine in ultra-­
280. Bullingham RE, Nicholls AJ, Kamm BR. Clinical rapid metabolizers due to CYP2D6 duplication.
pharmacokinetics of mycophenolate mofetil. Clin Pharmacogenomics J [Internet]. 2007 [cited 2022
Pharmacokinet. 1998;34(6):429–55. Mar 5];7(4):257–65. Available from: https://www.
281. Christie JD, Edwards LB, Aurora P, Dobbels F, Kirk nature.com/articles/6500406.
R, Rahmel AO, et al. The registry of the International 293. FDA. FDA Drug Safety Podcast: FDA restricts use of
Society for Heart and Lung Transplantation: twenty-­ prescription codeine pain and cough medicines and
sixth official adult lung and heart-lung trans- tramadol pain medicines in children; recommends
plantation report-2009. J Heart Lung Transplant. against use in breastfeeding women. FDA [Internet].
2009;28(10):1031–49. 2022 [cited 2022 Mar 6]; Available from: https://
282. Baraldo M, Sponga S, Livi U. Therapeutic drug www.fda.gov/drugs/fda-­drug-­safety-­podcasts/fda-­­
monitoring of micophenolate mofetil in cardiac drug-­safety-­podcast-­fda-­restricts-­use-­prescription-­
transplant patients by limited sampling strategy: an codeine-­pain-­and-­cough-­medicines-­and-­tramadol.
update. 2018. 294. Grond S, Sablotzki A. Clinical pharmacology of tra-
283. DeNofrio D, Loh E, Kao A, Korecka M, Pickering madol. Clin Pharmacokinet. 2004;43(13):879–923.
FW, Craig KA, et al. Mycophenolic acid concentra- 295. International Conference on Harmonisation Expert
tions are associated with cardiac allograft rejection. J Working Group. International conference on har-
Heart Lung Transplant. 2000;19(11):1071–6. monisation of technical requirements for registration
284. Dubrey SW, Holt DW, Banner N. Measurement of of pharmaceuticals for human use. ICH harmonised
mycophenolate mofetil plasma levels after heart tripartite guideline. Guideline for good clinical prac-
transplantation and a potential side effect of high tice. 1997 CFR ICH Guidel [Internet]. 1997 [cited
levels. Ther Drug Monit. 1999;21(3):325–6. 2022 Mar 30]; Available from: https://ci.nii.ac.jp/
285. Eisen H, Ross H. Optimizing the immunosuppres- naid/10020348247/.
sive regimen in heart transplantation. J Heart Lung 296. Winter ME. Basic clinical pharmacokinetics.
Transplant. 2004;23(5 Suppl):S207–13. Lippincott Williams & Wilkins; 2004.
286. Shaw LM, Korecka M, Venkataramanan R, 297. Steijns LSW, Bouw J, van der Weide J. Evaluation of
Goldberg L, Bloom R, Brayman KL. Mycophenolic fluorescence polarization assays for measuring val-
acid pharmacodynamics and pharmacokinetics pro- proic acid, phenytoin, carbamazepine and phenobar-
vide a basis for rational monitoring strategies. Am J bital in serum. Ther Drug Monit. 2002;24(3):432–5.
Transplant. 2003;3(5):534–42. 298. Roy SMN, Yetal SM, Vaidya VV, Joshi
287. Filler G. Value of therapeutic drug monitoring of SS. Determination and quantification of phenytoin
MMF therapy in pediatric transplantation. Pediatr in human plasma by liquid chromatography with
Transplant. 2006;10(6):707–11. electrospray ionization tandem mass spectrom-
288. Godron-Dubrasquet A, Woillard J-B, Decramer S, etry. E-J Chem [Internet]. 2008 [cited 2022 Mar
Fila M, Guigonis V, Tellier S, et al. Mycophenolic 30];5(1):169–76. Available from: https://www.
acid area under the concentration-time curve is hindawi.com/journals/jchem/2008/362512/.
associated with therapeutic response in childhood-­ 299. Patel JA, Clayton LT, LeBel CP, McClatchey
onset lupus nephritis. Pediatr Nephrol Berl Ger. KD. Abnormal theophylline levels in plasma by flu-
2021;36(2):341–7. orescence polarization immunoassay in patients with
289. Gammal RS, Crews KR, Haidar CE, Hoffman JM, renal disease. Ther Drug Monit. 1984;6(4):458–60.
Baker DK, Barker PJ, et al. Pharmacogenetics 300. Frank EL, Schwarz EL, Juenke J, Annesley TM,
for safe codeine use in sickle cell disease. Roberts WL. Performance characteristics of four
Pediatrics [Internet]. 2016 [cited 2022 Mar immunoassays for antiepileptic drugs on the
5];138(1):e20153479. Available from: https://www. IMMULITE 2000 automated analyzer. Am J Clin
ncbi.nlm.nih.gov/pmc/articles/PMC4925073/. Pathol. 2002;118(1):124–31.
290. Rodieux F, Vutskits L, Posfay-Barbe KM, Habre W, 301. Rane CT, Dalvi SS, Gogtay NJ, Shah PU, Kshirsagar
Piguet V, Desmeules JA, et al. When the safe alterna- NA. A pharmacoeconomic analysis of the impact
tive is not that safe: tramadol prescribing in children. of therapeutic drug monitoring in adult patients
Front Pharmacol [Internet]. 2018 [cited 2022 Mar with generalized tonic-clonic epilepsy. Br J Clin
5];9. Available from: https://www.frontiersin.org/ Pharmacol. 2001;52(2):193–5.
article/10.3389/fphar.2018.00148. 302. Bootman JL, Wertheimer AI, Zaske D, Rowland
291. Pratt VM, Scott SA, Pirmohamed M, Esquivel B, C. Individualizing gentamicin dosage regimens in
Kane MS, Kattman BL, et al., editors. Codeine ther-
232 J. Akingbasote et al.

burn patients with gram-negative septicemia: a cost- Clin Hypertens [Internet]. 2015 [cited 2022 Mar
-benefit analysis. J Pharm Sci. 1979;68(3):267–72. 31];17(9):713–9. Available from: https://www.ncbi.
303. Streetman DS, Nafziger AN, Destache CJ, Bertino nlm.nih.gov/pmc/articles/PMC4562815/.
AS. Individualized pharmacokinetic monitoring 306. McNeill RP, Barclay ML. Cost-effectiveness of
results in less aminoglycoside-associated nephrotox- therapeutic drug monitoring in inflammatory bowel
icity and fewer associated costs. Pharmacotherapy. disease. Curr Opin Pharmacol [Internet]. 2020
2001;21(4):443–51. [cited 2022 Mar 31];55:41–6. Available from:
304. Shaw LM, Kaplan B, Brayman KL. Prospective https://www.sciencedirect.com/science/article/pii/
investigations of concentration-clinical response S1471489220300874.
for immunosuppressive drugs provide the scientific 307. Dubinsky MC, Reyes E, Ofman J, Chiou C-F,
basis for therapeutic drug monitoring. Clin Chem. Wade S, Sandborn WJ. A cost-effectiveness analy-
1998;44(2):381–7. sis of alternative disease management strategies
305. Velasco A, Chung O, Raza F, Pandey A, Brinker S, in patients with Crohn’s disease treated with aza-
Arbique D, et al. Cost-effectiveness of therapeutic thioprine or 6-mercaptopurine. Am J Gastroenterol.
drug monitoring in diagnosing primary aldoste- 2005;100(10):2239–47.
ronism in patients with resistant hypertension. J
Recent Advances in Nanosensors
for Therapeutic Drug Monitoring
14
(TDM)

Percy Selasi Agogo-Mawuli


and David P. Siderovski

Abstract
14.1 Traditional TDM
Nanomaterials have at least one dimension and the Need
that is less than 100 nm in size. This chapter for Nanosensors
focuses on the recent development seen in the
use of nanomaterials and nanotechnology for One factor that significantly impacts the rational
therapeutic drug monitoring (TDM) in order prescribing process is therapeutic drug monitor-
to meet the urgent need of measuring drug ing. Despite the fact that, in general, one can eas-
concentrations in biological matrices like ily observe and measure the effects of many
plasma, serum, saliva, or urine, at a lower cost drugs that are administered for treatment pur-
and within a shorter time period than tradi- poses, the situation is different (and potentially
tional immunoassays or chromatographic life-threatening) for other specific drugs. In the
methods. Developments in the detection of case of the former group of drugs, their doses can
different types of medications (e.g., antibiot- be easily modified, or treatment can be halted in
ics, anticonvulsants, anticoagulants) are case of observed (and reversible) adverse events.
described, using both optical and electro- However, the latter group of drugs requires thera-
chemical approaches and including competing peutic drug monitoring to attain the desired ben-
nanosensor technologies for the same drug efits of the drugs with reduced risk. Some of
analyte. these medicines exhibit characteristics such as
large inter-individual variation between the drug
Keywords dosage and its benefits, which makes therapeutic
drug monitoring useful in patients taking such
Carbon nanotubes · Electrochemical determi- medications. It is generally not advisable nor
nation · Fluorescence · Nanoparticles · possible to take routine measurements of drug
Quantum dots concentration at the site of action; however, con-
sidering the fact that plasma concentration of a
drug can better predict the therapeutic outcome
P. S. Agogo-Mawuli · D. P. Siderovski (*) or adverse effects of the drug than the dosage, it
Department of Pharmacology & Neuroscience, has become necessary to monitor such drugs to
University of North Texas Health Science Center at improve outcomes [1]. The World Health
Fort Worth, Fort Worth, TX, USA Organization (WHO) has therefore proposed a
e-mail: david.siderovski@unthsc.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 233
S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_14
234 P. S. Agogo-Mawuli and D. P. Siderovski

list of medications that warrant therapeutic drug affinity, requirement of low sample volume, and
monitoring, as follows: phenytoin, amikacin, ease to conduct. The demerit in using immunoas-
cyclosporine, lithium, valproic acid, methotrex- says lies in the contribution of cross-reactants or
ate, digoxin, phenobarbital, gentamicin, carbam- other nonspecific constituents in the sample to
azepine, and vancomycin [2]. Therapeutic drug the observed analyte concentration that makes
monitoring involves multiple sequential stages this technique less specific [4]. A more recent
such as accurate determination of plasma con- development in TDM is the introduction of nano-
centration, interpretation of the obtained results, technology for drug monitoring, at a lower cost
and, finally, analysis of how much dose is and within a shorter time period than immunoas-
required for the patient to obtain benefits from says or chromatographic methods. In addition,
the medication without any adverse effects. This the use of nanosensors does not require highly
chapter focuses mainly on the stage of precisely trained personnel.
measuring the plasma concentration of the drugs
proposed by WHO, or measuring their concentra-
tion in other biological matrices like saliva and 14.3 Definition of Nanomaterials
urine, with the use of nanosensors (Table 14.1).
Nanomaterials (e.g., carbon nanotubes; Fig. 14.1)
refer to materials that have at least one dimension
14.2 Traditional TDM Practice of single or multiple phase that is less than
Versus Recent Advances 100 nm in size [5].

Therapeutic drug monitoring has evolved over


the years, beginning with the use of chromato- 14.4 Definition of Nanosensors
graphic methods. Gas chromatography and high-­
performance liquid chromatography (HPLC) Nanosensors are sensors that are made up of
have been widely applied to determine the con- nanomaterials or nanoparticles and engineered to
centration of drugs in body fluids, especially possess nanoscale dimensions in order to increase
plasma. Liquid chromatography with tandem their surface area for detection of a specific mol-
mass spectrometry (LC-MS/MS) methods were ecule, an environmental circumstance, or a bio-
later developed that exhibited high specificity in logical component [6]. These sensors thus exhibit
detecting the analyte. However, challenges improved selectivity and sensitivity compared to
remained, such as lower throughput and inability their macroscale analogs. Nanosensors are made
to get accurate results due to the interference of up of (1) a recognition element that is specific for
the matrix and co-eluting compounds during a particular analyte and helps to quantify them
mass spectrometry, among others [3]. HPLC and (2) a signal transducer that produces a quan-
techniques were then combined with simple tifiable signal based on the interaction between
detectors (diode-array detection, flame ionization the analyte and the recognition element [6].
detection, or ultraviolet detection) that helped These signals are further amplified and processed
relieve some burden off equipment cost, opera- to generate data that corresponds to the analyte
tion time, and the need for expertise. The diffi- concentration.
culty in resolving different analytes with similar
retention times was further worsened by unknown
constituents present in the biological matrices 14.5 Current Nanosensor
under study [4]. Immunoassays were later devel- Techniques for TDM
oped and have been widely employed in clinical
laboratories for therapeutic drug monitoring. Nanosensors are categorized into different types
This technique offers many advantages such as based on their signal transduction methods or the
affordability, high-throughput adaptability, high biological recognition element used (Table 14.1).
14 Recent Advances in Nanosensors for Therapeutic Drug Monitoring (TDM) 235

Table 14.1 TDM nanosensor design details discussed in this chapter


Biological
Detection matrix
Drug Nanosensor design Linear range reported limit reported investigated Reference
Gemcitabine Optical: silver-associated 0.003 μM–0.1 μM 0.002 μM Plasma and [20]
carbon quantum dots urine
(Ag-CQD)
Flucytosine Raman spectroscopy: 10 μg/mL–150 μg/mL 12.1 μg/mL Undiluted [21]
nitrocellulose/inkjet-­ serum and
fabricated paper sensor whole
blood
Linezolid Electrochemical: cobalt 1.0 × 10−10 M–1.0 × 10−4 M 1.7 × 10−11 M Plasma [22]
nanoparticle-incorporated
2-hydroxypropyl-β-­
cyclodextrin (HP-βCD)
Amikacin Optical: surface plasmon 0.33 ng/mL–5.77 ng/mL 0.13 ng/mL Serum [23]
resonance (SPR)
immunoassay
Gentamicin Optical: epicatechin coated 0 μM–100 μM 1.28 μM Serum and [24]
silver nanoparticles plasma
(Ag-NP)
Digoxin Electrochemical: silver 1 pM–0.1 μM 0.3 pM Plasma [27]
nanoparticle-decorated
graphene oxide composite
(AgNP-GO)
Digoxin Optical: aqueous gold N/A Colorimetric? Serum [28]
nanoparticles (AuNP) 571 pM
Fluorescence?
392 pM
Warfarin Electrochemical: magnetic 0.5–1000 μM 0.21 μM Urine and [31]
iron oxide nanoparticle-­ serum
modified carbon paste
Warfarin Electrochemical: cadmium 0.05–80 μM 8.5 nM Serum, [32]
sulfide (CdS) quantum dots breast milk,
on carboxylated multiwalled and urine
carbon nanotubes
(MWCNT)
Warfarin Electrochemical: manganese 0.10–447 μM 0.08 μM Serum and [35]
ferrite nanoparticles and a urine
MWCNT paste matrix
Warfarin Electrochemical: 0.031–0.616 ng/mL 0.024 ng/mL Serum [37]
molecularly imprinted
polymer (MIP) nanosensor
with MWCNT and gold
nanoparticles
Warfarin Electrochemical: MWCNT 0.008–800 μM 0.0033 μM Serum and [39]
with cobalt oxide urine
nanoparticles
Heparin Optical: rhodamine 0.01– 0.1 nM and 7.5 pM Serum [45]
B-labeled peptides 1.0–70.0 nM
Aspirin Electrochemical: carbon 1 pg/mL–1 μg/mL 0.03 pg/mL Urine and [51]
electrode with chitosan saliva
capped with gold
nanoparticles
(continued)
236 P. S. Agogo-Mawuli and D. P. Siderovski

Table 14.1 (continued)


Biological
Detection matrix
Drug Nanosensor design Linear range reported limit reported investigated Reference
Tramadol Electrochemical: MIPs on 3.5 × 10−9–1.0 × 10−2 M 2.04 × 10−9 M Urine [61]
decorated graphene
nanosheets with Ag-NPs
Sulfasalazine Electrochemical: nickel 5.0 × 10−7–8.0 × 10−4 M 9.0 × 10−8 M Urine [64]
oxide + carbon nanotube
(NiO/CNT) composite
Theophylline Electrochemical: screen-­ 1.0–700.0 μM 0.2 μM Urine [69]
printed carbon electrodes +
graphene quantum dots
Theophylline Optical: RNA aptamer + 50 nM–5 μM 47 nM Serum [72]
graphene oxide/cryonase-­
mediated fluorescence
amplification
Levothyroxine Electrochemical: melamine 10–120 nM 2.84 nM Serum [74]
Au-NPs + mercaptoethanol-­
modified MWCNT
Lithium Optical: ruthenium-based N/A N/A Serum [77]
metallacrown complexes
Carbamazepine Electrochemical: cerium-­ 0.05–100 μM 1.2 nM Urine [80]
doped zinc oxide (ZnO) and
reduced graphene oxide
Phenobarbital Optical: MIPs on green 0.4–34.5 mM 0.1 mM Plasma [88]
source carbon dots (GSCDs)
Phenobarbital Electrochemical: platinum 0.4–60 μM 0.1 μM Urine [93]
NPs + MWCNT
Valproic acid Optical: thioglycolic acid 0.3–7.5 mg/L 0.24 μg/mL Serum and [95]
(TGA)-capped CdTe urine
quantum dots
Phenytoin Optical: branched gold 67–670 ng/mL 21 ng/mL Plasma [96]
nanoparticles (B-AuNPs)

Fig. 14.1 Relative sizes of various objects to highlight describe materials like carbon nanotubes. The diameter of
the extreme scale of nanomaterials. “Nano-” is defined as a typical carbon nanotube is 109 shorter than a typical
one billionth (10−9) part of an arbitrary unit of measure- 4 year-old child
ment, hence the use of the term “nanomaterials” to
14 Recent Advances in Nanosensors for Therapeutic Drug Monitoring (TDM) 237

According to their signal transduction methods, ducting or metallic or carbonic polymer nanoma-
nanosensors are classified into optical, electro- terials, and/or quantum dots [13]. Various classes
chemical, nanomechanical, or piezoelectric sen- of biorecognition elements that can be incorpo-
sors [7]. Affinity and catalytic biosensors are rated onto an electrode (depending on the type of
differentiated based on the kind of biological rec- matrix and analyte) include redox reaction mole-
ognition element they possess: whereas catalytic cules, binding-only molecules, and conformation
biosensors depend on chemical reactions cata- changing molecules [12]. Examples of redox
lyzed by enzymes, affinity biosensors depend on reaction molecules are enzymes and redox-­
complex formation between the biorecognition reactive species. Some binding-only molecules
element and the analyte [8]. Detection of the that are used include molecularly imprinted poly-
analyte-­ receptor complex can be achieved via mers (MIP) and antibodies. Aptamers are confor-
labeling (e.g., fluorescent or enzymatic), or a mation changing molecules, often based on
change in the physical properties of the signal nucleic acids, that can also be used to modify an
transducer [9]. electrode to enhance its performance [12].
Electrochemical sensors are nanosensors that Optical sensors are sensor devices that deter-
determine the concentration of an analyte in a mine the concentration of an analyte based on the
medium or matrix based on the degree of change degree of change in properties of light such as
in current or voltage after an interaction between fluorescence, absorption, light scattering, or
a bioreceptor and the analyte [10]. Therefore, refractive index after a molecular recognition
nanosensors which produce signals that are type reaction [14]. Two categories of optical sen-
potentiometric, amperometric, or impedimetric sors have been described: (1) evanescent field-­
(which includes conductimetric) are electro- based sensors and (2) bio-optrodes. Evanescent
chemical in nature [11]. This kind of sensor has field-based sensors are characterized by produc-
been widely used in monitoring several drugs in tion of an evanescent wave due to transmission of
which some examples are listed below. For a suc- light by total internal reflection in an exposed
cessful electrochemical measurement, the setup fiber core. This generated evanescent wave can
requires electrodes, a potentiostat/galvanostat, be absorbed in the presence of an absorbing mol-
and a solution [12]. The three kinds of electrodes ecule in the field, resulting in diminished ampli-
used in the setup are pretreated and polished to tude of the wave being propagated in the fiber
enhance reproducibility and eliminate contami- core [15]. Examples of such sensors include total
nants. They include the reference electrode internal reflection fluorescence (TIRF) sensors,
(which creates an avenue for measuring the reflectometric interference spectroscopy (RIfS)
potential of other electrodes based on the stable sensors, surface-enhanced Raman scattering
potential it maintains), working electrode (where (SERS) sensors, and surface plasmon resonance
the reaction with analyte and electrochemical (SPR)-based sensors [8]. Bio-optrodes are built
measurement occurs), and counter/auxiliary elec- from an optical fiber with a sensing layer at the
trode (which completes the electrical circuit) end point of the fiber containing biorecognition
[12]. The analyte is present in the solution molecules and dyes. A reaction between the ana-
together with some ions that facilitate the flow of lyte and the constituents of the sensing layer
charges between the counter electrode and the causes a measurable change in the optical proper-
working electrode. The potentiostat connects to ties of the transducer. A fiber optic device is an
all the electrodes and measures the given response example of a bio-optrode [16].
[12]. Modification of the electrodes with nano- Piezoelectric sensors are sensors that possess
structures as well as biorecognition elements an oscillating piezoelectric crystal capable of
improves the overall performance of the sensor. vibrating at its natural frequency to produce reso-
The surface of the electrode can be modified with nance. A signal produced due to an interaction
nanostructures in many ways such as addition of between an analyte and a recognition element
graphene/graphene oxide carbon nanotubes, con- causes a change in frequency which in turn
238 P. S. Agogo-Mawuli and D. P. Siderovski

results in a change in electric current being mea- liver enzyme elevations, proteinuria, hematuria,
sured. This change in electric current determines severe hepatotoxicity like liver failure, and death.
the concentration of analyte detected [17]. Initial signs of microangiopathic hemolytic ane-
Nanomechanical sensors measure the concen- mia warrant the stoppage of gemcitabine treat-
tration of the target analyte, via either an optical ment [19].
or piezoelectric system, when their detection For the reasons outlined above, there is the
through molecular interaction on the surface of need to develop a suitable monitoring tool for
the sensor causes a cantilever flection [18]. Both detection of gemcitabine in body fluids for opti-
the piezoelectric and nanomechanical types of mum therapeutic outcome. Methods of detection
sensors are seldom used in therapeutic drug mon- that have previously been identified have posed
itoring, but they are popular in performing qual- several challenges due to how expensive, less sen-
ity control procedures in industry and protein sitive, and time-consuming they are. Therapeutic
evaluations, respectively. monitoring of gemcitabine using nanosensing
technology has of late shown a promising solution
to these aforementioned problems. Carbon quan-
14.6 Nanosensors for Clinical Use tum dots (CQDs) are fluorescent nanomaterials
that have favorable optical and electrical proper-
14.6.1 Nanosensor for Gemcitabine ties for the detection of gemcitabine [20]. The
ease of modification of this nanomaterial through
The anticancer chemotherapeutic agent gem- simple deposition or coating with metal particles
citabine (an analog of cytarabine) is primarily or organic molecules, as well as their biosafety
used to manage solid tumors occurring in the and simple preparation procedure, offers an
lung, bladder, pancreas, breast, and ovary [19]. advantage for extensive research and develop-
Gemcitabine is given by intravenous administra- ment into TDM nanosensors. Studies have inves-
tion in the hydrochloride form, infused over tigated the potential of CQDs associated to metal
30–60 min, and subsequent doses are adjusted nanoparticles (M-CQDs) to detect gemcitabine in
depending on both the therapeutic and toxico- the biological matrices of human plasma and
logic responses observed. Rapid clearance of urine. Silver-­associated carbon quantum dots
gemcitabine occurs after absorption in the blood (Ag-CQD) were seen to exhibit higher photosta-
stream. Metabolism of this drug occurs in the bility and detectable responses to gemcitabine
blood, kidney, liver, and other tissues by cytidine compared to other metals tested (e.g., Cu, Mn, Ni)
deaminase [19]. Almost the entire dose of gem- [20]. Optimal detection conditions were identified
citabine is eliminated in urine with 1% excreted as a pH of 6 and 7 min of incubation time, with a
in feces. Its half-life varies between 42 and linear range of drug detection from 0.003 μM to
94 min based on age and sex; clearance in men is 0.1 μM of gemcitabine [20]. Hence, Ag-CQD can
25% higher than in women [19]. Bone marrow be used to measure gemcitabine in nanomolar
suppression has been identified as the main concentrations. Subsequent tests performed to
adverse effect that limits the dose of gemcitabine; validate the application of Ag-CQD gemcitabine
however, this effect does not generally require sensor in human plasma and urine revealed no
therapy to be stopped. Interstitial pneumonitis, interference between the matrix and the different
acute respiratory distress syndrome, as well as concentrations of gemcitabine [20]. Therefore,
pulmonary fibrosis can occur with therapy. Such this Ag-CQD nanosensor design has the capabil-
pulmonary toxicity generally requires therapy to ity to meet the need to measure gemcitabine in
be stopped [19]. Caution must also be taken when ­biological specimens during cancer chemother-
using gemcitabine in patients with impaired renal apy treatment.
or hepatic function due to reports of transient
14 Recent Advances in Nanosensors for Therapeutic Drug Monitoring (TDM) 239

14.6.2 Nanosensors for Specific combined with amphotericin B, posing a chal-


Antibiotics lenge to determine the physiological load in real
time.
14.6.2.1 Nanosensor This justification above explains the need for
for the Antifungal real-time therapeutic drug monitoring of flucyto-
Flucytosine sine. Complicated microbiological assays, as
Flucytosine (5-fluorocytosine; 5FC) is an anti- well as methods like HPLC or LC/MS, have
fungal drug indicated for the treatment of sys- posed challenging in measuring flucytosine lev-
temic fungal infections. Cryptococcal meningitis els due to their time-consuming and expensive
and severe systemic candidiasis are commonly natures and their advanced training requirements.
treated with a combination therapy of flucytosine A dual substrate paper/membrane system utiliz-
and fluconazole or amphotericin B. The thera- ing inkjet-fabricated, surface-enhanced Raman
peutic range for flucytosine is narrow: spanning spectroscopy (SERS) nanosensors has been used
from 20 to 80 μg/mL [19]. Rapid and almost for rapid detection of flucytosine in undiluted
complete absorption of flucytosine occurs in the serum and whole blood [21]. Rapid monitoring
gastrointestinal tract, reaching a bioavailability of drugs from such biological matrices as serum
ranging from 78% to 89%. A trough plasma con- and whole blood requires physical separation of
centration of 25–50 μg/mL is targeted when the analyte from the blood components since
adjusting the dose in patients, especially patients these components can obscure signal detection
living with AIDS who are highly susceptible to by non-discriminately coating the surface of the
bone marrow toxicity [19]. Flucytosine can be paper-based SERS sensors. Nitrocellulose mem-
given by oral, intravenous, or topical administra- branes combined with paper SERS sensors in a
tion. Fungi such as Cryptococcus neoformans passive vertical flow scheme help to achieve this
and Candida spp. are known to develop resis- physical separation goal [21].
tance against flucytosine. However, less resis-
tance issues have been associated with application 14.6.2.2 Nanosensor for Linezolid
of the combination of flucytosine and amphoteri- Linezolid is used for the treatment of infections
cin B [19]. The distribution of flucytosine is caused by gram-positive bacteria occurring in the
extensive in the body tissues, with 65–90% of respiratory tract and the skin. It is active against
concentration in the serum present in the cerebro- nosocomial infections caused by multidrug-­
spinal fluid (CSF) [19]. The drug is filtered in the resistant bacteria such as methicillin-resistant
glomerulus, and about 90% is excreted in the Staphylococcus aureus (MRSA) and vancomycin-­
unchanged form; fluorouracil is a product of resistant enterococci [19]. It can be administered
metabolism of a minor amount of flucytosine. via the oral or intravenous route. It is rapidly
Orally administered flucytosine that does not get absorbed into the blood stream after 1–2 h of oral
absorbed in the gastrointestinal tract is elimi- administration. There is extensive distribution of
nated unchanged in the feces. This drug has an linezolid into the muscle, fat, cerebrospinal fluid,
elimination half-life of 2.5–6 h, a time span that lungs, bone, and skin blister fluids. It undergoes
rises with a decline in renal function. Combination extensive metabolism, with only 30% being
therapy with amphotericin B, poor renal func- excreted in urine in the unchanged form.
tion, as well as plasma concentrations exceeding Linezolid has an elimination half-life of 5–7 h
100 μg/mL has been associated with bone mar- [19]. Clearance of the drug is more rapid in chil-
row suppression, notably thrombocytopenia and dren compared to adults. Resistance in staphylo-
leucopenia [19]. Plasma concentrations greater cocci (including methicillin-resistant
than 100 μg/mL also pose a higher risk of hepato- Staphylococcus aureus, S. auricularis, and S.
toxicity and gastrointestinal toxicity [19]. ­epidermidis) and enterococci (Enterococcus fae-
Significant variations in renal clearance have cium and Enterococcus faecalis) has been
been observed among patients, especially when reported in its use [19].
240 P. S. Agogo-Mawuli and D. P. Siderovski

The variable pharmacokinetics of linezolid mL [19]. Amikacin has the tendency to cross the
and the emergence of resistance warrant the placenta but does not readily enter the cerebro-
need for therapeutic monitoring of linezolid. spinal fluid. Its half-life is 2–3 h. Almost the
One study reported utilizing nanoparticle- entire drug dose is filtered into the urine within
enhanced potentiometric ion-selective elec- 24 h [19]. The drug has a narrow therapeutic
trodes (ISE) for the detection of linezolid in range (1–30 μg/mL) and is associated with
human plasma. In addition to its reported advan- adverse effects such as irreversible, cumulative
tages of being sensitive, affordable, portable, ototoxicity and reversible nephrotoxicity [19].
and selective, little or no sample pretreatment Due to toxicities associated with amikacin
was found to be needed. According to the study, use, there is the need for therapeutic drug moni-
four fabricated sensors were used, namely, a toring in order to optimize therapy and improve
cationic exchanger phosphotungstate sensor, a patient outcomes. An SPR-based competitive
2-hydroxypropyl-β-­cyclodextrin (HP-βCD) sen- immunoassay has been developed for therapeutic
sor, a copper nanoparticle (NP)-incorporated drug monitoring of amikacin in serum [23]. This
HP-βCD sensor, and a cobalt NP-incorporated sensing technique employs an indirect competi-
HP-βCD sensor [22]. The cobalt NP-incorporated tive inhibition assay due to the low molecular
HP-βCD sensor was the most sensitive among weight of amikacin (585.6 Da), which can lead to
the four sensors tested. HP-βCD promotes rec- low detectability in the case of a direct assay
ognition of the molecular structure of linezolid given that SPR signal is proportional to the
as well as formation of inclusion complexes. molecular weight of the analyte. This technique
Moreover, incorporation of the metal oxide NP enables the use of a small volume of the patient
(cobalt) improved the sensor’s selectivity and sample, which is subsequently diluted for analy-
sensitivity, given the NP’s characteristics of sis [23]. Moreover, the indirect assay offers more
having rich electronic and chemical properties, advantage for the therapeutic monitoring of ami-
high surface area, thermal stability, and high kacin because the immobilization of antibodies
mechanical strength even though ultra-­to a solid surface in the direct assay poses many
lightweight. Its selectivity was maintained when challenges to accurate measurement of the ana-
used for monitoring linezolid in plasma in the lyte. Indiscriminate (random contact) attachment
presence of co-administered drugs (meropenem of the antibodies to a solid surface could tamper
and theophylline) [22]. with the native structure of the antibodies result-
ing in loss of affinity [23].
14.6.2.3 Nanosensor for Amikacin
Amikacin is a member of the group of antibacte- 14.6.2.4 Nanosensor for Gentamicin
rial agents classified as aminoglycosides. It is Gentamicin is an antibiotic that is used in the
used in the treatment of severe gram-negative treatment of systemic infections caused mainly
infections and is active against gentamicin- and by gram-negative organisms [19]. These diseases
tobramycin-resistant bacteria. Amikacin is com- include brucellosis, cystic fibrosis, pneumonia,
monly seen in neonatal therapy since newborns otitis media, meningitis, endocarditis, and septi-
are highly prone to infections [19]. Enzymes that cemia. Therapeutic drug monitoring of gentami-
are known to cause acquired aminoglycoside cin is paramount to prevent the incidence of
resistance by degrading other aminoglycosides antibiotic resistance as well as prevent adverse
are unable to break down amikacin. Hence, cross-­ effects that arise from overdosage such as irre-
resistance with other aminoglycosides does not versible ototoxicity and reversible nephrotoxic-
often occur, highlighting amikacin’s efficacy ity. Factors such as high doses over prolonged
against resistant strains. During treatment with courses, renal impairment, and age, each of
amikacin, care must be taken not to drop below which may predispose an individual to toxicity,
trough plasma concentrations of 5–10 μg/mL nor must be considered in therapeutic drug monitor-
exceed peak plasma concentrations of 30–35 μg/ ing of gentamicin.
14 Recent Advances in Nanosensors for Therapeutic Drug Monitoring (TDM) 241

Since there are no fluorophores or chromo- who respond well to treatment without any
phores in the molecular structure of gentamicin, a adverse effects depicting toxicity. However, fac-
nanosensor was developed using epicatechin tors such as poor compliance, inadequate
coated silver nanoparticles to determine the con- response, fluctuating renal function, lack of his-
centration of gentamicin in samples of human tory whether patient has previously taken a car-
serum and plasma [24]. The principle of measur- diac glycoside, confirmation of clinical toxicity,
ing gentamicin using this design is based on the and drug interactions all warrant the need for
fact that gentamicin can quench the absorbance therapeutic monitoring of digoxin. Digoxin tox-
intensity of silver nanoparticles at a pH ranging icity can result in cardiotoxicity, neurological
between 1 and 12. The detection limit for this symptoms like mental confusion and visual dis-
nanosensor was 1.28 μM, and the linear range turbances, as well as hypersensitivity reactions. It
was observed to be from 0 μM to 100 μM [24]. has a large volume of distribution with higher
concentrations in myocardium compared to the
plasma. Digoxin has an elimination half-life
14.6.3 Nanosensors for Digoxin ranging from 1.5 to 2 days. It is excreted in urine
in the unchanged form via tubular secretion and
Digoxin is a drug used for the management of glomerular filtration [19].
heart failure and cardiac arrhythmias. It is classi- 1. An electrochemical aptasensor using silver
fied as a cardiac glycoside sourced from the plant nanoparticle-decorated graphene oxide
Digitalis lanata, and it exerts its effects on the (AgNP-GO) has been investigated for use in
vascular smooth muscle and the heart [25, 26]. It analyzing digoxin concentration in biologi-
increases the myocardial force of contraction as cal samples [27]. A previous method of
well as reduces conduction of impulse to the employing an immunoassay for digoxin
heart through the atrioventricular node [19]. detection in the clinic posed challenges that
Moreover, digoxin increases vagal activity in included the interaction with digoxin-like
vascular smooth muscles. In a nutshell, digoxin molecules (e.g., ouabain). Aptamers are sin-
produces effects such as negative chronotropy, gle-stranded oligonucleotides that have
positive inotropy, and reduced atrioventricular shown great specificity and affinity for vari-
nodal activity. The therapeutic effect of digoxin ous targets including drugs. They provide
begins 2 h after oral administration and peaks benefits such as reproducibility for various
around the sixth hour. Steady-state plasma con- targets, great stability, and easily modified
centrations are reached between 1 and 3 weeks with electroactive enzymes and other suit-
based on the renal status [19]. Patient therapy able substances. Electrochemical aptasen-
begins with a loading dose of digoxin if “digitali- sors provide great benefit because of their
zation” of the patient (initial dose establishment simple operation, high sensitivity, low cost,
by fractionation) is needed, followed by a main- and great stability. An AgNP-GO nanocom-
tenance dose which is adjusted to individual posite was used as a redox tag for this digoxin
patients based on age, renal function, electrolyte sensing technique [27]. The surface area of
balance, lean body mass, disease nature, thyroid the biosensing platform was also increased
status, and degree of tissue oxygenation to with electrodeposited gold nanoparticles
achieve therapeutic efficacy. This drug can be (GNP) [27]. These strategies provided a dual
administered through the oral route, intravenous signal amplification, better detection limit,
route, or sometimes the intramuscular route. and greater dynamic range to the aptasensor.
Principally, it has a narrow therapeutic window It has been successfully used in detection of
between 0.5 and 2.0 ng/mL [26]. Confounding digoxin concentrations in human plasma
this narrow therapeutic window, there is the exis- samples without the need for pretreatment.
tence of inter-individual variation. Therapeutic Its linear dynamic range is reported as 1 pM
drug monitoring may not be needed in patients to 0.1 μM, with a detection limit of 0.3 pM
242 P. S. Agogo-Mawuli and D. P. Siderovski

[27]. There is also no need for continuous commonly monitored by checking the clotting
calibration considering its high degree of status of the patient’s plasma, and it is defined by
reproducibility. However, this biosensor can- an international normalized ratio (INR). Warfarin
not be used multiple times and must be dis- has a narrow therapeutic concentration ranging
posed after use. from 2.0 to 5.0 μg/ml and a long half-life between
2. An optical aptasensor based on aqueous gold 20 and 60 h [30]. Moreover, warfarin acts indi-
nanoparticles (AuNP) has independently been rectly to depress synthesis of vitamin K-dependent
developed for the determination of digoxin clotting factors II, VII, IX, and X. Therefore,
[28]. Due to the ability of aptamers to bind to there is the need to directly monitor its concentra-
a wide range of targets as well as exhibit high tion in plasma during treatment [19].
affinity and sensitivity to these targets, they 1. A magnetic iron oxide (Fe3O4) nanoparticle-­
were employed in this digoxin sensor. The modified carbon paste electrode (CPE) was
fluorescence produced by aptamers can be used to build an electrochemical sensor for
quenched through fluorescence resonance warfarin determination [31]. The electro-
energy transfer (FRET) when these aptamers chemical method is claimed to provide a
are adsorbed onto the AuNP surface [28]. cheap, sensitive, and simple sensor for drug
AuNPs were selected in fabricating this sen- detection. Fe3O4 nanoparticles were employed
sor because they are recognized as strong because they offer a large ratio of surface area
quenchers. Digoxin binding to the aptamers to volume as seen with other nanomaterials as
leads to AuNP aggregation (resulting in a col- well. Determination of warfarin was success-
orimetric change from red to blue) and recov- fully conducted in urine and serum with little
ery of the fluorescence produced by the interference. The linear range was 0.5–
aptamers. The detection limit for the colori- 1000 μM, and the limit of detection was
metric aptasensor was 571 pM while that of 0.21 μM [31].
the fluorescence quenching aptasensor was 2. A warfarin nanosensor has been developed in
392 pM [28]. Based on the outcome, the fluo- an independent effort based on covalent
rescence quenching aptasensor is said to be immobilization of cadmium sulfide (CdS)
more sensitive than the colorimetric aptasen- quantum dots onto carboxylated multiwalled
sor. This nanosensor design has subsequently carbon nanotubes and a chitosan composite
been applied to the detection of digoxin in film modified electrode [32]. Quantum dots
serum samples [28]. (QDs) are semiconductor nanocrystals that
possess electronic as well as optical character-
istics capable of being modified on their sur-
14.6.4 Nanosensors for Blood-­ face using various functional groups [33].
Thinning Agents QDs serve as a platform for the attachment of
multiwall carbon nanotubes (MWCNTs) to
14.6.4.1 Nanosensors for Warfarin improve electronic transmission as well as
Warfarin is an oral anticoagulant used to treat and electrode sensitivity. The presence of carbon
prevent venous thromboembolism [29]. Warfarin nanotubes conferred good stability, a wider
therapy is initiated shortly after heparin adminis- available surface area, and high conductivity
tration, and this combination is given up to at [34]. In addition to the low-cost advantage of
least 5 days before stopping heparin. It is also chitosan, this polycationic polymer substance
beneficial in stroke management and the preven- exhibits high permeability, good adhesive
tion of myocardial infarction. Warfarin-based properties, and acceptable film-forming
anticoagulant therapy must be monitored, and capacity. Warfarin determination was con-
doses individualized, due to increased risk of ducted in three potentially interfering biologi-
bleeding in patients, which could cause compli- cal matrices (viz., serum, breast milk, and
cations like anemia or hematomas. Warfarin is urine), where this method still exhibited
14 Recent Advances in Nanosensors for Therapeutic Drug Monitoring (TDM) 243

acceptable selectivity [32]. The sensor that cific binding sites that recognize the warfarin
was developed exhibited a wider linear range target. In addition, MIPs are cheap, highly
compared to previously reported sensors and stable, and robust and possess a strong affinity
good sensitivity as well as a low detection to their template molecule, warfarin. MIPs
limit: The linear range for the detection of had previously been used as a thin layer on the
warfarin with this method was from 0.05 to modified electrode [38]; the sensitivity of the
80 μM, and the detection limit was reported to nanosensor was further enhanced by the addi-
be 8.5 nM [32]. tion of MWCNT and Au nanoparticles (NP)
3. A nanosensor made up of MnFe2O4 and a on both sides of the MIP layer [37]. This form
multiwalled carbon nanotube (MWCNT) of nanosensor exhibited good selectivity to
paste matrix was built for the determination of warfarin when probing human serum
warfarin [35]. MnFe2O4 is a magnetic samples.
nanoparticle that possesses favorable proper- 5. A newer warfarin nanosensor was made by
ties such as excellent thermal stability, good incorporating cobalt oxide nanoparticles onto
magnetic property, good catalytic activity, and a multiwalled carbon nanotube-modified
excellent electronic conductivity [36]. These glassy carbon electrode [39]. The choice of
nanoparticles are combined with MWCNTs cobalt oxide nanoparticles enhanced the elec-
to promote high sensitivity, resistance to sur- trochemical response of warfarin [39]. Carbon
face fouling, low limits of detection, and nanotubes are commonly employed as a mod-
reduction of overpotentials. Previous warfarin ifier in such setups given their characteristics
electrochemical nanosensor design using such as large surface areas, good adsorptive
Fe3O4 magnetic particles [31] was considered ability, catalysis, improved mass transport,
by these designers [35] as limited by instabil- and high porosity [40]. Incorporating cobalt
ity in the external environment [35]. Therefore, oxide nanoparticles increased warfarin
the choice of manganese ferrite (MnFe2O4) adsorption that occurs as a result of cobalt
nanoparticles in their newly developed elec- ions and warfarin forming a complex [41].
trode was to help resolve stability issues expe- The electrochemical signal of warfarin was
rienced in previous nanosensors [35]. The amplified, and this nanosensor eventually
detection limit for the MWCNT/MnFe2O4/ widened the range of measurement as well:
CPE electrode was found to be 0.08 μM, and the limit of detection for this method was
the linear dynamic range was 0.10–447 μM 0.0033 μM, and the linear range was 0.008–
[35]. The only substance reported to interfere 800 μM [39]. Good selectivity as well as long-­
with this method was free cysteine, which is term stability was established in this
not abundant in biological matrices under nanosensor when used in the determination of
examination [35]. This procedure could there- warfarin without any form of interference
fore be considered for determination of warfa- [39], facilitating the measurement of warfarin
rin in serum and urine samples. in both human serum and urine samples [39].
4. A molecularly imprinted polymer (MIP)
nanosensor has also been described for warfa- 14.6.4.2 Nanosensor for Heparin
rin determination [37]. The sensitivity and Heparin functions as an anticoagulant by promot-
electronic transmission of the nanosensor ing antithrombin III activity in plasma. Hence,
were improved by incorporating multiwall the rate of inhibition by antithrombin III of blood
carbon nanotubes (MWCNTs) containing car- clotting factors such as thrombin and factor Xa is
boxylic functional groups onto a glassy car- potentiated by heparin in a dose-dependent man-
bon electrode [37]. The limit of detection for ner [19]. Heparin is used to treat and prevent
this method was 0.024 ng/mL, and the linear thromboembolic disorders such as pulmonary
range was 0.031–0.616 ng/mL [37]. embolism and deep vein thrombosis. It is highly
Molecularly imprinted polymers possess spe- bound to plasma proteins and excreted mainly in
244 P. S. Agogo-Mawuli and D. P. Siderovski

a metabolized form. Heparin therapy serves as a 14.6.4.3 Nanosensor for Aspirin


precursor to oral anticoagulation until the full Aspirin (also known as acetylsalicylic acid) has
effect of the oral anticoagulant is exerted. At this historically been used in low doses as an anti-
point, heparin can then be withdrawn [19]. thrombotic measure in the initial management of
Heparin is administered intravenously, and its cardiovascular diseases and prophylaxis of car-
effect is monitored daily by measuring activated diovascular diseases in high-risk patients, as well
partial thromboplastin time (APTT) [42, 43]. as for the treatment of mild to moderate pain.
However, this APPT method of monitoring is After its ingestion, acetylsalicylic acid is hydro-
indirect and may not provide accurate measure- lyzed into acetic acid and salicylic acid, and the
ments due to interference from other substances. latter moiety is responsible for aspirin’s pharma-
Moreover, the risks of hemorrhage and heparin-­ cological activity. It exhibits its action via inhibi-
induced thrombocytopenia warrant the need for tion of cyclooxygenase enzyme leading to a
heparin detection and quantification during treat- decreased production of prostaglandins and
ment [44]. thromboxanes [19]. Aspirin causes adverse
An optical nanosensor based on fluorescence effects such as NSAID-induced gastric ulcer-
has been developed for the detection of heparin. ation, increase in bleeding time, and hypersensi-
This nanosensor is made up of rhodamine tivity reactions. Salicylism, metabolic acidosis,
B-labeled peptides (RBP) that serve as sensitive and hyperventilation may also occur with high
bioreceptors for heparin given their provision of intoxication of aspirin, which can be controlled if
numerous heparin-binding sites [45]. The bind- dosage is reduced. Salicylate intoxication
ing to RBP of heparin quenches the fluorescence involves the ingestion of more than 125 mg/kg of
of RBP due to an induced conformational change aspirin, which warrants monitoring of plasma
of RBP from the unfolded form to the folded salicylate concentration to predict the severity of
form [45]. The RBP-based nanosensor was the poisoning.
reported to have two linear ranges of operation: An electrochemical nanosensor was built on a
0.01–0.1 nM and 1.0–70.0 nM; the limit of detec- screen-printed carbon electrode with chitosan
tion turned out to be 7.5 pM [45]. This method capped with gold nanoparticles (Cs + AuNPs)
was used for the determination of heparin in [51]. Its linear range in detection of aspirin was
human serum samples, exhibiting high selectivity from 1 pg/mL to 1 μg/mL. The limit of the detec-
and good sensitivity. This sensor performance tion was 0.03 pg/mL [51]. An electrochemical
was speculated to arise from the spatial confor- method of determination was chosen as it affords
mation assumed by the heparin and the electro- various advantages for this kind of drug nanosen-
static nature of the heparin/RBP binding sor design including cheaper costs, high sensitiv-
interaction [45]. An arginine-rich peptide ity, simple operation, and its capacity to analyze
sequence was specifically designed to serve as real samples in complex biological matrices [52–
the basis for the RBP heparin receptor because 60]. Chitosan, a polysaccharide, is a poor electri-
previous studies had shown that arginine was cal conductor and therefore needed to be attached
critical to nearly all heparin-binding peptides with conductive metallic loads to enhance its
from the G domain of the laminin α1 chain [46– conductivity. Gold nanoparticles were therefore
50]. This arginine-rich peptide was then conju- selected to augment this function, providing a
gated with the fluorophore rhodamine B, reduction in response times and improvement in
affording a measurable change in fluorescence signal-to-noise ratio. These nanoparticles also
when the peptide changed conformation upon promoted signal amplification by increasing the
binding heparin. adhesion that exists between the analyte and the
substrate. Screen-printed electrodes were consid-
ered to provide several convenience factors sup-
14 Recent Advances in Nanosensors for Therapeutic Drug Monitoring (TDM) 245

porting real-time monitoring that included low is used in this method as a conductive paste
cost, small volume of sample requirements, as binder. The specificity property conferred by car-
well as simplicity. Quantitative determination of bon paste electrodes, even in complex samples,
aspirin was carried out in both human urine and served as a central justification for its use in this
saliva [51]. method of determination [62, 63]; selection of
the type of modifier used for ultimate fabrication
of the carbon paste electrode was considered a
14.6.5 Nanosensor for Tramadol central mechanism by which the selectivity and
sensitivity of the nanosensor were improved.
Tramadol is an opioid drug that produces anal-
gesia mainly via opioid receptors. However, it
also exerts its actions via noradrenergic and 14.6.6 Nanosensor for Sulfasalazine
serotonergic means and therefore also consid-
ered a member of the serotonin-norepinephrine Sulfasalazine is a drug used in the treatment of
reuptake inhibitor (SNRI) class of antidepres- inflammatory bowel disease. It can be metabo-
sant drugs [19]. There is the need for this drug lized into mesalazine (5-aminosalicylic acid),
to be monitored in patients who have mild renal which is mainly responsible for its activity, and
impairment, consume large amounts of alcohol, into sulfapyridine, which has a significant impact
or have hepatic impairment. Tramadol also low- on the adverse reactions of sulfasalazine.
ers seizure threshold in patients with epilepsy; Sulfasalazine is also classified as a disease-­
hence, there is the need for dosage adjustment modifying antirheumatic drug (DMARD), which
for this potential adverse effect as well. helps to alleviate the joint pain, swelling, and
Metabolism of tramadol occurs in the liver by stiffness experienced in rheumatoid arthritis [19].
O-demethylation and N-demethylation via The small intestine absorbs about 15% of intact
CYP2D6 and CYP3A4 metabolizing enzymes. sulfasalazine, with some of it later undergoing
Sulfation or glucuronidation of tramadol can enterohepatic cycling. Most of the intact drug is
also occur. Its metabolism produces cleaved in the colon into mesalazine and sulfa-
O-desmethyltramadol, which is itself pharma- pyridine prior to absorption. Both mesalazine and
cologically active. Tramadol has an elimination sulfapyridine are extensively metabolized by
half-life of about 6 h, and its metabolites are acetylation. In addition, sulfapyridine is also
mainly excreted from the body in urine. metabolized by glucuronidation and hydroxyl-
An electrochemical nanosensor has been ation. Trace amounts of intact sulfasalazine is
described which has a carbon paste matrix com- excreted in urine in an unchanged form with sig-
posed of ionic liquid, decorated graphene nificant amounts excreted as metabolites.
nanosheets with silver nanoparticles, and graph- Therapeutic drug monitoring is therefore needed
ite powder [61]. Tramadol-imprinted polymer here since higher dosage of sulfasalazine is asso-
nanoparticles were combined with the carbon ciated with toxicity, especially in slow acetyl-
paste matrix to form a “nanosensing layer” of the ators with whom it is two to three times more
resultant potentiometric nanosensor [61]. probable to develop overt toxicity symptoms.
Quantitative determination of tramadol using this An electrochemical nanosensor made up of
method was conducted successfully in human ionic liquid and nickel oxide (NiO)/CNTs nano-
urine samples. The linear range for this electro- composite was used to fabricate a modified car-
chemical determination was found to be from bon paste electrode [64]. The linear range for this
3.5 × 10−9 to 1.00 ×10−2 M with a Nernstian slope nanosensor design was from 5.0 × 107 to
of 59.85 ± 0.13 mV per decade [61]. The limit of 8.0 × 10−4 M, and the limit of detection was
detection was 2.04 × 10−9 M [61]. Tramadol-­ 9.0 × 10−8 M [64]. The ionic liquid that was used
imprinted polymer nanoparticles function in this in this design as a conductive paste binder was
design as the sensing agent, while the ionic liquid reported to have high thermal stability, extensive
246 P. S. Agogo-Mawuli and D. P. Siderovski

electrochemical window, fast electron transfer, of 0.2 μM [69]. Merits of using screen-printed
and high conductivity [64]. Nickel oxide nanopar- carbon electrodes (SPCE) in these electro-
ticles were chosen as they were considered to chemical measurements have been described
possess satisfactory biological compatibility, as including affordability, quick response,
electron transfer capacity, and high chemical sta- small size, and simple fabrication [70, 71].
bility [65–67]. This nanosensor was used to Graphene quantum dots increase the surface
detect sulfasalazine in urine samples. area that has contact with the analyte.
Therefore, the electroactive analyte interacts
with an expanded electrochemical active sur-
14.6.7 Nanosensors for Theophylline face area [69].
2. A biosensor for detection of theophylline,
Theophylline is a xanthine used in the manage- based on an RNA aptamer for theophylline
ment of respiratory disorders like asthma and coupled with a graphene oxide (GO) and
chronic obstructive pulmonary disease. It is said cryonase-­ mediated amplified fluorescence
to exert its bronchodilatory effect via inhibition system, has been independently developed
of phosphodiesterase, leading to increased lev- [72]. Rather than establishing theophylline
els of cyclic adenosine monophosphate (cAMP) detection on the basis of an RNA aptamer-
[19]. Other schools of thought believe that the attuned nuclease, a GO/cryonase-­ mediated
action of theophylline arises from its antagonis- amplified fluorescence system was employed,
tic role on adenosine receptors as well as its to achieve signal amplification regardless of
anti-­inflammatory effect [68]. Theophylline is which nucleic acid molecular probe (NAMP)
rapidly absorbed and reaches peak serum con- was ultimately used in the design. GO has
centrations 1–2 h after oral administration. The affinity for both single-stranded and double-­
therapeutic window for theophylline is narrow, stranded NAMPs. Moreover, the ability of
ranging from 10 to 20 μg/ml [19]. Factors such cryonase to digest both DNA and RNA probes
as smoking, medications, age, and diet may enzymatically facilitates its universal applica-
modify the pharmacokinetics of theophylline. tion for all forms of NAMP sequences. This
Therefore, there is the need for the dose of the- method can be used to design new aptasensors
ophylline to be individualized during treatment for various analytes. The fluorescence pro-
based on plasma levels, adverse effects, and duced by dye-­labeled aptamers is quenched
therapeutic response. 40–60% of the drug is through fluorescence resonance energy trans-
bound to plasma proteins; moreover, variations fer (FRET) when these aptamers are adsorbed
in theophylline metabolism in different individ- onto the GO surface. GO was selected in fab-
uals affect the ultimate plasma concentration of ricating this nanosensor because it is recog-
theophylline. Products from theophylline nized as a strong fluorescence quencher.
metabolism are mainly excreted in urine, with However, this quenching is disrupted by the
only 10% excreted in the unchanged form in binding of theophylline to the aptamer, result-
adults [19]. Monitoring theophylline levels ing in the reoccurring of fluorescence pro-
would help prevent toxicity and, eventually, duced by the aptamers. Release of the aptamer/
serious adverse effects. theophylline complex from the GO surface
1. An electrochemical nanosensor design for the- becomes available for cryonase digestion.
ophylline has been described, using screen-­ Afterwards, the free theophylline binds to
printed electrodes (SPEs) modified with another aptamer to facilitate another cycle of
graphene quantum dots (GQDs) [69]. This cleavage. This cycle continues repeatedly
electrochemical method was tested on urine until all aptamer probes are consumed. This
samples; the linear dynamic range of this elec- cycling enables the activation of all fluoro-
trochemical nanosensor for theophylline was phores and consequently amplifies the fluores-
from 1.0 to 700.0 μM, with a limit of detection cent signals. The limit of detection of
14 Recent Advances in Nanosensors for Therapeutic Drug Monitoring (TDM) 247

theophylline was 47 nM [72]. This biosensor the detection of levothyroxine [74]. The linear
was used for determination of theophylline in dynamic range for the proposed nanosensor was
serum samples. from 10 to 120 nM, and the nanosensor’s limit of
detection was 2.84 nM [74]. Carbon nanotubes
were chosen as modifiers due to the fact that they
14.6.8 Nanosensor for Levothyroxine are relatively chemically unreactive and possess
good chemical stability, ordered structure, and
Levothyroxine (T4) is a drug used to treat dif- electrical conductivity [75, 76]. The incorpora-
fuse goiter and Hashimoto’s thyroiditis due to tion of gold nanoparticles helped to enhance the
its ­ability to suppress the production of thyroid-­ sensitivity as well as the selectivity of the electro-
stimulating hormone (TSH) [19]. Given its chemical nanosensor. The nanocomposite was
ability to suppress TSH secretion, T4 is also immobilized onto the surface of a glassy carbon
used in differential diagnosis of hyperthyroidism electrode (GCE), and this electrochemical
and beneficial in thyroid carcinoma treatment. method enabled by this nanosensor design was
This drug is essentially a thyroid hormone that subsequently applied in human blood serum.
is commonly given orally, intravenously, or
intramuscularly and does not immediately
change therapeutic response when its dose is 14.6.9 Nanosensor for Lithium
adjusted. It is often used as a replacement ther-
apy in hypothyroidism [73]. Moreover, its Lithium is a drug used to manage conditions such
absorption is greatly impaired with food. as bipolar disorder, recurrent unipolar depres-
During treatment, TSH levels are monitored, sion, and mania. It has a narrow therapeutic index
and the dosage of levothyroxine is adjusted that warrants the need for this drug to be moni-
accordingly to attain normal TSH levels follow- tored in patients taking it. While its mechanism is
ing total thyroidectomy. The physiological sig- unknown, it is said to be involved in a competi-
nificance of T4 speaks to the necessity in tion with sodium ions in various locations of the
monitoring its levels in serum to bring insight body [19]. Lithium is entirely absorbed in the
to the diagnosis of hypothyroidism and hyper- gastrointestinal tract and excreted mainly by the
thyroidism. Adverse effects are commonly kidneys into urine.
experienced in cases of T4 overdosage, which An optical nanosensor based on fluorescence
are similar to symptoms of hyperthyroidism spectroscopy was developed for the detection of
like insomnia, cardiac arrhythmias, muscle lithium ions. A 12-metallacrown-3 complex in
weakness, and tachycardia. Chronic intoxica- the lithium nanosensor played the role of recog-
tion results in events such as thyroid storm, car- nition component due to its high selectivity and
diac arrhythmias, heart failure, and death. Due affinity for lithium ions [77]. This nanosensor
to its narrow therapeutic index, extra precau- exhibited very good selectivity for lithium over
tions should be taken when administering T4 to Na+ and Mg2+ even while these latter ions are
patients with preexisting cardiovascular disor- more concentrated in biological matrices com-
ders, long-standing hypothyroidism, adrenal pared to lithium [77]. Prior to determination of
insufficiency, diabetes, and epilepsy to prevent lithium in human serum using this optical nano-
worsening their prognosis. sensor, a simple precipitation procedure was
A nanocomposite made up of melamine employed to remove the large proteins which
attached to gold nanoparticles by means of could have caused interference via autofluores-
mercaptoethanol-­ modified multiwalled carbon cence and competitive coordination to (arene) Ru
nanotubes (MWCNTs/CC-SH/AuNPs) was complexes that constituted the 12-metallacrown-­
developed as an electrochemical nanosensor for 3-based recognition component.
248 P. S. Agogo-Mawuli and D. P. Siderovski

14.6.10 Nanosensors oxide (rGO) [83]. rGO was chosen to reduce the
for Anticonvulsants aggregation of active materials and improve the
electrocatalytic properties of the nanosensor
14.6.10.1 Nanosensor design [80].
for Carbamazepine
Carbamazepine is an antiepileptic and psychotro- 14.6.10.2 Nanosensors
pic drug that is commonly used to manage gener- for Phenobarbital
alized tonic-clonic seizures as well as partial Phenobarbital is an antiepileptic medication that
seizures [19, 78]. Carbamazepine requires dose is prescribed to control seizures such as status epi-
adjustment from one patient to the other to lepticus and partial seizures, as well as general-
achieve plasma concentrations usually between 4 ized tonic-clonic seizures [19, 84, 85]. It has a
and 12μg/mL [79]. The dose must be adequate to half-life of about 75–120 h in adults with 45%–
control the seizure and yet minimize adverse 60% bound to plasma proteins. The dosage
effects like neurotoxicity. Carbamazepine under- required to bring the seizure under control varies
goes hepatic metabolism, with its metabolites from individual to individual with its usual thera-
almost entirely excreted in urine with trace peutic plasma concentration ranging from 15 to
amounts excreted in feces. Reports show that 40 μg/mL [19]. This therapeutic window may
70%–80% of the drug is bound to plasma pro- need to be increased with time due to the develop-
teins and elimination of the drug is slower in ment of tolerance with this drug [86]. Hence,
adults, resulting in longer retention of more there is the need for dose adjustment to be made
active metabolites; the half-life of carbamazepine based on the patient. Therapeutic drug monitoring
also shortens with repeated drug administration of phenobarbital helps to avoid adverse effects
because it can induce its own metabolism [19]. like cardiovascular and respiratory depression,
Based on the characteristics of this drug includ- ataxia, and coma that result from overdosage [87].
ing complex pharmacokinetics and its serious
adverse effects, therapeutic drug monitoring 1. A fluorescence nanosensor based on green
would help to optimize therapy in patients who source carbon dots (GSCDs) coated with
depend on it to live seizure-free. molecularly imprinted polymers (MIPs) was
A modified glassy carbon electrode which developed for the determination of phenobar-
comprises of a nanocomposite of cerium-doped bital [88]. The limit of detection for this nano-
zinc oxide (ZnO) and reduced graphene oxide sensor was 0.1 mM, and the linear range was
was developed for the electrochemical sensing of between 0.4 and 34.5 mM [88]. This optical
carbamazepine [80]. The linear range for carba- method of determination was conducted in
mazepine determination was from 0.05 to human blood plasma samples. The molecular
100 μM, and the limit of detection was 1.2 nM imprinting technique was used in building this
[80]. This nanosensor was reported to exhibit nanosensor, creating a recognition site with
high selectivity, long-term stability, high sensitiv- polymeric materials that is selective to the tar-
ity, and good reproducibility. This method of get, phenobarbital [89]. Moreover, as previ-
detection was successfully executed in urine ously reported, MIPs are cheap, are easy to
samples. ZnO has become an important metal prepare, are thermally stable in basic and
oxide that is used in electrochemical nanosensors acidic medium, and confer a high degree of
as a semiconductor for the determination of drugs selectivity to the proposed nanosensor [90].
[81, 82]. To increase the selectivity and sensitiv- The fluorescent characteristic of carbon dots,
ity of the nanosensor, ZnO was doped with a dif- together with their simplicity in terms of syn-
ferent rare earth metal (Cerium) [80]. The sensing thesis, surface functionalization, and good sta-
performance of the electrode was further bility, made these nanoparticles stand out as
improved by forming a nanocomposite of cerium-­ suitable for the development of this nanosen-
doped zinc oxide (ZnO) with reduced graphene sor [91, 92].
14 Recent Advances in Nanosensors for Therapeutic Drug Monitoring (TDM) 249

2. Independent of the optical design mentioned The extent of fluorescence of QDs is dependent
above, a modified multiwalled carbon nano- on the change in pH of the environment [95].
tube (MWCNT) paste electrode was set up Hence, the weakly acidic nature of valproic acid
with the incorporation of platinum (Pt) causes a pH change that results in fluorescence
nanoparticles for the detection of phenobarbi- quenching of CdTe QDs. The linear range of the
tal [93]. The linear range for phenobarbital for engineered nanosensor was from 0.3 to 7.5 mg/L,
this electrochemical nanosensor was from 0.4 and the detection limit was 0.24 μg/mL [95]. This
to 60 μM, and its limit of detection was 0.1 μM nanosensor was used for the detection of valproic
[93]. This e­ lectrochemical method was inves- acid in urine samples and human serum.
tigated in human urine samples. Carbon nano-
tubes are commonly employed as a modifier 14.6.10.4 Nanosensor for Phenytoin
in such setups due to their favorable character- Phenytoin is an antiepileptic drug that is used
istics such as large surface areas, good adsorp- to manage generalized tonic-clonic and partial
tive ability, catalysis, improved mass transport, seizures [19]. Treatment of patients with phenyt-
and high porosity [40]. Electrocatalytic pro- oin requires the dose to be individualized to con-
cesses and surface areas of the composite trol seizures due to the narrow therapeutic index
matrix were reported to improve significantly of this drug. The concentration range within
when Pt nanoparticles are combined with the which therapeutic outcomes can be effectively
carbon nanotubes, as compared to the indi- achieved is between 10 μg/mL and 20 μg/
vidual component properties [94]. mL. The half-life of phenytoin varies based on
the dose administered but averages about 22 h.
14.6.10.3 Nanosensor for Valproic Moreover, the potential for phenytoin to inhibit
Acid its own metabolism prolongs the time it takes to
Valproic acid is a drug used to treat primary gen- achieve steady-­state concentration.
eralized seizures and partial seizures. It has been Monitoring of phenytoin was reported to be
shown to be beneficial in the treatment of absence possible in plasma samples with a nanosensor
and myoclonic seizures. Despite the fact that its developed using branched gold nanoparticles
mechanism is not fully understood, valproic acid (B-AuNPs). The B-AuNPs possess a localized
is said to facilitate GABAergic transmission in surface plasmon resonance (SPR) property which
the central nervous system as well as inhibit helps in spectrophotometric determination of
sodium channels [19]. There is the need for dose phenytoin in situ. Phenytoin forms a complex
adjustment in patients taking valproic acid to with Au (III) leading to the aggregation of
gain adequate seizure control. The use of valproic B-AuNP. Therefore, at 540 nm, the intensity of
acid can result in adverse effects like liver dys- the characteristic SPR band of B-AuNP is
function which requires the discontinuation of reduced, followed by a change in color of the
treatment if left unchecked. The dose needs to be solution from red to blue [96]. The method is sen-
adjusted in patients that experience elevated liver sitive, rapid, and simple based on the detection
enzymes within the first few months of treatment limit of 21 ng/mL, the capacity to obtain results
because this adverse effect is dose related. The in less than 15 min, and being able to visually
metabolites of valproic acid are excreted entirely observe the color changes [96]. The low detec-
in urine. tion limit allows the plasma samples to be well
An optical method was developed for the diluted prior to determination to prevent interac-
determination of valproic acid. The nanosensor tions from species present in the sample.
was developed with thioglycolic acid (TGA)- However, interference studies revealed that drugs
capped CdTe quantum dots (QDs) [95]. These like lamotrigine and paracetamol can interfere
quantum dots are said to possess properties such with the detection of the analyte. The linear range
as high photoluminescence quantum efficiency, was determined to be from 67 to 670 ng/mL [96].
photostability, and tunable emission spectrum.
250 P. S. Agogo-Mawuli and D. P. Siderovski

References 20. Shekarbeygi Z, et al. Development of Ag nanoparticle-­


carbon quantum dot nanocomplex as fluorescence
sensor for determination of gemcitabine. Spectrochim
1. Ghiculescu R. Therapeutic drug monitoring: which
Acta A Mol Biomol Spectrosc. 2021;262:120148.
drugs, why, when and how to do it. Aust Prescr.
21. Berger AG, White IM. Therapeutic drug monitoring
2008;31:42–4.
of flucytosine in serum using a SERS-active mem-
2. Figueras A. Review of the evidence to include TDM in
brane system. In: Frontiers in biological detection:
the essential in vitro diagnostics list and prioritization
from nanosensors to systems, vol. IX. International
of medicines to be monitored. Geneva, Switzerland:
Society for Optics and Photonics; 2017.
WHO; 2019.
22. El-Sayed GM, et al. Nanoparticle-enhanced poten-
3. Mika A, Stepnowski P. Current methods of the analy-
tiometric ion-selective electrodes for therapeutic
sis of immunosuppressive agents in clinical materials:
drug monitoring of linezolid. J Electrochem Soc.
a review. J Pharm Biomed Anal. 2016;127:207–31.
2019;166(14):B1312.
4. Ates HC, et al. On-site therapeutic drug monitoring.
23. Losoya-Leal A, et al. Design of a surface plasmon
Trends Biotechnol. 2020;38(11):1262–77.
resonance immunoassay for therapeutic drug moni-
5. Iftikhar FJ, et al. Introduction to nanosensors. In:
toring of amikacin. Talanta. 2015;141:253–8.
New developments in nanosensors for pharmaceutical
24. Ikram F, Qayoom A, Shah MR. Synthesis of epicat-
analysis. Elsevier; 2019. p. 1–46.
echin coated silver nanoparticles for selective rec-
6. Sharma P, et al. A review on biosensors and nanosen-
ognition of gentamicin. Sensors Actuators B Chem.
sors application in agroecosystems. Nanoscale Res
2018;257:897–905.
Lett. 2021;16(1):1–24.
25. Kiani Z, et al. In vitro selection and characterization
7. Mehrotra P. Biosensors and their applications–a
of deoxyribonucleic acid aptamers for digoxin. Anal
review. J Oral Biol Craniofac Res. 2016;6(2):153–9.
Chim Acta. 2012;748:67–72.
8. Garzón V, et al. Optical biosensors for therapeutic
26. Cheng M-C, Chi K-M, Chang SY. Detection of digoxin
drug monitoring. Biosensors. 2019;9(4):132.
in urine samples by surface-assisted laser desorption/
9. Reder-Christ K, Bendas G. Biosensor applications
ionization mass spectrometry with dispersive liquid–
in the field of antibiotic research—a review of recent
liquid microextraction. Talanta. 2013;115:123–8.
developments. Sensors. 2011;11(10):9450–66.
27. Mashhadizadeh MH, Naseri N, Mehrgardi MA. A
10. Thevenot DR, et al. Electrochemical biosensors: rec-
digoxin electrochemical aptasensor using Ag nanopar-
ommended definitions and classification. Pure Appl
ticle decorated graphene oxide. Anal Methods.
Chem. 1999;71(12):2333–48.
2016;8(39):7247–53.
11. Grieshaber D, et al. Electrochemical biosensors-­
28. Emrani AS, et al. Sensitive and selective detec-
sensor principles and architectures. Sensors.
tion of digoxin based on fluorescence quench-
2008;8(3):1400–58.
ing and colorimetric aptasensors. Anal Methods.
12. Pollard TD, et al. Electrochemical biosensors: a
2015;7(8):3419–24.
nexus for precision medicine. Drug Discov Today.
29. Harrington RA, et al. Antithrombotic therapy for cor-
2021;26(1):69–79.
onary artery disease: the seventh ACCP conference
13. Wongkaew N, et al. Functional nanomaterials and
on antithrombotic and thrombolytic therapy. Chest.
nanostructures enhancing electrochemical biosen-
2004;126(3):513S–48S.
sors and lab-on-a-chip performances: recent prog-
30. Walfisch A, Koren G. The “warfarin window” in preg-
ress, applications, and future perspective. Chem Rev.
nancy: the importance of half-life. J Obstet Gynaecol
2018;119(1):120–94.
Can. 2010;32(10):988–9.
14. Gómez DR. Biosensores ópticos de alta sensibili-
31. Gholivand MB, Torkashvand M. Electrooxidation
dad basados en técnicas de modulación plasmónica.
behavior of warfarin in Fe3O4 nanoparticles modified
Universidade de Santiago de Compostela; 2012.
carbon paste electrode and its determination in real
15. Damborský P, Švitel J, Katrlík J. Essays Biochem.
samples. Mater Sci Eng C. 2015;48:235–42.
2016;60(1):91–100.
32. Gholivand MB, Mohammadi-Behzad L. An electro-
16. Kivirand K, et al. Analyzing the biosensor signal
chemical sensor for warfarin determination based on
in flows: studies with glucose optrodes. Talanta.
covalent immobilization of quantum dots onto car-
2015;131:74–80.
boxylated multiwalled carbon nanotubes and chitosan
17. Babacan S, et al. Evaluation of antibody immobiliza-
composite film modified electrode. Mater Sci Eng C.
tion methods for piezoelectric biosensor application.
2015;57:77–87.
Biosens Bioelectron. 2000;15(11–12):615–21.
33. Drbohlavova J, et al. Quantum dots—characteriza-
18. Carrascosa LG, et al. Nanomechanical biosen-
tion, preparation and usage in biological systems. Int
sors: a new sensing tool. TrAC Trends Anal Chem.
J Mol Sci. 2009;10(2):656–73.
2006;25(3):196–206.
34. Karuwan C, et al. Flow injection based microfluidic
19. Brayfield A. Martindale: the complete drug reference.
device with carbon nanotube electrode for rapid sal-
Pharmaceutical Press; 2014.
butamol detection. Talanta. 2009;79(4):995–1000.
14 Recent Advances in Nanosensors for Therapeutic Drug Monitoring (TDM) 251

35. Taei M, et al. Highly selective differential pulse from the G domain of the laminin α1 chain. J Biol
voltammetric determination of warfarin in pharma- Chem. 2001;276(25):22077–85.
ceutical and biological samples using MnFe2O4/ 51. Diouf A, et al. An electrochemical sensor based on
MWCNT modified carbon paste electrode. chitosan capped with gold nanoparticles combined
Microchem J. 2016;129:166–72. with a voltammetric electronic tongue for quantitative
36. Sun S, et al. Study on warfarin plasma concentration aspirin detection in human physiological fluids and
and its correlation with international normalized ratio. tablets. Mater Sci Eng C. 2020;110:110665.
J Pharm Biomed Anal. 2006;42(2):218–22. 52. Karimi-Maleh H, et al. A novel DNA biosensor based
37. Rezaei B, Rahmanian O, Ensafi AA. An electrochem- on a pencil graphite electrode modified with polypyr-
ical sensor based on multiwall carbon nanotubes and role/functionalized multiwalled carbon nanotubes for
molecular imprinting strategy for warfarin recogni- determination of 6-mercaptopurine anticancer drug.
tion and determination. Sensors Actuators B Chem. Ind Eng Chem Res. 2015;54(14):3634–9.
2014;196:539–45. 53. Gupta VK, et al. Mercury selective potentiometric sen-
38. Kan X, et al. Composites of multiwalled car- sor based on low rim functionalized thiacalix [4]-arene
bon nanotubes and molecularly imprinted poly- as a cationic receptor. J Mol Liq. 2013;177:114–8.
mers for dopamine recognition. J Phys Chem C. 54. Gupta VK, et al. Electrochemical analysis of some
2008;112(13):4849–54. toxic metals by ion–selective electrodes. Crit Rev
39. Gholivand MB, Solgi M. Sensitive warfarin sensor Anal Chem. 2011;41(4):282–313.
based on cobalt oxide nanoparticles electrodeposited 55. Vinod K. Determination of lead using a poly (vinyl
at multi-walled carbon nanotubes modified glassy chloride)-based crown ether membrane. Analyst.
carbon electrode (CoxOyNPs/MWCNTs/GCE). 1995;120(2):495–8.
Electrochim Acta. 2017;246:689–98. 56. Srivastava SK, et al. Caesium PVC–crown (dibenzo-­
40. Valcárcel M, Cárdenas S, Simonet B. Role of car- 24-­crown-8) based membrane sensor. In: Analytical
bon nanotubes in analytical science. Anal Chem. proceedings including analytical communications.
2007;79(13):4788–97. Royal Society of Chemistry; 1995.
41. Dehari D, et al. New complexes of Co(II) and 57. Karimi-Maleh H, et al. The determination of
Cd(II) using 4-Hydroxy-2-Oxo-2H chromene-3-­ 2-phenylphenol in the presence of 4-chlorophenol
carboxamide as ligand. Res J Appl Sci. 2013;8:112–5. using nano-Fe3O4/ionic liquid paste electrode as
42. Basu D, et al. A prospective study of the value an electrochemical sensor. J Colloid Interface Sci.
of monitoring heparin treatment with the acti- 2019;554:603–10.
vated partial thromboplastin time. N Engl J Med. 58. Tahernejad-Javazmi F, Shabani-Nooshabadi M,
1972;287(7):324–7. Karimi-Maleh H. 3D reduced graphene oxide/
43. Cho P, Bonilla-Linero F, Yim E. The activated coag- FeNi3-ionic liquid nanocomposite modified sensor;
ulation time test as a control for heparin therapy. an electrical synergic effect for development of tert-­
Hawaii Med J. 1971;30(6):474–7. butylhydroquinone and folic acid sensor. Compos Part
44. Capila I, Linhardt RJ. Heparin–protein interactions. B. 2019;172:666–70.
Angew Chem Int Ed. 2002;41(3):390–412. 59. Khodadadi A, et al. A new epirubicin biosensor based
45. Guo P, Wang Y, Zhuang Q. Highly sensitive and on amplifying DNA interactions with polypyrrole and
selective biosensor for heparin detection with rho- nitrogen-doped reduced graphene: experimental and
damine B-labelled peptides as fluorescent biorecep- docking theoretical investigations. Sensors Actuators
tors. Sensors Actuators B Chem. 2019;299:126873. B Chem. 2019;284:568–74.
46. Dai Q, et al. Ratiometric fluorescence sensor based 60. Karimi-Maleh H, et al. Amplified nanostructure elec-
on a pyrene derivative and quantification detection of trochemical sensor for simultaneous determination of
heparin in aqueous solution and serum. Anal Chem. captopril, acetaminophen, tyrosine and hydrochloro-
2011;83(17):6559–64. thiazide. Mater Sci Eng C. 2017;73:472–7.
47. Kim D-H, et al. Ratiometric detection of nanomolar 61. Bagheri H, et al. Determination of tramadol in phar-
concentrations of heparin in serum and plasma sam- maceutical products and biological samples using
ples using a fluorescent chemosensor based on pep- a new nanocomposite carbon paste sensor based
tides. Anal Chem. 2014;86(13):6580–6. on decorated nanographene/tramadol-imprinted
48. Sauceda JC, Duke RM, Nitz M. Designing fluorescent polymer nanoparticles/ionic liquid. Ionics.
sensors of heparin. Chembiochem. 2007;8(4):391–4. 2018;24(3):833–43.
49. Thirupathi P, et al. Pyrene excimer-based pepti- 62. Afkhami A, Shirzadmehr A, Madrakian
dyl chemosensors for the sensitive detection of T. Improvement in performance of a hyoscine butylbro-
low levels of heparin in 100% aqueous solutions mide potentiometric sensor using a new nanocompos-
and serum samples. ACS Appl Mater Interfaces. ite carbon paste: a comparison study with polymeric
2015;7(26):14243–53. membrane sensor. Ionics. 2014;20(8):1145–54.
50. Hoffman MP, et al. Cell type-specific differences in 63. Bagheri H, Shirzadmehr A, Rezaei M. Designing and
glycosaminoglycans modulate the biological activ- fabrication of new molecularly imprinted polymer-­
ity of a heparin-binding peptide (RKRLQVQLSIRT) based potentiometric nano-graphene/ionic liquid/­
252 P. S. Agogo-Mawuli and D. P. Siderovski

carbon paste electrode for the determination of 77. Rochat S, Grote Z, Severin K. Ruthenium-based
losartan. J Mol Liq. 2015;212:96–102. metallacrown complexes for the selective detection
64. Beitollahi H, Yoonesfar R. Sensitive detection of sul- of lithium ions in water and in serum by fluorescence
fasalazine at a carbon paste electrode modified with spectroscopy. Org Biomol Chem. 2009;7(6):1147–53.
NiO/CNT nanocomposite and ionic liquid in phar- 78. Mattson RH. Tricyclic anticonvulsants: efficacy in
maceutical and biological samples. Inorg Nano-Met clinical trials. Epilepsy Behav. 2002;3(3):S9–S13.
Chem. 2017;47(10):1441–8. 79. Mattson RH. Efficacy and adverse effects of estab-
65. Kim S-G, et al. A study on the chemical stability and lished and new antiepileptic drugs. Epilepsia.
electrode performance of modified NiO cathodes 1995;36:S13–26.
for molten carbonate fuel cells. Electrochim Acta. 80. Dhanalakshmi N, Priya T, Thinakaran N. Highly elec-
2004;49(19):3081–9. troactive Ce-ZnO/rGO nanocomposite: ultra-sensitive
66. Dong S, et al. Direct electrochemistry and electro- electrochemical sensing platform for carbamazepine
catalysis of hemoglobin in composite film based on determination. J Electroanal Chem. 2018;826:150–6.
ionic liquid and NiO microspheres with different mor- 81. Oyarzún DP, et al. Electrochemical synthesis, opti-
phologies. Biosens Bioelectron. 2011;26(10):4082–7. cal properties and morphological characterization of
67. Salimi A, et al. Immobilization of glucose oxidase on ZnO/Poly (N-PhMI-co-HEMA) nanocomposite. J
electrodeposited nickel oxide nanoparticles: direct Electroanal Chem. 2017;799:358–62.
electron transfer and electrocatalytic activity. Biosens 82. Zou Y, et al. MOF-derived porous ZnO/MWCNTs
Bioelectron. 2007;22(12):3146–53. nanocomposite as anode materials for lithium-ion bat-
68. Ruddarraju RR, et al. Design, synthesis, anticancer, teries. J Electroanal Chem. 2017;788:184–91.
antimicrobial activities and molecular docking studies 83. Dar G, et al. Ce-doped ZnO nanorods for the detection
of theophylline containing acetylenes and theophyl- of hazardous chemical. Sensors Actuators B Chem.
line containing 1, 2, 3-triazoles with variant nucleo- 2012;173:72–8.
side derivatives. Eur J Med Chem. 2016;123:379–96. 84. Lorenzo P, et al. Farmacología Básica y Clínica (18ª
69. Ganjali MR, et al. Highly sensitive determination of edición). Madrid: Editorial Médica Panamericana;
theophylline based on graphene quantum dots modi- 2008.
fied electrode. Int J Electrochem Sci. 2018;13(3):2448. 85. Kwan P, Brodie MJ. Drug treatment of epilepsy: when
70. Farghali R, Ahmed RA. Gold nanoparticles-modified does it fail and how to optimize its use? CNS Spectr.
screen-printed carbon electrode for voltammetric 2004;9(2):110–9.
determination of sildenafil citrate (Viagra) in pure 86. Leza J, et al. Farmacología Básica y Clínica (17va
form, biological and pharmaceutical formulations. Int Edición). Madrid: Editorial Médica Panamericana;
J Electrochem Sci. 2015;10(2):1494–505. 2005.
71. Ali TA, et al. New chemically modified screen-printed 87. Spiehler V, et al. Radioimmunoassay, enzyme
electrode for Co (II) determination in different water immunoassay, spectrophotometry, and gas-liquid
samples. Int J Electrochem Sci. 2014;9(4):1812–26. chromatography compared for determination of
72. Lou Y-F, et al. A universal aptasensing platform based phenobarbital and diphenylhydantoin. Clin Chem.
on cryonase-assisted signal amplification and gra- 1976;22(6):749–53.
phene oxide induced quenching of the fluorescence 88. Shariati R, et al. Application of coated green source
of labeled nucleic acid probes: application to the carbon dots with silica molecularly imprinted poly-
detection of theophylline and ATP. Microchim Acta. mers as a fluorescence probe for selective and sen-
2019;186(8):1–9. sitive determination of phenobarbital. Talanta.
73. Mandel SJ, Brent GA, Larsen PR. Levothyroxine 2019;194:143–9.
therapy in patients with thyroid disease. Ann Intern 89. Haupt K, Mosbach K. Molecularly imprinted poly-
Med. 1993;119(6):492–502. mers and their use in biomimetic sensors. Chem Rev.
74. Lotfi S, Veisi H. Synthesis and characterization of 2000;100(7):2495–504.
novel nanocomposite (MWCNTs/CC-SH/Au) and 90. Ensafi AA, Kazemifard N, Rezaei B. Development of
its use as a modifier for construction of a sensi- a selective prilocaine optical sensor based on molecu-
tive sensor for determination of low concentration larly imprinted shell on CdTe quantum dots. Sensors
of levothyroxine in real samples. Chem Phys Lett. Actuators B Chem. 2017;242:835–41.
2019;716:177–85. 91. Liu G, et al. In-situ hydrothermal synthesis of molec-
75. Baghayeri M, Amiri A, Farhadi S. Development of ularly imprinted polymers coated carbon dots for fluo-
non-enzymatic glucose sensor based on efficient load- rescent detection of bisphenol A. Sensors Actuators B
ing Ag nanoparticles on functionalized carbon nano- Chem. 2016;228:302–7.
tubes. Sensors Actuators B Chem. 2016;225:354–62. 92. Hou J, et al. Rapid microwave-assisted synthesis of
76. Huang K-J, et al. Electrochemical behavior and molecularly imprinted polymers on carbon quantum
voltammetric determination of norfloxacin at glassy dots for fluorescent sensing of tetracycline in milk.
carbon electrode modified with multi walled carbon Talanta. 2016;146:34–40.
nanotubes/Nafion. Colloids Surf B: Biointerfaces. 93. Raoof JB, Baghayeri M, Ojani R. A high sensitive
2008;64(2):269–74. voltammetric sensor for qualitative and quantitative
14 Recent Advances in Nanosensors for Therapeutic Drug Monitoring (TDM) 253

determination of phenobarbital as an antiepileptic 95. Sorouraddin M-H, et al. A new fluorimetric method
drug in presence of acetaminophen. Colloids Surf B: for determination of valproic acid using TGA-capped
Biointerfaces. 2012;95:121–8. CdTe quantum dots as proton sensor. J Lumin.
94. Jiang C, et al. Preparation of the Pt nanoparticles 2014;145:253–8.
decorated poly (N-acetylaniline)/MWNTs nanocom- 96. Khoubnasabjafari M, Samadi A, Jouyban A. In-situ
posite and its electrocatalytic oxidation toward form- microscale spectrophotometric determination of
aldehyde. Electrochim Acta. 2009;54(3):1134–40. phenytoin by using branched gold nanoparticles.
Microchim Acta. 2019;186(7):1–7.
Organ Toxicity
by Immunosuppressive Drugs
15
in Solid Organ Transplantation

George J. Dugbartey and Alp Sener

Abstract inhibitor, a purine synthesis inhibitor, and glu-


cocorticoid. Other centers have also used dual
Solid organ transplantation is the preferred therapy comprising inhibitors of mammalian
therapeutic option that confers signifcant sur- target of rapamycin and belatacept to achieve
vival advantage on patients suffering from end- the goal of immunosuppression. However,
organ dysfunction and provides improved chronic immunosuppression is associated with
quality of life together with its cost- post-transplant complications including organ
effectiveness. During perioperative and post- toxicity, resulting in signifcant mortality of
transplant periods, organ transplant recipients transplant recipients and hindering long-term
receive immunosuppressive therapy, which success of organ transplantation. This chapter
reduces the high risk of allograft rejection, min- discusses organ toxicity induced by commonly
imizes infections, improves allograft quality, used immunosuppressive drugs in solid organ
and prolongs recipient survival. While there is transplantation and strategies to reduce the bur-
no optimal immunosuppressive protocol in den of immunosuppression after solid organ
solid organ transplantation across transplant transplantation.
centers, some centers have adopted a triple
combination therapy consisting of a calcineurin Keywords

Allograft rejection · Immunosuppression ·


G. J. Dugbartey (*)
Department of Pharmacology and Toxicology, School
Immunosuppressive drugs · Organ toxicity ·
of Pharmacy, College of Health Sciences, University Solid organ transplantation
of Ghana, Accra, Ghana
Department of Surgery, Division of Urology, London
Health Sciences Center, Western University, 15.1 Introduction
London, ON, Canada
e-mail: gjdugbartey@ug.edu.gh
15.1.1 Historical Account
A. Sener of Immunosuppressive Drugs
Department of Surgery, Division of Urology, London
Health Sciences Center, Western University,
in Clinical Organ Transplantation
London, ON, Canada
Department of Microbiology & Immunology,
Solid organ transplantation is a revolutionary
Schulich School of Medicine & Dentistry, University feld in modern medicine that has become the
of Western Ontario, London, ON, Canada treatment of choice for patients with end-stage
e-mail: alp.sener@lhsc.on.ca

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 255
S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_15
256 G. J. Dugbartey and A. Sener

organ failure. It improves quality of life of trans- renal isotransplantation) for allotransplantation
plant recipients and confers signifcant survival [2]. Although TBI achieved immunosuppression,
advantage together with its cost-effectiveness it also resulted in high patient mortality due to
and can rightly be considered as one of the medi- overwhelming infections arising from profound
cal miracles of the twentieth century. This chal- bone marrow aplasia [2]. By the end of the mid-
lenging life-saving intervention has shown twentieth century, it became obvious that TBI
remarkable growth and has also made great could not be used as an immunosuppressive
strides over the past few decades to the present therapy in organ transplant recipients.
era, illustrating the evolution of a complex and Following the withdrawal of TBI,
technical procedure from its preclinical origin in 6-mecaptopurine (6-MP), an anticancer drug
the mid-twentieth century to becoming a routine which successfully suppressed the immune sys-
clinical practice today, with continuous refne- tem and prolonged the survival of organ grafts in
ment including the introduction of immunosup- previous experimental transplantation in rabbits
pressive drugs to prevent allograft rejection. and dogs [3, 4], entered into human clinical trials
Against the background of failure, the frst suc- with a 40–50% 1-year success rate [5]. Later,
cessful kidney transplantation was performed in 6-MP was replaced by its less toxic derivative
1954 in the United States, in which the donor and azathioprine, which is equally as effective as
recipient were identical twins [1]; hence, the 6-MP. Corticosteroids were also introduced to
transplant recipient did not require immunosup- treat the unavoidable allograft rejection and also
pression therapy. This outstanding clinical out- served as an adjunct to azathioprine therapy for
come paved way for research into maintenance immunosuppression. Development
immunosuppression, which suggests that organ of corticosteroid-resistant allograft rejection
transplantation could be extended beyond the soon after transplantation led to the introduction
boundaries of genetic pairs. Unlike isotransplan- of anti-thymocyte globulin, the frst immunosup-
tation in which the organ donor and recipient are pressive agent to treat corticosteroid-resistant
genetically identical, thus requiring no immuno- acute allograft rejection, with about 70% 1-year
suppression as seen in identical twins, allotrans- allograft survival rate in several clinical trials
plantation (the most common form of [6–8]. The “azathioprine era” ended in the early
transplantation) requires immunosuppression 1980s due to high mortality of transplant recipi-
since the donor and recipient are genetically ents (as high as 40% at 1-year post-transplant).
unidentical. It is important to note that immuno- This led to the arrival of a new era in clinical
suppression is greatest during peri-operative and transplantation, with the introduction of another
early post-transplant periods to reduce the high immunosuppressive drug, cyclosporine – a calci-
risk of allograft rejection, minimize infections, neurin inhibitor, producing over 80% 1-year
improve graft quality, and prolong recipient sur- allograft survival rate characterized by signif-
vival due to cold preservation injury of the cantly reduced renal graft loss from irreversible
allograft and the sudden exposure of recipient’s rejection and markedly improved quality of
immune system to abundance of foreign anti- other highly immunogenic solid organ allografts
gens. Later in the post-transplant period, immu- such as heart, lungs, liver, and intestine [2].
nosuppression is then reduced (maintenance Subsequent decades of the twentieth century
immunosuppression) to an optimal level depend- after the “cyclosporine era” produced a signif-
ing on allograft function and tolerability. Thus, cant armamentarium of immunosuppressive
while maintenance immunosuppression prevents agents with different effcacy and safety. These
allograft rejection, it is also a pharmacological new drugs include tacrolimus, everolimus, siro-
strategy to avoid organ toxicity. Total body irra- limus, and mycophenolate mofetil. In addition,
diation (TBI), a radiotherapy to suppress the monoclonal antibodies such as basiliximab (anti-
patient’s immune system and prevent allograft CD25/IL-2R), rituximab (anti-CD20), alemtu-
rejection, was identifed (after the frst successful zumab (anti-CD52), and belatacept
15 Organ Toxicity by Immunosuppressive Drugs in Solid Organ Transplantation 257

(anti-CD80/86) directed against T and B lym- purine synthesis inhibitor (antimetabolite) such
phocytes – the primary mediators of alloimmune as azathioprine or mycophenolate mofetil, and
response and effectors of the rejection process – glucocorticoid such as prednisone, prednisolone,
were also developed and introduced into clinical or methylprednisolone, all of which interfere
practice to induce immunosuppression or treat with different steps in the immune response to
allograft rejection episodes. All these new immu- the allograft. Some transplant centers have also
nosuppressive drugs that followed the “cyclo- adopted a double combination therapy compris-
sporine era” produced over 90% 1-year allograft ing a calcineurin inhibitor and other immunosup-
survival rate, thus facilitating major improve- pressive agents, while other centers have also
ment compared to the early era of clinical organ used inhibitors of mammalian target of rapamy-
transplantation. cin (mTOR; proliferation inhibitors) such as siro-
In the face of these great strides in clinical limus and everolimus, with belatacept. The
organ transplantation is the growing concern of classifcation of these drugs is based on their
long-term post-transplant complications, which mechanisms of action. Although induction and
hinders the long-term success of organ trans- maintenance immunosuppression therapy
plantation. Post-transplant complications reduces the rate of acute allograft rejection and
including organ toxicity due to prolonged expo- improves allograft half-life and patient survival,
sure and chronic immunosuppression are asso- it also has long-term adverse effect on virtually
ciated with signifcant mortality of transplant all organ systems. As illustrated in Fig. 15.1,
recipients even though the combination of chronic immunosuppression following organ
immunosuppressive drugs increases the effcacy transplantation has resulted in increasing evi-
of immunosuppression and allows maintenance dence of a number of post-transplant long-term
immunosuppression at reduced doses [9–12]. complications such as toxicity of organ systems,
This suggests the need for further studies to hypertension, dyslipidemia, opportunistic infec-
expand our understanding of the human immune tions, malignancies, and diabetes, all of which
system to develop newer and more effcient contribute to high post-transplant mortality
drugs that target newly discovered mechanisms [9–15].
and pathways of immune response to provide
immunotolerance without the burden of organ
toxicity and other side effects associated with 15.2.1 Immunosuppressant-Induced
current regimens. This chapter discusses organ Nephrotoxicity
toxicity induced by commonly used immuno-
suppressive drugs in solid organ transplantation Among the effects of immunosuppressive drugs
and strategies to reduce the burden of immuno- on organ systems after transplantation is nephro-
suppression after solid organ transplantation. toxicity, one of the most common and signifcant
complications which eventually leads to renal
graft loss in transplant recipients [16]. While
15.2 Immunosuppressant- calcineurin inhibitors such as cyclosporine and
Induced Organ Toxicity After tacrolimus are the main class of immunosuppres-
Solid Organ Transplantation sive agents used across the globe as part of immu-
nosuppressive regimens during peri- and
Optimal immunosuppressive protocol in solid post-transplant periods, they are major contribu-
organ transplantation varies widely across trans- tors to the development of chronic kidney disease
plant centers. As summarized in Table 15.1, the and renal failure after transplantation, with an
most frequently adopted regimen is a triple com- increased risk of mortality of transplant recipi-
bination therapy consisting of a calcineurin ents [17]. Long-term use of calcineurin inhibi-
inhibitor such as cyclosporine or tacrolimus, a tors, for example, has been reported to cause
258 G. J. Dugbartey and A. Sener

Table 15.1 Commonly used immunosuppressive agents in solid organ transplantation


Classifcation of
immunosuppressant Example Indication
Calcineurin inhibitors Cyclosporine, Voclosporine, Tacrolimus Maintenance of
immunosuppression
Purine synthesis inhibitors Azathioprine, Mycophenolate mofetil Maintenance of
(antimetabolite) Mycophenolate sodium immunosuppression
mTOR inhibitors (proliferation Sirolimus, Everolimus Maintenance of
inhibitors) immunosuppression
Corticosteroids Prednisone, Prednisolone, Induction of immunosuppression
Methylprednisolone
Antibodies Basiliximab, Daclizumab, Muromonab, Induction of immunosuppression
Alemtuzumab, Thymoglobulin

Fig. 15.1 Summary of clinical presentation of organ toxicity induced by immunosuppression therapy after solid organ
transplantation

acute allograft nephropathy. This includes There is also evidence of chronic allograft
impaired production of vasodilating mediators nephropathy induced by calcineurin inhibitors,
such as nitric oxide and prostanoid, increased which include irreversible renal pathological
sympathetic activation, and enhanced production changes such as arteriolopathy,
of vasoconstrictive mediators such as endothelin, glomerulosclerosis, damaged endothelium and
thromboxane, angiotensin II, and platelet- smooth muscle of afferent arterioles, tubular
activating factor [9, 18–25]. Such hemodynamic atrophy, and tubulointerstitial fbrosis character-
changes ultimately increase vascular resistance at ized by increased expression of transforming
the level of afferent arterioles to a greater extent growth factor-beta (TGF-β; a well-defned cen-
than efferent arterioles and altogether negatively tral mediator of renal fbrosis), which appear to
impact renal hemodynamics, resulting in vaso- be dependent on angiotensin II but independent
constriction and consequently reduced renal of the acute renal hemodynamic changes [26, 27]
blood fow and glomerular fltration rate in pre- (Fig. 15.1). At the molecular level, treatment
clinical and clinical studies [9, 18–25] (Fig. 15.1). with calcineurin inhibitors has been reported to
15 Organ Toxicity by Immunosuppressive Drugs in Solid Organ Transplantation 259

increase renal production of reactive oxygen spe- 1996 to 2005 involving 25,045 kidney transplant
cies (ROS; natural by-product of cellular oxida- recipients with well-functioning renal allograft
tion metabolism and destructive tissue mediator) showed that withdrawal of calcineurin inhibitors
with reduced levels of antioxidants (e.g., gluta- or mycophenolate mofetil from the immunosup-
thione). This observation correlated with pression regimens or reduction of their doses
increased expression of pro-apoptotic genes (e.g., below certain thresholds during early post-
p53, Bax and Fas-L) and decreased expression of transplant period was associated with signifcant
anti-apoptotic genes (e.g., Bcl-2), resulting in risk of allograft loss with a hazard ratio of 1.52–
apoptosis of renal tubular epithelial cells [28, 29]. 1.73 relative to continuing treatment [39]. These
Hauser et al. [30] also provided additional evi- clinical observations suggest that administration
dence of the nephrotoxic effect of calcineurin of calcineurin inhibitors is crucial especially dur-
inhibitors in which they showed in a clinical ing early post-transplant period and that their
study that renal allografts with increased expres- withdrawal may be considered a double-edge
sion of P-glycoprotein (a transmembrane drug sword. Thus, acute allograft rejection and
transporter whose function determines drug con- allograft loss will remain a major concern when
centration in plasma and tissues) are less vulner- considering withdrawal or discontinuation of cal-
able to immunosuppressant-induced cineurin inhibitors. It further suggests the need to
nephrotoxicity, suggesting that P-glycoprotein is develop pharmacodynamic monitoring tools and
a major determinant of calcineurin inhibitor- pharmacogenetic screening strategies to optimize
induced nephrotoxicity. individualization of immunosuppression therapy
Considering the nephrotoxic effect of calci- preferably during early post-transplant period
neurin inhibitors discussed above, there are a based on organ-specifc and patient-specifc fac-
number of suggestions including a dose reduc- tors such as age, race, comorbidities, genetic
tion or withdrawal of calcineurin inhibitors or polymorphism, immunologic risk, concomitant
their combination with signifcantly less toxic and previous pharmacotherapy, and overall
immunosuppressive agents such as sirolimus and immunosuppression burden. In summary,
everolimus or with non-nephrotoxic immunosup- immunosuppressant-induced nephrotoxicity is a
pressants such as mycophenolate mofetil as part major post-transplant complication, which places
of the immunosuppression regimen to reduce the a considerable metabolic burden on the renal
risk of post-transplant nephrotoxicity. While allograft and contributes in part to post-transplant
there is currently no general consensus on the renal dysfunction. Therefore, new agents will be
choice of combined immunosuppression therapy needed as an adjuvant to or replacement of cur-
to avoid allograft rejection and minimize nephro- rent calcineurin inhibitor to provide alternative
toxicity, such immunosuppressant combinations mechanisms of action and reduce
resulted in enhanced calcineurin inhibitor- nephrotoxicity.
induced nephrotoxicity in phase III clinical stud-
ies [31, 32]. This observation was attributed to
changes in renal tissue distribution of calcineurin 15.2.2 Immunosuppressant-Induced
inhibitors and inhibition of glycolytic pathway Neurotoxicity
by the anti-proliferation immunosuppressive
drugs [33]. Also, withdrawal of calcineurin Immunosuppressant-induced neurotoxicity is one
inhibitors during early post-transplant period in of the most signifcant post-transplant complica-
an attempt to start de novo transplant recipients tions with over 59% prevalence in organ transplant
on so-called “calcineurin inhibitor-free” immu- recipients. As shown in Fig. 15.1, chronic immu-
nosuppressive protocol resulted in signifcant nosuppression with calcineurin inhibitors, for
increase in acute and chronic allograft rejection example, is associated with hallucinations, sei-
[34–38]. In fact, a 9-year clinical study from zures, paresthesias, delusions, dizziness, hypore-
260 G. J. Dugbartey and A. Sener

sponsiveness, headaches, depression, stroke-like and increased the risk of developing metabolic,
episodes, progressive multifocal leukoencepha- neurodegenerative, and neuropsychiatric disor-
lopathy, and cortical blindness in organ transplant ders [52, 53]. Furthermore, anti-proliferation
recipients [40, 41]. Several mechanisms have agents (mTOR inhibitors) such as sirolimus have
been implicated in calcineurin inhibitor-induced been reported to increase ROS production via
neurotoxicity. While calcineurin inhibitors do not interaction with calcineurin inhibitors, leading to
readily cross the blood-brain barrier (BBB), they oxidative stress and its negative metabolic effects
alter its permeability by inducing apoptosis of in the cells of the CNS [54] as is also observed in
brain capillary endothelial cells and inhibiting nephrotoxicity. For this reason, further clinical
the function of P-glycoprotein and thereby and preclinical studies are needed to better under-
enhancing their diffusion across the BBB into the stand the pathogenesis and mechanisms as well
central nervous system (CNS) [42, 43]. In the as relationship between immunosuppressant-
CNS, calcineurin inhibitors selectively alter induced neurotoxicity and nephrotoxicity to
mitochondrial function in glial cells and oligo- allow proper clinical decisions to be taken on the
dendrocytes, resulting in increased mitochondrial choice of immunosuppression therapy during
ROS production and thereby inducing oxidative peri- and post-transplant periods to manage the
stress and ultimately damaging these cells that delicate balance between immunosuppression,
provide structural, trophic, and metabolic support neurotoxicity, and nephrotoxicity. This will
to neurons in the CNS [44–46]. Indeed, mito- require a close collaboration between neurolo-
chondrial dysfunction was reported in the brains gists and nephrologists.
of rats following administration of calcineurin
inhibitors such as cyclosporine, resulting in
reduced ATP production and increased ROS for- 15.2.3 Immunosuppressant-Induced
mation [41, 47]. Arnold et al. [48] also reported Cardiotoxicity
additional mechanism of calcineurin inhibitor-
induced neurotoxicity by altering excitability Cardiotoxic effects of immunosuppressive drugs
properties, causing neuronal membrane depolar- are less common, but serious adverse effects have
ization via modulation of receptors of excitatory been observed in both adult and pediatric trans-
and inhibitory neurotransmitters [49, 50]. As plant recipients with no previous risk factors for
with nephrotoxicity, calcineurin inhibitors also cardiac disease prior to organ transplantation.
activate vasoconstriction system and increase These cardiotoxic effects have manifested as
sympathetic activity, leading to inhibition of hypertrophic obstructive cardiomyopathy, dilated
nitric oxide-mediated and other vasodilation cardiomyopathy, and severe concentric left ven-
pathways. The consequent disruption of the tricular hypertrophy with congestive heart failure
endothelial function causes systemic hyperten- [55–59] (Fig. 15.1). Interestingly, most of the
sion, local ischemia, and cerebral edema [51]. known cases of immunosuppressant-induced
Besides calcineurin inhibitors, antimetabolites cardiotoxicity are associated with tacrolimus
(purine synthesis inhibitors) such as azathioprine therapy, while others are isolated with other
and mycophenolate mofetil also deplete purine immunosuppressants. In fve pediatric patients
stores in neuronal cells and inhibit DNA and without any history of cardiac diseases prior to
RNA synthesis and lymphocyte proliferation. In bowel and liver transplantation, Atkinson et al.
addition, administration of high doses of gluco- [55] observed various degrees of left ventricular
corticoids or prolonged administration at low hypertrophic obstructive cardiomyopathy and
doses has been reported to negatively affect the heart failure within 2 months of starting tacrolimus
hypothalamic-pituitary-adrenal axis (the main therapy after transplantation. Conversion to
physiological system that mediates the body’s cyclosporine resulted in improvement in their
stress response) and induced neuronal structural heart conditions. Several subsequent studies also
remodeling with synaptic loss, glial malfunction reported tacrolimus-related ventricular hypertrophy
15 Organ Toxicity by Immunosuppressive Drugs in Solid Organ Transplantation 261

characterized by thickening of left ventricular progression by sirolimus was shown in animal


wall and interventricular septum in adult and models to be due to inhibition of transforming
pediatric transplant recipients, which resolved growth factor-β pathway and fbrogenic activa-
after tacrolimus was discontinued [56–62]. tion of myofbroblasts [72]. Roberts et al. [73]
However, a unique case of tacrolimus-associated also observed cardiomegaly with preferential
hypertrophy was reported by McLeod et al. [63] septal hypertrophy at autopsy in pediatric and
in which they observed rapid progression from adult patients who received tacrolimus and cyclo-
cardiac hypertrophy to dilated cardiomyopathy sporine after liver transplantation and had no risk
with severely reduced systolic function in adult factors to cardiac diseases prior to transplanta-
liver transplant recipients shortly after post- tion. In addition, chronic immunosuppression
transplant initiation of tacrolimus therapy. with prednisone and cyclosporine caused mid-
Interestingly, the change in cardiac function apical hypertrophy, a phenotype of hypertrophic
remained the same even after tacrolimus with- cardiomyopathy in an adult heart transplant
drawal. The authors surmised that the progres- recipient [74]. Overall, the incidence of
sion to dilated cardiomyopathy could be due to immunosuppressant-induced cardiotoxicity,
tacrolimus, considering the short time period though less common, draws attention to the need
between initiation of tacrolimus-based immuno- for careful evaluation of transplant patients on
suppression and the sudden changes in cardiac these drugs, particularly tacrolimus. As only a
function. This conclusion, however, warrants few cases of cardiotoxicity by other immunosup-
investigation. The mechanism underlying pressive agents have been reported at single cen-
tacrolimus-induced hypertrophic cardiomyopa- ters, a larger dataset based on consistent
thy is unclear. Proposed mechanisms may be multicenter investigations is needed.
related to inappropriate intracellular calcium
handling. There are studies showing that FK506
binding protein (FKBP) in cardiac (and skeletal) 15.2.4 Immunosuppressant-Induced
muscle closes cardiac ryanodine receptor, block- Pulmonary Toxicity
ing calcium release from its storage in the sarco-
plasmic reticulum [64, 65]. Tacrolimus inhibits Immunosuppressant-induced pulmonary toxicity
the effect of FKBP by enhancing the opening of is one of the major causes of post-transplant com-
the ryanodine channels and subsequent calcium plications, accounting for 20% incidence of
release into the sarcoplasm of cardiomyocyte, allograft failure and mortality of transplant recip-
which induces cardiac hypertrophy [66]. Another ients [75–77]. It usually presents in the form of
study also proposed coronary artery arteritis and fatigue, fever, lymphocytic interstitial pneumoni-
tacrolimus-induced hypertension from sympa- tis, bronchiolitis obliterans, pulmonary alveolar
thetic activation [67]. proteinosis, organizing pneumonia,
Apart from tacrolimus, sirolimus and MMF bronchiectasis, dyspnea, nonproductive cough,
were recently reported to cause recurrent pericar- hemoptysis, alveolar hemorrhage, and progres-
dial effusion in a cohort of heart transplant sive pulmonary fbrosis with extensive bilateral
patients and required their discontinuation [68]. patchy or diffuse alveolo-interstitial infltrates as
However, another study reported that conversion revealed by imaging tests such as chest radio-
from calcineurin inhibitors to sirolimus improved graph and high-resolution computerized tomog-
diastolic function and flling pressure, attenuated raphy [78–82] (Fig. 15.1). There are published
myocardial fbrosis progression, and reduced left reports of isolated cases showing mTOR inhibi-
ventricular mass of cardiac allografts [69–71]. tors such as sirolimus and everolimus inducing
Considering these contradictory outcomes pulmonary toxicity in the absence of infectious
involving sirolimus, further studies are indicated causes (after bronchoscopy with bronchoalveolar
to clarify its mechanism of interaction with lavage) or other pulmonary diseases in recipients
MMF. The attenuation of myocardial fbrosis of deceased donor kidneys after conversion from
262 G. J. Dugbartey and A. Sener

calcineurin inhibitors, whereby dose reduction or recipients, often presenting as diarrhea, gastritis,
drug withdrawal and replacement with cortico- anorexia, ulcerations, gastric and duodenal ero-
steroid therapy resulted in clinical and radiologic sions, nausea, vomiting, gastroduodenitis, dys-
improvement in both adult and pediatric trans- pepsia, heartburn, necrosis, duodenal villus
plant recipients [83–88]. While the exact mecha- atrophy, and Crohn’s-like enterocolitis [99, 100]
nism of pulmonary toxicity by mTOR pathway (Fig. 15.1). Non-immunosuppressant co-
inhibition is unknown, some studies suggest a medications (e.g., antibiotics, diuretics) and con-
possible pathogenic mechanism including an current diseases (e.g., diabetes) also play a
idiosyncratic cell-mediated autoimmune contributing role in the pathogenesis and pro-
response after exposure of cryptic antigens or a T gression of GI toxicity [101, 102]. The GI tract is
cell-mediated delayed type of hypersensitivity involved in the metabolism of almost all immu-
reaction [83, 89]. nosuppressive agents; hence, it is exposed to
Besides mTOR inhibitors, tacrolimus and immunosuppressants and their metabolites. This
MMF have also been reported to cause dyspnea, makes the GI tract highly vulnerable to all forms
hemoptysis, and extensive pulmonary fbrosis at of GI disorders and complications, thereby affect-
1-month post-deceased donor kidney transplanta- ing the quality of life of organ transplant recipi-
tion, which was temporarily improved after dis- ents, which could lead to low patient compliance
continuation of the drugs. However, resumption and in turn increase the risk of allograft loss. In a
of MMF resulted in deterioration of respiratory meta-analysis of randomized controlled trials and
function and subsequent death from hypoxic a cross-sectional study with 543 kidney trans-
respiratory failure [90], suggesting that MMF, if plant recipients who underwent upper endoscopy
necessary, must be used with utmost care in due to GI complications, the incidence of erosive
transplant recipients with underlying pulmonary lesions in the upper GI tract increased when
conditions with poor baseline pulmonary func- tacrolimus was added to the immunosuppressive
tion. Other studies in adult and pediatric trans- regimens [103, 104]. Tacrolimus and cyclospo-
plant recipients also showed improvement in rine were also associated with acute pancreatitis
pulmonary conditions when MMF therapy was and fatality of kidney and heart transplant recipi-
stopped and replaced with azathioprine and corti- ents 2 years after transplantation [105–107]. In
costeroid therapy [91–98]. In summary, dose addition, administration of these calcineurin
reduction and conversion from one immunosup- inhibitors and sirolimus (an mTOR inhibitor)
pressive agent to another should be considered resulted in chronic severe diarrhea and constipa-
when unexplained pulmonary complaints develop tion in kidney transplant recipients [103, 108,
in patients post-transplant. Given that the afore- 109], which is attributed to altered expression of
mentioned examples represent single-center Na+/H+ exchanger 3 in apical membranes of the
experiences, a larger dataset based on consistent intestines and lipid malabsorption [110, 111]
multicenter trials would provide a more compre- with gastroduodenal ulcer bleeding in kidney and
hensive understanding and useful information liver transplant recipients who were on sirolimus
regarding the incidence of immunosuppressant- therapy [112, 113]. Interestingly, these symp-
induced pulmonary toxicity and its management. toms were reversed following sirolimus with-
drawal [113].
Antimetabolites (purine synthesis inhibitors)
15.2.5 Immunosuppressant-Induced such as mycophenolate and azathioprine have
Gastrointestinal Toxicity also been implicated in GI toxicity following
their administration in organ transplant recipi-
Gastrointestinal (GI) toxicity commonly occurs ents. Mycophenolate comes as mycophenolate
after solid organ transplantation and is one of the mofetil (MMF) and enteric-coated mycopheno-
major causes of allograft loss, morbidity, and late sodium (EC-MPS; to reduce incidence of
mortality of immunosuppressed organ transplant GI adverse effect), which are hydrolyzed to the
15 Organ Toxicity by Immunosuppressive Drugs in Solid Organ Transplantation 263

active drug mycophenolic acid (MFA) by ester- 131]. Azathioprine administration also resulted
ases in the stomach, small intestine, liver, blood, in acute pancreatitis as observed in tacrolimus
and other tissues after oral administration. In and cyclosporine administration [132]. Besides
three clinical trials involving kidney transplant MMF and azathioprine, a meta-analysis also
recipients on either MMF-cyclosporine or revealed a signifcant increase in the risk of GI
MMF-tacrolimus therapy, the incidence of hemorrhage or perforation in corticosteroid-
severe diarrhea was reported during the frst treated patients [133]. Corticosteroid therapy
post-transplant year [114–117], with the occur- also caused acute pancreatitis and colonic mala-
rence of duodenal villus atrophy and erosive koplakia, a chronic granulomatous disease in a
infammation in the small intestine, in which the kidney transplant recipient [134, 135]. In two
patients responded following MMF dose reduc- phase III clinical trials, diarrhea, constipation,
tion or withdrawal [118, 119]. There are also nausea, and ulcerative colitis were observed in
reports of colonic and oral ulcerations and kidney transplant recipients who were on belata-
enterocolitis similar to Crohn’s disease in kid- cept-based immunosuppression, which was
ney and liver transplant recipients, which were comparable to those placed on cyclosporine-
linked to direct GI mucosal injury by the active based immunosuppression therapy [136–139].
drug, MPA, and were resolved following dis- As GI toxicity is commonly associated with
continuation of MMF [120–122]. The mecha- chronic immunosuppression following organ
nism of action of MPA is linked to its transplantation, it is important to develop an
anti-proliferative properties, by selectively effective approach to reduce these adverse events
inhibiting inosine monophosphate dehydroge- to prevent allograft rejection and subsequent loss.
nase, the main enzyme in de novo pathway of Also, a systemic and individualized approach
synthesis purine in GI epithelial cells for growth should be developed to optimize the management
and replication [123]. This suggests that MPA of transplant recipients’ GI toxicity, which over-
could inhibit GI epithelial cell growth and repli- laps with non-immunosuppressant co-
cation and consequently disrupt fuid absorption medications and comorbidities. In the absence of
in the GI tract, resulting in diarrhea. The enteric- critical illness, a watchful waiting approach
coated form of mycophenolate, EC-MPS, which should be considered to determine whether the
was developed as an alternative to MMF with GI disorder will spontaneously resolve without
the goal of reducing GI adverse events, indeed pharmacological intervention, as was observed in
resulted in improved GI tolerability after con- 65% of 130 kidney transplant recipients with
version from MMF to EC-MPS in kidney trans- diarrhea [128]. In cases where a transplant
plant recipients [124, 125]. This observation recipient develops severe or prolonged GI com-
suggests that indeed EC-MPS could serve as a plications requiring pharmacological treatment,
useful alternative in transplant recipients who the underlying cause should be determined.
are intolerant to MMF. However, comparative Moreover, once a GI complication occurs at a
studies of EC-MPS with equipotent dose of suffcient post-transplant period such that any
MMF showed no difference in GI adverse event possible transplantation-induced adverse effects
profle in other kidney transplant recipients can be eliminated, other possible etiologies such
[126–128]. This contradictory outcome war- as non-iatrogenic causes should be considered
rants further investigations. As with MMF, aza- before altering the patient’s immunosuppressive
thioprine administration also led to severe therapy. In the absence of non-iatrogenic etiolo-
persistent diarrhea with severe small-bowel vil- gies, the patients’ immunosuppressive regimen
lus atrophy, gastroenteritis, and malabsorption should be modifed in a controlled manner with
in kidney transplant recipients, which were careful evaluation of each component of the
reversed upon discontinuation of the drug [129– regimen.
264 G. J. Dugbartey and A. Sener

15.2.6 Immunosuppressant-Induced bile duct stones, and sledge formation resulting


Hepatotoxicity from reduced bile acid secretion in a subset of
kidney transplant recipients [153–155]. Animal
Following organ transplantation, transplant studies also revealed sinusoidal dilatation and
recipients often have more aggressive liver dis- congestion, infltration, hydropic degeneration,
eases compared to their non-transplant counter- and loss of glycogen storage in the liver after
parts. For example, 20% of transplant recipients cyclosporine treatment [148]. Mechanistically,
with hepatitis C (HCV) disease recurrence prog- cyclosporine decreased the levels and activities
ress to cirrhosis within 5 years of liver transplan- of hepatic antioxidants such as glutathione, cata-
tation [140]. Immunosuppressants and other lase, and superoxide dismutase while increasing
concomitant drugs used by transplant recipients hepatic ROS production with increased liver
often cause hepatotoxicity characterized by enzymes and total bilirubin levels [148]. Also,
increased levels of liver enzymes and conjugated tacrolimus-induced hepatotoxicity characterized
hyperbilirubinemia, with histological patterns by hyperbilirubinemia, increased levels of liver
such as cholestatic hepatitis, sinusoidal obstruc- enzymes, centrilobular cholestasis, hepatocellu-
tion, granulomatous infammation, macro- and/or lar necrosis, and cholestatic jaundice was reported
micro-vesicular steatosis with or without steato- in previous studies in kidney transplant recipients
hepatitis, hepatocellular necrosis ranging from after all other possible etiologies of hepatic dys-
single-cell dropout to broad necrosis, veno- function including viral hepatitis were ruled out.
occlusive disease, or a combination of any of However, dose reduction or conversion to other
these injury patterns [141–151] (Fig. 15.1). Dual immunosuppressive drugs such as prednisolone
therapy with cyclosporine and MMF or tacroli- resulted in complete resolution of the liver condi-
mus with MMF signifcantly increased hepatic tion [147, 152, 156–158]. Although the exact
apoptotic markers and contributed to infamma- molecular mechanism underlying tacrolimus-
tion and the development of liver graft injury induced hepatotoxicity is unknown, a preclinical
after liver transplantation [149]. The authors sup- study in rats showed inhibition of biliary excre-
ported their observation with results from their tion of glutathione and bicarbonate following
in vitro study in which they treated primary tacrolimus administration [159].
mouse hepatocytes with various combinations of Contrary to the hepatoprotection by sirolimus
cyclosporine, tacrolimus, and MMF and observed and MMF in transplant recipients [149, 152],
signifcant reduction in hepatocyte viability and MMF administration in a subset of liver trans-
increased expression of hepatic apoptotic mark- plant recipients induced hepatotoxicity as
ers such as cleaved caspases 3 and PARP. However, evidenced by hepatic mitochondrial stress and
sirolimus/MMF combination prevented apopto- abnormalities similar to those observed in pri-
sis and enhanced hepatocyte viability [149], mary and secondary mitochondrial abnormalities
which also confrms a previous study in which [151]. This observation was supported by an
sirolimus/MMF combination reversed hepatotox- in vitro study by the same authors in which MMF
icity caused by tacrolimus and cyclosporine at induced various stress changes in hepatic mito-
17 weeks after bilateral lung transplant [152]. chondria of mice and markedly increased mito-
This suggests that withdrawal of calcineurin chondrial number, size, and number of lipid
inhibitors from the immunosuppression regimens droplets per cell compared to untreated mice
could protect against hepatotoxicity in transplant [151]. However, reduction or withdrawal of
recipients. MMF resulted in improvement of liver function.
It is worth noting that calcineurin inhibitors Mycophenolate sodium, another form of MMF
have been previously implicated in the develop- that has been proven to be as safe and effective as
ment of hepatotoxicity after organ transplanta- MMF [126, 127], also induced centrilobular cho-
tion. Cyclosporine, for example, was reported to lestatic hepatitis in a kidney transplant recipient,
have caused development of cholestatic hepatitis, which was completely reversed after withdrawal
15 Organ Toxicity by Immunosuppressive Drugs in Solid Organ Transplantation 265

of the drug [145]. Sirolimus administration also the goal of individualizing organ-specifc immu-
produced sinusoidal congestion with elevated nosuppression and limiting the impact of chronic
levels of liver enzymes in liver and kidney trans- immunosuppression should be considered at all
plant recipients after exclusion of possible etiolo- transplant centers. This suggests development of
gies. A quick normalization of liver function was tools such as peripheral and intra-graft expres-
observed after sirolimus was discontinued [143, sion markers of immune activation as tools to
144]. Everolimus, another immunosuppressant in carefully guide patient selection and closely
the class of mTOR inhibitors with sirolimus, also monitor drug minimization trials including
increased the levels of liver enzymes within a allograft function. Also, there is the need to
week of therapy in a liver transplant recipient, develop newer and more effcient drugs that tar-
with corresponding histopathological changes, get novel mechanisms and pathways of both cel-
notably the presence of eosinophils, rare acido- lular and humoral mechanisms of adaptive
phil bodies, and focal mild portal infammation immune response including mechanisms related
after other possible etiologies of liver dysfunc- to ischemia-reperfusion injury in order to provide
tion including concomitant medications have immunotolerance without the burden of organ
been ruled out [150]. Finally, azathioprine use toxicity and other complications induced by the
post-transplant has also been linked to elevated currently used immunosuppressive regimens. At
levels of liver enzymes and induced hepatotoxic- the same time, these proposed drugs should
ity characterized by jaundice, chronic hepatitis, achieve acceptable graft rejection rate and pro-
chronic active hepatitis, chronic persistent hepa- long survival of organ transplant recipients. In
titis, and cirrhosis in kidney transplant recipients, addition, while there are reports of rare cases of
which were enhanced by hepatitis B virus or immunotolerance using existing drugs (with
hepatitis C virus. However, azathioprine with- inexplicable underlying mechanisms), reliable
drawal normalized the liver condition and levels methods and assays should be developed to iden-
of liver enzymes [141, 142, 146]. Taken together, tify organ transplant recipients who can be
immunosuppressant-induced hepatotoxicity is a weaned of immunosuppression since the current
common complication in organ transplant recipi- clinical practice requires that organ transplant
ents. Therefore, dose reduction or early identif- recipients are bound to lifelong immunosuppres-
cation and withdrawal of a suspected sion. Furthermore, high-throughput “omic” tech-
immunosuppressive agent from the treatment niques such as genomics, proteomics, and
regimen are important to avoid progression or metabolomics should be considered in identify-
deterioration of the liver condition. Thus, close ing allograft injury-specifc mechanisms in order
monitoring of liver tests in organ transplant recip- to develop biomarkers for acute and chronic
ients and early evaluation of unexplained persis- rejection as well as tolerance. Finally, future
tent liver function test abnormalities using biopsy investigations should consider polymorphism
and possibly electron microscopic examination studies involving gene interaction with immuno-
should be requested. suppressive agents as an additional tool towards
the management of organ transplant recipients. In
conclusion, individualization of immunosuppres-
15.3 Reducing the Burden sion, development of novel drugs to provide
of Immunosuppression After immunotolerance without organ toxicity, identif-
Solid Organ Transplantation cation of biomarkers of allograft rejection, and
close monitoring of drug minimization trials may
As discussed in the above section, minimization prevent immunosuppressant-induced organ tox-
of immunosuppressive drugs in the treatment icity and facilitate lasting tolerance of the human
algorithm following organ transplantation with immune system in the future.
266 G. J. Dugbartey and A. Sener

References pathophysiology, incidence and management. Drug


Saf. 1999;21(6):471–88.
17. Ojo AO, Held PJ, Port FK, et al. Chronic renal fail-
1. Morris PJ. Transplantation – a medical miracle of
ure after transplantation of a nonrenal organ. N Engl
the 20th century. N Engl J Med. 2004;351:2678–80.
J Med. 2003;349(10):931–40.
2. Sayegh MH, Carpenter CB. Transplantation 50 years
18. Curtis JJ, Luke RG, Dubovsky E, Diethelm AG,
later – progress, challenges, and promises. N Engl J
Whelchel JD, Jones P. Cyclosporin in therapeutic
Med. 2004;351:2761–6.
doses increases renal allograft vascular resistance.
3. Schwartz R, Eisner A, Dameshek W. The effect of
Lancet. 1986;2(8505):477–9.
6-mercaptopurine on primary and secondary immune
19. Weir MR, Klassen DK, Shen SY, Sullivan D,
responses. J Clin Invest. 1959;38:1394–403.
Buddemeyer EU, Handwerger BS. Acute effects
4. Schwartz R, Dameshek W. Drug-induced immuno-
of intravenous cyclosporine on blood pressure,
logical tolerance. Nature. 1959;183:1682–3.
renal hemodynamics, and urine prostaglandin
5. Murray JE, Merrill JP, Harrison JH, Wilson RE,
production of healthy humans. Transplantation.
Dammin GJ. Prolonged survival of human-kidney
1990;49(1):41–7.
homografts by immunosuppressive drug therapy. N
20. Van Buren DH, Burke JF, Lewis RM. Renal function
Engl J Med. 1963;268:1315–23.
in patients receiving long-term cyclosporine therapy.
6. Hamilton D. Kidney transplantation: a history. In:
J Am Soc Nephrol. 1994;4(8 Suppl):S17–22.
Morris PJ, editor. Kidney transplantation: principles
21. Bennett WM. Clinical algorithms for cyclospo-
and practice. 5th ed. Philadelphia: W.B. Saunders;
rine management based on pathophysiologic
2001. p. 1–8.
constructs of nephrotoxicity. Transplant Proc.
7. Hamilton D. Reaching for the impossible: the
1994;26(5):2583–4.
quest for tissue replacement. In: Ginns LG, Cosimi
22. Andoh TF, Burdmann EA, Lindsley J, Houghton
AB, Morris PJ, editors. Transplantation. Boston:
DC, Bennett WM. Functional and structural charac-
Blackwell Science; 1999. p. 1–19.
teristics of experimental FK 506 nephrotoxicity. Clin
8. Tilney NL. Transplant: from myth to reality. New
Exp Pharmacol Physiol. 1995;22(9):646–54.
Haven, CN: Yale University Press; 2003.
23. Neuhaus P, Blumhardt G, Bechstein WO, et al.
9. Vaziri ND, Ni Z, Zhang YP, Ruzics EP, Maleki P,
Comparison of FK506- and cyclosporine-based
Ding Y. Depressed renal and vascular nitric oxide
immunosuppression in primary orthotopic liver
synthase expression in cyclosporine-induced hyper-
transplantation. A single center experience.
tension. Kidney Int. 1998;54(2):482–91.
Transplantation. 1995;59(1):31–40.
10. Mingos MA, Kane GC. Sirolimus-induced intersti-
24. Remuzzi G, Perico N. Cyclosporine-induced renal
tial pneumonitis in a renal transplant patient. Respir
dysfunction in experimental animals and humans.
Care. 2005;50(12):1659–61.
Kidney Int Suppl. 1995;52:S70–4.
11. Karolin A, Genitsch V, Sidler D. Calcineurin
25. Pirsch JD, Miller J, Deierhoi MH, Vincenti F, Filo
inhibitor toxicity in solid organ transplantation.
RS. A comparison of tacrolimus (FK506) and cyclo-
Pharmacology. 2021;106(7–8):347–55.
sporine for immunosuppression after cadaveric renal
12. Faravelli I, Velardo D, Podestà MA, Ponticelli
transplantation. FK506 Kidney Transplant Study
C. Immunosuppression-related neurological dis-
Group. Transplantation. 1997;63(7):977–83.
orders in kidney transplantation. J Nephrol.
26. Avdonin PV, Cottet-Maire F, Afanasjeva GV,
2021;34(2):539–55.
Loktionova SA, Lhote P, Ruegg UT. Cyclosporine
13. Krämer BK, Zülke C, Kammerl MC, Schmidt C,
A up-regulates angiotensin II receptors and calcium
Hengstenberg C, Fischereder M, Marienhagen
responses in human vascular smooth muscle cells.
J, European Tacrolimus vs. Cyclosporine
Kidney Int. 1999;55(6):2407–14.
Microemulsion Renal Transplantation Study Group.
27. Burdmann EA, Andoh TF, Yu L, Bennett
Cardiovascular risk factors and estimated risk for
WM. Cyclosporine nephrotoxicity. Semin Nephrol.
CAD in a randomized trial comparing calcineurin
2003;23(5):465–76.
inhibitors in renal transplantation. Am J Transplant.
28. Wang C, Salahudeen AK. Lipid peroxidation accom-
2003;3(8):982–7.
panies cyclosporine nephrotoxicity: effects of vita-
14. Heisel O, Heisel R, Balshaw R, Keown P. New onset
min E. Kidney Int. 1995;47(3):927–34.
diabetes mellitus in patients receiving calcineurin
29. Shihab FS, Bennett WM, Yi H, Andoh TF. Effect of
inhibitors: a systematic review and meta-analysis.
pirfenidone on apoptosis-regulatory genes in chronic
Am J Transplant. 2004;4(4):583–95.
cyclosporine nephrotoxicity. Transplantation.
15. Wimmer CD, Rentsch M, Crispin A, et al. The
2005;79(4):419–26.
janus face of immunosuppression – de novo malig-
30. Hauser IA, Schaeffeler E, Gauer S, et al. ABCB1
nancy after renal transplantation: the experience
genotype of the donor but not of the recipient is a
of the Transplantation Center Munich. Kidney Int.
major risk factor for cyclosporine-related nephrotox-
2007;71(12):1271–8.
icity after renal transplantation. J Am Soc Nephrol.
16. Olyaei AJ, de Mattos AM, Bennett
2005;16(5):1501–11.
WM. Immunosuppressant-induced nephropathy:
31. Kahan BD. Effcacy of sirolimus compared with aza-
thioprine for reduction of acute renal allograft rejec-
15 Organ Toxicity by Immunosuppressive Drugs in Solid Organ Transplantation 267

tion: a randomised multicentre study. The Rapamune 46. Jin KB, Choi HJ, et al. The production of reactive
US Study Group. Lancet. 2000;356(9225):194–202. oxygen species in tacrolimus-treated glial cells.
32. Kahan BD, Kaplan B, Lorber MI, Winkler M, Transplant Proc. 2008;40(8):2680–1.
Cambon N, Boger RS. RAD in de novo renal 47. Christians U, Gottschalk S, Miljus J, et al.
transplantation: comparison of three doses on Alterations in glucose metabolism by cyclosporine
the incidence and severity of acute rejection. in rat brain slices link to oxidative stress: inter-
Transplantation. 2001;71(10):1400–6. actions with mTOR inhibitors. Br J Pharmacol.
33. Klawitter J, Bendrick-Peart J, Rudolph B, et al. 2004;143(3):388–96.
Urine metabolites refect time-dependent effects of 48. Arnold R, Pussell BA, Pianta TJ, Lin CS, Kiernan
cyclosporine and sirolimus on rat kidney function. MC, Krishnan AV. Association between calcineurin
Chem Res Toxicol. 2009;22(1):118–28. inhibitor treatment and peripheral nerve dysfunc-
34. Keitel E, Bittar AE, Losekann A, et al. Elective tion in renal transplant recipients. Am J Transplant.
cyclosporine withdrawal in renal transplant recipi- 2013;13(9):2426–32.
ents maintained on triple therapy. Transplant Proc. 49. Sander M, Lyson T, Thomas GD, Victor
1992;24(6):2606–8. RG. Sympathetic neural mechanisms of
35. Kasiske BL, Heim-Duthoy K, Ma JZ. Elective cyclosporine-induced hypertension. Am J Hypertens.
cyclosporine withdrawal after renal transplantation. 1996;9(11):121S–38S.
A meta-analysis. JAMA. 1993;269(3):395–400. 50. Gold BG. FK506 and the role of immuno-
36. Groth CG, Bäckman L, Morales JM, et al. Sirolimus philins in nerve regeneration. Mol Neurobiol.
(rapamycin)-based therapy in human renal trans- 1997;15(3):285–306.
plantation: similar effcacy and different toxicity 51. Hoorn EJ, Walsh SB, McCormick JA, Zietse R,
compared with cyclosporine. Sirolimus European Unwin RJ, Ellison DH. Pathogenesis of calcineu-
Renal Transplant Study Group. Transplantation. rin inhibitor-induced hypertension. J Nephrol.
1999;67(7):1036–42. 2012;25(3):269–75.
37. Kreis H, Cisterne JM, Land W, et al. Sirolimus in 52. Sousa N, Almeida OF. Disconnection and reconnec-
association with mycophenolate mofetil induc- tion: the morphological basis of (mal)adaptation to
tion for the prevention of acute graft rejection stress. Trends Neurosci. 2012;35(12):742–51.
in renal allograft recipients. Transplantation. 53. Moisiadis VG, Matthews SG. Glucocorticoids
2000;69(7):1252–60. and fetal programming part 1: outcome. Nat Rev
38. Han WK, Bonventre JV. Biologic markers for the Endocrinol. 2014;10:391–402.
early detection of acute kidney injury. Curr Opin Crit 54. Klawitter J, Gottschalk S, Hainz C, Leibfritz D,
Care. 2004;10(6):476–82. Christians U, Serkova NJ. Immunosuppressant
39. Opelz G, Döhler B. Effect on kidney graft survival neurotoxicity in rat brain models: oxidative stress
of reducing or discontinuing maintenance immu- and cellular metabolism. Chem Res Toxicol.
nosuppression after the frst year posttransplant. 2010;23(3):608–19.
Transplantation. 2008;86(3):371–6. 55. Atkison P, Joubert G, Barron A, et al. Hypertrophic
40. Bechstein WO. Neurotoxicity of calcineurin inhibi- cardiomyopathy associated with tacrolimus
tors: impact and clinical management. Transpl Int. in paediatric transplant patients. Lancet.
2000;13(5):313–26. 1995;345(8954):894–6.
41. Serkova N, Christians U. Transplantation: toxico- 56. Pappas PA, Weppler D, Pinna AD, et al. Sirolimus
kinetics and mechanisms of toxicity of cyclospo- in pediatric gastrointestinal transplantation: the
rine and macrolides. Curr Opin Investig Drugs. use of sirolimus for pediatric transplant patients
2003;4(11):1287–96. with tacrolimus-related cardiomyopathy. Pediatr
42. Kochi S, Takanaga H, Matsuo H, et al. Induction Transplant. 2000;4(1):45–9.
of apoptosis in mouse brain capillary endothelial 57. Jarzembowski TM, John E, Panaro F, et al. Reversal
cells by cyclosporin A and tacrolimus. Life Sci. of tacrolimus-related hypertrophic obstructive
2000;66(23):2255–60. cardiomyopathy 5 years after kidney transplant
43. Dohgu S, Yamauchi A, Nakagawa S, et al. Nitric in a 6-year-old recipient. Pediatr Transplant.
oxide mediates cyclosporine-induced impairment of 2005;9(1):117–21.
the blood-brain barrier in cocultures of mouse brain 58. Turska-Kmieć A, Jankowska I, Pawłowska J, et al.
endothelial cells and rat astrocytes. Eur J Pharmacol. Reversal of tacrolimus-related hypertrophic cardio-
2004;505(1–3):51–9. myopathy after conversion to rapamycin in a pedi-
44. Stoltenburg-Didinger G, Boegner F. Glia toxicity in atric liver transplant recipient. Pediatr Transplant.
dissociation cell cultures induced by cyclosporine. 2007;11(3):319–23.
Neurotoxicology. 1992;13(1):179–84. 59. Kakhi S, Phanish MK, Anderson L. Dilated car-
45. McDonald JW, Goldberg MP, Gwag BJ, Chi SI, diomyopathy in an adult renal transplant recipient:
Choi DW. Cyclosporine induces neuronal apoptosis recovery upon tacrolimus to sirolimus switch: a case
and selective oligodendrocyte death in cortical cul- report. Transplant Proc. 2020;52(9):2758–61.
tures. Ann Neurol. 1996;40(5):750–8.
268 G. J. Dugbartey and A. Sener

60. Uemoto S, Inomata Y, Egawa H, et al. Effects of FK506 (tacrolimus) or cyclosporine A after liver
tacrolimus on cardiac function after living related transplant. Transplantation. 2002;74(6):817–21.
liver transplantation in pediatric patients. Paper 74. Ananthasubramaniam K, Garikapati K, Williams
presented at 15th Annual Meeting of the American CT. Progressive left ventricular hypertrophy after
Society of Transplant Physicians; May 26–30, 1996; heart transplantation: insights and mechanisms
Dallas, TX. suggested by multimodal images. Tex Heart Inst J.
61. Dehghani SM, Haghighat M, Imanieh MH, et al. 2016;43(1):65–8.
Tacrolimus related hypertrophic cardiomyopa- 75. Canet E, Osman D, Lambert J, et al. Acute respira-
thy in liver transplant recipients. Arch Iran Med. tory failure in kidney transplant recipients: a multi-
2010;13(2):116–9. center study. Crit Care. 2011;15(2):R91.
62. Liu T, Gao Y, Gao YL, et al. Tacrolimus-related 76. Pencheva VP, Petrova DS, Genov DK, Georgiev
hypertrophic cardiomyopathy in an adult cardiac OB. Risk factors for lung diseases after renal trans-
transplant patient. Chin Med J. 2012;125(7):1352–4. plantation. J Res Med Sci. 2015;20(12):1127–32.
63. McLeod J, Wu S, Grazette L, Sarcon A. Tacrolimus- 77. Ulas A, Kaplan S, Zeyneloglu P, Torgay A, Pirat A,
associated dilated cardiomyopathy in adult patient Haberal M. Acute respiratory failure in renal trans-
after orthotopic liver transplant. J Investig Med High plant recipients: a single intensive care unit experi-
Impact Case Rep. 2017;5(2):2324709617706087. ence. Exp Clin Transplant. 2015;13 Suppl 3:44–7.
64. Jayaraman T, Brillantes AM, Timerman AP, et al. 78. Garrean S, Massad MG, Tshibaka M, Hanhan Z,
FK506 binding protein associated with the calcium Caines AE, Benedetti E. Sirolimus-associated inter-
release channel (ryanodine receptor). J Biol Chem. stitial pneumonitis in solid organ transplant recipi-
1992;267(14):9474–7. ents. Clin Transpl. 2005;19(5):698–703.
65. Lam E, Martin MM, Timerman AP, et al. A novel 79. Jiménez Pérez M, Olmedo Martín R, Marín García
FK506 binding protein can mediate the immu- D, Lozano Rey JM, de la Cruz LJ, Rodrigo López
nosuppressive effects of FK506 and is associated JM. Pulmonary toxicity associated with siroli-
with the cardiac ryanodine receptor. J Biol Chem. mus therapy in liver transplantation. Gastroenterol
1995;270(44):26511–22. Hepatol. 2006;29(10):616–8.
66. Atkison PR, Joubert GI, Guiraudon C, Armstrong 80. Hashemi-Sadraei N, Sadrpour S, Baram D,
R, Wall W, Asfar S, Grant D. Arteritis and Miller F, Nord EP. Sirolimus-associated diffuse
increased intracellular calcium as a possible mech- alveolar hemorrhage in a renal transplant recipi-
anism for tacrolimus-related cardiac toxicity in ent on long-term anticoagulation. Clin Nephrol.
a pediatric transplant recipient. Transplantation. 2007;68(4):238–44.
1997;64(5):773–5. 81. Pérez MJ, Martín RO, García DM, Rey JM, de la
67. Baruch Y, Weitzman E, Markiewicz W, Eisenman Cruz LJ, Rodrigo López JM. Interstitial pneumonitis
A, Eid A, Enat R. Anasarca and hypertrophic car- associated with sirolimus in liver transplantation: a
diomyopathy in a liver transplant patient on FK506: case report. Transplant Proc. 2007;39(10):3498–9.
relieved after a switch to Neoral. Transplant Proc. 82. Feagans J, Victor D, Moehlen M, Florman SS,
1996;28(4):2250–1. Regenstein F, Balart LA, Joshi S, Killackey MT,
68. Sallam K, Bhumireddy GP, Evuri VD, et al. Slakey DP, Paramesh AS. Interstitial pneumonitis in
Sirolimus adverse event profle in a non-clinical the transplant patient: consider sirolimus-associated
trial cohort of heart transplantation patients. Ann pulmonary toxicity. J La State Med Soc.
Transplant. 2021;26:e923536. 2009;161(3):166.
69. Kushwaha SS, Raichlin E, Sheinin Y, et al. Sirolimus 83. Pham PT, Pham PC, Danovitch GM, et al. Sirolimus-
affects cardiomyocytes to reduce left ventricular associated pulmonary toxicity. Transplantation.
mass in heart transplant recipients. Eur Heart J. 2004;77(8):1215–20.
2008;29(22):2742–50. 84. Das BB, Shoemaker L, Subramanian S, Johnsrude
70. Raichlin E, Chandrasekaran K, Kremers WK, et al. C, Recto M, Austin EH. Acute sirolimus pulmonary
Sirolimus as primary immunosuppressant reduces toxicity in an infant heart transplant recipient: case
left ventricular mass and improves diastolic func- report and literature review. J Heart Lung Transplant.
tion of the cardiac allograft. Transplantation. 2007;26(3):296–8.
2008;86(10):1395–400. 85. Gupte GL, Mahadevan S, Clarke JR, Alton H, Beath
71. Alnsasra H, Asleh R, Oh JK, et al. Impact of siroli- SV. Sirolimus-related pulmonary toxicity mimick-
mus as a primary immunosuppressant on myocardial ing 'asthma like' symptoms. World J Gastroenterol.
fbrosis and diastolic function following heart trans- 2007;13(38):5151–3.
plantation. J Am Heart Assoc. 2021;10(1):e018186. 86. Rodríguez-Moreno A, Ridao N, García-Ledesma P,
72. Chen G, Chen H, Wang C, et al. Rapamycin ame- et al. Sirolimus and everolimus induced pneumoni-
liorates kidney fbrosis by inhibiting the activation tis in adult renal allograft recipients: experience in a
of mTOR signaling in interstitial macrophages and center. Transplant Proc. 2009;41(6):2163–5.
myofbroblasts. PLoS One. 2012;7(3):e33626. 87. Errasti P, Izquierdo D, Martín P, et al. Pneumonitis
73. Roberts CA, Stern DL, Radio SJ. Asymmetric car- associated with mammalian target of rapamy-
diac hypertrophy at autopsy in patients who received cin inhibitors in renal transplant recipients:
15 Organ Toxicity by Immunosuppressive Drugs in Solid Organ Transplantation 269

a single-center experience. Transplant Proc. 102. Bunnapradist S, Neri L, Wong W, et al. Incidence
2010;42(8):3053–4. and risk factors for diarrhea following kidney trans-
88. Depuydt P, Nollet J, Benoit D, Praet M, Caes F. Fatal plantation and association with graft loss and mor-
acute pulmonary injury associated with everolimus. tality. Am J Kidney Dis. 2008;51(3):478–86.
Ann Pharmacother. 2012;46(3):e7. 103. Webster AC, Woodroffe RC, Taylor RS, Chapman
89. Morelon E, Stern M, Israël-Biet D, et al. JR, Craig JC. Tacrolimus versus ciclosporin as
Characteristics of sirolimus-associated inter- primary immunosuppression for kidney transplant
stitial pneumonitis in renal transplant patients. recipients: meta-analysis and meta-regression of
Transplantation. 2001;72(5):787–90. randomised trial data. BMJ. 2005;331(7520):810.
90. Takahashi K, Go P, Stone CH, et al. Mycophenolate 104. Telkes G, Peter A, Tulassay Z, Asderakis A. High
mofetil and pulmonary fbrosis after kidney frequency of ulcers, not associated with Helicobacter
transplantation: a case report. Am J Case Rep. pylori, in the stomach in the frst year after kid-
2017;18:399–404. ney transplantation. Nephrol Dial Transplant.
91. Gross DC, Sasaki TM, Buick MK, Light JA. Acute 2011;26(2):727–32.
respiratory failure and pulmonary fbrosis second- 105. Yoshimura N, Nakai I, Ohmori Y, et al. Effect of
ary to administration of mycophenolate mofetil. cyclosporine on the endocrine and exocrine pan-
Transplantation. 1997;64(11):1607–9. creas in kidney transplant recipients. Am J Kidney
92. Morrissey P, Gohh R, Madras P, Monaco Dis. 1988;12(1):11–7.
AP. Pulmonary fbrosis secondary to administra- 106. Ogunseinde BA, Wimmers E, Washington B, Iyob
tion of mycophenolate mofetil. Transplantation. M, Cropper T, Callender CO. A case of tacrolimus
1998;65(10):1414. (FK506)-induced pancreatitis and fatality 2 years
93. Shrestha NK, Mossad SB, Braun W. Pneumonitis postcadaveric renal transplant. Transplantation.
associated with the use of mycophenolate mofetil. 2003;76(2):448.
Transplantation. 2003;75(10):1762. 107. Im MS, Ahn HS, Cho HJ, Kim KB, Lee HY. Diabetic
94. Pijnenburg MW, Cransberg K, Wolff E, Bouquet J, ketoacidosis associated with acute pancreatitis in a
Merkus PJ. Bronchiectasis in children after renal or heart transplant recipient treated with tacrolimus.
liver transplantation: a report of fve cases. Pediatr Exp Clin Transplant. 2013;11(1):72–4.
Transplant. 2004;8(1):71–4. 108. Alkhatib AA. Sirolimus-induced intractable
95. Rook M, Postma DS, van der Jagt EJ, et al. chronic diarrhea: a case report. Transplant Proc.
Mycophenolate mofetil and bronchiectasis in kid- 2006;38(5):1298–300.
ney transplant patients: a possible relationship. 109. Malinowski M, Martus P, Lock JF, Neuhaus P,
Transplantation. 2006;81(2):287–9. Stockmann M. Systemic infuence of immunosup-
96. Reynolds BC, Paton JY, Howatson AG, Ramage pressive drugs on small and large bowel transport and
IJ. Reversible chronic pulmonary fbrosis associ- barrier function. Transpl Int. 2011;24(2):184–93.
ated with MMF in a pediatric patient: a case report. 110. Dias VC, Madsen KL, Mulder KE, Keelan M,
Pediatr Transplant. 2008;12(2):228–31. Yatscoff RW, Thomson AB. Oral administra-
97. Boddana P, Webb LH, Unsworth J, Brealey M, tion of rapamycin and cyclosporine differentially
Bingham C, Harper SJ. Hypogammaglobulinemia alter intestinal function in rabbits. Dig Dis Sci.
and bronchiectasis in mycophenolate mofetil-treated 1998;43(10):2227–36.
renal transplant recipients: an emerging clinical phe- 111. Yang J, Zhao X, Patel A, et al. Rapamycin inhi-
nomenon? Clin Transpl. 2011;25(3):417–9. bition of mTOR reduces levels of the Na+/
98. Gorgan M, Bockorny B, Lawlor M, Volpe J, H+ exchanger 3 in intestines of mice and
Fiel-Gan M. Pulmonary hemorrhage with capil- humans, leading to Diarrhea. Gastroenterology.
laritis secondary to mycophenolate mofetil in a 2015;149(1):151–62.
heart-transplant patient. Arch Pathol Lab Med. 112. van Gelder T, ter Meulen CG, Hené R, Weimar W,
2013;137(11):1684–7. Hoitsma A. Oral ulcers in kidney transplant recipi-
99. Simmons WD, Rayhill SC, Sollinger HW. ents treated with sirolimus and mycophenolate
Preliminary risk-beneft assessment of mycophe- mofetil. Transplantation. 2003;75(6):788–91.
nolate mofetil in transplant rejection. Drug Saf. 113. Smith AD, Bai D, Marroquin CE, et al.
1997;17(2):75–92. Gastrointestinal hemorrhage due to complicated gas-
100. Arns W. Noninfectious gastrointestinal (GI) com- troduodenal ulcer disease in liver transplant patients
plications of mycophenolic acid therapy: a con- taking sirolimus. Clin Transpl. 2005;19(2):250–4.
sequence of local GI toxicity? Transplant Proc. 114. Sollinger HW. Mycophenolate mofetil for the
2007;39(1):88–93. prevention of acute rejection in primary cadav-
101. Ekberg H, Kyllönen L, Madsen S, Grave G, Solbu eric renal allograft recipients. U.S. Renal
D, Holdaas H. Increased prevalence of gastrointes- Transplant Mycophenolate Mofetil Study Group.
tinal symptoms associated with impaired quality of Transplantation. 1995;60(3):225–32.
life in renal transplant recipients. Transplantation. 115. Placebo-controlled study of mycophenolate
2007;83(3):282–9. mofetil combined with cyclosporin and corticoste-
270 G. J. Dugbartey and A. Sener

roids for prevention of acute rejection. European 129. Marbet U, Schmid I. Severe life-threatening diarrhea
Mycophenolate Mofetil Cooperative Study Group. caused by azathioprine but not by 6-mercaptopurine.
Lancet. 1995;345(8961):1321–5. Digestion. 2001;63(2):139–42.
116. A blinded, randomized clinical trial of mycopheno- 130. Ziegler TR, Fernández-Estívariz C, Gu LH, Fried
late mofetil for the prevention of acute rejection in MW, Leader LM. Severe villus atrophy and
cadaveric renal transplantation. The Tricontinental chronic malabsorption induced by azathioprine.
Mycophenolate Mofetil Renal Transplantation Gastroenterology. 2003;124(7):1950–7.
Study Group. Transplantation. 1996;61(7):1029–37. 131. Weclawiak H, Ould-Mohamed A, Bournet B,
117. Squiffet JP. Dose optimization of mycopheno- et al. Duodenal villous atrophy: a cause of chronic
late mofetil when administered with a low dose of diarrhea after solid-organ transplantation. Am J
tacrolimus in cadaveric renal transplant recipients. Transplant. 2011;11(3):575–82.
Transplantation. 72(1):63–9. 132. Ledder O, Lemberg DA, Day AS. Thiopurine-
118. Squiffet JP, Bäckman L, Claesson K. European induced pancreatitis in infammatory bowel
Tacrolimus-MMF Renal Study Group. diseases. Expert Rev Gastroenterol Hepatol.
Transplantation. 2001;72(1):63–9. 2015;9(4):399–403.
119. Kamar N, Faure P, Dupuis E, et al. Villous atro- 133. Narum S, Westergren T, Klemp M. Corticosteroids
phy induced by mycophenolate mofetil in renal- and risk of gastrointestinal bleeding: a sys-
transplant patients. Transpl Int. 2004;17(8):463–7. tematic review and meta-analysis. BMJ Open.
120. Golconda MS, Valente JF, Bejarano P, Gilinsky N, 2014;4(5):e004587.
First MR. Mycophenolate mofetil-induced colonic 134. Nitsche C, Maertin S, Scheiber J, Ritter CA, Lerch
ulceration in renal transplant recipients. Transplant MM, Mayerle J. Drug-induced pancreatitis. Curr
Proc. 1999;31(1–2):272–3. Gastroenterol Rep. 2012;14(2):131–8.
121. Naranjo J, Poniachik J, Cisco D, et al. Oral ulcers pro- 135. Bae GE, Yoon N, Park HY, et al. Silent colonic mala-
duced by mycophenolate mofetil in two liver trans- koplakia in a living-donor kidney transplant recipi-
plant patients. Transplant Proc. 2007;39(3):612–4. ent diagnosed during annual medical examination.
122. Dost D, van Leerdam ME, van Dekken H, et al. Korean J Pathol. 2013;47(2):163–6.
Crohn's-like enterocolitis associated with mycophe- 136. Durrbach A, Pestana JM, Pearson T, et al. A
nolic acid treatment. Gut. 2008;57(9):1330. phase III study of belatacept versus cyclospo-
123. Allison AC, Kowalski WJ, Muller CD, Eugui rine in kidney transplants from extended criteria
EM. Mechanisms of action of mycophenolic acid. donors (BENEFIT-EXT study). Am J Transplant.
Ann N Y Acad Sci. 1993;696:63–87. 2010;10(3):547–57.
124. Burg M, Säemann MD, Wieser C, Kramer S, Fischer 137. Vincenti F, Charpentier B, Vanrenterghem Y, et al.
W, Lhotta K. Enteric-coated mycophenolate sodium A phase III study of belatacept-based immunosup-
reduces gastrointestinal symptoms in renal transplant pression regimens versus cyclosporine in renal trans-
patients. Transplant Proc. 2009;41(10):4159–64. plant recipients (BENEFIT study). Am J Transplant.
125. Ortega F, Sánchez-Fructuoso A, Cruzado JM, 2010;10(3):535–46.
et al.; MYVIDA Study Group. Gastrointestinal 138. Motohashi R, Ikeuchi H, Hiromura K, et al. Two
quality of life improvement of renal transplant cases of ulcerative colitis developing in rheumatoid
recipients converted from mycophenolate mofetil arthritis patients during abatacept therapy. Scand J
to enteric-coated mycophenolate sodium drugs Gastroenterol. 2014;49(10):1270–1.
or agents: mycophenolate mofetil and enteric- 139. Bozon A, Jeantet G, Rivière B, et al. Stricturing
coated mycophenolate sodium. Transplantation. Crohn's disease-like colitis in a patient treated
2011;92(4):426–32. with belatacept. World J Gastroenterol.
126. Budde K, Curtis J, Knoll G, et al.; ERL B302 Study 2017;23(48):8660–5.
Group. Enteric-coated mycophenolate sodium 140. Neumann UP, Berg T, Bahra M, et al. Fibrosis pro-
can be safely administered in maintenance renal gression after liver transplantation in patients with
transplant patients: results of a 1-year study. Am J recurrent hepatitis C. J Hepatol. 2004;41(5):830–6.
Transplant. 2004;4(2):237–43. 141. Kowdley KV, Keeffe EB. Hepatotoxicity of trans-
127. Salvadori M, Holzer H, De Mattos A, et al. Enteric- plant immunosuppressive agents. Gastroenterol Clin
coated mycophenolate sodium is therapeutically N Am. 1995;24(4):991–1001.
equivalent to mycophenolate mofetil in de novo renal 142. Pol S, Cavalcanti R, Carnot F, et al. Azathioprine
transplant patients. Am J Transplant. 2004;4:231–6. hepatitis in kidney transplant recipients. A predispos-
128. Kamar N, Oufroukhi L, Faure P, et al. Questionnaire- ing role of chronic viral hepatitis. Transplantation.
based evaluation of gastrointestinal disorders in 1996;61(12):1774–6.
de novo renal-transplant patients receiving either 143. Neff GW, Ruiz P, Madariaga JR, et al. Sirolimus-
mycophenolate mofetil or enteric-coated myco- associated hepatotoxicity in liver transplantation.
phenolate sodium. Nephrol Dial Transplant. Ann Pharmacother. 2004;38(10):1593–6.
2005;20(10):2231–6. 144. Niemczyk M, Wyzgał J, Perkowska A, Porowski
D, Paczek L. Sirolimus-associated hepatotox-
15 Organ Toxicity by Immunosuppressive Drugs in Solid Organ Transplantation 271

icity in the kidney graft recipient. Transpl Int. 153. Lorber MI, Van Buren CT, Flechner SM, Williams
2005;18(11):1302–3. C, Kahan BD. Hepatobiliary complications of cyclo-
145. Loupy A, Anglicheau D, Mamzer-Bruneel MF, et al. sporine therapy following renal transplantation.
Mycophenolate sodium-induced hepatotoxicity: frst Transplant Proc. 1987;19(1 Pt 2):1808–10.
report. Transplantation. 2006;82(4):581. 154. Soresi M, Sparacino V, Pisciotta G, et al. Effects of
146. Contreras AM, Monteón FJ, Flores MR, Mendoza- cyclosporin A on various indices of cholestasis in
Sánchez F, Ruiz I. Drug-related hepatotoxicity in kidney transplant recipients. Minerva Urol Nefrol.
a renal transplant recipient with long-term survival 1995;47(2):65–9.
and hepatitis C. Ann Hepatol. 2007;6(1):70–3. 155. Yuan QS, Zheng FL, Sun Y, Yu Y, Li Y. Rescue
147. Yadav DK, Gera DN, Gumber MR, et al. Tacrolimus- therapy with tacrolimus in renal graft patients with
induced severe cholestasis complicating renal trans- cyclosporine A-induced hepatotoxicity: a prelimi-
plantation. Ren Fail. 2013;35(5):735–7. nary study. Transplant Proc. 2000;32(7):1694–5.
148. Akbulut S, Elbe H, Eris C, et al. Effects of antioxi- 156. Vallet-Pichard A, Rerolle JP, Fontaine H, et al.
dant agents against cyclosporine-induced hepatotox- Veno-occlusive disease of the liver in renal
icity. J Surg Res. 2015;193(2):658–66. transplant patients. Nephrol Dial Transplant.
149. Lim EJ, Chin R, Nachbur U, et al. Effect of immuno- 2003;18(8):1663–6.
suppressive agents on hepatocyte apoptosis post-liver 157. Ganschow R, Albani J, Grabhorn E, Richter A,
transplantation. PLoS One. 2015;10(9):e0138522. Burdelski M. Tacrolimus-induced cholestatic syn-
150. Patel S, Mendler MH, Valasek MA, Tsunoda drome following pediatric liver transplantation and
SM. Drug-induced liver injury associated with the steroid-resistant graft rejection. Pediatr Transplant.
use of everolimus in a liver transplant patient. Case 2006;10(2):220–4.
Rep Transpl. 2018;2018:7410508. 158. Taniai N, Akimaru K, Ishikawa Y, et al.
151. Warren M, Mitsinikos T, Yanni G, Sasaki M, Sasaki Hepatotoxicity caused by both tacrolimus and cyclo-
AT, Thomas D. Mycophenolate mofetil hepatotox- sporine after living donor liver transplantation. J
icity associated with mitochondrial abnormality Nippon Med Sch. 2008;75(3):187–91.
in liver transplant recipients and mice. J Pediatr 159. Sanchez-Campos S, Lopez-Acebo R, Gonzalez
Gastroenterol Nutr. 2021;73(4):463–70. P, Culebras JM, Tuñon MJ, Gonzalez-Gallego
152. Sacher VY, Bejarano PA, Pham SM. Tacrolimus J. Cholestasis and alterations of glutathione metab-
induced hepatotoxicity in a patient with bilateral olism induced by tacrolimus (FK506) in the rat.
lung transplant. Transpl Int. 2012;25(10):e111–2. Transplantation. 1998;66(1):84–8.
Artificial Intelligence-Based
Techniques to Assess Drug Toxicity
16
in Drug-Induced Liver Injury
(DILI) Disease

Munish Puri

Abstract Keywords

Drug-induced liver injury (DILI) is difficult to Automated machine learning · Computational


detect, and a rare condition which is respon- biomarker · Diagnostic support system ·
sible for drug removal from the market. Drug-induced liver injury · Feature-based
DILI poses a great drug safety concern for the detection
Foods and Drug Authority (FDA) and pharma
industry. It is challenging for pathologists to
differentially diagnose DILI injury patterns 16.1 Introduction
and to assess hepatotoxicity and other closely
related liver disease phenotypes such as non-­ Drug-induced liver injury (DILI) or hepatotoxic-
alcoholic steatohepatitis (NASH). Early detec- ity is a common cause of acute liver failure. DILI
tion of DILI can save a person from morbidity is a rare disease, responsible for the withdrawal
associated with this condition and also cost of drugs from the market due to its late detection.
of hospitalization. It is generally difficult to High cost of drug development process and a
detect complex and overlapping DILI injury long time to reach the market create disappoint-
patterns by human eyes under microscope. ment and waste of scientific efforts. The cause of
Computational pathology and artificial intel- DILI-related injury is poorly understood and still
ligence can be helpful for diagnosing DILI unknown today. It’s very hard to detect an agent
injury patterns, liver toxicity, and disease responsible for DILI and challenging to assess
management. Deep learning models in artifi- the injury levels induced on the liver, because of
cial intelligence can be tested on the whole multiple drugs taken by patients. DILI is a grow-
slide digital images (WSI) of liver biopsy to ing concern in the drug development research
classify the DILI injury patterns such as fibro- community because of the increasing number of
sis and steatosis. drugs used in medical care and the increasing
number of individuals who takes them routinely.
Hepatotoxicity is the highest concern of adverse
drug reactions in the pharmaceutical industry [1].
DILI injury is induced by prescription drugs
M. Puri (*) taken together with over-the-counter drugs and
Artificial Intelligence, Immunology, AbbVie alternative medicines, such as herbal products
Pharmaceutical, Worcester, MA, USA and supplements.
e-mail: munish.puri@abbvie.com

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 273
S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_16
274 M. Puri

There is no treatment available to cure injured associated phenotypes. Clinical features associ-
liver other than discontinuation of drug and ated with DILI etiologies described in patients
removal of offending agents which can be helpful are closely related to herbal medications and
to avoid risk involved in pre-existing liver disease dietary supplements [4]. DILI risk factors related
patients. In clinical practice, pathologists study to patients include age, gender, alcohol, genetics,
liver biopsy under microscope to assess DILI drug-dose, and metabolism.
injury patterns and score affected liver. There is a
great inter- and intra-variability of observations
among the pathologists to read and diagnose 16.3 LiverTox
DILI injury and to assess the state of disease
severity. Other major challenges are overlapping National Institute of Health’s (NIH) LiverTox
DILI injury patterns with other common liver website is the important resource to study liver
diseases. toxicity. LiverTox is a freely available resource
Digital pathology is a potential area of interest which provides clinical and research information
for pathologists to understand DILI injury pat- on DILI drugs, herbals, and dietary supplements
terns using machine learning imaging algorithms. responsible for causing DILI liver injury.
Artificial intelligence and machine learning tech- LiverTox platform is designed by the National
nologies are rapidly gathering attention in the Institute of Diabetes and Digestive and Kidney
digital imaging space due to its pattern recogni- Diseases (NIDDK) and widely used by the medi-
tion features. Deep learning, a sub-type of artifi- cal community, mostly physicians, clinicians,
cial intelligence algorithm, successfully tested on patients, researchers, and academicians for the
millions of images (with great accuracy and effi- updated DILI-related hepatotoxicity information
ciency) for pattern recognition [2]. Artificial [5]. The LiverTox can be accessed at https://
intelligence can be used to assess DILI-induced www.ncbi.nlm.nih.gov/books/NBK547852/
hepatotoxicity, disease severity, and DILI pattern maintained by NCBI National Library of medi-
recognition on liver digital biopsy images as an cine website. The information documented in
AI-based diagnostic tool. LiverTox is taken from scientific literature, pub-
lic databases, various drug development studies,
and clinical trials.
16.2 Etiology DILI liver injury relies on diagnosis of exclu-
sion and is very closely associated with other
DILI is classified as intrinsic and idiosyncratic. liver diseases. There is no diagnosis available for
Intrinsic DILI is predictable and dose-dependent, DILI-related injury, other than causality assess-
whereas idiosyncratic DILI is unpredictable and ment and disease management which is again
dose-independent and has variable latency challenging and relies on the timing of onset of
period. The etiology of DILI remains complex allergic reactions and starting and stopping of
and unclear and makes it harder for a pathologist medication. There is no treatment available to
to perform differential diagnosis to classify if reverse DILI injury but discontinuation of the
these are drug-induced patterns [3]. In a liver drug and toxins.
biopsy, DILI injury patterns are widely resem-
bled with commonly expressed histologic pat-
terns of chronic hepatitis and fatty liver disease 16.3.1 Phenotype of DILI
ranging from inflammation, necrosis, and fibro-
sis. Pathogenesis of DILI is complex and unpre- DILI is categorized by phenotypes, clinical find-
dictable and could be related to the activation of ings, and symptoms, which are closely associ-
the immune system. Liver biopsy is not recom- ated with other liver disease and hepatotoxicity
mended for DILI diagnosis but can be useful in patterns. LiverTox broadly categorizes DILI
excluding other etiologies to assess the closely expression into 12 phenotypes, namely, acute
16 Artificial Intelligence-Based Techniques to Assess Drug Toxicity in Drug-Induced Liver Injury (DILI… 275

hepatic necrosis, acute hepatitis, non-alcoholic Table 16.1 DILI grading system
fatty liver (NAFL), and others which could be Score Condition Assessment
helpful in diagnosis, disease management, cau- 1+ Mild Raised serum aminotransferase or
alkaline phosphatase levels or both,
sality assessment, and understanding the disease
but total serum bilirubin <2.5 mg/
pathogenesis. These phenotypes have distinct dL and no coagulopathy (INR <1.5)
immunologic features and adverse outcomes 2+ Moderate Raised serum aminotransferase or
which may lead to liver failure and cirrhosis. alkaline phosphatase levels or both
DILI diagnosis is hard due to its resembling and and total serum bilirubin level
≥2.5 mg/dL or coagulopathy (INR
overlapping phenotypes with other liver diseases ≥1.5) without hyperbilirubinemia
and fatty liver, especially in patients with pre- 3+ Moderate Raised serum aminotransferase or
existing disease conditions and who take multi- to severe alkaline phosphatase levels and
ple drugs of different doses. total serum bilirubin level ≥2.5 mg/
dL and hospitalization (or
preexisting hospitalization is
prolonged) because of the
16.3.2 Classification of DILI Drugs drug-induced liver injury
4+ Severe Raised serum aminotransferase or
There is no clear-cut categorization recom- alkaline phosphatase levels and
serum bilirubin ≥2.5 mg/dL and at
mended for classification of direct DILI or idio- least one of the following:
syncratic DILI. Based on the level of injury Prolonged jaundice and symptoms
induced by DILI drugs, frequency of drug taken, beyond 3 months, or signs of
commonly or uncommonly, reported injury lev- hepatic decompensation (INR ≥1.5,
ascites, encephalopathy), or other
els, etc., NIDDK created a “Likelihood Score” on organ failure believed to be related
five-point categorization of the likelihood that a to drug-induced liver injury
medication is associated to DILI liver injury. The 5+ Fatal Death or liver transplantation for
network can be accessed at https://dilin.org/ and drug-induced liver injury
is known as Drug-Induced Liver Injury Network Adopted from LiverTox
a
The international correction ratio (INR) is a calculation
(DILIN) [6].
used to monitor individuals who are being treated with
DILIN collects and analyzes cases of severe blood-thinning medication
injury and documents toxin agents that can cause
DILI injury in normal or overdose conditions.
Based on the five-point system adopted by 1. Roussel Uclaf Causality Assessment Method
DILIN, tabulated in Table 16.1, the severity of (RUCAM): the Roussel Uclaf Causality
DILI is assessed as mild, transient, liver and other Assessment Method (RUCAM) was devel-
organ failure, jaundice, and any hospitalization oped in 1989 by the Council for International
conditions. Organizations of Medical Scientists (CIOMS)
for the diagnostic criteria or DILI injury
assessment [8].
16.4 Liver Causality Assessment 2. Digestive Disease Week Japan 2004 scale
Tools (DDW-J): DDW-J is superior to CIMOS and
M & V scales and includes in vitro Drug
DILI casualty is assessed and weighted on other Lymphocyte Stimulation Test (DLST) [9]
important factors which include hypersensitivity, 3. Clinical Diagnostic Scale (CDS). CDS is
recurrence, and drug allergies. Commonly used more stringent tool in assessing DILI in com-
scale for capturing adverse drug reactions known parison to RUCAM and DDW-J [10].
as Naranjo Probability Scale [7] is also used for
causality assessment. Broadly categorized, there Mostly all three assessment tools behave simi-
are three liver-specific causality assessment tools larly in selection of variables and exhibitions of
currently available in clinical practice for the casualty assessment with minor variations which
diagnosis of drug-related DILI:
276 M. Puri

may likely benefit for improvement by using bodies (ANAs), smooth muscle antibodies
data-driven and computer-based techniques. (SMAs), and elevated immunoglobulin G (IgG)
levels is diagnostically challenging for DILI dis-
ease management.
16.5 Computational Pathology DILI drugs in patients with non-alcoholic
for DILI Injury Assessment fatty liver disease (NAFLD) share common
pathophysiological features and induce macrove-
Deep learning algorithms are tested to under- sicular steatosis which can trigger the inflamma-
stand chemical or molecular behavior of DILI tions and hepatotoxicity. Steatosis manifests as
drugs [11]. Very few studies are available on the accumulation of droplets of triglycerides
computational pathology and DILI detection on inside the cytoplasm of hepatocytes.
histology images [12]. Biopsy tissue glass slides Hepatotoxicity in NAFLD exhibits necroinflam-
scanned digitally at higher magnification provide mation and fibrosis which belongs to pharmaco-
a rich source of disease phenotypic information logical class of few of the DILI drugs such as
which can be helpful to assess the DILI injury acetaminophen, halothane, methotrexate, rosigli-
patterns. Computational pathology and artificial tazone, and tamoxifen [16]. Obesity is also
intelligence [13] used with feature engineering, a closely associated with NAFL characterized by
technique in computer vision to extract morpho- accumulation of hepatic lipids which may prog-
metric disease information from digital images, ress towards NAFLD and later to non-alcoholic
can be helpful for a computer algorithm to extract steatohepatitis (NASH), an advanced form of
enriched disease information which is challeng- NAFLD. NAFLD tends to develop in patients
ing for a human pathologist to interpret. Image who are overweight or obese or have diabetes,
magnification and staining variability add another high cholesterol, or high triglycerides. In a patho-
level of complexity. Artificial intelligence and logical setting, the NASH scoring system com-
deep learning techniques could be incorporated monly considers main histopathological features
in this space of imaging-based diagnostic tools to such as ballooning degeneration of hepatocytes
analyze DILI injury patterns. (liver cells), inflammation, steatosis, and fibrosis.
Liver fibrosis developed by deposition of an
extracellular matrix is a major parameter guiding
16.6 Liver Toxicity and Fatty Liver the prognosis of liver diseases.
Necrosis is another phenotype induced by the
Liver is the largest organ in the body performing DILI drug. Acetaminophen (paracetamol,
more than 500 tasks; major functions include N-acetyl-p-aminophenol; APAP) is a widely used
detoxification, protein synthesis, and bile produc- over-the-counter analgesic drug and considered
tion. Liver cells are known as hepatocytes. safe at therapeutic doses; acetaminophen pro-
Toxicity caused to hepatocytes is called hepato- duces a hepatic necrosis that can be fatal if taken
toxicity which is induced by the intake of toxins in higher doses [17]. Some other drugs such as
or overdose of prescribed drugs. One of the phe- bromobenzene, beryllium, quinidine, Iodoform,
notypes of liver toxicity is fatty liver. In some ferrous sulphate also cause zonal hepatotoxic
patients, autoimmune hepatitis and DILI injury injury that spreads from central vein to portal
patterns present and manifest very closely associ- vein [18].
ated overlapping pathological features [14]. DILI
present with autoimmune features is known as
AI-DILI. Patients with DILI features show posi- 16.7 Artificial Intelligence in DILI
tive markers for autoimmunity with IgG immu- Pattern Detection
noglobulin. Few drugs are listed here that carry
similar features: nitrofurantoin, minocycline, Artificial intelligence is a computer-based tech-
methyldopa, or hydralazine [15]. The presence of nique used for process automation and pattern
autoimmune features such as antinuclear anti- recognition which is gathering attention of the
16 Artificial Intelligence-Based Techniques to Assess Drug Toxicity in Drug-Induced Liver Injury (DILI… 277

medical community and widely used in medicine open-source toxicogenomics database imaging
for various applications [19]. Artificial neural drug dataset popularly known as the
network (ANN) is the basic machine learning Toxicogenomics Project—Genomics Assisted
architecture in AI which mimics the human brain Toxicity Evaluation Systems (TG-GATEs) [27].
to learn, process, and perform a computational Machine learning algorithm was able to detect
task with minimal human interference. ANN is necrotic DILI injury levels induced by the use of
used as a computational tool for process automa- commonly overdosed DILI drugs on biopsy
tion and high throughput in the drug development images using mathematical geometry of fractals
pipeline, extensively used and adopted in the and lacunarity. Fractal-based texture analysis is
pharma industry [20, 21]. ANN can be used on used in detecting tumor geometry and growth in
histology digital images which are scanned at digital mammogram images [28].
high magnification. Digitally scanned biopsy
images provide rich disease information in vary-
ing morphology in the shape of imaging features 16.7.1 AuML for DILI Pattern
to assess disease diagnosis, severity, and progres- Detection
sion [22].
Some important areas of bioinformatics such A study is designed as a proof of concept (PoCP)
as computational pathology, machine learning to test the AutoML AI algorithm for DILI pattern
(ML), morphology feature engineering, and data detection. The histology image dataset used in
integration [23] have been widely accepted in the this study is taken from a public open source
digital pathology community. Computational which is available at Gtex biobank maintained by
analysis is helpful to assess phenotype disease the National Institutes of Health’s Genotype-­
progression, in finding hotspots of disease activ- Tissue Expression (GTEx) project [29].
ity [24], in reducing inter-/intra-observer vari- Liver toxicity image dataset is divided into
ability [25], and for accurate disease diagnosis four classes as fibrosis, steatosis, necrosis, and
[26]. In this range of computational tools used in normal liver (2000 images in each class) as
biomedicine, automated machine learning shown in Fig. 16.1.
(AutoML) is another fully automated computa- AuML model is set to divide the dataset into
tional model (with no human interference) which training the model as 80% training data, 10%
works on the principle of learning from experi- validation, and 10% out-of-box testing. AuML
ence and mistakes and in which ANN is designed model performance is tested by the confusion
by another neural network. The AutoML model matrix which is created between true and pre-
of ML is highly successful in industry and in dicted labels, as shown in Fig. 16.2; the matrix
accurately predicting outcomes and pattern rec- shows how often the model classifies each label
ognition. AuML is a Google’s machine learning correctly (in blue) and which labels are most
(ML) model supported with the latest graphical often confused for that label (in orange). The ML
processing units (GPUs). AuML is available in model is able to classify true and predicted values
open-source public domain for high-performance with 98.5% accuracy in case of normal, 95.6% in
artificial intelligence model processing and com- case of steatosis, 94.9% in necrosis, and 92.7% in
puting designed for non-ML experts on the con- fibrosis.
cept where an artificial neural network is designed AuML model is validated on blinded test
by another neural network with minimal human images (which the model has never seen during
interference. In a DILI hepatotoxicity study, an training). Model can perform 98% accurately in
AutoML model is tested to classify DILI injury detecting test images of necrosis, fibrosis, steato-
patterns on whole slide pathology images on sis, and normal. Few prediction examples are
commonly used DILI drugs [12]. shown in Fig. 16.3 where randomly picked test
In this study, a deep learning model was images are tested in all four classes to validate the
designed and tested on the publicly available model. Figure 16.4 shows the model evaluation
278 M. Puri

Fig. 16.1 Image dataset: Four classes of images (2000 & s1 (Steatosis). Two images of each class are shown here
images in each class are set for training ML model); F1 in the bottom rows
(Fibrosis), N1 (Necrosis), Normal (Normal liver images),

matrix and the test images which have mixed model is learning the features of input image and
classes, i.e., necrosis with fibrosis and necrosis pushing data towards prediction, shown in
with steatosis. In most of the cases, the model can Figs. 16.6 and 16.7.
predict accurately. Figure 16.5 shows the In fatty liver, steatosis develops in stages
­accuracy curve of ML model during training and which can provide information in diagnosis
validation. severity for the liver disease. In pathology lab,
progression of steatosis is assessed on H & E
images under microscope. This process is highly
16.7.2 Deep Learning Model for DILI error-prone and subject to inter-/intra-observer
variability. High-throughput computer-aided
Deep learning model in AI is made up of three image analysis tools and machine learning algo-
major layers as input layer, hidden layer, and out- rithms are widely accepted in pathology labs for
put layer. Data handling architecture layers in scoring stages of steatosis.
between input and output layers are referred to as In deep learning, there are two ways to
“hidden layers,” of which deep neural networks automate the process, by classification or by seg-
have many hidden layers; originally, “deep” mentation technique. In steatosis segmentation,
means having more than one hidden layer. The a deep learning model is proposed as DeEp
DILI image data processing happens in these LearnINg stEATosis sEgmentation
three layers. The original input data is first fed to (DELINEATE) both at patch-wise and whole
the “input layer,” and the “output layer” pushes slide for steatosis prediction analysis [30].
out data that represents the model’s prediction. Proposed steatosis scoring method presents a
Learning by ML model during the training pro- strong correlation with pathologist annotations.
cess can be seen through internal hidden deep In case of DILI, it’s hard to implement preclinical
layer architecture. For example, here layer 5 models into clinical practice; drugs that cause
learning is picked in the case of fibrosis and DILI in humans typically do not show clear hepa-
steatosis that illustrates how a deep learning totoxicity in animals. Molecular structure-based
16 Artificial Intelligence-Based Techniques to Assess Drug Toxicity in Drug-Induced Liver Injury (DILI… 279

Fig. 16.2 Confusion matrix illustrates how ML model predicts the true class correctly (blue) and incorrectly (orange)
as other class

Fig. 16.3 Prediction on test images (a–d); prediction for fibrosis, steatosis, necrosis, and normal liver on test images
by ML model

deep learning model was proposed and tested as 16.8 Conclusion


UGRNN neural network architecture [31]. It’s
always a good practice to utilize the transfer Studies that assess DILI provide considerable
learning technique in deep learning, instead of insights into the importance of hepatotoxicity in
developing a new deep learning model from drug development. DILI is responsible for drug
scratch. For scoring developing stages of fibrosis, removal from the market, even in the final stages
different machine learning-based models using of drug approval, which poses a great concern for
pre-trained AlexNet-CNN were tested which FDA and pharma industry. LiverTox and DILIN
automatically score liver fibrosis stages with a are the open-source resources available for the
level of accuracy similar to pathologists [32]. updated DILI-related information. Liver function
280 M. Puri

Fig. 16.4 Model evaluation matrix and prediction: (a) recall change by adjusting threshold. ROC receiver oper-
Threshold slider to adjust precision and recall. (b) ROC ating curve. (d) Prediction on necrosis, fibrosis. (e)
precision recall curve at 0.5 threshold. (c) Average preci- Prediction on necrosis, steatosis, and fibrosis
sion, precision, and recall at 0.5 threshold. Precision and

Fig. 16.5 Accuracy curve: ML model accuracy during training and validation
Fig. 16.6 Fibrosis learning representation: hidden layer 5 learning process in deep learning model, input image (left)
and internal learning images (right)

Fig. 16.7 Steatosis learning representation: hidden layer 5 learning process in deep learning model, input image (left)
and internal learning images (right)
282 M. Puri

is highly complex, and it is hard to differentially 3. Kleiner DE. Drug-induced liver injury: the hepatic
pathologist’s approach. Gastroenterol Clin N Am.
diagnose hepatotoxicity injury patterns. 2017;46(2):273–96.
Unknown pathogenesis and disease etiologies are 4. Lee BM, Lee WC, Jang JY, Ahn P, Kim JN, Jeong
poorly understood which makes the pathologists’ SW, et al. Clinical features of drug-induced liver
task complex and challenging. An integrated injury according to etiology. J Korean Med Sci.
2015;30(12):1815–20.
approach will be helpful to adopt computational 5. LiverTox: clinical and research information on drug-­
pathology in the clinical setting for DILI detec- induced liver injury. Bethesda: National Institute of
tion at early stages. Computational pathology Diabetes and Digestive and Kidney Diseases; 2012.
with artificial intelligence can add valuable con- 6. DILIN | Drug Induced Liver Injury Network [Internet].
[cited 2021 Nov 18]. Available from: https://dilin.org/.
tributions in helping pathologists in diagnosing DILI 7. Naranjo CA, Busto U, Sellers EM, Sandor P, Ruiz I,
injury patterns and for better prognosis and dis- Roberts EA, et al. A method for estimating the proba-
ease management. bility of adverse drug reactions. Clin Pharmacol Ther.
1981;30(2):239–45.
8. Rochon J, Protiva P, Seeff LB, Fontana RJ,
Key Points Liangpunsakul S, Watkins PB, et al. Reliability of
the Roussel Uclaf Causality Assessment Method
• DILI is a rare condition, a common cause of for assessing causality in drug-induced liver injury.
acute liver failure, and responsible for drug Hepatology. 2008;48(4):1175–83.
9. García-Cortés M, Stephens C, Lucena MI, Fernández-­
withdrawal from the market. Castañer A, Andrade RJ. Causality assessment
• No treatment is available, but discontinuation methods in drug induced liver injury: strengths and
of the drug is often the best approach to reduce weaknesses. J Hepatol. 2011;55(3):683–91.
injury. 10. Tillmann HL, Suzuki A, Barnhart HX, Serrano J,
Rockey DC. Tools for causality assessment in drug-­
• NIH LiverTox website is a great resource for induced liver disease. Curr Opin Gastroenterol.
updated information on DILI drugs, herbals, 2019;35(3):183–90.
dietary supplements, and toxins that cause 11. Xu Y, Dai Z, Chen F, Gao S, Pei J, Lai L. Deep learn-
DILI-associated hepatotoxicity. ing for drug-induced liver injury. J Chem Inf Model.
2015;55(10):2085–93.
• DILI diagnosis is challenging because of 12. Puri M. Automated machine learning diagnostic sup-
overlapping symptoms with other common port system as a computational biomarker for detect-
liver disease phenotypes. ing drug-induced liver injury patterns in whole slide
• DILI is a disease of exclusion, which makes liver pathology images. Assay Drug Dev Technol.
2020;18(1):1–10.
it challenging for human pathologists to assess 13. Cui M, Zhang DY. Artificial intelligence and compu-
DILI related injury. tational pathology. Lab Investig. 2021;101(4):412–22.
• Artificial intelligence has a great potential for 14. Febres-Aldana CA, Alghamdi S, Krishnamurthy K,
detecting DILI injury patterns. Poppiti RJ. Liver fibrosis helps to distinguish auto-
immune hepatitis from DILI with autoimmune fea-
• AuML architecture of ML model is a resource tures: a review of twenty cases. J Clin Transl Hepatol.
for a non-ML expert to run a deep learning 2019;7(1):21–6.
model on diagnosing DILI injury patterns. 15. de Boer YS, Kosinski AS, Urban TJ, Zhao Z, Long
N, Chalasani N, et al. Features of autoimmune hepa-
titis in patients with drug-induced liver injury. Clin
Gastroenterol Hepatol. 2017;15(1):103–112.e2.
16. Allard J, Le Guillou D, Begriche K, Fromenty B. Drug-­
Bibliography induced liver injury in obesity and nonalcoholic fatty
liver disease. Adv Pharmacol. 2019;85:75–107.
1. Opar A. Overtaking the DILI Model-T. Nat Rev Drug 17. Davidson DG, Eastham WN. Acute liver necro-
Discov. 2012;11(8):585–6. sis following overdose of paracetamol. Br Med J.
2. Chen M, Zhang B, Topatana W, Cao J, Zhu H, 1966;2(5512):497–9.
Juengpanich S, et al. Classification and mutation 18. Chan AWH, Quaglia A, Haugk B, Burt A. Atlas of
prediction based on histopathology H & E images in liver pathology. Springer New York: New York; 2014.
liver cancer using deep learning. NPJ Precis Oncol. 19. Hamet P, Tremblay J. Artificial intelligence in medi-
2020;4:14. cine. Metab Clin Exp. 2017;69S:S36–40.
16 Artificial Intelligence-Based Techniques to Assess Drug Toxicity in Drug-Induced Liver Injury (DILI… 283

20. Artificial neural network for drug design, delivery and tal whole slide images informs conversion for clinical
disposition. Amsterdam: Elsevier; 2016. diagnosis. Acad Pathol. 2019;6:2374289519859841.
21. Puri M, Solanki A, Padawer T, Tipparaju SM, Moreno 26. Puri M, Tipparaju S, Moreno W, Bui M, Lloyd
WA, Pathak Y. Introduction to artificial neural network M. Computational analysis for highly accurate diag-
(ANN) as a predictive tool for drug design, discovery, noses. 2014.
delivery, and disposition. In: Artificial neural network 27. Igarashi Y, Nakatsu N, Yamashita T, Ono A, Ohno
for drug design, delivery and disposition. Amsterdam: Y, Urushidani T, et al. Open TG-GATEs: a large-­
Elsevier; 2016. p. 3–13. scale toxicogenomics database. Nucleic Acids Res.
22. Puri M, Lloyd M, Bui M. Role of an artificial neu- 2015;43(Database issue):D921–7.
ral network classifier in nuclear pleomorphic fea- 28. Guo Q, Shao J, Ruiz VF. Characterization and clas-
ture analysis of histopathological images of breast sification of tumor lesions using computerized fractal-­
cancer. In: Artificial neural network for drug design, based texture analysis and support vector machines
delivery and disposition. Amsterdam: Elsevier; 2016. in digital mammograms. Int J Comput Assist Radiol
p. 377–91. Surg. 2009;4(1):11–25.
23. US10733726B2 – Pathology case review, analysis 29. GTEx Consortium. The Genotype-Tissue Expression
and prediction - Google Patents [Internet]. [cited 2021 (GTEx) project. Nat Genet. 2013;45(6):580–5.
Nov 24]. Available from: https://patents.google.com/ 30. Roy M, Wang F, Vo H, Teng D, Teodoro G, Farris AB,
patent/US10733726B2/en. et al. Deep-learning-based accurate hepatic steatosis
24. Puri M, Hoover SB, Hewitt SM, Wei B-R, Adissu quantification for histological assessment of liver
HA, Halsey CHC, et al. Automated computational biopsies. Lab Investig. 2020;100(10):1367–83.
detection, quantitation, and mapping of mitosis in 31. Lusci A, Pollastri G, Baldi P. Deep architectures and
whole-slide images for clinically actionable surgi- deep learning in chemoinformatics: the prediction of
cal pathology decision support. J Pathol Inform. aqueous solubility for drug-like molecules. J Chem
2019;10:4. Inf Model. 2013;53(7):1563–75.
25. Wei B-R, Halsey CH, Hoover SB, Puri M, Yang HH, 32. Yu Y, Wang J, Ng CW, Ma Y, Mo S, Fong ELS, et al.
Gallas BD, et al. Agreement in histological assess- Deep learning enables automated scoring of liver
ment of mitotic activity between microscopy and digi- fibrosis stages. Sci Rep. 2018;8(1):16016.
Drug Dose and Therapy
Individualization
17
Ashley Mason, Gavin Lockard, Vance Cantrell,
Snow Pinxue Li, Kirtan Patel, Sierra Klein,
Andre Elder, Melissa Sur, and Charles Preuss

Abstract ter explores the effects of patients’ comorbidi-


ties and pharmacogenetics, specifically in
This chapter discusses how drug dosing regard to cytochrome P450 (CYP450) metab-
depends on a variety of factors including those olism, infection, obesity, and renal insuffi-
of the drug itself and those of the recipient. ciency in order to highlight how important it is
Principles of pharmacokinetics and pharma- to understand a patient’s individualism when
codynamics can aid drug dosing, as well as prescribing based on treatment algorithms.
prevent drug toxicity and overdose, which is
especially important as the amount of opioid Keywords
and over-the-counter medication overdoses is
on the rise within the United States and world- Enzyme polymorphisms · Opioid overdose ·
wide. Utilizing drug dosing algorithms and Medication overdose · Therapy individualiza-
understanding the varied dosing of the com- tion · Pharmacodynamics · Drug dosing
mon medications warfarin, glipizide, codeine,
and clozapine can help clinicians make
informed decisions on patient care. This chap- 17.1 Introduction
to Pharmacodynamics
Gavin Lockard, Vance Cantrell, Snow Pinxue Li, Kirtan
and Pharmacokinetics
Patel, Sierra Klein, Andre Elder and Melissa Sur contrib-
uted equally with all other contributors. There are multiple subspecialties of medical sci-
ence that focus on the importance of drug dosing.
Pharmacokinetics is the study of how drug dos-
A. Mason (*) · G. Lockard · V. Cantrell · S. P. Li ·
K. Patel · S. Klein · A. Elder · M. Sur
age is affected by its absorption, distribution,
University of South Florida Morsani College of metabolism, and excretion. Alongside influenc-
Medicine, MD Program, Tampa, FL, USA ing drug delivery methodologies, pharmacoki-
e-mail: mason16@usf.edu; gavinlockard@usf.edu; netics determines drug dosing, as different drugs
cantrell20@usf.edu; pinxue@usf.edu; kpatel37@usf.
edu; klein37@usf.edu; adelder@usf.edu;
will have differing bioavailability. Bioavailability
msur@usf.edu is a ratio of the amount of drug available after
C. Preuss
oral consumption versus IV administration; in
University of South Florida Morsani College of drugs with lower bioavailability, physicians will
Medicine, Department of Molecular Pharmacology & have to prescribe more of the drug when given
Physiology, Tampa, FL, USA orally than when given intravenously [1]. Oral
e-mail: cpreuss@usf.edu
drugs can undergo first-pass metabolism, where
© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 285
S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_17
286 A. Mason et al.

they must pass through the liver before being based pharmacokinetic) models, and propagation
absorbed into the body. IV drugs do not undergo models of the diseases to see which drugs directly
this first-pass metabolism and thus do not require affect them [6].
high dosage [2]. Pharmacokinetics also includes Overall, drug individualization is rooted in an
the study of therapeutic indexes, or the ranges understanding of not only drug profiles, but the
within which a drug is clinically effective but patient who will be receiving that drug as well as
nontoxic. Understanding therapeutic indexes is the environmental factors that might affect that
essential to preventing overdose as well as mak- drug’s ultimate effectiveness for that particular
ing sure sufficient dosage is given for an actual patient. This chapter will explore concepts of
effect on the disease process to occur [3]. drug dosing, adjusting prescription practices, and
Other areas of medical science that home in medication prescription individualization in
on drug personalization are pharmacogenetics order to reiterate the importance of drug therapy
and pharmacogenomics. Pharmacogenetics personalization.
explores how individual genes affect a person’s
response to drugs, while pharmacogenomics
explores how a person’s entire genome can pro- 17.2 Influence
vide a spectrum of quantifiable and qualitative of Pharmacodynamics
responses to particular subcategories of drugs and Pharmacokinetics
[4]. Specific genetic polymorphisms are found in on Dosing
specific ethnic groups or are associated with fam-
ily histories, and understanding these differences By definition, pharmacokinetics means looking
can help with adjusting dosages or preventing at how a medication is processed throughout the
adverse outcomes. Specific patients have differ- body, while pharmacodynamics is looking at how
ent isoenzymes, or enzymes with the same action the body is affected by the medication [7]. Both
but slightly different structure or ability to cata- of these processes greatly impact how much of a
lyze a reaction. Most notable of these enzymes is certain drug should be given to a patient and
cytochrome P450, found in the liver, kidneys, and whether or not it should be given at all.
GI tract and throughout the body. Different per- There are four steps that contribute to pharma-
mutations of these cytochrome enzymes are asso- cokinetics – how the drug is absorbed into the
ciated with increased bleeding with warfarin body, how it gets to different parts of the body,
administration, reduced metabolism of lornoxi- how it is processed by the body itself, and how it
cam, and poor metabolism of proton pump inhib- leaves the body [7]. Multiple factors are involved
itors [4]. in each of the steps to further contribute to dos-
Another helpful tool in the clinician’s arsenal ing. For example, it is important to consider if a
to individualize prescribing practices is targeted medication is long-acting or short-acting which
therapy. Targeted therapy is the usage of drugs is based off how it is metabolized. Giving too
that will respond to a specific enzyme mutation, high of a dose of a long-acting drug can be toxic
genetically polymorphic protein, or other unique if it’s in the body for longer than it is supposed to
aspects of the disease of interest. For instance, be. Furthermore, anything that changes the abil-
erlotinib is a tyrosine kinase inhibitor that can ity of a part of the body to absorb a medication
specifically be used for patients whose cancer can affect the pharmacokinetics, such as a sur-
cells express an overactive epidermal growth fac- gery in the gastrointestinal tract which may
tor receptor (EGFR). By being “targeted” to a impact absorption of oral drugs. A condition that
specific mutation or abnormality, these drugs are is detrimental to blood flow can alter how the
less likely to have dangerous off-target side drug is distributed throughout the body if it is
effects and are better suited for that individual’s taken in an intravenous method. A disorder in the
disease and prognosis [5]. Other important tools body’s ability to break down the medication can
include genetic profiling, PBPK (physiology-­ have an effect on the metabolism, while some-
17 Drug Dose and Therapy Individualization 287

thing that influences the end of the gastrointesti- ingly so that the patient is still receiving an
nal tract can affect excretion. In these ways and amount that is clinically effective as a treatment.
more, the differences in pharmacokinetics among On the other hand, if a drug is slower in leaving
people can be markedly significant, making it the system, it is necessary to recognize that hav-
more difficult to generalize a treatment strategy. ing too high of a dose present in the body at once
Thus, it is important to consider the specific may lead to toxicity.
patient as much as possible in making the deci- Another consideration is volume of distribu-
sions regarding dosing. tion which can be related to weight if it is a drug
Therapeutic drug monitoring is a system that normally disburses in the fat tissue [9]. When
greatly impacted by these processes. It requires put together, these are important factors that may
knowing how much of the drug is in the plasma change how much of the drug is given to the
to decide what patients’ accurate dosing will be, patient, although it does require a knowledge of
thus making it a more precise way of treating properties of the drug itself as well. Not all drugs
patients [8]. The Bayesian method of predicting need to consider all of the same factors when
dosing influences therapeutic drug monitoring. It being dosed, as exemplified with the volume of
works specifically with pharmacokinetics by distribution. Some common calculations relating
looking at the differences between the concentra- to weight when dosing are total body weight,
tion of the drug in the patient and how much it ideal body weight, adjusted body weight, lean
should be and the characteristics of the patients body weight, and body surface area [9].
and the general population [8]. The comparison
to average makes it easier to differentiate the
patient’s specific characteristics so that the ther- 17.2.2 Dosing Algorithm
apy can be further tailored to them in the best as Population Based
way possible.
There are numerous factors that play a part in
developing a dosing algorithm for each drug. For
17.2.1 Drug Dosing and Weight example, age, gender, weight, clinical conditions
present in the patient, and characteristics of the
As mentioned previously, several factors must be drug itself are all considered when figuring out
considered when dosing drugs, and one of those how much of a drug a patient should receive. As
considerations is the weight of the patient. such, various methods can contribute to figuring
Because weight can fluctuate throughout the out the ideal dosing for drugs in different
years and it can be so varied among people, it is populations.
important to think about when trying to individu- One of the aforementioned methods is through
alize the treatments of patients to what suits them modelling and simulation [10]. This allows for
best over the period of their care. A heavier or more information to be collected regarding how a
lighter weight makes enough of an important dif- drug works without doing an extensive clinical
ference when trying to understand how much of a trial, thus making the process a lot faster [10].
drug should be administered in a specific case. The efficiency can greatly contribute to develop-
When thinking of obese patients, the ratio of ing dosing algorithms for large populations, as it
the weight of fat tissue to weight of lean tissue quickly generates the amount of data needed for
changes so that there are more of both types of patient dosing. Furthermore, it is less common to
tissues in the body [9]. How fast the drug leaves find information regarding doses that are in
the body is proportional to how much lean tissue between the ones that are already suggested,
there is, so having more would mean that the which modelling and simulation can find [10].
drug would leave faster [9]. If a drug is taken out This allows information to be gained in a new
of the system at a more rapid rate, it may be realm of doses for medications. Moreover, it can
important to recognize that and adjust accord- be used to look at how the safety and efficacy of
288 A. Mason et al.

a drug are impacted by prognostic factors [10]. tase (VKOR encoded by vitamin K epoxide
This is extremely important in coming up with reductase complex 1 (VKORC1) gene). Vitamin
guidelines for dosing, as it greatly changes which K is then further reduced to vitamin K hydroqui-
medications a patient can or cannot take and how none, now ready for use with gamma-glutamyl
much they should be using. These nuances in carboxylase [14] (Fig. 17.2).
guidelines are essential and can have life-­ It is now obvious that the role of vitamin K in
threatening effects if not considered. the conversion of glutamate to gamma-­
carboxyglutamate is absolutely necessary in the
clotting cascade, as demonstrated in the produc-
17.3 Examples of Commonly tion of clotting factor IIa (thrombin), but recall
Individualized Medications that the vitamin K-dependent factors also include
and Their Pharmacology VII, IX, X, and proteins C and S. Without vita-
min K, blood clotting is severely impaired, but
17.3.1 Warfarin this is taken advantage of in medications used to
prevent inappropriate clotting in hypercoagulable
In the event of bleeding, a thrombus is formed via patients. Warfarin is a clinically important vita-
the following sequence of events: vasoconstric- min K antagonist and, by way of its similar ring
tion, platelet plug formation, and activation of the structure to vitamin K, binds to and inhibits
coagulation cascade to ultimately form a cross-­ VKOR [15].
linked fibrin mesh which stops the bleeding. In Warfarin is commonly taken by patients to
the coagulation cascade, vitamin K is critical for prevent venous thromboembolism and to reduce
the biosynthesis of clotting factors II, VII, IX, the risk of ischemic cerebrovascular accident in
and X and antithrombotic proteins C and S [11, patients with atrial fibrillation [16]. Warfarin
12]. To demonstrate the role of vitamin K, the response is monitored clinically to maintain a
prothrombinase complex will be discussed. This patient’s international normalized ratio (INR)
complex requires factor II (prothrombin) as the between 2.0 and 3.0 for most medical conditions,
substrate, factor Xa as the protease, factor Va as but between 2.5 and 3.5 for patients with cardio-
the cofactor, calcium, and anionic phospholipids genic embolus, antiphospholipid syndrome, or
such as phosphatidylserine found on the activated mechanical heart valve. Healthy patients fall
platelet plasma membrane [12]. Vitamin K is between 1 and 2. The INR measures the extrinsic
functionally necessary for the enzyme gamma-­ pathway of coagulation and is necessary in place
glutamyl carboxylase in the conversion of gluta- of the prothrombin time, because of differences
mate to gamma-carboxyglutamate. in manufacturers of biological tissue factor [17,
Gamma-carboxyglutamate residues in the 18]. Recall that the extrinsic pathway involves
579-amino-acid-containing-prothrombin chelate tissue factor and factor VIIa converting X to Xa
calcium involved with the platelet plasma mem- [18, 19].
brane. This allows for factor Xa to cleave pro- It is especially critical to monitor a patient’s
thrombin at codons Arg271 and Arg320 from the initial response to warfarin as there is wide vari-
remainder of the protein, to form thrombin (IIa) ability in each patient’s pharmacokinetics and
[13] (Fig. 17.1). pharmacodynamics with this drug, and the INR
Gamma-glutamyl carboxylase is a vitamin K can help direct the clinician to prescribe the
hydroquinone-dependent hepatic enzyme resid- appropriate dose. Of note, since gamma-­
ing in the endoplasmic reticulum that converts carboxylation of glutamate residues occurs
the prozymogen clotting factors into gamma-­ shortly after protein translation, warfarin therapy
carboxylated prozymogens at the time of protein must be bridged with another anticoagulant to
synthesis. Upon this conversion, vitamin K epox- allow pre-existing clotting factors (especially
ide is released which is then reduced to vitamin K factor X and prothrombin) to naturally deplete.
via the hepatic enzyme vitamin K epoxide reduc- Prothrombin has the longest half-life at 3 days
17 Drug Dose and Therapy Individualization 289

Fig. 17.1 This figure demonstrates the conversion of a cofactor) to cleave prothrombin at sites Arg271 and
prothrombin to thrombin. Gamma-carboxyglutamate resi- Arg320, resulting in the thrombin product. Of note,
dues on prothrombin chelate calcium associated with the thrombin is composed of two protein chains connected by
platelet plasma membrane, allowing factor Xa (with Va as a disulfide bridge. Note this figure is not to scale

Fig. 17.2 This figure demonstrates gamma-glutamyl car- min K-dependent enzyme; this figure shows vitamin K’s
boxylase’s role in the conversion of glutamate to gamma-­ role and how warfarin inhibits it and thus the clotting
carboxyglutamate which is critical for proper clotting factor
factor function. Gamma-glutamyl carboxylase is a vita-
290 A. Mason et al.

[20, 21]. Thus bridging must last for 3 days after will not saturate at physiological concentrations
warfarin initiation. Interestingly, proteins C and of glucose and would otherwise starve the
S, which naturally inhibit factors V and VIII, are remainder of the body.
also vitamin K-dependent; therefore, there is an Upon phosphorylation via glucokinase, the
initial paradoxical procoagulant effect of warfa- glycolysis, citric acid cycle, and oxidative phos-
rin that is short-lived and rarely clinically signifi- phorylation pathways are completed resulting in
cant [22]. an increased intracellular adenosine triphosphate
VKORC1 and CYP2C9 are key in warfarin (ATP)/adenosine diphosphate ratio. The increased
pharmacogenetics and individual patient ATP inhibits ATP-sensitive potassium channels
response. Warfarin primarily inhibits vitamin K located on the cell membrane, preventing hyper-
epoxide reductase. Single-nucleotide polymor- polarization and subsequently resulting in depo-
phisms in VKORC1 have been found to alter war- larization of the cell. Voltage-gated calcium
farin sensitivity [23]. Additionally of note, channels open, and increased intracellular cal-
warfarin is a racemic drug. Cytochrome P450 cium concentrations result in exocytosis of
2C9 (CYP2C9) primarily metabolizes S-warfarin, insulin-­containing granules (Fig. 17.3).
and studies demonstrate that CYP2C9 polymor- Increased insulin release benefits patients with
phisms affect patient response [24]. In terms of type II diabetes mellitus. Glipizide is a sulfonyl-
clinical significance, it is recommended that if urea medication that inhibits ATP-sensitive
testing of these genetics is not performed, then a potassium channels in pancreatic beta cells, thus
clinician should begin a patient on a low starting inducing insulin release [31]. Glipizide is metab-
dose and then increase this dose slowly while olized by the cytochrome P450 2C9 (CYP2C9).
monitoring the INR carefully. If testing is com- Two variants, CYP2C9*2 (R144C) and
pleted and shows variations, a safer, lower dose CYP2C9*3 (I359L), have been shown to cause
can be initiated [24]. However, routine genetic reduced enzymatic activity and thus decreased
testing is not recommended at this time [26]. glipizide metabolism [32]. One study showed
that patients with two variant alleles were 3.4×
more likely to reach a target hemoglobin Alc of
17.3.2 Glipizide <7, compared to patients with two wildtype
alleles, and that patients with variant alleles were
Pancreatic beta cells are constantly measuring less likely to fail sulfonylurea monotherapy33. In
blood glucose concentrations so that they are able patients carrying the CYP2C9*3 allele, oral
to release commensurate amounts of insulin. This clearance of glipizide is reduced, resulting in
is accomplished via the glucokinase receptor, increased plasma drug concentrations. In one
present only in the liver and pancreas [27, 28]. study, sulfonyl-treated patients with the
First, glucose enters the pancreatic beta cell via CYP2C9*3/*3 or *2/*3 genotypes had a 5.2×
the low affinity GLUT2 transporter, which will increased risk of a severe hypoglycemic incident
increase with chronic hyperglycemia [29, 30]. [33]. Knowing a patient’s pharmacogenomics
The rate-controlling step in glucose-stimulated can assist a physician in choosing a medication or
insulin release involves glucokinase phosphory- a certain dosage to avoid adverse effects and
lating glucose to glucose-6-phosphate [28]. achieve an ideal treatment goal.
Glucokinase differs from hexokinase in terms of
enzyme kinetics; glucokinase is low affinity and
of high maximum velocity, and hexokinase is the 17.3.3 Codeine
opposite, though both catalyze glucose to
glucose-­6-phosphate. The low affinity of gluco- Codeine is a mild/moderate agonist of opioid
kinase is critical to allow the remainder of the receptors. Clinical indications may include noci-
body to obtain glucose via the high-affinity hexo- ceptive pain, psychogenic pain, and breakthrough
kinase, because glucokinase, with its high Vmax, pain. Opioid receptors are Gi-protein coupled
17 Drug Dose and Therapy Individualization 291

Fig. 17.3 This figure demonstrates the biochemistry of nels, preventing hyperpolarization of the cell.
insulin secretion in a pancreatic beta cell. Glucose under- Subsequently, the cell depolarizes, activating voltage-­
goes uptake via GLUT2 and is converted by glucokinase gated calcium channels and resulting in an influx of cal-
to glucose-6-phosphate, which then undergoes typical cium. Increased intracellular calcium causes exocytosis of
metabolism resulting in ATP production. The increased insulin granules
ATP: ADP ratio inhibits ATP-sensitive potassium chan-

receptors (GiPCR) located in areas including the SNARE proteins and thus fuse intracellular vesi-
brain, spinal cord, and gastrointestinal tract and cles to the cell membrane for exocytosis of
are approximately found in the same areas that neurotransmitters.
endorphins are synthesized. Endorphins are Activation of opioid receptors located within
endogenous peptides that act on opioid receptors the periaqueductal gray (PAG) reduces emotional
[33]. There are three clinically important catego- response to pain by reducing signals from the
ries of opioid receptors with similar structure and PAG to the forebrain and amygdala and reduces
function though different distribution: mu, delta, perception of pain by inhibiting the inhibitory
and kappa []. gamma-aminobutyric acid neurons within the
GiPCRs inhibit adenylate cyclase which sub- PAG, which subsequently stimulates endorphin
sequently decreases concentrations of cyclic ade- release onto spinal cord neurons [36].
nosine monophosphate (cAMP). This directly Codeine is a prodrug and is clinically effective
reduces neurotransmitter release. Opioid recep- once converted into morphine via CYP2D6. Per
tors also have two other mechanisms; they reduce the Clinical Pharmacogenetics Implementation
the likelihood of action potentials by hyperpolar- Consortium (CPIC), in CYP2D6 ultra-rapid
izing neurons by stimulating potassium efflux, metabolizers, codeine should be avoided second-
and they reduce neurotransmitter release by ary to the risk of toxicity [37, 38]. In CYP2D6
inhibiting voltage-gated calcium channels [35]. poor metabolizers, codeine should be avoided
Recall that calcium influx is critical to bind to secondary to lack of efficacy [25].
292 A. Mason et al.

17.3.4 Clozapine clozapine use [45]. If a clinician were to identify


a patient at risk, they would likely avoid clozap-
Antipsychotics are often used to treat severe psy- ine in the treatment of schizophrenia and select
chotic disorders such as schizophrenia. Clinically another antipsychotic for treatment.
effective concentrations of antipsychotics corre-
late with dopamine D2 receptor antagonism.
Antipsychotics inhibit D2 receptors along the 17.4 Drug Dosing in Patients
mesocortical (ventral tegmental area releases with Comorbidities
dopamine onto frontal cortex), mesolimbic (ven-
tral tegmental area releases dopamine onto 17.4.1 Involvement of CYP450
nucleus accumbens), nigrostriatal (substantia in Drug Metabolism
nigra releases dopamine onto striatum), and
tuberoinfundibular (hypothalamus releases dopa- Cytochrome P450 is a collection of enzymes,
mine on pituitary gland) pathways, though thera- mainly found in the liver but also in the small
peutic effects arise from antagonism of the intestine, lungs, kidneys, and placenta, involved
mesolimbic and mesocortical pathways. D2 is a in the metabolism of drugs and toxins. There are
Gi/o-protein coupled receptor, which will inhibit six specific enzymes in this family, CYP1A2,
adenylate cyclase and subsequently decrease CYP2C9, CYP2C19, CYP2D6, CYP3A4, and
concentrations of cAMP. CYP3A5, which are involved in the metabolism
Clozapine was the first developed second-­ of 90% of drugs. The activity of these enzymes
generation antipsychotic, so-called atypical can be affected in comorbid conditions in the
because it is less likely to cause extrapyramidal patient which may require adjusting the dosages
symptoms like the typical antipsychotics, e.g., of drugs that are metabolized by it. This section
haloperidol. This is possibly due to its transient will look at drug dosing in patients with
binding with D2 receptors. Clozapine also binds comorbidities, specifically infection, obesity,
­
to D1; D3; D4; D5; histamine H1; acetylcholine hepatic insufficiency, renal insufficiency, and
muscarinic M1; serotonin 5-HT2A, 5-HT2C, pregnancy.
5-HT6, and 5-HT7 receptors; and alpha 1 adreno-
receptors [39].
Studies have shown clozapine to be more 17.4.2 Infection
effective than other typical antipsychotics in
treatment-resistant patients [40], and in treating Infection and inflammatory states have been
schizophrenic patients with persistent suicidal shown to decrease the activity of CYP450. The
ideation, clozapine reduces suicide attempts [41]. interferon (IFN) hypothesis states that depression
However, use is restricted to the aforementioned of CYP450 enzymes is a common property of
patients due to its potential severe side effect pro- interferons and factors that induce interferons.
file including agranulocytosis [42], seizures [43], Studies have shown that many inflammatory
and myocarditis [41], among others. cytokines are also involved in the depression of
HLA-DQB1 is a genetic marker found to be CYP450 during inflammatory states. Due to the
disproportionately represented in patients who inhibition of the CYP450 system, infection and
later resulted with a leukocyte count <500/μL inflammatory states are thus implicated in
while being treated with clozapine, compared to impaired drug metabolism [46]. In an infection,
patients treated for at least 1 year with clozapine drug concentrations may be increased due to not
and with normal leukocyte and absolute neutro- being able to be metabolized by CYP450.
phil counts. PGxPredict:Clozapine (Clinical Current viral infections that have been found
Data, Inc., New Haven, CT) is a tool that will uti- to depress the CYP450 system in humans include
lize this marker to allow for the identification of hepatitis A, influenza A and B, adenovirus, HSV,
patients at risk of agranulocytosis secondary to and HIV. Specifically, influenza A has been
17 Drug Dose and Therapy Individualization 293

shown to decrease clearance of theophylline in rather drug specific. Sufentanil has been shown
the pediatric asthmatic population. This is likely to increase in the Vd as TBW increases, but remi-
due to the increase in endogenous IFN, which is fentanil has shown a Vd similar to hydrophilic
released in response to influenza, which depresses drugs, as its Vd positively correlates with IBW,
CYP1A2, the primary enzyme responsible for the rather than TBW [48].
metabolism of theophylline [46]. The metabolism of drugs in obese patients
can be altered as there are changes in concentra-
tions and activities of CYP450 enzymes.
17.4.3 Obesity Specifically, CYP3A4 has been shown to be
downregulated in obesity, resulting in a decreased
Drug dosing for patients based on weight is clearance of alprazolam, midazolam, fentanyl,
dependent on four main pharmacokinetic factors: carbamazepine, and cyclosporine. The enzyme
absorption, distribution, metabolism, and elimi- CYP2E1 has been shown to be upregulated in
nation. There are many biometrics that a clinician obesity, resulting in increased metabolism of
can use to quantify the size of a patient: body chlorzoxazone, enflurane, sevoflurane, halo-
mass index (BMI), total body weight, ideal body thane, and acetaminophen. The phase II conju-
weight, body surface area, and lean body weight gating enzyme UGT (uridine
[48]. 5′-diphospho-glucuronosyltransferase) has been
Oral drug absorption has not been shown to be shown to be upregulated in obesity, resulting in
significantly affected in obese patients. Animal increased glucuronidation of acetaminophen,
and human studies have found that some paren- oxazepam, and lorazepam [49]. Obesity is also
teral drug absorption is negatively affected by an associated with increased concentrations of
increase in BMI. Specifically, subcutaneous insu- pseudocholinesterase, resulting in increased
lin and enoxaparin injections are associated with metabolism of succinylcholine [1].
delayed systemic absorption, as weight increases. The changes in elimination of drugs in obe-
Obesity is also associated with gastric bypass sity are largely due to changes in kidney func-
surgery, which can lead to a decrease in absorp- tion. Initially, obesity is associated with an
tion of certain nutrients and drugs, specifically increase in glomerular filtration rate (GFR),
those that require an acidic environment or which would result in an increase in clearance.
require duodenal intestinal transporters. However, obesity is an independent risk factor
Specifically, cyclosporine, tacrolimus, thyroxine, for focal and segmental glomerulosclerosis,
phenytoin, and rifampin have been shown in case which decreases GFR, thereby decreasing drug
reports to have diminished absorption following clearance. The changes in volume of distribu-
gastric bypass surgery [48]. tion will also affect the renal elimination of a
Volume of distribution (Vd) of a drug mainly drug, as with an increase in Vd, there is less
appears to rely on the lipophilicity or hydrophi- serum drug concentration, leading to decreased
licity of the drug, as it relates to obesity. In gen- excretion [48].
eral, charged hydrophilic drugs, such as lithium,
are contained to the water compartments of the
body, and their distributions are not significantly 17.4.4 Renal Insufficiency
affected by changes in adipose tissue.
Hydrophilic drugs, such as lithium, are better Patients with renal insufficiency, whether it be
dosed based on the ideal body weight (IBW) acute or chronic, require special attention when it
rather than the total body weight. Furthermore, it comes to drug dosing. The pharmacokinetic
can be predicted that lipophilic drugs could be parameters that typically change in decreased
better dosed based on total body weight (TBW), kidney function include volume of distribution
as they are more likely to deposit in adipose tis- (Vd) and the clearance (CL). These changes are
sue, but it is not a generalized rule, and it is important because the therapeutic index window
294 A. Mason et al.

can be easily exceeded when changes in Vd or CL 17.5 Implications of Incorrect


are not taken into account in drug dosing, which Drug Dosing
could result in drug toxicity. Conversely, under-
dosing due to these changes, in an effort to pro- 17.5.1 Drugs with a Significant Risk
tect from adverse reactions, could result in of Overdose: Opioids
therapeutic failure, which is particularly impor-
tant in acute situations, such as infection or Opioids are powerful drugs that can dramatically
immunosuppression [50]. reduce the acute or chronic pain a patient experi-
Changes in the Vd are unpredictable in chronic ences both quickly and effectively. However, the
kidney disease (CKD) and vary between drugs. statistics on the dangers of opioid prescriptions
Fluid expansion and edema, due to protein loss for long-term pain management are startling and
during CKD, could lead to increase in Vd, worth addressing here. According to the Centers
although some studies have shown no change at for Disease Control and Prevention [52], one out
all. This change in fluid expansion most likely of every four patients receiving long-term opioid
affects hydrophilic drugs rather than lipophilic therapy, e.g., oxycodone, develops opioid sub-
drugs [51]. stance use disorders (SUDs). In recent years,
Initial dosing for acute kidney injury (AKI) there have been 38 overdose deaths a day involv-
and CKD depends on the type of drug as well as ing prescription opioid use [53].
the condition it treats. A higher loading dose Therefore, it is strongly recommended that
might be necessary if there is significant fluid opioid therapy be a last resort treatment option
overload as a result of the AKI or the CKD, espe- when other treatment options have not been
cially if the drug is hydrophilic or if the disease effective in controlling chronic pain [54]. To mit-
that is being treated is a life-threatening infection igate the inherent risk of opioid use for patients,
[51]. The maintenance dosing of a drug will cor- the CDC and other governmental and profes-
relate with the degree of reduced kidney function. sional entities have outlined guidelines that medi-
If kidney function is reduced <50%, maintenance cal providers should consider when determining
dosing is not changed. If kidney function is the appropriateness of opioid prescriptions for
reduced ≥50%, maintenance dosing is adjusted patients. Table 17.1 is an adapted summary of the
according to the eq. MD = CL * target concentra- main points set forth by the CDC [54].
tion [51]. Morphine milligram equivalents (MME)
AKI poses high variability between patients, were created to help providers keep track of
making drug dosing and maintenance difficult to patient opioid dosing and to help weigh the
achieve for clinicians in a short time. An increase potential risks of a current treatment plan with
in Vd is very common in AKI, resulting in many the potential benefits [55]. It is recommended
loading doses for antimicrobials to be insufficient that physicians and providers should keep
for the sepsis it is attempting to combat. Methods patients undergoing long-term opioid therapy
used to quantify AKI are plasma creatinine con- for chronic pain below 50 MME/day [54].
centration and urine output. Quantification of According to the CDC, going above 50 MME/
plasma creatinine can be deceiving as IV fluid day has shown little additional efficacy in reduc-
and dialysis could act to slow the increase in con- ing pain but has shown great increases in patient
centration. Two other methods of measuring GFR odds for prescription opioid overdose and opi-
during a single point in time are measuring cre- oid SUD [54]. To calculate MME, multiply the
atinine clearance (CrCL) in patients without total dosage of a given medication within a
anuria over 2–12 hours or measuring GFR as an 24-hour period by that medication’s relative
exogenous compound. AKI has also been associ- strength which is its conversion factor (CF)
ated with the decrease in function of certain [55]. Sum all values of MME together for each
CYP450 enzymes, specifically CYP3A4/5, with medication prescribed for the total MME for
the substrate midazolam [51]. that patient:
17 Drug Dose and Therapy Individualization 295

Total Dosage  CF  MME the UK) [60, 61]. Teens are more likely to abuse
dextromethorphan or any OTC medication more
MME1  MME 2  MME 3  Total MME than seniors [61]. Females are more likely to
abuse barbiturates and sedatives more than males
Table 17.2 contains a number of common opi- [61]. US patient populations are less likely to
oids, their conversion factors, and example abuse codeine-containing products than patients
prescriptions: in European countries [60].
Keep in mind that there are a number of mobile- Unfortunately, statistical studies on the preva-
based applications that can assist physicians in cal- lence of OTC drug overdose are still not as robust
culating and managing these values for patients. as the data available on opioid prescription over-
Every patient undergoing long-term opioid doses [62].
therapy for chronic pain should also be given nal- Intentional OTC overdosing is done for vari-
oxone to help prevent accidental overdose [56, ous reasons. Aside from recreational use, some
57]. There are several different forms of nalox- individuals do so to commit suicide through the
one available. For example, there are nasal ingestion of either diphenhydramine or acetamin-
sprays, auto-injectors, and syringes with ampules ophen [63], while others misuse medications like
[58]. Each form of naloxone is straightforward loperamide in an attempt to stave off opioid with-
and user-friendly; however, training (and mainte- drawal symptoms and accidentally reach a toxic
nance training) should take place for both patients threshold [64, 65].
and physicians in anticipation of emergencies. The below table contains commonly misused
OTC medications, known or estimated lethal
thresholds, and a non-exhaustive list of treatment
17.5.2 Drugs with a Significant Risk options available (Table 17.3).
of Overdose: OTC Medications For treatment advice on OTC medication
overdoses or poison-related emergencies, contact
There is an abundance of OTC medications that the national Poison Help hotline (USA) for assis-
have high abuse potentials [59], for example, tance (1-800-222-1222).
codeine-containing products, acetaminophen, Medical community awareness and action on
antihistamines, pseudoephedrine, dextrometho- medication abuse do have a positive impact on
rphan, and laxatives to name a few [59, 60]. patient populations. For instance, abuse trends
Abuse, misuse, and overdose trends for OTC have declined significantly for drugs like dextro-
medications appear to be based on both patient methorphan due in part to medical community
population demographics (e.g., age, sex, etc.) and advocacy, pharmacy retail policy changes, and
geographic region (e.g., the USA compared to FDA involvement [71].

Table 17.1 Summary of 12 CDC opioid recommendations 2016


1. Use alternative and non-opioid 5. Start at the lowest effective 9. Review prescription drug monitoring
Rx therapies first dose. Stay well below 90 program (PDMP) data at the start and
MME then every 3 months
2. Set realistic goals with patients 6. Acute pain opioid Rx should 10. Perform urine drug testing at the start
and commit to reduction and be less than 3 days and then annually to rule out illicit
discontinuation plans drug use
3. Routinely go over patient and 7. Follow up within 3 weeks of 11. Avoid concurrent use of
physician responsibilities start of opioid therapy and benzodiazepine and opioid drug
then every 3 months or less prescriptions when possible
4. Prescribe immediate-release 8. Prescribe naloxone when at 12. If opioid SUD is determined,
opioids and avoid extended-­ 50 MME, or if other risk recommend behavioral therapy with
release when possible factors are present buprenorphine or methadone treatment
Adapted from CDC Guideline for Prescribing Opioids for Chronic Pain — United States, 2016 [54]
296 A. Mason et al.

Table 17.2 MME doses for commonly prescribed opioids and example prescriptions
CDC MME recommendation Prescription and MME example
Conversion factor MME
Opioid (CF) Dosage Frequency example
Codeine 0.15 15 mg 1 tablet PO prn every 9
6 hours
Fentanyl 2.4 12.5 mcg/h 1 patch every 3 days 30
transdermal
Hydrocodone 1 2.5 mg/325 mg 1 tablet PO prn every 15
(acetaminophen) 4 hours
Hydromorphone 4 2 mg 1 tablet PO prn every 32
6 hours
Methadone 4 5 mg 3 tablets PO daily 60
Morphine 1 60 mg 1 tablet PO prn every 60
6 hours
Oxycodone 1.5 5 mg/325 mg 1 tablet daily PO q6hr 30
(acetaminophen)
Oxymorphone 3 5 mg 1 tablet PO prn every 60
6 hours
Tapentadol 0.4 250 mg 1 tablet PO prn every 120
12 hours
Adapted from CDC Guideline for Prescribing Opioids for Chronic Pain — United States, 2016 [54]

Table 17.3 Commonly abused OTC drugs, estimated minimum toxic dose, and available treatment options
Drug name Estimated minimum toxic dose Emergency treatment options
Acetaminophen Adults: 7 g–10 g Activated charcoal
Children: 150 mg/kg; 200 mg/kg in healthy N-Acetylcysteine (NAC)
children aged 1–6 years [66] Stomach pump
Antihistamines Adults: 20–40 mg/kg or 3–5 times usual dose Activated charcoal
Children: At or over 7.5 mg/kg [67, 68] IV fluid for tachycardia
Urine catheterization
Stomach pump
Pseudoephedrine Not well characterized. One case study suggests Activated charcoal
3–4 times therapeutic dose, or over 1 gram [69] Closely monitor vital signs
Sudden drop in BP, tachycardia, febrile, and
altered mental status may occur
Stomach pump [67]
Dextromethorphan Adults: 20–30 mg/kg Activated charcoal
Exceeding 300 mg can cause dissociative Benzodiazepines, e.g., lorazepam if patient
psychosis; >600 mg can cause comas and becomes combative
eventual death [69, 70] Cold IV, inspired oxygen
Naloxone
Loperamide Adults: >100 mg with median dose around ACLS for cardiac dysrhythmias
250 mg [64, 65] Activated charcoal
Naloxone

17.5.3 Overdose Prevention as Part practices. Overdose has its origin in misuse of
of Drug Individualization substances, poor patient understanding of medi-
cations, and socioeconomical disparities. It is
Individualization of drug prescription practices influenced through individual perceptions of
can have a dramatic effect on reducing overdose drugs’ usage and role for treatments, social
risk and as such is firmly rooted in the necessity behavior behind drug usage, and lack of general
of patient education and responsible prescription knowledge about drugs due to lack of either edu-
17 Drug Dose and Therapy Individualization 297

cation in schools or education when receiving before taking them concurrently. Although drug
prescriptions [72]. packaging has changed to attempt to draw
One area where individualization is lacking attention to those active ingredients, education
within the pharmaceutical injury is with over-the-­ in the form of public service announcements
counter medications (OTC), as these come with and warnings about common toxicities, such as
“one size fits all” packaging. Because they are acetaminophen poisoning, can help to reduce
not as tightly regulated and do not require a pre- overdose risk [73].
scription, patients will often take over-the-­ Opioid misuse is another area where drug
counter medications without consideration of individualization is necessary to prevent over-
their own individual genetics and comorbidities dose and toxicity. Individualizing how opioids
or the pharmacodynamics of those drugs. Thus, are prescribed can have dramatic effects on
education about the associated risk of OTC medi- decreasing the harmful effects of opioid over-
cations is a necessity, as it can show patients how dose, as well as the associated elevated risk of
their own individual characteristics (such as renal developing blood-borne infections and the terato-
failure, previous liver damage, a history of hyper- genic effects on unborn fetuses [74]. Increased
coagulability) can be affected by those drugs. opioid dosage increases overdose risk; thus, one
Patients might see OTC medications as safe to should try to limit the daily maximal dosage of
take as they do not require prescription or control opioids their patients take. In addition, patients
by a medical professional and tend to form cogni- on extended-release or long-acting opioids are
tive shortcuts in their understanding of medica- more likely to overdose, as well as have depres-
tions’ mechanisms (and adverse effects). One sion, drug use disorder, or chronic pain [75].
such shortcut of understanding is that medicine is Specific drugs are also associated with higher
seen as having a targeted approach, only treating rates of opioid overdose specifically oxycodone,
the medical ailment of interest and not affecting hydromorphone, and methadone. Another neces-
other parts of the body [73]. For instance, a sity is to cross-reference the patient’s history for
patient might believe that acetaminophen only medications that may react with opioids or
treats their headache and might not consider that increase risk of drug misuse. Sedative hypnotics
it will pass through and be metabolized by the (especially benzodiazepines, e.g., diazepam)
liver, thus missing the critical side effect of have increased risk of overdose due to their
potential liver damage. These misunderstandings intrinsic CNS effects [75].
persist through varied medication types and are Another aspect of overdose that requires indi-
compounded by the variation between brand vidualization is in the realm of intentional over-
names and off-brand medications that all have dose, or attempted suicide by consumption of
similar active ingredients. drugs. Opioid drugs, both illicitly obtained and
Alternatively, patients may take concurrent prescribed, are commonly utilized in intentional
OTC medications that have the same major overdoses. OTC drugs and controlled substances
active ingredient due to lack of medical under- that were prescribed for mental health disorders
standing about those ingredients. This adverse are also used for intentional overdose. Opioids
event, termed “double dosing,” is a common are the most often cause of death due to inten-
mistake. Patients may utilize pain relief medi- tional overdose, followed by barbiturates, antide-
cations at the same time as cold relief medica- pressants, antidiabetics, and alcohol [76].
tions or alternatively may utilize similar drugs Notably, of these classes, alcohol is readily avail-
in the belief it will help relieve symptoms faster. able with little to no regulation, and the other
Often, patients without medical training will drugs are prescribed to patients with an increased
not actively look at active ingredients listed on risk of suicide (e.g., antidepressants, which are
drug packaging; in stark contrast, those with prescribed to lower depression’s increased risk of
medical training have the foreknowledge to suicide). Although both teenagers and adults
consider the ingredients in various medications attempt suicide, adults have more lethal drugs at
298 A. Mason et al.

their disposal, meaning their success rate with utilization of prescriptions that are optimized to
intentional overdose is higher [76]. the patient and that also address comorbidities
Alongside the access to these medications and social predeterminants of health.
with known toxicities, physicians must focus on
the risk of suicide in their individual patients.
Alcohol and opioid use disorders significantly References
increase the risk for suicidal ideation, attempts,
and deaths, with any substance use (including 1. Offerman S, Rosenthal W. Bioavailability.
Encyclopedia of molecular pharmacology. Berlin,
marijuana, cocaine, amphetamines) increasing Heidelberg: Springer; 2008. https://doi-­org.ezproxy.
the risk for suicide ideation [77]. Risk factors for lib.usf.edu/10.1007/978-­3-­540-­38918-­7_5268.
suicide include job loss, divorce, mental illness, 2. Atkinson AJ. Individualization of drug therapy:
physical illness, chronic pain, stress, advanced an historical perspective. Transl Clin Pharmacol.
2014;22(2):52–4. https://doi.org/10.12793/
age, and financial difficulties. Male and Caucasian tcp.2014.22.2.52.
patients also have a higher incidence of suicide 3. Pater, C. Individualizing therapy – in search
attempts [78]. Prevention of overdose-related of approaches to maximize the benefit of drug
suicide, therefore, must be a multifaceted treatment (II). Trials. 2004;5:7. https://doi.
org/10.1186/1468-­6708-­5-­7.
approach, focusing both on the suicidal ideation 4. Shastry B. Pharmacogenetics and the concept of
and the access to dangerous medications. individualized medicine. Pharmacogenomics J.
Disposing properly of leftover drugs, locking up 2006;6:16–21. https://doi.org/10.1038/sj.tpj.6500338.
dangerous medications, and educating patients 5. Smith J. Erlotinib: small-molecule targeted therapy in
the treatment of non-small-cell lung cancer. Clin Ther.
on which drugs might have dangerous interac- 2005;27(10):1513–34. https://doi.org/10.1016/j.
tions if in the hands of a person who has suicide clinthera.2005.10.014. PMID: 16330289.
risk factors can all help to reduce availability of 6. Lesko LJ, Schmidt S. Individualization of drug ther-
drugs for suicide attempts. Design of drug pack- apy: history, present state, and opportunities for the
future. Clin Pharmacol Ther. 2012;92(4):458–66.
aging can also reduce risk, as blister packing will https://doi.org/10.1038/clpt.2012.113.
slow the ability of persons to open the lethal 7. Negus SS, Matthew LB. Pharmacokinetic-­
drugs, causing them to instead choose an easier Pharmacodynamic (PKPD) analysis with
to use drug (and often a less toxic drug) for the drug discrimination. Curr Top Behav
Neurosci. 2018;39:245–59. https://doi.
overdose in the moment [76]. org/10.1007/7854_2016_36.
8. Fuchs A. Implementation of Bayesian therapeu-
tic drug monitoring in modern patient care. Diss.
17.6 Conclusion Université de Lausanne, Faculté de biologie et méde-
cine; 2015.
9. Barras M, Legg A. Drug dosing in obese adults. Aust
Although adjusting drug dosing to individual Prescr. 2017;40(5):189–93. https://doi.org/10.18773/
patients’ needs requires an understanding of austprescr.2017.053.
complex topics such as pharmacokinetics, phar- 10. Gobburu JV, Marroum PJ. Utilisation of
pharmacokinetic-­ pharmacodynamic modelling and
macogenetics, and drug biochemistry, it provides simulation in regulatory decision-making. Clin
an essential tool for effectively addressing Pharmacokinet. 2001;40(12):883–92. https://doi.
patients’ maladies. In addition, understanding org/10.2165/00003088-­200140120-­00001.
how these factors differ between patients can pre- 11. Vermeer C, De Boer-Van den Berg MA. Vitamin
K-dependent carboxylase. Haematologia (Budap).
vent undesired toxicities, side effects, and over- 1985;18(2):71–97. PMID: 3896983.
dose. As discussed in this chapter, a thorough 12. Sims PJ, Wiedmer T, Esmon CT, Weiss HJ, Shattil
understanding of the more common medications SJ. Assembly of the platelet prothrombinase com-
associated with differing pharmacokinetics and plex is linked to vesiculation of the platelet plasma
membrane. Studies in Scott syndrome: an isolated
metabolism ensures therapeutic doses are pre- defect in platelet procoagulant activity. J Biol Chem.
scribed accurately. As medicine becomes more 1989;264(29):17049–57. PMID: 2793843.
personalized, these concepts enable the clinical
17 Drug Dose and Therapy Individualization 299

13. Chinnaraj M, Chen Z, Pelc LA, et al. Structure of 27. Matschinsky FM. Banting Lecture 1995. A lesson
prothrombin in the closed form reveals new details on in metabolic regulation inspired by the glucokinase
the mechanism of activation. Sci Rep. 2018;8:2945. glucose sensor paradigm. Diabetes. 1996;45(2):223–
https://doi.org/10.1038/s41598-­018-­21304-­1. 41. https://doi.org/10.2337/diab.45.2.223. PMID:
14. Suttie JW. Vitamin K-dependent carboxylase. 8549869.
Annu Rev Biochem. 1985;54:459–77. https://doi. 28. Matschinsky F, Liang Y, Kesavan P, et al. Glucokinase
org/10.1146/annurev.bi.54.070185.002331. PMID: as pancreatic beta cell glucose sensor and diabetes
3896125. gene. J Clin Invest. 1993;92(5):2092–8. https://doi.
15. Liu S, Li S, Shen G, et al. Structural basis of antago- org/10.1172/JCI116809. PMID: 8227324; PMCID:
nizing the vitamin K catalytic cycle for anticoagula- PMC288386.
tion. Science. 2021;371:eabc5667. 29. Yasuda K, Yamada Y, Inagaki N, et al. Expression of
16. Shapiro SS. Treating thrombosis in the 21st century. GLUT1 and GLUT2 glucose transporter isoforms
N Engl J Med. 2003;349:1762–4. in rat islets of Langerhans and their regulation by
17. Smith SA, Morrissey JH. Properties of recom- glucose. Diabetes. 1992;41(1):76–81. https://doi.
binant human thromboplastin that determine the org/10.2337/diab.41.1.76. PMID: 1370154.
International Sensitivity Index (ISI). J Thromb 30. Liang Y, Cushman SM, Whitesell RR, Matschinsky
Haemost. 2004;2:1610. FM. GLUT1 is adequate for glucose uptake in
18. Morrissey JH. Tissue factor: an enzyme cofactor and GLUT2-deficient insulin-releasing beta-cells.
a true receptor. Thromb Haemost. 2001;86(1):66–74. Horm Metab Res. 1997;29(6):255–60. https://doi.
PMID: 11487043. org/10.1055/s-­2007-­979032. PMID: 9230344.
19. Butenas S. van’t Veer C, Mann KGEvaluation of the 31. Aguilar-Bryan L, Nichols CG, Wechsler SW, et al.
initiation phase of blood coagulation using ultra- Cloning of the beta cell high-affinity sulfonylurea
sensitive assays for serine proteases. J Biol Chem. receptor: a regulator of insulin secretion. Science.
1997;272(34):21527–33. https://doi.org/10.1074/ 1995;268(5209):423–6. https://doi.org/10.1126/sci-
jbc.272.34.21527. PMID: 9261172. ence.7716547. PMID: 7716547.
20. Lind SE, Callas PW, Golden EA, et al. Plasma levels 32. Kirchheiner J, Roots I, Goldammer M, Rosenkranz
of factors II, VII and X and their relationship to the B, Brockmöller J. Effect of genetic polymorphisms
international normalized ratio during chronic warfarin in cytochrome p450 (CYP) 2C9 and CYP2C8 on the
therapy. Blood Coagul Fibrinolysis. 1997;8:48. pharmacokinetics of oral antidiabetic drugs: clinical
21. Zivelin A, Rao LV, Rapaport SI. Mechanism of the relevance. Clin Pharmacokinet. 2005;44(12):1209–25.
anticoagulant effect of warfarin as evaluated in rabbits https://doi.org/10.2165/00003088-­200544120-­00002.
by selective depression of individual procoagulant PMID: 16372821.
vitamin K-dependent clotting factors. J Clin Invest. 33. Holstein A, Plaschke A, Ptak M, et al. Association
1993;92:2131. between CYP2C9 slow metabolizer genotypes and
22. Clouse LH, Comp PC. The regulation of hemo- severe hypoglycaemia on medication with sulpho-
stasis: the protein C system. N Engl J Med. nylurea hypoglycaemic agents. Br J Clin Pharmacol.
1986;314(20):1298–304. https://doi.org/10.1056/ 2005;60(1):103–6. https://doi.org/10.1111/j.1365-­-
NEJM198605153142006. PMID: 3010107. 2125.2005.02379.x. PMID: 15963101; PMCID:
23. Sconce EA, Khan TI, Wynne HA, et al. The impact PMC1884896.
of CYP2C9 and VKORC1 genetic polymorphism and 34. Hughes J, Smith T, Kosterlitz H, Fothergill L, Morgan
patient characteristics upon warfarin dose require- B, Morris H. Identification of two related pentapep-
ments: proposal for a new dosing regimen. Blood. tides from the brain with potent opiate agonist activ-
2005;106(7):2329–33. https://doi.org/10.1182/blood-­ ity. Nature. 1975;258:577–80.
2005-­03-­1108. Epub 2005 Jun 9. PMID: 15947090. 35. McCleane G, Smith H. Opioids for persistent noncan-
24. Wadelius M, Chen LY, Eriksson N, et al. Association cer pain. Med Clin N Am. 2007;91:177–97.
of warfarin dose with genes involved in its action and 36. Li C, Sugam J, Lowery-Gionta E, et al. Mu opioid
metabolism. Hum Genet. 2007;121(1):23–34. https:// receptor modulation of dopamine neurons in the peri-
doi.org/10.1007/s00439-­006-­0260-­8. Epub 2006 Oct aqueductal gray/dorsal raphe: a role in regulation of
18. PMID: 17048007; PMCID: PMC1797064. pain. Neuropsychopharmacology. 2016;41:2122–32.
25. Oates JT, Lopez D. Pharmacogenetics: an important https://doi.org/10.1038/npp.2016.12.
part of drug development with a focus on its applica- 37. Kelly LE, Rieder M, van den Anker J, et al.
tion. Int J Biomed Investig. 2018;1(2):111. More codeine fatalities after tonsillectomy
26. Ansell J, Hirsh J, Hylek E, Jacobson A, Crowther in North American children. Pediatrics. 2012
M, Palareti G. Pharmacology and management May;129(5):e1343–7. https://doi.org/10.1542/
of the vitamin K antagonists: American College peds.2011-­2538. Epub 2012 Apr 9. PMID:
of Chest Physicians Evidence-Based Clinical 22492761.
Practice Guidelines (8th edition). Chest. 2008;133(6 38. Gasche Y, Daali Y, Fathi M, et al. Codeine intoxica-
Suppl):160S–98S. https://doi.org/10.1378/chest.08- tion associated with ultrarapid CYP2D6 metabolism.
­0670. PMID: 18574265. N Engl J Med. 2004;351(27):2827–31. https://doi.
300 A. Mason et al.

org/10.1056/NEJMoa041888. Erratum in: N Engl J 51. Roberts DM, Sevastos J, Carland JE, Stocker SL,
Med. 2005;352(6):638. PMID: 15625333. Lea-Henry TN. Clinical pharmacokinetics in kidney
39. Gardner DM, Baldessarini RJ, Waraich P. Modern disease: application to rational design of dosing regi-
antipsychotic drugs: a critical overview. CMAJ. mens. Clin J Am Soc Nephrol. 2018;13(8):1254–63.
2005;172(13):1703–11. https://doi.org/10.1503/ https://doi.org/10.2215/CJN.05150418.
cmaj.1041064. PMID: 15967975; PMCID: 52. Centers for Disease Control and Prevention. CDC
PMC1150265. guideline for prescribing opioids for chronic pain.
40. Siskind D, McCartney L, Goldschlager R, Kisely 2016. Retrieved from https://www.cdc.gov/drugover-
S. Clozapine v. first- and second-generation antipsy- dose/pdf/guidelines_at-­a-­glance-­a.pdf.
chotics in treatment-refractory schizophrenia: sys- 53. Centers for Disease Control and Prevention. Wide-­
tematic review and meta-analysis. Br J Psychiatry. ranging online data for epidemiologic research (CDC
2016;209(5):385–92. https://doi.org/10.1192/bjp. WONDER). Atlanta: CDC, National Center for
bp.115.177261. Epub 2016 Jul 7. PMID: 27388573. Health Statistics; 2020. Retrieved from https://www.
41. Meltzer HY, Alphs L, Green AI, et al. International cdc.gov/drugoverdose/deaths/prescription/maps.html.
Suicide Prevention Trial Study Group. Clozapine 54. Dowell D, Haegerich TM, Chou R. CDC guideline
treatment for suicidality in schizophrenia: for prescribing opioids for chronic pain—United
International Suicide Prevention Trial (InterSePT). States, 2016 [published correction appears in MMWR
Arch Gen Psychiatry. 2003;60(1):82–91. https://doi. Recomm Rep. 2016;65(11):295]. MMWR Recomm
org/10.1001/archpsyc.60.1.82. Erratum in: Arch Gen Rep. 2016;65(1):1–49.
Psychiatry. 2003;60(7):735. PMID: 12511175. 55. Von Korff M, Saunders K, Thomas Ray G, Boudreau
42. Munro J, O’Sullivan D, Andrews C, et al. Active mon- D, Campbell C, Merrill J, Sullivan MD, Rutter CM,
itoring of 12,760 clozapine recipients in the UK and Silverberg MJ, Banta-Green C, Weisner C. De facto
Ireland. Beyond pharmacovigilance. Br J Psychiatry. long-term opioid therapy for noncancer pain. Clin
1999;175:576–80. https://doi.org/10.1192/ J Pain. 2008;24(6):521–7. https://doi.org/10.1097/
bjp.175.6.576. PMID: 10789357. AJP.0b013e318169d03b.
43. Devinsky O, Honigfeld G, Patin J. Clozapine-related 56. American Medical Association Opioid Task Force.
seizures. Neurology. 1991;41(3):369–71. https://doi. Opioid Task Force 2019 Progress Report. 2019.
org/10.1212/wnl.41.3.369. PMID: 2006003. Retrieved from https://www.end-­opioid-­epidemic.
44. Haas SJ, Hill R, Krum H, et al. Clozapine-associated org/wp-­content/uploads/2019/06/AMA-­Opioid-­Task-­
myocarditis: a review of 116 cases of suspected myo- Force-­2019-­Progress-­Report-­web-­1.pdf.
carditis associated with the use of clozapine in Australia 57. Centers for Disease Control and Prevention. Life-­
during 1993-2003. Drug Saf. 2007;30(1):47–57. saving naloxone from pharmacies. 2019. https://www.
https://doi.org/10.2165/00002018-­200730010-­00005. cdc.gov/vitalsigns/naloxone/index.html
PMID: 17194170. 58. American Medical Association Opioid Task Force.
45. Nnadi CU, Malhotra AK. Individualizing antipsy- Help save lives: co-prescribe naloxone to patients at
chotic drug therapy in schizophrenia: the prom- risk of overdose. 2017. Retrieved https://www.end-­­
ise of pharmacogenetics. Curr Psychiatry Rep. opioid-­epidemic.org/wp-­content/uploads/2017/08/
2007;9(4):313–8. AMA-­O pioid-­Task-­F orce-­n aloxone-­o ne-­p ager-­
46. Renton KW. Hepatic drug metabolism and immuno- updated-­August-­2017-­FINAL-­1.pdf.
stimulation. Toxicology. 2000;142(3):173–8. https:// 59. Sansgiry SS, Bhansali AH, Bapat SS, Xu Q. Abuse of
doi.org/10.1016/s0300-­483x(99)00142-­0. over-the-counter medicines: a pharmacist’s perspec-
47. Renton KW. Alteration of drug biotransformation tive. Integr Pharma Res Pract. 2016;6:1–6. https://doi.
and elimination during infection and inflammation. org/10.2147/IPRP.S103494.
Pharmacol Ther. 2001;92(2–3):147–63. https://doi. 60. Algarni M, Hadi MA, Yahyouche A, Mahmood
org/10.1016/s0163-­7258(01)00165-­6. S, Jalal Z. A mixed-methods systematic review of
48. Zuckerman M, Greller HA, Babu KM. A review the prevalence, reasons, associated harms and risk-­
of the toxicologic implications of obesity. J Med reduction interventions of over-the-counter (OTC)
Toxicol. 2015;11(3):342–54. https://doi.org/10.1007/ medicines misuse, abuse and dependence in adults.
s13181-­015-­0488-­6. J Pharm Policy Pract. 2021;14(1):76. https://doi.
49. Brill MJ, Diepstraten J, van Rongen A, van Kralingen org/10.1186/s40545-­021-­00350-­7. PMID: 34517925;
S, van den Anker JN, Knibbe CA. Impact of obesity PMCID: PMC8439034.
on drug metabolism and elimination in adults and 61. Lessenger J, Feinberg S. J Am Board Fam Med.
children. Clin Pharmacokinet. 2012;51(5):277–304. 2008;21(1):45–54. https://doi.org/10.3122/
https://doi.org/10.2165/11599410-­000000000-­00000. jabfm.2008.01.070071.
50. Lea-Henry TN, Carland JE, Stocker SL, Sevastos J, 62. Cooper RJ. Over-the-counter medicine abuse – a
Roberts DM. Clinical pharmacokinetics in kidney dis- review of the literature. J Subst Use. 2013;18(2):82–
ease: fundamental principles. Clin J Am Soc Nephrol. 107. https://doi.org/10.3109/14659891.2011.615002.
2018;13(7):1085–95. https://doi.org/10.2215/ 63. Hedegaard H, Bastian BA, Trinidad JP, Spencer M,
CJN.00340118. Warner M. Drugs most frequently involved in drug
overdose deaths: United States, 2011–2016. National
17 Drug Dose and Therapy Individualization 301

Vital Statistics Reports, vol. 67, no 9. Hyattsville: exposure calls. Clin Toxicol. 2018;56(7):656–63.
National Center for Health Statistics; 2018. https://doi.org/10.1080/15563650.2017.1416124.
64. Eggleston W, Palmer R, Dubé P, Thornton S, Stolbach 72. Overdose, Drug (OD). In: Korsmeyer P, Kranzler
A, Calello D, Marraffa J. Loperamide toxicity: recom- HR, editors. Encyclopedia of drugs, alcohol &
mendations for patient monitoring and management. addictive behavior. 3rd ed., vol. 3. Macmillan
Clin Toxicol. 2019. https://doi.org/10.1080/15563650 Reference USA; 2009, p. 190. Gale health and
.2019.1681443. wellness. link.gale.com/apps/doc/CX2699700338/
65. Lee VR, Vera A, Alexander A, Ruck B, Nelson H W R C ? u = t a m p 4 4 8 9 8 & s i d = b o o k m a r k -­
LS, Wax P, Campleman S, Brent J, Calello HWRC&xid=193d470c. Accessed 9 Oct. 2021.
DP. Loperamide misuse to avoid opioid withdrawal 73. Catlin JR, Pechmann C, Brass EP. Dangerous double
and to achieve a euphoric effect: high doses and high dosing: how naive beliefs can contribute to unin-
risk. Clin Toxicol. 2019;57(3):175–80. https://doi.org tentional overdose with over-the-counter drugs. J
/10.1080/15563650.2018.1510128. Public Policy Mark. 2015;34(2):194–209. https://doi.
66. Farrell S. Acetaminophen toxicity. [Updated: Oct 05, org/10.1509/jppm.14.061.
2021]. Medscape. Retrieved from https://emedicine. 74. Cerdá M, Krawczyk N, Hamilton L, Rudolph KE,
medscape.com/article/820200-­overview. Friedman SR, Keyes KM. A critical review of the
67. Manning B. Chapter 18. Antihistamines. In: Olson social and behavioral contributions to the overdose
KR, editor. Poisoning & drug overdose, 6th edn. epidemic. Annu Rev Public Health. 2021;42:95–
McGraw Hill; 2012. https://accessmedicine. 114. https://doi.org/10.1146/annurev-­publhealth-­
mhmedical.com/content.aspx?bookid=391&sectio 090419-­102727. Epub 2021 Nov 30. PMID:
nid=42069832. Accessed 21 Dec 2021. 33256535.
68. Borowy CS, Mukherji P. Antihistamine toxicity. 75. Park TW, Lin LA, Hosanagar A, Kogowski A, Paige
[Updated 2021 Mar 12]. In: StatPearls [Internet]. K, Bohnert AS. Understanding risk factors for opioid
Treasure Island: StatPearls Publishing; 2021. overdose in clinical populations to inform treatment
Available from: https://www.ncbi.nlm.nih.gov/books/ and policy. J Addict Med. 2016;10(6):369–81. https://
NBK482318/. doi.org/10.1097/ADM.0000000000000245.
69. Petekkaya S, Ayaz N, Dogan M, Oruc M, Oner B, 76. Miller TR, Swedler DI, Lawrence BA, et al. Incidence
Gokturk C, Celebi A, Budak A, Soylu O, Celbis and lethality of suicidal overdoses by drug class.
O. Suicidal death from pseudoephedrine sulfate over- JAMA Netw Open. 2020;3(3):e200607. https://doi.
dose: a case report. Ulutas Med J. 2015;1:119–21. org/10.1001/jamanetworkopen.2020.0607.
https://doi.org/10.5455/umj.20151110010021. 77. Rizk MM, Herzog S, Dugad S, Stanley B. Suicide
70. Journey J, Agrawal S, Stern E. Dextromethorphan risk and addiction: the impact of alcohol and opioid
toxicity. [Updated 2021 Jun 28]. In: StatPearls use disorders [published online ahead of print, 2021
[Internet]. Treasure Island: StatPearls Publishing; Mar 14]. Curr Addict Rep. 2021:1–14. https://doi.
2021. Available from: https://www.ncbi.nlm.nih.gov/ org/10.1007/s40429-­021-­00361-­z.
books/NBK538502/. 78. O’Rourke MC, Jamil RT, Siddiqui W. Suicide screen-
71. Karami S, Major J, Calderon S, McAninch J. Trends ing and prevention. In: StatPearls [Internet]. Treasure
in dextromethorphan cough and cold products: 2000– Island: StatPearls Publishing; 2021. PMID: 30285348.
2015 National Poison Data System intentional abuse
Models for Drug Individualization:
Patient to Population Level
18
Sierra Klein, Ashley Mason, Gavin Lockard,
Vance Cantrell, Snow Pinxue Li, Kirtan Patel,
Andre Elder, Melissa Sur, and Charles Preuss

Abstract individuals and populations, clinicians can


begin to optimize drug therapy, taking the
Multiple drug dosing models have been devel- next step forward into personalized medicine
oped which try to address optimal drug ther- which will optimize drug efficacy and safety.
apy for the individual patient based on
population data. Drug therapy individualiza- Keywords
tion is based on many characteristics, such as
ethnicity, age, gender, pregnancy, and renal Therapeutic drug monitoring · Drug individu-
function, all of which require a clinician’s alization · Pharmacogenetics ·
understanding of each factor. Drug dosing Pharmacokinetics/pharmacodynamics
models such as pharmacokinetic/pharmaco- modeling · Physiologically based pharmaco-
dynamic (PK/PD) and physiologically based kinetic modeling
pharmacokinetic modeling (PBPK) have been
developed in order to gradually replace large
and expensive clinical studies. By understand- 18.1 Historical Perspectives
ing how pharmacotherapy differs between on Therapy Individualization

Drug therapy individualization is as old as medi-


Ashley Mason, Gavin Lockard, Vance Cantrell, Snow
Pinxue Li, Kirtan Patel, Andre Elder and Melissa Sur con- cine itself. Hippocrates himself purported the
tributed equally with all other contributors. importance of understanding a patient’s health
and mind and how their individuality is influ-
S. Klein (*) · A. Mason · G. Lockard · V. Cantrell · enced by the social and natural environment [1].
S. P. Li · K. Patel · A. Elder · M. Sur The Hippocratic tradition was, and still is, firmly
University of South Florida Morsani College of rooted in exploring how psychological factors,
Medicine, MD Program, Tampa, FL, USA
e-mail: klein37@usf.edu; mason16@usf.edu;
mind, body, spirit, and social and natural environ-
gavinlockard@usf.edu; cantrell20@usf.edu; pinxue@ ments influence one’s health [1]. Therapy indi-
usf.edu; kpatel37@usf.edu; adelder@usf.edu; vidualization is the process of adjusting dosage,
msur@usf.edu medication type, drug packaging, drug delivery
C. Preuss methods, and other aspects of medications to the
University of South Florida Morsani College of patient’s individual physical, emotional, and
Medicine, Department of Molecular Pharmacology &
Physiology, Tampa, FL, USA
social needs. Individualization of drug therapy is
e-mail: cpreuss@usf.edu multifaceted and must encompass the complexi-

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 303
S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_18
304 S. Klein et al.

ties of medicinal prescription as well as social or at all to that therapy. An all too harrowing les-
and personal norms of the patient. son in drug resistance is seen with the history of
Individualization involves integrating pharma- antibiotic resistance to penicillin, as over time
cology, physiology, and psychology, all of which overprescription and inter-bacterial transfer of
can differ between different patients and patient resistance genes against the antibiotic have led to
populations. In addition, patients with a wide significantly decreased utility of penicillin, the
variety of comorbidities such as dementia, cere- once so-called miracle drug. To address this
bral palsy, gastro-esophageal reflux, and even a resistance, antibacterial, antiviral, and anti-­
history of myocardial infarction may encounter protozoal drugs require individualization based
difficulties with taking medications on schedule, on the minimum inhibitory concentration (MIC)
adjusting to the route of administration of that of that drug necessary to eliminate the pathogen
drug, following instructions on how to take those of interest.
drugs, and avoiding drugs that will worsen their Utilization of a patient’s history is also
ongoing medical complaints. extremely helpful for individualizing prescrip-
At its core, therapy individualization is the tion practices; patients may have a family or per-
backbone of prescribing medications and other sonal history of sensitivity to therapies. Adjusting
treatments responsibly and successfully. As such, to these factors can help ensure the drug is more
personalized medicine is rooted in a strong his- effective and less dangerous to the patient. Side
torical tradition. As early as 2600 B.C., the effects of medications change prescription prac-
Egyptian physician Papyrus Ebers described how tices; in HAART (highly active anti-retroviral
to treat individual patients for asthma, cancer, therapy, a combination of anti-retroviral drugs
and a wide range of other maladies [2]. Testing with varying mechanisms) treatment of HIV,
the efficacy of individualized therapy appeared medications are often chosen based on the toler-
within the Bible’s parable of Daniel testing ability of side effects. While one patient may not
Nebuchadnezzar’s diet and adjusting it based on be concerned about hair loss, another patient
the king’s responses [3]. In the eighteenth and might prefer drugs that have more severe symp-
nineteenth centuries, apothecary shops formed, toms (such as nausea) in exchange for preventing
mimicking modern-day pharmacies and bringing that hair loss.
with them principles of changing prescription Within the patient history, drug individualiza-
practices based on the patient’s individual needs tion further requires an encompassing under-
[2]. Physicians of the time would adjust medica- standing of the patient’s socioeconomic status.
tions based on their patient’s “humors,” partici- Patients may be at an increased risk of nonadher-
pating in bloodletting and other practices based ence for a drug based on their access to medical
on their presentation and history. One great leap care and personal history, as well as the socioeco-
in medicine individualization came at the turn of nomic burden of that drug. Among patients with
the twenty-first century with the completion of cardiovascular risk, higher education, greater
the human genome project. Knowing the genetic income, less financial strain, and being employed
code provided a basis from which to explore why were associated with better self-rated health,
certain drugs affected certain patients differently while financial strain was associated with poorer
and therefore opened the door for true one-on-­ medication adherence [4]. For these circum-
one prescription practices in medicine, tailored to stances, the prescribing clinician must adjust
a person’s complete genetic profile [3]. accordingly and consider other routes of
In practice, drug individualization is also part prescription.
of the stewardship of medications for future gen- Due to the diverse implications of drug indi-
erations to use. Resistance is the process by vidualization, multiple models have been devel-
which pathogens, tumors, and disease processes oped, addressing the different ways individuals
such as autoimmunity over time acclimate to a may be affected by therapy from the individual
particular therapy and no longer respond as well level up to the population level. Drug therapy
18 Models for Drug Individualization: Patient to Population Level 305

requires individualization based on race, ethnic- American descent can be low renin producers
ity, age, gender, pregnancy state, religious beliefs,
[6]. Renin is an important contributor to the
and many more aspects, all of which require a renin-angiotensin-aldosterone system (RAAS).
physician’s understanding of the patient as a This system is designed to elevate blood pressure
whole. To aid this understanding, PK/PD, PBPK, through regulation of blood volume and systemic
and other diverse models have been developed in vascular resistance. Due to low blood pressure,
order to gradually replace in vivo studies with lower sodium concentrations detected in the dis-
less ethically challenging systems of understand- tal convoluted tubule of the nephron, or beta-­
ing drug utilization in the body. This chapter adrenergic receptor activation, the
explores drug individualization from a patient juxtaglomerular cells in the kidney are activated,
level to the broader level of population-based leading to the cleavage of prorenin to active
modeling. By understanding how therapy differs renin. While in the blood, renin cleaves angioten-
between individuals and populations, clinicians sinogen to angiotensin 1. Then, the enzyme
can begin to optimize that therapy, taking the angiotensin-converting enzyme (ACE) is respon-
next step forward into the future of personalized sible for converting inactive angiotensin 1 into
medicine. active angiotensin II. Angiotensin II then acts on
the kidneys, adrenal cortex, systemic arterioles,
and the brain with the primary goal of increasing
18.2 Patient Populations sodium and water to increase blood pressure [7].
and Drug Individualization ACE inhibitors or angiotensin II blockers (ARBs)
are generally indicated as first-line treatment for
When a patient presents to a provider, it is imper- hypertension and work by preventing the activity
ative to remember that they are influenced by of this RAAS. However, patients who self-­
broader factors that can directly influence their identify as being of African-American descent
appropriate pharmacological treatment. Each have been shown to be lower renin producers
individual may identify as being a part of many and, therefore, have lower activation of the RAAS
different patient populations, including their race pathway. Because of this, individuals who self-­
and ethnicity, age, gender, pregnancy state, and identify as being of African-American descent
more, that can impact the appropriate pharmaco- have negative feedback on their RAAS. Utilizing
logical strategy. Further, patients have unique an ACE inhibitor or an ARB in a patient who self-­
comorbidities and manifestations of their dis- identifies as being of African-American descent
eases, as well as individual differences in the can lead to a reduced blood pressure response or
metabolism of drugs. It is the challenge of the even resistance to the effects of the medication
clinician to combine the broader, biopsychoso- [6]. Therefore, thiazide diuretics, such as
cial patient population influences with the unique chlorthalidone, or calcium channel blockers,
physiology and desires of the patient sitting in such as amlodipine, are instead recommended as
front of them to create the most appropriate, indi- first-line monotherapy for this patient population
vidualized, and effective treatment strategy for [8].
each patient [5]. The following paragraphs dis- Hepatitis C is another chronic disease in which
cuss the influence patient population characteris- incidence and treatment have traditionally been
tics can have on treatment strategy. impacted by race. Unfortunately, those who self-­
identify as African-American are significantly
more likely to suffer from this chronic disease.
18.2.1 Race and Ethnicity Despite only being 13% of the US population,
23% of HCV cases in the United States were in
Traditionally, race has been a relevant variable in African-American individuals [9]. Further, race
the treatment algorithm of a variety of disorders. has been classically thought to influence differ-
Individuals who self-identify as being of African-­ ing immune responses and, therefore, differing
306 S. Klein et al.

responses to pharmacological treatment. It is important to emphasize that race has no


Specifically, those of African-American descent biological definition, and therefore, the scientific
were found to have a higher prevalence of the community is moving away from including race
non-CC interleukin IL28B genotype, which led as a prominent factor in clinical situations. In
to a decrease in effectiveness of pegylated inter- many instances, the factor of race fails to recog-
feron treatment. Thankfully, the newer treatments nize social and environmental factors that also
for hepatitis C were more effective in the African-­ influence outcomes [13]. In addition, it must be
American population than interferon treatment, noted that there are many differences between
but the disparity is still present. Researchers have individuals of the same race. An example of this
found that patients who identify as African-­ is showcased through allopurinol-induced severe
American had a higher rate of relapse when cutaneous adverse reactions (SCARS), which
treated with sofosbuvir/ledipasvir compared to have been shown to potentially be associated
those who did not identify as African-American. with the HLA-B*5801 allele. Since research has
Therefore, race can be an important consider- demonstrated that the allele prevalence varies
ation in the treatment of hepatitis C [10]. among those of different races and ethnicities,
The treatment of asthma is also classically the American College of Rheumatology recom-
associated with race and ethnicity differences. mends that those of Southeast Asian and African-­
Unfortunately, there are also disparities in the American descent should be tested for this allele.
prevalence of asthma, with the incidence being However, this fails to consider the influence of
higher in those from those from African-­ geography within racial groups. In Switzerland, a
American and Puerto Rican descent compared to country that is considered smaller and more
those from European descent. Pharmacological homogenous, researchers found variability across
treatment effectiveness also differs between the country. Residents in the city of Basel had a
patient populations. Short-acting β-agonists higher frequency of this allele than the entire US
(SABAs) are commonly utilized in the treatment African-American population, demonstrating
of asthma, working by activating the that within a relatively homogenous ethnic popu-
β2-adrenergic receptor in the respiratory tract. lation, there was incredible variability based on
Through cAMP preventing intracellular calcium geography. Further, it is recommended that those
release and decreasing extracellular calcium of Southeast Asian descent are screened.
entry, administration of SABAs leads to the However, Japan, a country in Asia, has been
overall effect of bronchodilation and relaxation shown to have an even more decreased preva-
of the airways, which can help attenuate an lence of the HLA-B*5801 variation than
asthma attack. However, they were found to be Caucasian individuals from the United States
less effective in African-American children and [14]. It is evident that race is not the only factor a
those of Puerto Rican descent [11]. Further, an clinician must consider when prescribing phar-
inhaled corticosteroid in conjunction with a maceuticals, and clinicians must be sure to not
short-acting bronchodilator is often recom- generalize individuals into categories based
mended for asthma. When looking at impact on solely on their race or ethnicity.
children suffering from asthma, researchers
found that this combination led to an increased
bronchodilator response only among those who 18.2.2 Age
identified as Mexican American and not those
who identified as African-American or Puerto When prescribing medications, it is important to
Rican [12]. Therefore, when a patient presents keep in mind that drug regimens, dosages, and
with asthma, a clinician should consider how pharmacokinetics can differ based on the age of
their race and ethnicity could influence their the patient. Therefore, to practice individualized
treatment. medicine, a clinician must often adjust the dos-
age and even sometimes the medication recom-
18 Models for Drug Individualization: Patient to Population Level 307

mended for the age of the particular patient being at risk for toxicity due to the buildup of chloram-
treated. From birth to middle age to elderly phenicol. This can lead to the infamous “gray
adults, there can be significant differences in baby syndrome,” which can lead to complica-
drug pharmacokinetics that are crucial for a phy- tions such as gray skin, abdominal distention,
sician to consider. When considering a drug with and respiratory and cardiovascular collapse [17].
poor solubility or one that is slowly released over Further, the composition of the intestinal bacteria
time, a clinician must consider drug absorption in is also believed to differ with age. This becomes
the patient. Younger children have been shown to clinically relevant with digoxin, a drug with a
have decreased intestinal transit time, which can very narrow therapeutic index. Therefore, the
decrease drug absorption of drugs such as the- correct dosing of digoxin is very important in
ophylline. For newborns, a clinician must con- order to avoid toxicity. As children age, the inac-
sider the impact of gastric pH. At birth, it is tivation of digoxin leads to its excretion in the gut
thought that gastric pH is neutral, and then there lumen to increase. Therefore, neonates require a
is a decrease to acidic pH typically characteristic higher loading dose than older patients of digoxin
of gastric pH. For these newborn babies, this neu- [15]. Neonatal dosing recommendations for sev-
tral pH can lead to higher concentrations of acid-­ eral drugs are listed in Table 18.1.
labile drugs, such as penicillin, and lower Age is also an important consideration for
concentrations of weakly basic drugs, such as drug metabolism when considering the elderly.
itraconazole. Further, there are differences in The Beers Criteria for Potentially Inappropriate
drug distribution between children and adults Medication Use in Older Adults was created by
based on their body composition. Infants tend to the American Geriatrics Society. The purpose of
have higher levels of fat, and therefore lipophilic the Beers Criteria is to provide recommendations
drugs like diazepam have a larger volume of dis- to clinicians in the formulation of an appropriate
tribution in these patients than in older children pharmacological plan for patients ages 65 years
and adults. Further, extracellular water tends to and older, as their age directly impacts their phar-
decrease during development. Therefore, infants macokinetics, side effects, and drug-related prob-
have a higher volume of distribution of water-­ lems. As individuals age, the harm outweighs the
soluble drugs and must be given higher doses of benefit for many pharmacological interventions,
water-soluble drugs [15]. and the Beers Criteria aims to mitigate this. An
Drug metabolism can also differ based on age. example of a drug recommended to be avoided in
The protein content within the liver changes the elderly is that of chlorpropamide, a long-­
throughout life, increasing from birth to a maxi- acting sulfonylurea. The half-life is extended in
mum in a 30-year-old adult, which can lead to a the elderly, which can lead to the adverse effects
lower first-pass effect. However, hepatic clear- of SIADH and hypoglycemia [26]. It is essential
ance and blood flow through the liver are higher to apply the Beers Criteria along with what would
in children compared to adults, which can lead to be best for the individual patient to truly practice
a greater first-pass effect [16]. Therefore, it is drug individualization.
important to look at the mechanism of each indi-
vidual drug to properly individualize drug regi-
mens for each stage of life [15]. 18.2.3 Sex
Chloramphenicol, a broad-spectrum antibi-
otic, is an example of a drug in which under- Sex, defined as the biological classification at
standing the lower enzyme concentrations in birth due to anatomical and chromosomal differ-
neonates is crucial. Chloramphenicol is typically ences, influences drug individualization. Even
metabolized by glucuronidation through the though the Institute of Medicine of the National
UDP-glucuronyltransferase enzyme and then Academy of Science advocated for the impor-
renally excreted. Due to lower concentrations of tance of analyzing sex differences in drug devel-
this enzyme in the liver, premature neonates are opment, the FDA guidelines specifying that both
308 S. Klein et al.

Table 18.1 Neonatal dosing recommendations for common drugs


Drug name Indication Dosing recommendations Adverse effects
Ampicillin Neonatal infections, especially No specific dosing in neonates [18] Rashes [19]
Group B Streptococcus spp.
and Listeria spp.
Cefotaxime Sepsis, meningitis, Lower dosage (4 mg/kg/day) in Vomiting, diarrhea, rash
3rd-generation cephalosporin premature and full-term infants less [19]
[19] than 1 week old; increase to
6–7.5 mg/kg/day at 1-week
postnatal age [18]
Digoxin Heart disease in children [19] Increased dosing in younger EKG changes, GI distress,
children; decrease loading doses CNS changes, arrhythmia
from 45 μg kg−1 in infants to 35 μg [20]
kg−1 in preschool children [15]

Famotidine GERD (gastro-esophageal Utilize body size-based dosing [22]. Agitation, headache [21]
(Pepcid) reflux) [21] A dosage of 0.5 mg/kg is typically
followed [21]
Furosemide Fluid overload, management Decreased dosing in comparison to Dehydration, hyponatremia,
of PDA (patent ductus adults, as t1/2 of furosemide is hypokalemia, rash,
arteriosus), cardiac failure, greater in neonates [23] ototoxicity, nephrotoxicity,
pulmonary edema [19] nephrocalcinosis [19]
Gentamicin Gram-negative sepsis due to Lower dosage (5 mg/kg/day) in Hearing loss, reduced renal
Pseudomonas aeruginosa, premature and full-term infants less function and renal failure
Escherichia coli, Klebsiella than 1 week old; increase to 7.5 mg/ [19]
pneumoniae, and Enterobacter kg/day at 1-week postnatal age [18]
spp. [18]
Ibuprofen Closure of PDA, pain, fever In infants <6 months of age, Reduced urine output,
[19] ibuprofen should be prescribed platelet dysfunction [19]
based on body weight at 5–10 mg/
kg [24]
Metronidazole Necrotizing enterocolitis, No specific dosing in neonates [18] Gastrointestinal
anaerobic infections [19] disturbances, peripheral
neuropathy, neutropenia
[19]
Phenytoin Neonatal seizures 10–20 mg/kg/day, greater than Irritation at injection site,
unresponsive to phenobarbital doses for adults [25] gastrointestinal distress,
[19] hypotension, coma,
respiratory depression [19]
Adapted from Refs. [15, 18, 19, 21–25]

sexes be included in clinical trials, and the sion and metabolic rate are thought to be signifi-
requirement to label drugs if there are differences cantly greater than in males. Therefore, drugs
noted between male and female patients, there is dependent on CYP3A4 can have a higher clear-
still a gap in knowledge about the differences in ance in women than men [28]. Further, women
male and female pharmacology due partly to lack have decreased concentrations of the MDR1
of inclusion of female patients in clinical trials. product, hepatic P-glycoprotein, in comparison
Pharmacokinetics and pharmacodynamics differ to men. Verapamil, a non-dihydropyridine cal-
between men and women, influencing recom- cium channel blocker, is a substrate for
mended drug individualization strategies for P-glycoprotein and is metabolized by CYP3A4.
these patients [27]. The enzyme CYP3A4 is a Females can have a shorter half-life and mean
crucial component of the cytochrome P450 fam- residence time for intravenous verapamil com-
ily, which is essential for first-pass metabolism pared to males, which is clinically relevant when
through the liver. In females, CYP3A4 expres- considering the dosage [29]. Anatomical differ-
18 Models for Drug Individualization: Patient to Population Level 309

ences between males and females also can impact to lead to an increase in the expression of certain
drug individualization. Women have higher lev- hepatic enzymes, like CYP2D6. The addition of a
els of body fat compared to males, which is placenta and fetus mean additional components
important when considering lipid-soluble drugs. are participating in metabolizing medications
Vecuronium, a skeletal muscle relaxant, was [33]. An example of a medication with known
found to have a longer duration of action in complications during pregnancy is lithium which
women than men, and it is hypothesized to occur is used to treat bipolar disorder. This medication
because of their higher fat content [27]. is excreted primarily renally. In pregnancy, GFR
The physiology of women at different stages and renal excretion increase, so lithium blood
of life significantly influences how drugs affect concentrations decrease. This can be dangerous,
these patients. Exogenous hormones are com- as it can lead to sub-therapeutic concentrations in
monly prescribed in the form of oral contracep- pregnant women suffering from bipolar disorder
tives. Through competitive inhibition and/or [34]. Lithium has been described as a teratogen,
estradiol downregulating expression, oral contra- with the potential to lead to cardiac concerns and
ceptives lead to decreased concentrations of increased weight in neonates [35]. Table 18.2
enzymes in the cytochrome P450 family [30]. lists several drugs which can be teratogenic and/
Tizanidine, an α2-adrenergic agonist with signifi- or toxic during pregnancy.
cant first-pass metabolism, can have increased
plasma concentrations in patients taking oral con-
traceptives. This is believed to occur through inhi- 18.2.4 Other Considerations
bition of CYP1A2 presystematic metabolism,
and researchers found that it led to decreased sys- As the past section demonstrated, it is crucial to
tolic and diastolic blood pressures when com- consider the different patient populations and bio-
pared to those not taking oral contraceptives. physical factors that influence patient health
However, differences exist between women tak- when considering drug individualization.
ing oral contraceptives containing gestodene and However, it is essential to not broadly group
ethinyl estradiol, with some patients having patients into one category and forget that there are
greater reductions in blood pressure than others. so many factors that go into the unique identity of
This emphasizes the need to look at each aspect each patient. If an African-American middle-­aged
of individual patients, and not to generalize pregnant woman presents to the clinic for blood
patients into one group [31]. pressure medication, the clinician must consider
Pregnancy leads to numerous changes that the implications of her sex, race, age, pregnancy
impact drug regimens. For one, some medica- state, and comorbid conditions on her drug regi-
tions can be teratogenic and must be avoided in mens. The process of creating individualized
pregnant women to avoid adverse effects on the pharmacological treatment strategies is difficult,
baby. An example of this is isotretinoin, a deriva- but it is absolutely crucial so that each patient can
tive of vitamin A utilized to treat severe acne. have the most effective and safe care possible.
Isotretinoin is so teratogenic that two forms of
birth control are recommended before prescrib-
ing it in women. It can lead to significant neuro- 18.3 Models for Drug
cognitive deficits and congenital deficits that can Individualization
significantly harm the fetus [32]. Further, preg-
nancy leads to a number of physiological changes 18.3.1 Current Practices for Drug-­
that impact drug pharmacokinetics. Pregnancy Resistant Problems
changes the composition of the body, leading to
increased total body water, extracellular fluid, Most simply put, drug dose individualization
and plasma volume. Further, there is an increase hinges on deciding upon the most efficient phar-
in cardiac output and GFR. Pregnancy also tends macotherapy which both treats the disease to the
310 S. Klein et al.

best of the clinician’s ability, maximizing thera- et al. showed that there are already more and
peutic effect while minimizing the risk of adverse more physicians moving to fluoroquinolones as
events or side effects. A hugely prominent field in first-line empiric therapy despite the 95% clinical
which pharmacotherapy individualization has cure rate shown with TMP-SMX, a shift that may
immediate and tangible benefits is in the pre- eventually encourage resistance to these essential
scribing of antimicrobials. antimicrobials.
With antimicrobial agents, the shift towards It would be remiss to discuss antimicrobial
an individualized, efficient regimen is best therapy individualization without mentioning the
achieved by expeditiously moving from empiric challenges in that pursuit; namely, microbial data
therapy to definitive therapy. One serious concern is often not available for 48–72 hours, and many
alleviated by moving patients to definitive ther- cases are not even indicated for culture. However,
apy is apprehension regarding proliferation of there are clinically applicable suggestions
antibiotic-resistant organisms. The current ­allowing clinicians to move away from a one-
empiric therapy for acute uncomplicated cystitis size-fits-­all empirical formula towards what David
is trimethoprim-sulfamethoxazole (TMP-SMX) Paterson calls in his 2008 study on antibiotic
as first-line treatment; however, resistance to resistance in Gram-negative bacilli the “individu-
TMP-SMX among uropathogens has been on the alizing initial empirical therapy” [39]. One
rise since the early 1990s [38]. This begs the important and often overlooked tool is surveil-
question, at what point does resistance become lance programs; essentially, clinicians have the
too widespread, at what point do the empiric rec- data to determine if they should deviate from the
ommendations change? Research by Hooton generically recommended first-line empiric drug

Table 18.2 Examples of teratogenic/toxic medications during pregnancy


Medication name Medication type Effects
Chloramphenicol Antibiotic Gray baby syndrome, bone marrow suppression [36]
Coumarin derivatives Anticoagulant Skeletal abnormalities, nasal hypoplasia, CNS malformations,
(e.g., warfarin) intracranial hemorrhage in the fetus [36]
Diethylstilbestrol Estrogen derivative for Development of clear cell adenocarcinoma of the vagina and
(DES) miscarriage prevention cervix in mothers; genital tract abnormalities in infants [36]
Glucocorticoids, e.g., Anti-inflammatory, Cleft palate [37]
prednisone anti-autoimmunity
steroids
Lisinopril, captopril Angiotensin-converting Fetal hypotension, fetal kidney hypoperfusion, pulmonary
enzyme inhibitor hypoplasia, anuria [37]
Lithium Treatment for bipolar Cardiac abnormalities, increased birth weight [35]
disorder; mood stabilizer
Losartan, valsartan Angiotensin receptor Neonatal oliguria, anuria, hypotension, renal tubular dysgenesis
blocker (ARB) [37]
Misoprostol Prostaglandin E1 analog Vascular disruptions (Moebius syndrome, terminal limb
defects), induces abortions during the first and second
trimesters. Safe during delivery to induce labor [37]
Quinolones and Antimicrobials Renal, cardiac, and CNS toxicity; organ agenesis and
fluoroquinolones carcinogenesis in fetuses, articular cartilage damage [36]
Streptomycin Antibiotics Ototoxic, can damage CN VIII, leading to deafness in newborns
Tetracyclines Antimicrobials Liver necrosis, bone and teeth defects, suppression of skeletal
bone growth and hypoplasia of tooth enamel [36]
Valproate/valproic acid Antiepileptic Cognitive defects, neural tube defects, cardiac abnormalities,
fetal valproate syndrome [36]
Vitamin A (retinal/ Vitamin supplement Neural tube defects (exencephaly, spina bifida, hydrocephalus),
isotretinoin) thymic and cardiovascular abnormalities [36]
Adapted from Refs. [36, 37]
18 Models for Drug Individualization: Patient to Population Level 311

if resistance is suspected in their area. One exam- 18.3.2 Physiologically Based


ple noted by Neuhauser et al. is rising levels of Pharmacokinetic (PBPK)
fluoroquinolone resistance among Pseudomonas Modeling
aeruginosa [40]. As such, if fluoroquinolone
resistance is demonstrated locally, greater phar- Pivotal in the development of drug individualiza-
macotherapy individualization can be achieved tion models was the advent of physiologically
by re-evaluating local empiric treatment. Paterson based pharmacokinetic/pharmacodynamic
also notes that a clinical knowledge regarding the (PBPK/PD) modeling. This synthetic representa-
infection’s port of entry can give hints guiding tion of drug performance relies on drug mecha-
individualization of empiric treatment. In his nism data combined with organ system
research on Gram-negatives and antibiotic resis- physiology in a single organism to develop prop-
tance, he delineates that regimens targeting ositional concentrations over time within the
Gram-negative infections must assess whether respective compartments of an organ or tissue
treating Pseudomonas aeruginosa is necessary [41]. This is essential to drug therapy individual-
[39]. Where clinical knowledge comes onto the ization because it can account for known differ-
field is knowing that Pseudomonas aeruginosa ences in physiology not necessarily seen in drug
infection most commonly presents through trials, such as pregnancy, age, concurrent drugs,
ventilator-­associated pneumonia and neutropenic or genetic polymorphisms altering physiology or
fever and often in undifferentiated fever in the a combination of the above – all of which will be
critically ill. All of these allow for individualizing discussed in this chapter.
empiric therapy before having knowledge direct- Due to fetal sensitivity and altered maternal
ing to definitive therapy. Figure 18.1 summarizes physiology, pregnancy is not an ideal time to
stages in addressing microbial drug resistance. introduce pharmacological intervention and even
less of an ideal time to evaluate drug efficacy. As
such, patients are often prescribed based on pro-
tocols derived from studies excluding pregnant
patients, which may lead to dosing below thera-
peutic concentrations [42]. To examine the physi-
ologic variables that must be accounted for
during pregnancy, a study can be done that

Form a broad differential diagnosis utilizing common presentations


and screen for common drug-resistant strains (e.g. Pseudomonas
aeruginosa).

Look at geographically drug-resistant strains of bacteria, viruses, or


fungi

Prescribe empiric therapy based on available guidelines

Perform bacterial, fungal, or viral culture and determine minimal


inhibitory concentration required for treatment.

Adjust therapy accordingly by withdrawing unnecessary drugs;


monitor for development of drug resistance or adverse effects.

Fig. 18.1 Stages to addressing drug-resistant problems


312 S. Klein et al.

devises a PBPK model for pregnant patients pharmaceutical companies, due to a combination
regarding the metabolism of a model compound of decreased finances in the pediatric drug market
bisphenol A (BPA). It alters endocrine function and difficulty recruiting children for these studies
with pathological results, ranging from reproduc- [46]. Consequently, dosing is often based on
tive harm to increasing obesity risk to causing body size. As such, PBPK modeling to specify
developmental delay [43]. As such, it is a com- drug regimens in pediatric populations is a prom-
pound of interest regarding maternal-fetal metab- ising alternative. This was the approach of one
olism. BPA is also an excellent substance to make team creating a PBPK model for theophylline
a maternal-fetal PBPK model after because there and midazolam disposition from birth to adult-
is already maternal BPA exposure, allowing for hood. In order to build this model, the researchers
comparison between measured results in human relied on physiological data for neonates, 0.5-, 1-,
subjects and the projected results based on a 2-, 5-, 10-, and 15-year-old children. Additionally,
PBPK model. Ideally, this type of modeling could volume and weight data were included, such as
be used to devise dosing regimens for drugs body weight, organ weight, extracellular fluid
whose metabolism is altered by pregnancy to bet- (ECF) and vascular volume, cardiac output, and
ter tailor dosing schemes. To begin individualiz- glomerular filtration rate (GFR). All data was
ing drug dosing in pregnant patients based on a collected for both sexes. The goal was to deter-
PBPK model, it is essential to know how the mine the volume of distribution at steady state
compound is metabolized in a healthy adult with (Vdss), drug half-life (t1/2), and total and renal
no additional considerations. For the researchers clearance of both drugs (CL & CLR) correspond-
that devised a maternal-fetal PBPK model for ing to age.
BPA, this meant modeling based on rapid hepatic The results of the study correlated quite
and intestinal metabolism of BPA followed by closely with the published data for the goal values
excretion in the urine [43]. Adding the consider- the model predicted. Specifically, the Vdss of
ations for a pregnant patient meant accounting theophylline was reasonably close to the pub-
for transfer across the placenta, which was dem- lished values. However, there were no published
onstrated by ex vivo studies to rapidly diffuse values for term neonates; the Vdss for midazolam
[44]. This was implemented by the researchers as based on published results was more variable, but
a first-order kinetic following a simple diffusion the Vdss predicted by the PBPK model was
process. Further concerns for a pregnant subject within the reported ranges for the ages studied.
included in this model include changes in mater- Since the organ volume/functionality and physi-
nal plasma, hematocrit, fat, and amniotic fluid ological data were collected to represent both
volume with further sub-compartments for the sexes, the model was also able to predict a lower
fetal liver, kidneys, plasma, and brain. These fetal Vdss for theophylline and higher Vdss for mid-
sub-compartments are important to include azolam in women as opposed to men, findings
because despite low exposure since some drugs which are congruent with the literature [47, 48].
may not diffuse or accumulate as readily, fetal This was an excellent use of PBPK modeling
metabolic pathways may not be adequately pre- because there was already existing literature
pared to deal with them, regardless of concentra- against which to verify the results and test the
tions in the fetal sub-compartments. One such validity of PBPK modeling.
example is gray baby syndrome in children The motivation behind studying drug-drug
whose UDP-glucuronyl transferase is not suffi- interactions (DDIs) is to determine potential
ciently developed to handle the toxicity of chloram- reactions, from increasing potency through
phenicol. BPA is also metabolized by ­synergy to rendering one therapeutic ineffective.
UDP-glucuronyl transferase [45]. Potential DDIs are difficult to study in vivo in a
The challenges in studying drug safety and human model, as there are differences in physio-
efficacy in children are numerous. This is further logical environment between humans that can
compounded by a lack of interest on behalf of influence DDIs. As such, PBPK modeling is an
18 Models for Drug Individualization: Patient to Population Level 313

excellent alternative; with unparalleled safety azepines. The observation was most true for fen-
due to the virtual study population, PBPK model- tanyl, which showed the greatest increase in AUC
ing can evaluate potential DDIs in the context of when combined with an overdose of benzodiaz-
different genetic, demographic, or even ethnic epines [50]. The team also found that the AUC of
populations. Accounting for interactions in all of fentanyl was highest when combined with alpra-
the drugs currently prescribed in the context of zolam, a prediction which explained why fen-
all of these variables amounts to an implausible tanyl and alprazolam made up the greatest
amount of in vivo studies. Therefore, the FDA proportion of drug-related deaths in which a ben-
states that data from PBPK modeling of potential zodiazepine was combined with an opioid [51].
DDIs can be used in lieu of prospective DDI The last of the major uses for PBPK modeling
studies in certain circumstances [49]. is in determining potential drug outcomes based
The goal of the 2019 study by Ji et al. was to on genotype. This is very impactful in the realm
study the DDIs between opioids and benzodiaz- of drug therapy individualization because geno-
epines. These two drug classes were selected typic variations in alleles responsible for drug
because of the rise in opioid prescription rates in metabolism can severely alter the necessary dos-
the past decade alongside the fact that benzodiaz- ages. The goal of one study was to determine the
epines are rarely prescribed alone. This combina- pharmacokinetics of celecoxib in regard to
tion can be deadly due to their synergistic genetic polymorphisms in CYP2C9, the major
respiratory depression, but the exact mechanism pathway of celecoxib metabolism, to reduce
was unknown. This is too dangerous to be studied adverse events in celecoxib treatment. This study
in vivo in a human model. The study by Ji et al. was carried out with the goal of determining how
was spurred by previous studies in animal models genetic differences impact drug dosing. This
or in vitro studies that had attempted to evaluate study conducted by Kim et al. first evaluated the
pharmacokinetic interactions [50]. The research metabolism of celecoxib in 39 individuals of
team chose to evaluate interactions between ben- known CYP2C9*1/*1 or CYP2C9*1/*3 geno-
zodiazepines, including alprazolam, diazepam, types. This data was then used to verify the data
midazolam, and triazolam, and opioids, includ- generated from a PBPK model. Once validated,
ing fentanyl, oxycodone, and buprenorphine. the model was expanded to include
Once the research team was able to make a work- CYP2C9*1/*13 and CYP2C9*3/*3 genotypes
ing model for each individual drug, they evalu- which could be validated based on data from
ated their model against the literature for the area other studies. Once completed, the PBPK model
under the curve (AUC), Cmax, and tmax to verify determined statistically significant differences in
their results. All of these were within acceptable AUC0–48, AUCinf, Cmax, and CL/F (oral clear-
limits except for the AUC and tmax of buprenor- ance) as confirmed by the group’s in vivo phar-
phine, which fell slightly below the lower refer- macokinetic study and the studies they found.
ence value. Additionally, there was The research team found a higher AUC0–48,
pharmacokinetic (PK) data available for fentanyl. AUCinf, and Cmax but lower CL/F in the
In summation, the model was still deemed valid CYP2C9*1/*3 genotype [52] which paralleled
due to the model’s projected data falling within their observations in their own pharmacokinetic
the confidence interval of each drug’s study.
concentration-­time curve. With a working model,
the research team then used it to evaluate each of
the drugs in combination. In an interesting pre- 18.3.3 Pharmacogenetics
diction, the model showed no PK interactions
between opioids and what the team deemed nor- Just as a person’s genes can impact the manifes-
mal doses of benzodiazepines and only showed tations of a condition in an individual’s body,
PK interactions when a normal dose of opioids genes can also play an essential role in the impact
was combined with a severe overdose of benzodi- medications have on a person. Variations in the
314 S. Klein et al.

genes could lead to differences in characteristics significant concern to patients and decrease their
of a patient’s conditions that could later impact overall quality of life. Disregarding this informa-
their prognosis and how the drugs function in the tion can lead to dangerous effects on the patient.
body. Further, they can more directly impact how Table 18.3 lists common drug-metabolizing
the drug works in the body, so it is crucial to polymorphisms.
understand exactly how it does so.
The patients’ genes can affect the enzymes
that break down and transport the drug, impact- 18.4 Challenges
ing how the drug leaves the body and the mainte- to Individualization
nance dose [53]. As a result of enzyme-substrate
connections being extremely specific, the small- Despite the advantages offered by individualiza-
est change in the enzyme can cause a significant tion, many challenges still exist. The following
transformation in the function of the enzyme. If section explains the specifics of each type of
the drug is metabolized by the enzyme in a differ- challenge faced by various modes of drug indi-
ent manner than normal, it is essential to include vidualization and discusses some options to
that when figuring out the accurate dose for the address them.
patient to take. In terms of moving around the
body, if the medication is not able to get to its
intended target in the right amount of time or 18.4.1 Body Mass and Body Surface
goes to the wrong place, the efficacy of the drug Area Dosing
can be considerably altered.
A patient’s genes can impact how that indi- Drug dosage models are generally prescribed on
vidual reacts to a particular drug, as genes can a fixed dosage or based on readily obtained phys-
affect the proteins the drug comes into contact ical parameters of the patient, including but not
with [53]. Eventually, these interactions will limited to age and weight. Such individualization
cause a modification in whether more or less of methods include approaches such as body mass
the drug should be given at a time. Some exam- dosage and body surface area dosage [56]. As
ples of genes affecting drugs could be cyto- their names suggest, body mass dosing is pre-
chrome genes like CYP2C9 with irbesartan and scribed on a basis of patient weight, whereas
celecoxib or CYP2D6 with metoprolol [53]. In all body surface area dosage is prescribed on the
of these examples, the genotypes affect how the estimated surface area of a patient calculated
drug leaves the body, which in turn can impact through their height and weight. To date, these
dosing [53]. are the most commonly utilized individualized
Furthermore, genetics can be important in drug dosing methods with significant impacts on
how drugs work with each other [53]. This can the current practice of drug individualization and
especially be of concern in patients with comor- will thus be the focus of discussion in this
bidities and long-standing diseases for which subsection.
they take many medications. For example, in the Dosing based on body mass comes with inher-
case of hypertension, a patient could be pre- ent issues as weight may not accurately represent
scribed a combination of medications that work a patient’s status. Research in the 1940s first indi-
best together to help lower the patient’s blood cated that body mass dosing suffers gross inac-
pressure, while being careful not to prescribe curacies, particularly among younger populations
combinations that do not work as well. Drugs [57] with frequently reported cases of overexpos-
that do not interact well together can cause side ing patients to the drug in question [58]. To try
effects and toxicities that can be avoided by and address the issues arising from such a unidi-
understanding the individual pharmacogenetics mensional individualization approach, body sur-
of the patient. Even a seemingly small side effect face area was proposed as an alternative [59] to
continuing over a long period of time can be a incorporate additional factors such as height and
18 Models for Drug Individualization: Patient to Population Level 315

Table 18.3 Some of the common enzymatic genetic polymorphisms that affect common drugs
Important polymorphic
Enzyme variations Drugs affected Effects
N-Acetyltransferase NAT1 and NAT2 (A and B) Caffeine, isoniazid, Fast acetylators have only low
nitrazepam, concentrations of active drug; slow
sulfonamides acetylators have higher
concentrations of active drug
CYP2D6 80 genotypic variants, with Over 70 drugs, including Extensive metabolizers (autosomal
autosomal recessive alleles antidepressants, dominant wild-type gene) have
corresponding to poor antiarrhythmic agents, increased breakdown of drugs; poor
metabolizers. beta-blockers, opioids, metabolizers have a 10- to 100-fold
CYP2D6*2×n CYP2D6*4, and neuroleptic agents lower breakdown of drugs.
CYP2D6*5, CYP2D6*10, Ultra-metabolizers have increased
CYP2D6*17 blood concentrations of morphine, as
well as other opioid receptor
agonists, resulting in greater risk for
opioid-related adverse effects. In
contrast, UMs require higher
dosages of mirtazapine, a tetracyclic
antidepressant, to achieve
therapeutic effects
CYP2C9 CYP2C9*2, CYP2C9*3 NSAIDs, phenytoin, Increased warfarin activity in
ARBs, sulfonylureas, extensive metabolizers, leading to
warfarin increased INR. Patients with
CYP2C9 as well as VKORC1 have
increased risk for major bleeding
events
CYP2C8 CYP2C8*3 Arachidonic acid, Inhibited by ketoconazole and
paclitaxel, rosiglitazone, gemfibrozil in extensive
zopiclone metabolizers
CYP2C19 CYP2C19*2, CYP2C19*3, Diazepam, Increased adverse drug reactions
CYP2C19*17 S-mephenytoin, with utilization of prodrugs (drugs
moclobemide, PPIs, where the enzyme activates the
proguanil, SSRIs drug); higher rates of eradication of
H. pylori when utilizing triple
therapy (PPIs + clarithromycin +
amoxicillin). Carriers of the
CYP2C19*7 allele have improved
effect with clopidogrel treatment,
while CYP2C19*2 and CYP2C19*3
carriers have reduced effects
CYP2B6 CYP2B6*6 Methadone CYP2B6*6 carriers have slower
metabolic capacity, resulting in
higher blood concentrations of
methadone and efavirenz (a NNRTI
for HIV treatment)
TPMT (thiopurine TPMT*3A Mercaptopurine, In children with ALL, high TPMT
methyltransferase) TPMT*3C thioguanine azathioprine activity causes a poor clinical
response, while low TPMT is
associated with severe to fatal
myelosuppression
UGT1A1 UGT1A1*28 Irinotecan UGT1A1*28 carriers have increased
risk of ADRs when treated for colon
cancer with irinotecan
Adapted from Refs. [54, 55]
316 S. Klein et al.

size of the patient. However, continued research tions towards what factors contribute most to
suggests that dosing by body surface area fre- drug toxicity and response. Therefore, all
quently underexposes a patient to the prescribed approaches suffer from the central issue stem-
drug [58]. Such data indicates both models dis- ming from unique or unexpected patient response
play inaccuracies when extrapolated outside of due to special metabolisms or changing statuses.
the narrow patient population used in the original Dosing equations extrapolate poorly in pediatric
clinical tests to determine individualized drug situations [56] in the case of abacavir or acyclovir
dosing. This weakness of individualized drug [62] and for pregnant women in the case of tinza-
dosing approaches becomes especially concern- parin, whose manufacturer suggested weight-­
ing when it comes to treating patient populations adjusted doses were not effective at
that may be prone to obesity or when a popula- anticoagulating most pregnant women against
tion’s distribution density of BMI shifts over venous thromboembolisms [63]. Additionally, in
time, as clinicians may continue to use outdated special circumstances, factors such as kidney and
dosing guidelines based on BMI ranges mea- liver functionality can also become as relevant to
sured in a different era. the dosage. Overall, due to varying success in
In light of the issues generated by the use of application, not all clinicians will agree on the
body mass or body surface area to estimate dos- best model for each drug, and significant debate
age, studies have assessed the benefits of incor- still exists across many clinical situations for the
porating additional patient parameters such as appropriate selection of individualized drug dos-
age and variations on body weight (ideal, ing approach [58].
adjusted, fat-free, and lean) into dosing estima- A key advantage of body mass/body surface
tions and comparing such individualized area dosing is its relative ease of use and wide
approaches against the usage of fixed dosages applicability; however, it fails to accurately rep-
[56, 60]. Study results indicate that drug dosing resent the unique variations of each patient’s
may need to be adjusted based on differing body and how they individually react to drugs.
parameters according to the drug in question, as The inclusion of the extremes of weight subsets
different medications show ideal performance of the population into clinical drug testing [56]
under different models. For example, for drugs can build more accurate models for drug response
such as hydrocortisone, vancomycin, linezolid, outside of average weight ranges. This inclusion
and aprotinin, weight-based dosing is recom- of diverse subsets of the populations can be
mended. For cyclosporine microemulsion, applied for all “special populations” such as
recombinant activated Factor VII, and epoetin α, pediatrics, pregnant women, and special condi-
fixed dosing shows superior results. For cases of tions relevant to the drug metabolism. Though
atracurium and rocuronium, ideal body weight ideal, this proposed inclusion of population sub-
calculations are preferred. For intravenous busul- sets could provide significant challenge in the
fan and dalteparin, it has been found that age-­ current process of bringing drugs to market: the
based dosing is most appropriate [56]. Certain necessity of recruiting adequate number (N) of
patterns that begin to emerge suggest that lean patients to rule out the variations in personal
body mass may be a good predictor of kidney response and testing drugs on sensitive popula-
clearance [61], and total body weight is a better tions such as those with extreme comorbidities,
metric to use for lipophilic drugs [58]. Such data pregnant women, or pediatric populations. Such
indicates that individualized drug dosing may alternative chart creation is further complicated
need to be approached on a drug-based basis as when more than one significant factor is present
clinical researchers come to understand the on the same patient. This is best illustrated by the
underlying mechanisms as to why certain param- decades-long debate between clinicians on how
eters work better for dosing than others. However, best to dose pediatric patients who are also obese
all methods are ultimately approximations of [64–66].
patient individuality based on clinicians’ assump-
18 Models for Drug Individualization: Patient to Population Level 317

A particular concern in generating individual- pregnant women from initial testing. Such exclu-
ization approaches is the standardization of sions of sensitive populations may affect accurate
methods used to make the measurements that prediction of drug dosage in relevant populations
establish body mass or body surface area param- later in clinical settings. The use of indirect test-
eters for the patient. Equipment differences, such ing with population pharmacokinetics, allometric
as using manual scale in the clinic versus using scaling, and in silico modeling to estimate the
weight-detecting gurneys, may create significant applicable dose to pediatric populations [67] has
differences, especially in patients outside of aver- been proposed to circumvent this issue. Statistical
age range. Though dosing based on measurable tools such as population pharmacokinetics
factors like body weight or body surface area is (PopPK) modeling analyze drug concentration
one of the least complex of the individualization variation in the wider population through meta-­
models discussed, issues with standardization analysis of established clinical data. The inclu-
have resulted in numerous instances of non-­ sion of clinical data supplements the weaknesses
compliance and non-standardization in real-life of the PK/PD patient testing models; however, it
practice [56], such as in the vancomycin trial requires long-term usage and detailed metabolite
where the addition of electronic weight-based data collection among all patients receiving the
checking increased percentage in appropriate drug over a long period of time. This exposes
dosing in ED patients. Therefore, in the valida- patients to subpar dosing regimens during the
tion of individualization methods, it is not only time enough data is generated to supplement the
important to ensure accurate prediction of dosage original model. Allometric scaling uses large
and drug response but is essential to ensure the populations of animal models to overcome the
basis of the methods is generated on reliable limitations of PopPK by replacing human patients
datasets. with animal subjects. However, the high cost and
limited equivalence of animal models to human
patient response make such studies inferior to
18.4.2 PK/PD and PBPk Modeling PopPK supplementation. In silico modeling uti-
lizes computer models to replace in vitro experi-
For drugs with extremely narrow therapeutic mentation. Though ideal, massive strides in
indexes, extreme care must be taken to dose computing and biological modeling must be
patients accurately. Dosing strategies based on made before such methods may be considered
body mass and body surface area only apply to viable.
drugs that are affected significantly by weight PK/PD modeling allows for rudimentary pre-
with few additional limitations, such as signifi- dictions of drug-drug interactions [69], mitigat-
cant interactions with other drugs. Thus, for ing the daunting task of creating dosing curves
drugs with highly specific therapeutic indexes, for all possible drug combinations. However,
pharmacokinetic/pharmacodynamic models have because PK/PD modeling relies heavily on a
been developed to provide more accurate dosing mechanistic understanding of drug metabolites, it
regimens. cannot predict dosages and interactions with
PK/PD modeling seeks to mechanistically drugs whose mechanisms are unknown or
model the path across compartments (PK) and unclear. The accuracy of PK/PD interaction mod-
response (PD) to a drug. The data used to build eling is dependent on understanding of metabo-
PK/PD equations rely on administering the drug lites including intermediates and their distribution
to a small group of participants from whom in the body, as well as all receptors that interact
metabolites and drug concentrations are sampled with the drugs and their distribution across differ-
to determine drug elimination rates [67, 68]. ent compartments. Therefore, PK/PD in the pres-
Although this method takes drug mechanisms ent day suffers for being unable to account for
and metabolism into consideration, ethical con- interactions with common medications with
siderations exclude populations like children and unknown mechanisms, such as ketamine, lith-
318 S. Klein et al.

ium, and general anesthetics [70–72]. These 18.4.3 TDM


challenges are gradually being overcome as
in vitro and in vivo experimentations using small Therapeutic drug monitoring (TDM) is the prac-
to large models aim to elucidate drug actions at tice of monitoring concentrations or clinical
all levels of biology [73, 74]. responses of a drug (and/or its metabolites in the
Standardization of data collection also con- relevant compartments). This process is similar
tributes to the complexity of PK/PD modeling to the test done in a clinical trial for determining
and is vital to creating accurate and reliable PK/PD models, but instead adjusts the patient’s
mechanistic profiles of drugs. Recent PK/PD data dose of the drug in real time to achieve the desired
of mass antiviral agent testing to combat the concentrations for optimal effect [77].
recent COVID-19 pandemic has highlighted TDM circumvents many of the difficulties of
many issues such as inconsistencies in methodol- other models: eliminating the need to consider
ogy, lack of demographic data, or limitations in drug loss during administration by focusing on
patient populations and controls due to co-­ final drug concentration and ignoring differences
administration of other agents [75]. Supplemental in patient metabolism by measuring drug concen-
tools to PK/PD modeling such as PopPK rely tration in the desired compartment (i.e., plasma).
heavily on accumulation of clinical data, making Clinical, biochemical, and plasma drug concen-
standardization of data collection and faithful trations may all be valid options regarding the
reporting indubitably essential to actively characteristics of the drug of interest. The diffi-
improving PK/PD model accuracy and clinical culties in applying TDM successfully stem from
relevance. deciding which measurement to use, as TDM
PK/PD modeling is highly specific and there- was first developed based on blood pressure med-
fore may have issues with real-world complica- ications, where it is relatively easy to monitor and
tions when it comes to dosing in the clinical precisely titrate concentration of the drug over
setting. The original model is unable to account time to create the desired blood pressure.
for physiological realities such as changes in However, for other drugs, differences in patient
blood flow, which affects concentration of the response may not be due to differences in enzy-
drug to different compartments. Additional mod- matic concentrations or efficiencies in metaboliz-
els such as physiologically based pharmacoki- ing the drug, but rather due to factors like
netic model (PBPK) are in development to differing receptors or secondary signaling, mak-
provide a more granular version of PK/PD to ing the concentration of the drug irrelevant to the
account for additional factors and nested com- optimal therapeutic dose. This makes TDM less
partments that can predict drug flow across effective in cases where the physiological
organelles. However, such methods may worsen response is not as clear, immediate, and measur-
the issues of complexity that already exist within able as blood pressure.
PK/PD modeling, especially during stages of The TDM requirement of measurement of
drug development where efficacy of a particular drug concentration in relevant compartments
drug may not be certain [76]. Since PK/PD dos- may provide real-world difficulties, such as sam-
ing calculations are so specific, real-world limita- pling from difficult to assay areas such as CSF,
tions of drug delivery can also influence patients’ hepatic circulation, or intracellular concentra-
actual exposure to a drug. Variations in formula- tion. Downstream difficulties in storage, trans-
tions of certain commercially available drugs, the port, and consistent measurement in the
need to compound drugs in house, or loss of for- laboratory provide further challenges in
mulation in IV or PICC lines can negate much of ­accurately measuring TDM. At this point in time,
the work done in refining models to ensure ade- generalized methods of analysis include anti-
quate dosage. body-based and chromatography-based methods,
which specialize in measuring protein-­based and
synthetic-based compounds, respectively. Further
18 Models for Drug Individualization: Patient to Population Level 319

assessment of drug interactions using TDM is 3. Phillips CJ. Precision medicine and its imprecise his-
tory. Harv Data Sci Rev. 2020;2(1). https://doi.org/10
reliant on understanding of the drug’s PK and .1162/99608f92.3e85b56a.
PD, such that that effective use of TDM must be 4. Osborn CY, Kripalani S, Goggins KM, Wallston
based on previous work with PK/PD models. KA. Financial strain is associated with medication
Therefore, TDM may serve to supplement rather nonadherence and worse self-rated health among car-
diovascular patients. J Health Care Poor Underserved.
than supplant PK/PD modeling, given its need of 2017;28(1):499–513. https://doi.org/10.1353/
stringent patient monitoring requires personnel hpu.2017.0036.
and clinical environments that can support the 5. Denford S, Frost J, Dieppe P, Cooper C, Britten
testing regimens. N. Individualisation of drug treatments for patients
with long-term conditions: a review of concepts. BMJ
Open. 2014;4(3):e004172. Published 2014 Mar 26.
https://doi.org/10.1136/bmjopen-­2013-­004172.
18.5 Conclusion 6. Helmer A, Slater N, Smithgall S. A review of ACE
inhibitors and ARBs in black patients with hyper-
tension. Ann Pharmacother. 2018;52(11):1143–51.
Despite these growing areas of study and tools https://doi.org/10.1177/1060028018779082.
for pharmacotherapy, the practice of personaliz- 7. Fountain JH, Lappin SL. Physiology, renin angio-
ing therapy is often left in the hands of the pre- tensin system. [Updated 2021 Jul 22]. In: StatPearls
scribing physician and clinical team. Existing [Internet]. Treasure Island: StatPearls Publishing;
2021. Available from: https://www.ncbi.nlm.nih.gov/
data on the drug profiles of many medications is books/NBK470410/.
based on randomized control trials, which will 8. Flack JM, Sica DA, Bakris G, et al. Management
use the same dosage across multiple people from of high blood pressure in Blacks: an update
differing backgrounds. Although this is helpful of the International Society on Hypertension
in Blacks consensus statement. Hypertension.
for blanket prescription practices, physicians are 2010;56(5):780–800. https://doi.org/10.1161/
often left to fine-tune the medications to the HYPERTENSIONAHA.110.152892.
patient’s individual needs through adjusting dos- 9. Wilder J, Saraswathula A, Hasselblad V, Muir
age, delivery, or personal preferences of the A. A systematic review of race and ethnicity in
hepatitis C clinical trial enrollment. J Natl Med
patient for one drug over another [2]. Therapy Assoc. 2016;108(1):24–9. https://doi.org/10.1016/j.
individualization must encompass the patient’s jnma.2015.12.004.
individual physical characteristics, such as age, 10. Benhammou JN, Dong TS, May FP, et al. Race affects
weight, gender, or ethnicity; their medical his- SVR12 in a large and ethnically diverse hepatitis
C-infected patient population following treatment
tory, including drug history, history of renal or with direct-acting antivirals: analysis of a single-­center
hepatic failure, and comorbidities; genetic poly- Department of Veterans Affairs cohort. Pharmacol
morphisms of the patient; family history; and Res Perspect. 2018;6(2):e00379. Published 2018 Feb
sociocultural understandings. By having a core 22. https://doi.org/10.1002/prp2.379.
11. Zhang E, Levin AM, Williams LK. How does race
understanding of these factors, physicians can and ethnicity effect the precision treatment of asthma?
adjust to changing knowledge of drug prescrip- Expert Rev Precis Med Drug Dev. 2019;4(6):337–56.
tion practices and most importantly prescribe https://doi.org/10.1080/23808993.2019.1690396.
medications not based on a formula, but rather on 12. Samedy-Bates LA, Oh SS, Nuckton TJ, et al.
Racial/ethnic-specific differences in the effects
the individual needs of the patient. of inhaled corticosteroid use on bronchodilator
response in patients with asthma. Clin Pharmacol
Ther. 2019;106(5):1133–40. https://doi.org/10.1002/
References cpt.1555.
13. Reddick B. Fallacies and dangers of practic-
ing race-based medicine. Am Fam Physician.
1. Kleisiaris CF, Sfakianakis C, Papathanasiou IV. Health
2021;104(2):122–3.
care practices in ancient Greece: the Hippocratic
14. Goodman CW, Brett AS. Race and
ideal. J Med Ethics Hist Med. 2014;7:6–6.
pharmacogenomics-­ personalized medicine or mis-
2. Lesko LJ, Schmidt S. Individualization of drug ther-
guided practice? JAMA. 2021;325(7):625–6. https://
apy: history, present state, and opportunities for the
doi.org/10.1001/jama.2020.25473.
future. Clin Pharmacol Ther. 2012;92(4):458–66.
15. Batchelor HK, Marriott JF. Paediatric pharmaco-
https://doi.org/10.1038/clpt.2012.113.
kinetics: key considerations. Br J Clin Pharmacol.
320 S. Klein et al.

2015;79(3):395–404. https://doi.org/10.1111/ 28. Wolbold R, Klein K, Burk O, et al. Sex is a major


bcp.12267. determinant of CYP3A4 expression in human
16. Evans WE, Relling MV, de Graaf S, et al. Hepatic drug liver. Hepatology. 2003;38(4):978–88. https://doi.
clearance in children: studies with indocyanine green org/10.1053/jhep.2003.50393.
as a model substrate. J Pharm Sci. 1989;78(6):452–6. 29. Dadashzadeh S, Javadian B, Sadeghian S. The effect
https://doi.org/10.1002/jps.2600780605. of gender on the pharmacokinetics of verapamil and
17. Oong GC, Tadi P. Chloramphenicol. [Updated 2021 norverapamil in human. Biopharm Drug Dispos.
Sep 29]. In: StatPearls [Internet]. Treasure Island: 2006;27(7):329–34. https://doi.org/10.1002/bdd.512.
StatPearls Publishing; 2021. Available from: https:// 30. Moyer AM, Matey ET, Miller VM. Individualized
www.ncbi.nlm.nih.gov/books/NBK555966/. medicine: sex, hormones, genetics, and adverse drug
18. Rivera-Chaparro ND, Cohen-Wolkowiez M, reactions. Pharmacol Res Perspect. 2019;7(6):e00541.
Greenberg RG. Dosing antibiotics in neonates: review Published 2019 Dec 6. https://doi.org/10.1002/
of the pharmacokinetic data. Future Microbiol. prp2.541.
2017;12(11):1001–16. https://doi.org/10.2217/ 31. Granfors MT, Backman JT, Laitila J, Neuvonen
fmb-­2017-­0058. PJ. Oral contraceptives containing ethinyl estradiol
19. Sande A, Burgoine K, La Valle S, Talewoya and gestodene markedly increase plasma concentra-
S. Neonatal drug formulary. Neonatal Unit tions and effects of tizanidine by inhibiting cytochrome
(NNU) Mbale Regional Referral Hospital. 2017. P450 1A2. Clin Pharmacol Ther. 2005;78(4):400–11.
https://gh.bmj.com/content/bmjgh/3/1/e000586/ https://doi.org/10.1016/j.clpt.2005.06.009.
DC2/embed/inline-­s upplementary-­m aterial-­2 . 32. Choi JS, Koren G, Nulman I. Pregnancy and isotreti-
pdf?download=true. noin therapy. CMAJ. 2013;185(5):411–3. https://doi.
20. Mutlu M, Aslan Y, Kader Ş, Aktürk-Acar F, Dilber org/10.1503/cmaj.120729.
E. Clinical signs and symptoms of toxic serum digoxin 33. Mauvais-Jarvis F, Berthold HK, Campesi I, et al. Sex-
levels in neonates. Turk J Pediatr. 2019;61(2):244– and gender-based pharmacological response to drugs
249. https://doi.org/10.24953/turkjped.2019.02.013. [published correction appears in Pharmacol Rev.
PMID: 31951334. 2021;73(2):860]. Pharmacol Rev. 2021;73(2):730–
21. Orenstein SR, Shalaby TM, Devandry SN, Liacouras 62. https://doi.org/10.1124/pharmrev.120.000206.
CA, Czinn SJ, Dice JE, Simon TJ, Ahrens SP, Stauffer 34. Poels EMP, Bijma HH, Galbally M, Bergink V. Lithium
LA. Famotidine for infant gastro-­ oesophageal during pregnancy and after delivery: a review. Int J
reflux: a multi-centre, randomized, placebo-­ Bipolar Disord. 2018;6(1):26. Published 2018 Dec 2.
controlled, withdrawal trial. Aliment Pharmacol https://doi.org/10.1186/s40345-­018-­0135-­7.
Ther. 2003;17(9):1097–107. https://doi.org/10.1046/ 35. Giles JJ, Bannigan JG. Teratogenic and developmental
j.1365-­2036.2003.01559.x. PMID: 12752346. effects of lithium. Curr Pharm Des. 2006;12(12):1531–
22. Wang AD, Green D, Seo S, Fisher J, Mulberg A, 41. https://doi.org/10.2174/138161206776389804.
McCune S, Burckart G. A survey of neonatal phar- 36. Tsamantioti ES, Hashmi MF. Teratogenic medica-
macokinetic and pharmacodynamic studies in tions. [Updated 2021 Aug 9]. In: StatPearls [Internet].
pediatric drug development. Clin Pharmacol Ther. Treasure Island: StatPearls Publishing; 2021.
2015;98(3):328–35. https://doi.org/10.1002/cpt.149. Available from: https://www.ncbi.nlm.nih.gov/books/
23. Pacifici GM. Clinical pharmacology of furosemide NBK553086/.
in neonates: a review. Pharmaceuticals (Basel). 37. Bacino CA. Birth defects: causes. UptoDate. 2021.
2013;6(9):1094–129. Published 2013 Sep 5. https:// https://www-uptodate-com.ezproxy.hsc.usf.edu/con-
doi.org/10.3390/ph6091094. tents/birth-defects-causes?search=teratogenic%20
24. Ziesenitz VC, Zutter A, Erb TO, van den Anker d r u g s% 2 0 i n % 2 0 p r eg n a n cy & so u r c e = se a r c h _
JN. O-23 Ibuprofen in infants younger than 6 result&selectedTitle=2~150&usa
months: what is the efficacy and safety profile? ge_type=default&display_rank=2#H1671907210.
Arch Dis Child. 2017. https://doi.org/10.1136/ 38. Hooton TM, Besser R, Foxman B, Fritsche TR,
archdischild-­2017-­esdppp.23. Nicolle LE. Acute uncomplicated cystitis in an era of
25. Cheng A, Banwell B, Levin S, Seabrook JA, Freeman increasing antibiotic resistance: a proposed approach
D, Rieder M. Oral dosing requirements for phenytoin to empirical therapy. Clin Infect Dis. 2004;39(1):75–
in the first three months of life. J Popul Ther Clin 80. https://doi.org/10.1086/422145.
Pharmacol. 2010;17(2):e256–61. Epub 2010 Jul 6. 39. Paterson DL. Impact of antibiotic resistance in gram-­
PMID: 20664118. negative bacilli on empirical and definitive antibiotic
26. American Geriatrics Society 2019. Updated AGS therapy. Clin Infect Dis. 2008;47:S14–20. https://doi.
Beers Criteria® for potentially inappropriate org/10.1086/590062.
medication use in older adults. J Am Geriatr Soc. 40. Neuhauser MM, Weinstein RA, Rydman R,
2019;67:674–94. https://doi.org/10.1111/jgs.15767. Danziger LH, Karam G, Quinn JP. Antibiotic resis-
27. Soldin OP, Mattison DR. Sex differences in phar- tance among gram-negative bacilli in US intensive
macokinetics and pharmacodynamics. Clin care units: implications for fluoroquinolone use.
Pharmacokinet. 2009;48(3):143–57. https://doi. JAMA. 2003;289(7):885–8. https://doi.org/10.1001/
org/10.2165/00003088-­200948030-­00001. jama.289.7.885.
18 Models for Drug Individualization: Patient to Population Level 321

41. Kuepfer L, Niederalt C, Wendl T, Schlender JF, 2019;196:1–8, . ISSN 0376-8716. https://doi.
Willmann S, Lippert J, Block M, Eissing T, Teutonico org/10.1016/j.drugalcdep.2018.12.004.
D. Applied concepts in PBPK modeling: how to 52. Kim YH, Kang P, Cho CK, Jung EH, Park HJ, Lee
build a PBPK/PD model. CPT Pharmacometrics YJ, Bae JW, Jang CG, Lee SY. Physiologically
Syst Pharmacol. 2016;5(10):516–31. https://doi. based pharmacokinetic (PBPK) modeling for pre-
org/10.1002/psp4.12134. Epub 2016 Oct 19. PMID: diction of celecoxib pharmacokinetics according to
27653238; PMCID: PMC5080648. CYP2C9 genetic polymorphism. Arch Pharm Res.
42. Abduljalil K, Badhan RKS. Drug dosing dur- 2021;44(7):713–24. https://doi.org/10.1007/s12272-­­
ing pregnancy-­ opportunities for physiologically 021-­01346-­2. Epub 2021 Jul 25. PMID: 34304363.
based pharmacokinetic models. J Pharmacokinet 53. Westervelt P, Cho K, Bright DR, Kisor DF. Drug-gene
Pharmacodyn. 2020;47(4):319–40. https://doi. interactions: inherent variability in drug maintenance
org/10.1007/s10928-­020-­09698-­w. Epub 2020 Jun dose requirements. P T. 2014;39(9):630–7.
26. PMID: 32592111. 54. Johansson I, Ingelman-Sundberg M. Genetic poly-
43. Sharma RP, Schuhmacher M, Kumar V. The develop- morphism and toxicology—with emphasis on cyto-
ment of a pregnancy PBPK Model for Bisphenol A chrome P450. Toxicol Sci. 2011;120(1):1–13. https://
and its evaluation with the available biomonitoring doi.org/10.1093/toxsci/kfq374.
data. Science of The Total Environment, Volume. 55. Shenfield GM. Genetic polymorphisms, drug metabo-
2018;624:55–68 . ISSN 0048-9697. https://doi. lism and drug concentrations. Clin Biochem Rev.
org/10.1016/j.scitotenv.2017.12.023. 2004;25(4):203–6.
44. Morck TJ, Sorda G, Bechi N, Rasmussen BS, Nielsen 56. Pan SD, Zhu LL, Chen M, Xia P, Zhou Q. Weight-­
JB, Ietta F, Rytting E, Mathiesen L, Paulesu L, based dosing in medication use: what should we
Knudsen LE. Placental transport and in vitro effects know? Patient Prefer Adherence. 2016;10:549–60.
of Bisphenol A. Reprod Toxicol. 2010;30:131–7. Published 2016 Apr 12. https://doi.org/10.2147/PPA.
https://doi.org/10.1016/j.reprotox.2010.02.007. S103156.
45. Jiang HM, Fang ZZ, Cao YF, Hu CM, Sun XY, Hong 57. Mueller JA, Patel T, Halawa A, Dumitrascu A,
M, Yang L, Ge GB, Liu Y, Zhang YY, Dong Q, Liu Dawson NL. Warfarin dosing and body mass index.
RJ. New insights for the risk of bisphenol A: inhi- Ann Pharmacother. 2014;48(5):584–8. https://doi.
bition of UDP-glucuronosyltransferases (UGTs). org/10.1177/1060028013517541. Epub 2014 Feb 20.
Chemosphere. 2013;93(6):1189–93. https://doi. PMID: 24558184.
org/10.1016/j.chemosphere.2013.06.070. Epub 2013 58. Pai MP. Drug dosing based on weight and body surface
Aug 12. PMID: 23948605. area: mathematical assumptions and limitations in
46. Conroy S, McIntyre J, Choonara I, Stephenson obese adults. Pharmacotherapy. 2012;32(9):856–68.
T. Drug trials in children: problems and the way for- https://doi.org/10.1002/j.1875-­9114.2012.01108.x.
ward. Br J Clin Pharmacol. 2000;49(2):93–7. https:// Epub 2012 Jun 18. PMID: 22711238.
doi.org/10.1046/j.1365-­2125.2000.00125.x. PMID: 59. Crawford JD, Terry ME, Rourke GM. Simplification
10671901; PMCID: PMC2014901. of drug dosage calculation by application of the sur-
47. Björkman S, Wada DR, Berling B-M, Benoni face area principle. Pediatrics. 1950;5:783–90.
G. Prediction of the disposition of midazolam in sur- 60. Barras M, Legg A. Drug dosing in obese adults. Aust
gical patients by a physiologically based pharmacoki- Prescr. 2017;40(5):189–93. https://doi.org/10.18773/
netic model. J Pharm Sci. 2001;90:1226–41. https:// austprescr.2017.053.
doi.org/10.1002/jps.1076. 61. Morgan DJ, Bray KM. Lean body mass as a predic-
48. Nafziger AN, Bertino JS Jr. Sex-related differences in tor of drug dosage. Implications for drug therapy. Clin
theophylline pharmacokinetics. Eur J Clin Pharmacol. Pharmacokinet. 1994;26:292–307.
1989;37(1):97–100. https://doi.org/10.1007/ 62. Cella M, et al. A model-based approach to dose selec-
BF00609434. PMID: 2591473. tion in early pediatric development. Clin Pharmacol
49. Yeo K. How PBPK modeling can replace drug-drug Ther. 2010;87(3):294–302.
interaction studies – ACRP. [online]. 2021. Available 63. Gibson PS, Newell K, Sam DX, et al. Weight-adjusted
at: https://acrpnet.org/2020/11/10/how-­pbpk-­ dosing of tinzaparin in pregnancy. Thromb Res.
modeling-­can-­replace-­drug-­drug-­interaction-­studies/. 2013;131:e71–5.
50. Ji B, Liu S, Xue Y, He X, Man VH, Xie XQ, Wang 64. Matson KL, Horton ER, Capino AC, Advocacy
J. Prediction of drug-drug interactions between opi- Committee for the Pediatric Pharmacy Advocacy
oids and overdosed benzodiazepines using physi- Group. Medication dosage in overweight and obese
ologically based pharmacokinetic (PBPK) modeling children. J Pediatr Pharmacol Ther. 2017;22(1):81–3.
and simulation. Drugs R D. 2019;19(3):297–305. https://doi.org/10.5863/1551-­6776-­22.1.81.
https://doi.org/10.1007/s40268-­019-­00282-­3. PMID: 65. Gade C, Christensen HR, Dalhoff KP, Holm JC, Holst
31482303; PMCID: PMC6738369. H. Inconsistencies in dosage practice in children
51. Dai Z, Abate MA, Smith GS, Kraner JC, Mock with overweight or obesity: a retrospective cohort
AR. Fentanyl and fentanyl-analog involvement study. Pharmacol Res Perspect. 2018;6(3):e00398.
in drug-related deaths. Drug Alcohol Depend. https://doi.org/10.1002/prp2.398. PMID: 29721323;
PMCID: PMC5909170.
322 S. Klein et al.

66. Burke CN, Voepel-Lewis T, Wagner D, Lau I, Baldock 72. Lugli KA, Yost CS, Kindler CH. Anaesthetic
A, Malviya S, Nafiu O. A retrospective description mechanisms: update on the challenge of unravel-
of anesthetic medication dosing in overweight and ling the mystery of anaesthesia. Eur J Anaesthesiol.
obese children. Paediatr Anaesth. 2014;24(8):857–62. 2009;26(10):807–20. https://doi.org/10.1097/
https://doi.org/10.1111/pan.12396. Epub 2014 Apr 8. EJA.0b013e32832d6b0f.
PMID: 24708463. 73. Jusko WJ, Hui CK. Physiologic indirect response mod-
67. Hawcutt DB, Smyth RL. One size does not fit all: els characterize diverse types of pharmacodynamic
getting drug doses right for children. Arch Dis Child. effects. Clin Pharmacol Ther. 1994;56(4):406–19.
2008;93(3):190–1. 74. Mager DE, Sukyung W, Jusko WJ. Scaling phar-
68. US Food and Drug Administration. Guidance for macodynamics from in vitro and preclinical animal
industry. E11 clinical investigation of medicinal prod- studies to humans. Drug Metab Pharmacokinet.
ucts in the pediatric population. December 2000. 2009;24(1):16–24.
69. Wicha SG, et al. A general pharmacodynamic interac- 75. Venisse N, Peytavin G, Bouchet S, et al. Concerns
tion model identifies perpetrators and victims in drug about pharmacokinetic (PK) and pharmacokinetic-­
interactions. Nat Commun. 2017;8(1):1–11. pharmacodynamic (PK-PD) studies in the new
70. Shaldubina A, Agam G, Belmaker RH. The mecha- therapeutic area of COVID-19 infection. Antivir
nism of lithium action: state of the art, ten years Res. 2020;181:104866. https://doi.org/10.1016/j.
later. Prog Neuro-Psychopharmacol Biol Psychiatry. antiviral.2020.104866.
2001;25(4):855–66. https://doi.org/10.1016/s0278-­­ 76. Hussey E. Pharmacokinetics and pharmacody-
5846(01)00154-­3. PMID: 11383981. namics (PK/PD). 2021. Retrieved 21 December
71. Strasburger SE, et al. What is the mechanism of 2021, from https://www.nuventra.com/services/
Ketamine’s rapid-onset antidepressant effect? A con- pharmacokinetics-­pharmacodynamics/.
cise overview of the surprisingly large number of pos- 77. Schumacher GE. Therapeutic drug monitoring.
sibilities. J Clin Pharm Ther. 2017;42(2):147–54. Norwalk: Appleton & Lange; 1995.
Toxicity Evaluation
of Nanomedicine
19
Archna Panghal and Swaran Jeet Singh Flora

Abstract ing/concerns due to associated toxicity. The


unknown biodistribution pattern and unde-
Nanomedicine is the application of nanotech- fined fate in biological system also contribute
nology to achieve innovation in healthcare. It to their miscellaneous actions on organs. The
uses the properties developed by a material at small size of nanoparticles allows them to
its nanometric scale 10−9 m which often differ breach the biological barriers such as blood-­
in terms of physics, chemistry, or biology brain barrier (BBB), blood-testis barrier
from the same material at a bigger scale. (BTB), or other barriers resulting in organ tox-
Nanomedicine is understood to be a key icity such as neurotoxicity, reproductive toxic-
enabling instrument for personalized, targeted ity and other toxicities. Although size and
and regenerative medicine by delivering the shape are strong determinants of toxicity of
next level of new drugs, treatments and nanomedicines, other factors such as chemical
implantable devices to clinicians and patients, composition, charge and structure also should
for real breakthroughs in medicine. Several not be ignored as they influence the interac-
areas of medical care are already benefiting tion of nanomedicine with biological targets.
from the advantages that nanotechnology can Although the knowledge regarding molecular
offer. Researchers are still fighting against mechanisms of nanomedicine-induced toxic-
numerous serious and complex illnesses like ity is still in its infancy, mechanisms such as
cancer, cardiovascular diseases, multiple scle- oxidative stress, inflammation, apoptosis and
rosis, Parkinson’s disease and infectious dis- deoxyribonucleic acid (DNA) damage are
eases like human immunodeficiency virus considered key players in toxicity. Although
(HIV). This chapter discusses the mechanisms nanomedicine toxicity has caught the atten-
and strategies for improving the safety of tion of researchers in the last decade, the cur-
nanoparticles including important consider- rent knowledge is inadequate and requires
ations for nanoparticle safety assessment. extensive future studies. In our opinion we
Despite success of nanomedicines in bio- need to assess risk potential of nanomedicine,
medical sciences, there are several shortcom- limitations of experimental methods and pre-
cise characterization of nanomaterials.
A. Panghal · S. J. S. Flora (*)
Department of Pharmacology and Toxicology,
National Institute of Pharmaceutical Education and
Research, Mohali, India

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 323
S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_19
324 A. Panghal and S. J. S. Flora

Keywords actions on organs. The small size allows them to


breach the biological barriers such as blood-brain
Nanomedicine · Nanoparticle · Nanosafety · barrier (BBB), blood-testis barrier (BTB), or
Nanotoxicity · Organ toxicity · Safety · other barriers resulting in organ toxicity such as
Toxicity neurotoxicity, reproductive toxicity and other
toxicities [6]. Although size and shape are strong
determinants of toxicity of nanomedicines, other
19.1 Introduction factors such as chemical composition, charge and
structure also should not be ignored as they influ-
Pharmaceutical applications of nanotechnology ence the interaction of nanomedicine with bio-
have largely transformed healthcare system in the logical targets [7, 8]. For instance, metal ions
twenty-first century. It includes the development such as silver (Ag), titanium (Ti) and cadmium
of nanomedicines which deal with the biological (Cd) present in nanomedicines are inherently
systems in dimensions even less than 100 nm by toxic, whereas metals such as iron (Fe) and zinc
multidisciplinary approaches. The term nano- (Zn) are biologically tolerable, but might induce
medicine has been interchangeably used with toxicity at concentration beyond physiological
nanopharmaceuticals since its advent [1]. levels [9, 10]. Although the knowledge regarding
Nanomedicines are synthesized by involving molecular mechanisms of nanomedicine-induced
nanomaterials which provide them edge over toxicity is still in its infancy, mechanisms such as
conventional therapeutics by modulating several oxidative stress, inflammation, apoptosis and
properties such as solubility and surface to vol- DNA damage are considered key players involved
ume ratio. Generally, they are nanomaterials in toxicity [11]. Genotoxicity and cytotoxicity
which have noteworthy applications in biomedi- have been frequently reported with metal oxide
cal science such as diagnosis, monitoring and and carbon-based nanomedicine [12]. Polymer-
therapeutic implications for a particular condi- and silica-based nanomedicines have been found
tion by targeting a specific organ or tissue [2]. to be relatively less toxic and more biocompati-
Nano-size, enhanced permeation rate, site-­ ble; however, ROS formation is considered to be
specific active as well as passive targeting, eco- involved in toxicity to some extent [13]. Although
nomic suitability and higher safety and efficacy nanomedicine toxicity has caught the attention of
have revolutionized use of nanomedicines in bio- researchers in the last decade, the current knowl-
medical field [1, 3]. In future, the practice of edge is mere a drop in the ocean and thus more
nanomedicines in healthcare system will flourish needs to be explored to understand the toxicity of
due to their breakthrough success and as an addi- nanomedicines in depth. One of the most proba-
tional revenue-generating system for pharmaceu- ble reasons of lagging behind in assessing risk
tical players. However, there are few factors potential of nanomedicine might be limitations of
which need to be considered to improve current experimental methods and precise characteriza-
generation of nanomedicines. These factors tion of nanomaterials.
include inculcating properties to evade innate Identifying the toxicity potential of nanomedi-
defence system of body, traceability within bio- cine at an early stage has become a key issue in
logical system, effective pharmacokinetic profile, their wide applications. The limitations of nano-
biodegradability and reduced toxicity [4]. medicines need to be overcome by effective mon-
Despite worth mentioning success of nano- itoring and defining their fate in biological system
medicines in biomedical system, still there are to ensure safety [14]. Since physico-chemical
several concerns due to associated toxicity owing profile determines the toxicity of nanomedicine,
to their specific physico-chemical and pharmaco- thorough characterization is indispensable to
kinetic profile [5]. Further, the unknown biodis- understand their interaction with cellular targets.
tribution pattern and undefined fate in biological Further, multidisciplinary studies need to be car-
system also contribute to their miscellaneous ried out in addition to implementation of
19 Toxicity Evaluation of Nanomedicine 325

advanced testing procedures to explore the tox- disease and rare genetic disorders [19]. Clinically
icities of nanomedicines. Validated testing used nanoformulations significantly enhanced
­procedures such as in vitro and in vivo shall be biological half-life, improved bioavailability and
upheld for benefit-risk assessment of nanomedi- limited the adverse effects as compared to con-
cines. Further, adequate data generation on ventional medicines. The statistics shows that not
ADME profile and acute and chronic toxicity via only the approval rate of nanomedicine is increas-
in vivo studies might prove beneficial in limiting ing, but new generation of nanomedicines is also
the toxicity of nanomedicine [15]. Although making their way to the clinic [18]. Besides con-
OECD guidelines provide elaborated protocols ventional molecules, nanomedicines also have
which need to be followed to determine toxicity potential to deliver DNA, siRNA, miRNA and
of nanomedicines on regular basis, yet they need peptides-based moiety. The approval of first
to be refined to focus more on hazards and bio- siRNA-encapsulating nanodrug Onpattro for
safety of nanomaterials [16]. Sensors (such as transthyretin amyloidosis in 2018 provided oppor-
biosensors and chemical sensors)-based approach tunities for nucleic acid delivery [20]. One of the
for real time toxicity assessment and monitoring most noteworthy accomplishments has been the
of nanomedicines might prove highly advanta- development of first polymer-based biodegrad-
geous over conventional approaches. able nanoparticle competent of targeting multi-
Additionally, implication of more advanced tech- drug-resistant microbes by acting on bacterial cell
niques including proteomics, genomics and walls and membranes [21]. Further, the most
metabolomics might be of critical interest to thrilling progress appears to be the in vivo appli-
understand the risk potential more precisely [17]. cation of DNA nanorobots in oncology for shrink-
This chapter focuses on risk potential of nano- ing the tumours by cutting blood supplies [22].
medicine along with major molecular pathways
involved in their toxicity. Further, conventional
and novel approaches implemented in detection
of toxicity due to nanomedicines also have been
summarized.

19.2 Breakthrough Success


of Nanomedicine

Implication of nanotechnology in the field of


pharmaceuticals has been a breakthrough step for Application of nanomedicine
development of several therapeutics of interest.
The roadmap for the use of nanomedicines for
mankind is depicted in Fig. 19.1. With the advent
of nanotechnology, several nanomedicines based
on dendrimers, micelles, liposomes, protein-drug In vitro safety screening
conjugates, nanoparticles (NPs) and hydrogels In vivo preclinical safety and biodistribution
have been developed [18]. The first landmark
achievement was the approval of liposomal for-
mulation of doxorubicin (Doxil) by USFDA in
1995. More than 50 nanomedicines are in clinical
use currently (Table 19.1). These marketed medi- Design and development of nanomedicine
cations are used in several medical conditions
such as cancer, neurodegenerative disorders, Fig. 19.1 A brief roadmap for nanomedicine-based
anaesthesia, rheumatoid arthritis, cardiovascular application in humans
326 A. Panghal and S. J. S. Flora

Table 19.1 FDA-approved nanomedicines [24, 25]


Approval
Drug name Company Applications/indications year
INFeD Allergan Iron-deficient anaemia 1992
Epaxal Crucell, Berna Biotech Hepatitis A 1993
Oncaspar Servier Pharmaceuticals Acute lymphoblastic leukaemia 1994
Doxil Janssen Kaposi sarcoma, ovarian cancer, multiple myeloma 1995
Abelcet Sigma-Tau Fungal infections 1995
Pharmaceuticals
Amphotec Ben Venue Laboratories Fungal infections 1996
Inc.
DaunoXome Galen Kaposi sarcoma 1996
Copaxone Teva Multiple sclerosis 1996
Dexferrum American Regent Iron-deficient anaemia 1996
AmBisome Gilead Sciences Fungal/protozoal infections 1997
Inflexal Crucell, Berna Biotech Influenza 1997
Ferrlecit Sanofi Iron deficiency in chronic kidney disease 1999
Curosurf Chiesi USA Respiratory distress syndrome 1999
DepoCyt SkyePharma Neoplastic meningitis 1999
Visudyne Bausch and Lomb Macular degeneration, myopia, ocular histoplasmosis 2000
Myocet Elan Pharmaceuticals Metastatic breast cancer 2000
Venofer American Regent Iron deficiency in chronic kidney disease 2000
Peglntron Merck Hepatitis C infection 2001
Neulasta Amgen Neutropenia 2002
Eligard Tolmar Prostate cancer 2002
Emend Merck Antiemetic 2003
Mepact Takeda Pharmaceutical Non-metastatic osteosarcoma 2004
Ltd.
DepoDur SkyPharma Inc. Pain management 2004
Tricor Lupin Atlantis Hyperlipidaemia 2004
Abraxane Celgene Lung cancer, metastatic breast cancer, metastatic 2005
pancreatic cancer
Focalin XR Novartis Psychostimulant 2005
Cimzia UCB Crohn’s disease, rheumatoid arthritis, ankylosing 2008
spondylitis
Feraheme AMAG Iron deficiency in chronic kidney disease 2009
Exparel Pacira Pharmaceuticals Pain management 2011
Inc.
Marqibo Acrotech Biopharma Acute lymphoblastic leukaemia 2012
Injectafer American Regent Iron deficiency anaemia 2013
Plegridy Biogen Multiple sclerosis 2014
Onivyde Ipsen Metastatic pancreatic cancer 2015
ADYNOVATE Takeda Haemophilia 2015
Vyxeos Jazz Pharmaceuticals Acute myeloid leukaemia 2017
Onpattro Alnylam Pharmaceuticals Transthyretin-mediated amyloidosis 2018

The impact of nanotechnology in the biomedical in May 2020 with the initiation of three clinical
field is evident by the fact that more than 247 new trials [23]. Taken together, landmark escalation in
clinical trials started in the beginning of 2018. the entry of nanomedicines in the clinical trials
Further, a quest for developing lipid-based nano- followed by clinics is itself proof of breakthrough
medicines of COVID-19 vaccines already started success in improving mankind health.
19 Toxicity Evaluation of Nanomedicine 327

19.3 Benefit-Risk Assessment tion. Route of administration is also a contributing


of Nanotherapeutics factor in toxicity profile of nanomedicines.
Increased endocytosis rate due to distinct
Emergence of nanomedicine has provided new physico-chemical properties of nanomedicines
opportunities for improving the poor drug solubil- initiates inflammatory and pro-oxidant responses
ity and drug targeting. Along with the better safety which further trigger the activation of different
and efficacy, nanoformulations also allow dose molecular pathways leading to toxicity at organ,
reduction which improves patient compliance and tissue, cellular and molecular levels.
reduces the toxic effects owing to accumulation of
high volume. Nanomedicines improve dissolution
rate, solubility and intracellular uptake of drugs 19.4.1 Hepatotoxicity
by increasing surface area. Multifunctional nano-
medicines have high therapeutic index due to bet- The liver has been recognized as a vital organ
ter efficacy of drug delivery and release at the where >90% nanomaterials are accumulated
specific site [26]. Nanoformulations have poten- from systemic circulation. Meanwhile, accumu-
tial to deliver the drugs across physiological barri- lated nanomaterials interact with the hepatic cells
ers such as BBB, blood-lung barrier and BTB by and, therefore, lead to the structural and func-
facilitating the active as well as passive transport tional perturbations in the liver. Kupffer cells are
across tight junctions [27, 28]. resident macrophages in the liver which elicit
Besides the widespread use, knowledge on the immune response; however, they are not efficient
safety and toxicity profile of many nanoformula- enough to prevent the nanomaterials which get
tions is still lacking. The most often reported access through open sinusoidal fenestrations [32,
adverse effect after injecting nanotherapeutics is 33]. Recently, it was reported that cerium oxide
hypersensitivity reaction which perhaps may be (CeO2) and titanium oxide (TiO2) NPs injected
due to activation of the immune system [29]. into mice were detected into sinusoids and
Increased surface area improves solubility on one Kupffer cells up to 6 months post-exposure [34].
side, whereas it creates problems like inter-­ NPs may induce hepatic toxicity by activating the
particular friction, sticking and varied physical inflammatory signalling as evident by the ele-
and chemical interactions on another side. Further, vated pro-inflammatory and reduced anti-­
their small size contributes to high clearance rate inflammatory cytokine level in response to nickel
from the body which restricts their use for thera- oxide (NiO) NPs [35]. Serum parameters such as
peutic purposes. Another major downside of total bilirubin, alkaline phosphatase and aspartate
nanomedicines is that they do not have common aminotransferase serve as good biomarkers to
properties other than their size; therefore, each identify toxicity potential of nanomedicines [35,
nanomedicine has to be evaluated individually 36]. NPs induce hepatic toxicity via different
[30]. Nanoformulations elevate ROS level and mechanisms depending upon their size, chemical
oxidative stress which are major culprit factor for composition and physical characteristics. For
inducing several disorders such as metabolic, car- instance, Ag NPs manifest their hepatic toxicity
diovascular and neurological disorders [31]. by inducing necrosis and haemorrhage in hepato-
Further, NPs have tendency to cause inflamma- cytes and endothelial damage in portal vein [37].
tory responses and immune and systemic effects. TiO2 NPs trigger infiltration of inflammatory
cells, increase collagen deposition and induce
fibrosis [38]. Gold (Au) nanorods elicit hepatic
19.4 Toxicity Profile toxicity by activating hepatic macrophages which
of Nanomedicine subsequently aggravate hepatitis and liver dam-
age [39]. Poly-amidoamine (PAMAM) den-
In biomedical context, nanotoxicology deals with drimers have been reported to cause hepatic
the negative impact of nanomedicines on biologi- toxicity by inhibiting cell growth and inducing
cal system due to their unique surface modifica- mitochondrial injury, apoptosis and Akt/mTOR-­
328 A. Panghal and S. J. S. Flora

mediated autophagy [40]. Although the role of to cause shedding of epithelial cells, Leydig cell
inflammation, apoptosis and oxidative stress has dysfunctions and germ cell apoptosis in in vivo
been elucidated in nanomedicine-induced hepatic study [49, 50].
toxicity, in-depth understanding of metabolic Several developmental toxicities such as
changes in terms of energy, protein and lipid early miscarriages, retarded embryo and neona-
metabolism in in vivo and in vitro system is still tal growth rate have been identified against
lacking. nanotherapeutics exposure. Since foetus has not
developed adequate defence system to counter
the toxic insults, therefore it is more susceptible
19.4.2 Reproductive for nanomedicine-induced malformations [51].
and Developmental Toxicity Being small in size, nanomedicines have inher-
ent properties of crossing the placenta which is
The available data on reproductive nanotoxicity considered as the main reason behind the
is inadequate to make a conclusive remark on nanotherapeutics-­induced developmental toxic-
nanomedicine-associated risk potential to ity in embryo and foetus. The toxicity pattern
gonadal health. The scarcity of the research in involving direct interaction of nanomaterials
this field can be understood by the fact that out of with the developing embryo is regarded as direct
more than 60 countries having their own nano- effects of nanomedicines. Besides this, the con-
technology research programmes, just 29 coun- cept of indirect effect has also emerged in recent
tries have undertaken research activities in this years where the nanomaterials trigger the
domain [41]. The potential of nanostructures to release of mediators without their translocation
breach the BTB has raised concern about harmful across the placenta which results in toxicity [52,
effects of nanotherapeutics on male reproductive 53]. Besides physico-chemical factors, gesta-
system [42]. Metallic NPs made up of Ti, Ag, Zn tional stage also affects nanomedicine-associ-
and Au have been frequently reported to hamper ated developmental toxicity which makes it
normal reproductive functions in both male and complicated to predict the adverse effects [54].
female rodent models [43–45]. Decreased sperm For instance, oral administration of zinc NPs in
counts, defective oogenesis, hormonal dysfunc- mice during gestational day (GD) 7–16
tions and perturbations in hypothalamus-­ decreased foetal viability and growth, whereas
pituitary-­gonadal axis are few of the adverse no such serious effects except slight change in
effects of nanotherapeutics observed in in vivo placenta weight were noticed when adminis-
models [45, 46]. The complex interplay between tered during GD 1–10 [55]. Further, nano-engi-
oxidative stress, inflammation and apoptosis is neered formulations interfere with uterine
the most accepted mechanism for nanomedicine-­ contractility and placental vasculature [56, 57].
induced reproductive toxicity. For instance, long-­ Previous findings suggest that nanomedicines
term TiO2 NPs (10 mg/kg) exposure in female accumulate inside tissue and, therefore, alter the
mice induced inflammation and oxidative stress, transport of crucial micronutrients across the
upregulated Cyp17a1 gene expression and down- placenta as evident by the findings of Blum
regulated apoptosis regulating genes as well as et al. [58]. Upon accumulation in tissues, nano-
also [47]. Further, exposure of male mice to 50, medicines may activate oxidative stress and
150 and 450 mg/kg ZnO NPs for 14 days resulted inflammation which in turn trigger the release of
in apoptosis and endoplasmic reticulum stress-­ inflammatory cytokines in systemic circulation
mediated azoospermia and testicular toxicity in a which are transported to the tissues relevant to
dose-dependent manner [48]. Besides action on the pregnancy such as uterus, placenta and foe-
the germ cells, nanomaterials may also induce tus. These cytokines may trigger the immune
toxicity by affecting other cells such as Leydig activation in target organs which leads to the
cells and Sertoli cells. Ag NPs have been reported developmental toxicity [59].
19 Toxicity Evaluation of Nanomedicine 329

19.4.3 Pulmonary Toxicity Additionally, kidneys are highly vascular organs


and able to concentrate nanomaterials reaching to
Lungs are one of the most common routes of them via systemic circulation and therefore are
entry of NPs in the body and therefore have high highly susceptible to nanotoxicity [67].
chances of accumulation of nanomaterials. After Nanomedicines elicit their toxic response in kid-
inhalation, NPs enter into interstitial spaces fol- neys in terms of glomerular swelling, necrosis of
lowed by their infiltration to alveoli. NPs ranging epithelial cells and basal membrane degeneration
in the size of approximately 20 nm are more which are more frequently correlated with oxida-
likely to get deposited in alveoli as compared to tive stress, increased apoptosis, inflammation and
smaller and larger size particles [60]. Airway DNA damage [68]. Several studies suggested
inflammation in response to inhalation of nano- harmful effects of carbon nanotubes (CNTs) on
materials is one of the most common adverse kidneys as evident by glomerular degeneration
effects. The degree of inflammatory response of and tubular necrosis. Reddy et al. reported cyto-
airways differs depending upon the type of nano- toxicity in human embryonic kidney cell line
materials. For instance, single-walled carbon (HEK293) due to oxidative stress induced by car-
nanotubes induced higher interstitial inflamma- bon nanotubes [69]. Carbon NPs such as fuller-
tion in mice lungs as compared to carbon black enes and CNTs exhibit potentially deleterious
and quartz NPs after 7 and 90 days of intra-­ effects on kidneys by inducing alterations in
tracheal administration [61]. NPs elicit their toxic trans-epithelial electrical resistance and expres-
effects on lungs via initiation of several cellular sion of barrier proteins [70]. Long-term Ag NPs
and molecular cascades such as activation of ER exposure leads to inflammation and necrosis-­
stress, mitogen-activated protein kinases mediated ultrastructural changes in the kidney
(MAPK), nuclear factor-kB (NF-kB), tyrosine [71]. Copper NPs were found to cause redox
kinases and numerous other inflammatory and imbalance which ultimately leads to ROS pro-
fibrotic genes [62, 63]. Dokka et al. reported that duction, poor podocyte activity and apoptosis
the polyvalent cationic liposomes-induced pul- [72]. Further, ZnO NPs also have been reported
monary toxicity is mediated by reactive oxygen to induce renal toxicity by inducing hypoxia
species and that toxicity potential owes to the cat- inducing factor-1α (HIF-1α), apoptosis and
ionic charge and not to the liposome [64]. Further, autophagy in in vitro and in vivo models [73].
cationic PAMAM dendrimers have been reported Although liposomal amphotericin B is less toxic
to induce acute lung injury in mice by inducing as compared to conventional formulations, yet it
Akt-TSC2-mTOR-mediated autophagy as evi- has been associated with several side effects out
dent by amelioration of lung injury in response to of which nephrotoxicity is more pronounced.
autophagy inhibitor 3-methyladenin [65]. Approximately, 43% of persons consuming
Intraperitoneal administration of mesoporous 15 mg/kg/day liposomal amphotericin B have
silica NPs at 25, 50, 100 and 200 mg/kg dose in been reported to suffer from renal toxicity [74].
Wistar rats induced pulmonary toxicity by atten- In a recent study, authors reported that intrave-
uating redox imbalance and TNF-α activation nous injection of iron (Fe) NPs (encoated in poly-
[66]. meric micelles) at dose 5 and 15 mg/kg in mice
induces nephrotoxicity by inducing oxidative
stress [75]. Further, Yousef et al. found synergis-
19.4.4 Nephrotoxicity tic effects of aluminium oxide (Al2O3) and ZnO
NPs on hepato-renal toxicity in terms of increased
After the liver, kidneys are other prominent tar- DNA damage and pro-oxidant levels along with
gets for nanomaterials-induced toxicity as they decrease in antioxidants levels in male Wistar
are involved in their clearance from body. rats [76].
330 A. Panghal and S. J. S. Flora

19.4.5 Neurotoxicity 19.4.6 Cardiotoxicity

Nanomedicines get access to the central nervous In recent years, the potential of nanotherapeutics
system (CNS) by surpassing the to exert adverse effects on the cardiovascular sys-
BBB. Additionally, nanotherapeutics may also tem (CVS) has gained attention of research com-
get entry into the brain via trigeminal nerve and munity. Although the accumulating evidences
olfactory nerve post-inhalation [77]. Upon their indicate ability of nanomaterials to cause patho-
entry into the brain, nanomaterials have potential physiological changes in CVS, the precise
to get accumulated in several regions such as molecular mechanisms involved in cardiovascu-
olfactory lobes, striatum and cerebral cortex lar toxicity are not fully elucidated [86].
[78]. Nanomedicines interact with native cells of Translocation of NPs into systemic circulation,
the brain including neurons and glial cells which induction of oxidative/inflammatory response
initiates a cascade of disrupted events eventually and activation of neurogenic pathways are con-
leading to the neurotoxicity [79]. Neurological sidered most fundamental mechanisms involved
disturbances induced by nanomaterials are in nanotherapeutics-induced cardiovascular dys-
highly dependent on their physiochemical prop- functions [87]. TiO2 NPs have been reported to
erties such as size, shape, charge and surface cause arrhythmia, myocardial fibre disarrange-
coating as reflected in several in vivo and in vitro ment, decreased cardiac output, heart fragmenta-
studies [78, 80]. The most common mechanisms tion and hypertrophy of cardiomyocytes in
involved in nanomedicines-induced neurotoxic- several in vitro and in vivo studies [88, 89].
ity include oxidative stress, glial cell activation, Further, Feng et al. reported systolic and sarco-
inflammation, apoptosis and autophagy. Further, meric disorders, interstitial oedema and myocar-
neurotoxic response may be elicited in terms of dial apoptosis in response to acute SiO2 NPs
neuronal structural and physiological changes treatment in rats [90]. Furthermore, exposure of
and neurotransmitters dysbalance [81]. Ag NPs in mice via nasal route induced oxidative
Conventionally used evaluation methods of neu- stress and pulmonary as well as systemic circula-
rotoxicity are not much efficient and reliable to tion. In addition, NPs got access to systemic cir-
fully understand the molecular mechanisms of culation followed by their accumulation in the
nano-neurotoxicity. In an in vivo study, heart subsequently causing lipid peroxidation,
polysorbate-­ encapsulated chitosan NPs have thrombosis and apoptosis of cardiomyocytes
been found to induce neurotoxicity by increasing [90]. Thompson et al. reported the role of adren-
inflammation and oxidative stress which owes to ergic signalling and mitochondrial membrane
their accumulation in the frontal cortex and cer- permeability in CNTs-induced cardiac injury in
ebellum [82]. Liposomal formulations have been mice model [91]. In the view of current scientific
reported to possess intrinsic neurotoxic proper- literature, it can be concluded that high dose and
ties in the form of neuroinflammation and necro- chronic exposure to nano-sized particles seem to
sis as proved by Huo et al. in glioma cells and cause more cardiac damage as compared to acute
rats [83]. Cationic dendrimers exert toxic effects exposure. Extensive research to determine car-
on human neural progenitor cells by altering diovascular toxicity against chronic exposure to
mitochondrial activity, apoptosis, cell differenti- different-sized NPs might be helpful to under-
ation and redox balance; however, these effects stand the pattern of toxicity.
are influenced by surface group charge and den-
sity [84]. Further, the intranasal administration
of NPs leads to their accumulation in the brain 19.4.7 Genotoxicity
which subsequently causes cognitive impair-
ment, plasticity loss, neurotransmitter imbalance In recent years, NPs-induced genotoxicity has
and synaptic changes [85]. gained the interest of researchers. It has been
reported that cationic liposomes, dendrimers and
19 Toxicity Evaluation of Nanomedicine 331

supermagnetic iron oxide NPs cause genotoxicity 19.5.1 Oxidative Stress


even at non-cytotoxic doses which renders scientific
community to ponder on their risk-benefit profile Nanomedicines-associated toxicity is more
[92]. The need of determining the genotoxicity closely related to extensive generation of ROS
potential of nanomedicines has led to the arising of in cellular environment as evident by several
nanogenotoxicology branch which specifically studies conducted in vivo and in vitro. Elevated
deals with the assessment of impact of nanothera- ROS level disrupts redox homeostasis leading to
peutics on DNA. Contrary to cytotoxicity, genotox- oxidative stress which is highly deleterious for
icity of nanodrugs has been studied in limited cellular functioning [97]. Nanodrugs-mediated
research works [93, 94]. Determination of genotox- excessive ROS generation is attributed to the
icity potential of nanocarriers meant for delivery of presence of reactive functional groups on their
non-cytotoxic drugs is of utmost importance to surface which get involved in interaction with
design safer nanocarriers. The physico-chemical cellular targets. Mild oxidative stress increases
properties of nanocarriers such as size, charge, biosynthesis of antioxidant enzymes via upregu-
composition and molecular weight are the key lation of Nrf2 expression. Moderate oxidative
determinants for their genotoxicity profile as stress leads to pro-inflammatory response by
reported by Shah et al. They reported that cationic triggering activation of mitogen-activated pro-
nanocarriers induced more micronucleus formation tein kinase (MAPK) and nuclear factor kappa B
in cells as compared to anionic nanocarriers [92]. (NF-­κB). Further, extreme oxidative stress
Further, Bahadori et al. investigated DNA-damaging causes mitochondrial membrane damage and
potential of lipid-­based and polymeric micelles and electron chain perturbations which in turn lead
reported that later one was more genotoxic as com- to apoptotic cell death [98]. Several NPs have
pared to lipid-based micelles owing to its synthetic been reported to induce biological toxicity by
nature [95]. Cationic micelles and liposomes have elevating ROS levels [31]. The oxidative stress
been found to cause DNA damage as well as alter has been found to play key role in CuO, ZnO,
the gene expressions in the spleen, lungs and liver as Ag2O3, SiO2 and TiO2 NPs-induced biological
reported by Knudsen et al. [96]. toxicity [99–102]. Terada et al. reported that
cationic lipids induced cytotoxicity in mouse
macrophage cell line RAW264 via oxidative
19.5 Insights Into Mechanisms stress and apoptosis; therefore, oxidative stress
of Toxicity may be involved in toxicity of lipid-based
nanodelivery systems such as liposomes and
Understanding of the molecular mechanisms micelles [103]. Further, the complex interplay
involved in nanotherapeutic toxicity is most cru- between oxidative stress and autophagy has
cial for their safe designing to reduce the adverse been found culprit in PAMAM dendrimers-
biological impact. The synthetic strategies need induced neuronal cytotoxicity as proved by Li
to be oriented towards minimizing the side effects et al. [104]. Furthermore, Mukherjee et al. also
considering the mechanisms of toxicity. The reported the role of oxidative stress, glutathione,
detailed mechanisms of cytotoxicity, genotoxic- apoptosis and inflammation in PAMAM den-
ity and immunotoxicity of nanomedicines have drimers-induced cytotoxicity in human kerati-
not been fully elucidated yet. Here, we focus on nocytes cells [105]. Findings of Rasras et al.
some of the most important mechanisms such as suggest that CNTs can cause mitochondrial dys-
oxidative stress, inflammation, apoptosis, necro- functions by generation of ROS as evident by
sis, autophagy and DNA damage which contrib- increased malondialdehyde levels and decreased
ute to nanomedicine toxicity profile and have antioxidants level [106].
been depicted in Fig. 19.2.
332 A. Panghal and S. J. S. Flora

Production of
Nanoparticles ROS
interference
with lysosomes

DNA aberration
Mitochondrial
due to
dysfunction by
nanoparticles
nanoparticles

Cell membrane
damage by Damaged and
nanoparticles oxidized
proteins

Alteration in
cytoplasm
membrane due
to adhesion of
nanoparticles

Fig. 19.2 An illustrative representation of various mechanisms of cellular toxicity

19.5.2 Inflammation NPs have been frequently reported to induce


in vivo and in vitro inflammatory response via
Several studies have demonstrated the induction ROS-mediated NF-κB activation which leads to
of inflammatory response with nanomedicine release of TNF-α, IL-8, IL-2 and IL-6 [102, 112].
exposure. For instance, carbon-based nanothera- Further, TiO2 NPs have been found to cause bron-
peutics (CNTs and fullerenes) trigger inflamma- chial inflammation via p38/ERK MAPK
tory response in various cells and tissues such as pathway-­ mediated release IL-8 [113].
pulmonary airways, human keratinocytes and Interestingly, certain surface properties, steric
macrophages [63, 107]. Recent computational effects and shape of nanocarriers are key deter-
studies suggest that innate immune response minants of immunogenic response of the formu-
which is associated with the release of chemo- lations. For instance, rod-shaped NPs have shown
kines and interleukins is activated with the expo- high potential to induce macrophagic inflamma-
sure of NPs as they are considered as foreign tion due to their high uptake as compared to
invaders by the Toll-like receptors [108]. Further, spherical- and star-shaped NPs [114].
liposomes and other lipid-based nanoformula-
tions have been reported to induce inflammatory
response by activation of complement signalling 19.5.3 Apoptosis
pathways [109]. Recently, Hu et al. proved the
crucial role of ROS in triggering inflammation NPs-induced apoptosis is dependent on physico-­
and apoptosis in keratinocytes with the therapy chemical profile of NPs. For instance, metal NPs
with PEGylated liposomal doxorubicin [110]. such as TiO2, Ag2O3, ZnO and CuO NPs have
Fascinatingly, oxidative stress may also direct the diverse apoptotic potency [115]. Nanotherapeutics
induction of inflammatory response by promot- can induce apoptosis either by mitochondrial
ing the NF-κB- and MAPK-mediated release of pathways or by activating the ER stress signal-
pro-inflammatory cytokines [111]. Metal oxide ling. Excessive ROS generation triggered by
19 Toxicity Evaluation of Nanomedicine 333

nanomedicines activates intrinsic and extrinsic both beneficial and detrimental role; therefore, it
apoptotic pathways in mitochondria [116, 117]. contributes to the therapeutic as well as toxic
Mitochondria perturbations-mediated apoptosis effects of nanodrugs. Multifaceted interaction of
is the most important pathway that contributes to autophagy with other molecular pathways ren-
toxic effects of several metal NPs [118, 119]. Ag ders it difficult to understand the individual
NPs have been reported to induce apoptosis by effects of autophagy in vitro and in vivo [131].
activation of TP53 protein which in turn upregu- Autophagy has been implicated in multiple organ
lates BAX and BAD expression and downregu- toxicity such as neurotoxicity, hepatotoxicity,
lates BCL2 family protein expression causing nephrotoxicity and pulmonary toxicity with the
alterations in mitochondrial membrane permea- exposure to nanomaterials [132–135]. The accu-
bility, release of cytochrome c and cell blebbing mulating evidences suggest that inhibition of
[120]. Moreover, Ag NPs trigger apoptosis via pro-survival mechanisms of autophagy is the
ROS-mediated activation of most probable cause of nanomaterials-induced
JUN/c-JNK-dependent pathways [116]. Recently, cytotoxicity as evident by the disrupted autopha-
Hanna et al. reported role of mitochondria-­ gic flux and accumulation of autophagosomes
mediated apoptosis in ovarian cancer cell cyto- [126]. For instance, acute exposure of rats to
toxicity with exposure to folic acid-coated TiO2 polyalkylsulfonated fullerene caused lysosomal
NPs [121]. Elevated ROS level is involved in dis- accumulation along with blockade of lysosomal
ruption of calcium homeostasis which triggers trafficking [136].
ER stress. Recent scientific evidences suggest
long-term ER stress-mediated misfolded protein
accumulation as an important aspect involved in 19.5.5 Necrosis
nanomaterials-induced apoptosis [122]. ER
stress-mediated apoptosis has been found with Necrosis is one of the most frequently encoun-
the exposure to several NPs such as ZnO NPs tered cell death modalities in response to nano-
exposure to human umbilical vein endothelial material exposure. The necrotic effects of NPs
cells (HUVECs) and PLGA and gold NPs expo- are conflicting because on one side loss of cell
sure to leukaemia cells [123–125]. viability without deep focus on the exact mecha-
nism of cell death is reported, whereas on the
other side apoptosis leading to necrosis has been
19.5.4 Lysosomal Dysfunction reported in plenty of literature [137]. Size, struc-
and Autophagy ture and surface charge of the NPs are the most
crucial properties to determine the cell death
Since NPs are sequestered in lysosomes, lyso- mode with their exposure. For instance, anatase-­
somal dysfunction has been recognised as one of shaped TiO2 NPs induced necrosis, whereas
the outcomes of NP exposure [126]. Alterations rutile-shaped NPs resulted in apoptosis in mouse
in the permeability of lysosomal membrane keratinocytes [138]. Charged gold NPs induced
potential have been associated with the toxicity apoptotic cell death, whereas neutral NPs elicited
of CNTs and cationic nanospheres in human response via necrosis as reported by Schaeublin
fibroblasts and macrophages, respectively [127, et al. [139]. In the preview of current knowledge,
128]. Further, PAMAM dendrimers have been it is evident that the more accurate and precise
reported to cause lysosomal membrane permea- studies are required to fully delineate the role of
bilization leading to mitochondrial dysfunctions necrosis in nanomedicines-induced cytotoxicity
as evident by altered mitochondrial membrane [140, 141]. Till now, pro-oxidants-mediated
potential and apoptosis [129]. ZnO NPs-induced ­oxidative stress is considered as the main reason
lung injury in rat has also been credited to lyso- for the nanomedicines-induced necrosis.
somal dysfunctions and uptake by macrophages Excessive ROS reduces mitochondrial membrane
[130]. Autophagy induced by nanomedicines has potential and ultimately leads to the necrotic cell
334 A. Panghal and S. J. S. Flora

death [142]. Cationic nanocarriers have been rial is still in its infancy. Additionally, nanomedi-
reported to induce necrosis via impairment of cines have been reported to induce epigenetic
Na+/K+ ATPase pump which subsequently changes in genetic material such as DNA meth-
resulted in inflammatory response [143]. Further, ylation and histone modifications; however, the
the release of von Willebrand factor and release knowledge of their mechanisms is still lacking.
of pro-­ inflammatory cytokines are the most The in vitro and in vivo evidences suggest that
potential mechanisms of endothelial cell necrosis NPs can induce the following epigenetic changes:
in response to nanodelivery systems [142]. (i) alterations in DNA methylation in non-­specific
Another most potential mechanism involved in and gene-specific manner; (ii) increased phos-
nanomedicines-­ induced necrosis is the rapid phorylation of histone proteins; (iii) increased
lysosomal degradation of NPs followed by desta- acetylation of H3 and H4 histones; and (iv) ele-
bilization of lysosomes and the cytosolic release vated HDAC2 protein levels and decreased levels
of toxic mediators [144]. of HDAC1 and HDAC6 proteins [149–151]. The
lack of deep information about the impact of
nanomedicines on epigenome is also a limiting
19.5.6 DNA Damage factor to fully elucidate the genotoxicity potential
of nanotherapeutics.
NPs have been recognised as genotoxic agents
since long back. They have been reported to
induce DNA damage in terms of chromosomal 19.6 Evaluation Techniques
fragmentation, DNA strand breakages and the of Nanomedicine Toxicity
gene mutations. Nanogenotoxicology is a field
which is specifically dedicated to the study of The propensity of nanomedicines to induce
genotoxic potential of nanomaterials including several toxic effects paves the way for the need to
nanodelivery systems and nanomedicines [93]. evaluate their toxicity potential. The combination
Nanomedicines might be inducing genotoxicity of various conventional in vitro and in vivo tech-
by acting at two fronts either by directly inducing niques along with the new emerging techniques
DNA damage or by inhibiting the DNA repair such as sensors-based tests is employed to hunt
mechanisms [145]. For instance, Demir et al. for the nanomedicines-induced specific toxicity
reported alterations in DNA repair system in biomarkers as well as to determine the toxicity
human lymphoblastoid cell line with ZnO NPs profile of new nanotherapeutics. The assay meth-
exposure [146]. Further, impairment of DNA ods, advantages and disadvantages of several
repair mechanisms was observed by Kruszewski nanotoxicity detection techniques are summa-
et al. in human cell lines exposed to Ag NPs rized in Table 19.2.
[147]. The previous literature has suggested that
metal NPs induce DNA fragmentation and for-
mation of oxidation-induced DNA adducts [148]. 19.6.1 In Vitro Approaches
Nanotherapeutics can induce genotoxicity either for Toxicity Evaluation
by direct mechanisms or by indirect mechanisms.
The direct mechanisms involve direct interaction In vitro toxicity assessment is an effective method
of nanomedicines with the genetic material post to understand the toxic mechanisms of
their diffusion through nuclear membrane, nanotherapeutics-­induced cytotoxicity; however,
whereas indirect mechanisms are triggered with these methods are not complete substitutes for
inflammatory response involving macrophages in vivo methods of toxicity evaluation. The most
and interleukins [145]. Although the mechanisms common assays carried out to determine toxicity
of nanomedicines-induced genotoxicity are potential of nanomedicines include alamar blue
known to an extent, the knowledge regarding assay, 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl
interaction of nanomedicines with genetic mate- tetrazolium bromide (MTT) assay, lactate dehy-
19 Toxicity Evaluation of Nanomedicine 335

Table 19.2 Advantages and disadvantages of several approaches which are usually carried out for nanotoxicity evalu-
ation [152, 157–159]
Assays/techniques Methods Advantages Disadvantages
In vitro
Cell viability MTT, MTS, XTT, alamar blue Fast, sensitive NPs may interfere with
assay signal transduction
Cell death Trypan blue assay, LDH assay, Differs between different Cross-reaction, non-­
(apoptosis, necrosis caspase 3/7 activation, FACS, modes of cell death specificity, non-definitive
and autophagy) ELISA, Annexin-V assay, comet for individual protein
assay, TUNEL assay, expression and activity
immunoblotting,
immunofluorescence
Oxidative stress ROS assay, MDA assay, oxidants/ Fast and reliable for Non-specific, properties of
antioxidants ratio studying basic mechanisms NPs may interfere with the
of toxicity results
Genotoxicity Comet assay, micronuclei presence, Rapid, facile and reliable to Need to be performed
TUNEL assay identify genotoxic agents under stringent conditions
to prevent exogenous DNA
damage
In vivo
Pharmacokinetic Biodistribution and clearance Characterize NPs in terms Not sensitive enough to
assays of uptake, localization and detect extremely low levels
biodistribution of drugs, rigorous
compartmental modelling
Histopathology H & E staining, PAS staining, PSR Provides cell-specific Rigorous procedure,
staining, Masson trichrome staining toxicity effects of NPs on false-positive and
an organ false-negative outcomes
are encountered
Sensors
Chemical sensors Ion-selective electrodes, optodes, Detection limit is low (in Non-ion specificity
fluorescent sensors, anodic ppb)
stripping voltammetry Rapid and selective
discrimination among
metal NPs
Biosensors FRET, electrical impedance sensing Monitor cellular dynamics High cost, less specificity
at single cell level, and resolution,
real-time monitoring in immunogenic
heterogenous cell
population
Nanochemical CNTs-coated interdigitated Highly sensitive and Labelling process can
sensors electrode, NPs-coated ion selective, monitor drug modify surface and
electrodes and fluorescent sensors translocation in real functions of NPs, difficulty
manner, able to interact in finding the adequate
with target via multiple labelling techniques
interaction sites
Nanobiosensors NPs-coated FRET and electrical Portable and highly Immunogenic, sometimes
impedance sensing sensitive, able to interact biomolecules lose their
with target via multiple properties due to labelling
interaction sites
Omics
Genomics/ miRNA profiling Better platform to study Lack of correlation to
miRNomics genetic changes, rapid in vivo findings
screening
(continued)
drogenase (LDH) leakage assay, apoptotic and Determination of NPs’ potential to breach and
necrotic test, endotoxin signalling and oxidative interact with cellular barriers is an important
stress assays [152]. aspect to evaluate their toxicity. Assays used to
336 A. Panghal and S. J. S. Flora

Table 19.2 (continued)


Assays/techniques Methods Advantages Disadvantages
Transcriptomics DNA microarray, NGS, subtraction Only one type of Lack of direct correlation
hybridization biomolecules has to be between changes in mRNA
extracted and analysed expression and phenotypic
changes
Proteomics Shotgun method, LC-MS, Determine protein Different types of
MALDI-TOF, protein microarray, expression and characterize protocols have to follow
gel electrophoresis, NMR and expressed proteins for different types of
X-ray crystallography proteins, and samples are
prone to contamination
Metabolomics NMR, LC-MS, GC-MS, HRMS In-expensive analysis, Organism non-specific
non-invasive sampling metabolites, different
detection techniques
required for different
metabolites
Computational
QSAR Hansch analysis model, 3D-QSAR, Rapid, in-expensive, less Large dataset required,
nano-QSAR (1D, 2D and 3D) resources required non-specific due to
unknown exact structure of
NPs
Molecular docking Conformational search algorithm Rapid, in-expensive, less Lack of quality database,
such as GA, MC, IC resources required, standardization and
provides insight into the scoring functions, less
drug-biomolecule correlation to in vivo
results due to target
flexibility
Abbreviations: MTT 3-[4,5-dimethylthiazole-2-yl]-2,5-diphenyltetrazolium bromide, MTS (3-(4,5-dimethylthiazol-­2-
yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium), XTT 2,3-bis (2-methoxy-4-nitro-5-­sulfophenyl)-
5-[(phenylamino) carbonyl] -2H-tetrazolium hydroxide, LDH lactate dehydrogenase, FACS fluorescence-activated cell
sorting, ELISA enzyme-linked immunosorbent assay, TUNEL terminal deoxynucleotidyl transferase-mediated dUTP
nick-end labelling, MDA malondialdehyde, H & E haematoxylin and eosin, PAS periodic acid-Schiff, PSR picrosirius
red, FRET fluorescence resonance energy transfer, miRNA microRNA, NGS next-­generation sequencing, LC-MS liquid
chromatography-mass spectrometry, MALDI-TOF matrix-assisted laser desorption/ionization-­time of flight, NMR
nuclear magnetic resonance, GC-MS gas chromatography-mass spectrometry, HRMS high-resolution mass spectrome-
try, QSAR quantitative structure activity relationship, GA genetic algorithm, MC Monte Carlo, IC incremental
construction

carry out for determining cellular infiltration are Although analysis of caspases by immunoflu-
required for quantitative analysis of particles orescence, immunoblotting and affinity tests with
within cells and qualitative identification of nano- attached reported moieties offers new approaches
material’s destination. Nanotherapeutics tagged for determination of apoptotic activities of nano-
with fluorescent tags such as fluorescein isothio- therapeutics, however none of these caspase-­
cyanate (FITC), Alexa dyes with fluorescence-­ based assays are suitable for estimating activities
activated cell sorting (FACS), confocal laser of individual caspases. The ability of caspases to
scanning microscopy (CLSM), and imaging flow cleave multiple substrates is the main limitation
cytometry (IFC) serves the need for quantitative for establishing apoptotic potential [153].
and qualitative determination of nanomaterials Further, assays such as Annexin-V assay, comet
inside cellular system [152]. Most of the conven- assay, TdT-mediated dUTP-biotin nick end label-
tional cytotoxicity assays involve chemical ling (TUNEL) assay, and DNA laddering assay
reagents for the evaluation of cellular metabolic efficiently serve for the purpose of identification
disruption. These chemical reagents and cell cul- of apoptotic cell death [15].
ture media interact with the NPs under test and Additionally, quantification of oxidative stress
therefore can influence the test results and give and inflammatory biomarkers such as ROS, lipid
false-positive or false-negative outcomes. peroxidation, antioxidant levels and inflamma-
19 Toxicity Evaluation of Nanomedicine 337

tory cytokines reveals the in vitro cytotoxicity-­ of nanotherapeutics is highly dependent on the
inducing potential of nanomedicines. Quantitative exposure conditions and dose and duration of
luciferase assays along with inflammatory cyto- treatment. The toxicity potential of nanomedi-
kine estimation and ROS determining assays are cines curtails their use in single and low dosages,
a wholesome approach for recognizing the and therefore, chronic application of nanomedi-
in vitro cytotoxicity of nanomedicines [154]. cines is still a dream to come true. Fonseca-­
Endotoxin-based detection approaches exploit Gomes and coworkers studied the biodistribution
the use of Limulus amoebocyte lysate (LAL) and toxicity potential of polymeric and lipid-­
assay which constitutes the gel clot assay, the based NPs in chronic disease in vivo model and
coagulogen-based (turbidity) assay and the chro- interestingly reported that repeated dose expo-
mogenic assay [155]. Remarkably, the arising sure of NPs was not associated with side effects
literature suggests that LAL assay-based [165]. The effects of chronic exposure to
approaches are not accurate for the toxicity PEGylated copper indium sulphide/zinc sulphide
assessment of NPs. Advanced approaches take quantum dots were evaluated in BALB/c mice to
consideration of toll-like receptor (TLR) 4 as a determine their in vivo biocompatibility [166].
reporter protein which has come to way as HEK-­ The in vivo approach of toxicity evaluation has
Blue™ commercial kits [156]. an edge over the in vitro method in terms of
reproducibility and clinical relevance. Since the
physiology of animal models is closer to humans,
19.6.2 In Vivo Approaches the data obtained by in vivo assessment
for Toxicity Evaluation approaches is more realistic and relevant as com-
pared to in vitro methods [167]. Another major
Accomplishing preclinical studies to evaluate the advantage of in vivo methods is that they provide
in vivo toxicity is a pre-requisite for any new wholesome picture of toxic effects of nanomedi-
chemical entity and nanomedicine to make their cines on an organ which can be observed by his-
way to clinical trials [160]. In vivo studies endow topathological evaluations [168].
with the information about the lethal dose (LD50),
maximum tolerated dose (MTD), no observed
adverse effect level (NOAEL) and acute and 19.6.3 Sensor-Based Techniques
chronic toxicity profile of the drug candidate and
should be carried out in rodents as well as non-­ Sensor (chemical sensors, biosensors and
rodents [161, 162]. Taking consideration of cur- nanosensors)-based approaches of toxicity
rent literature, invertebrates such as assessment are a hot topic in the field of monitor-
Caenorhabditis elegans and Drosophila melano- ing and risk assessment of nanomedicines.
gaster, zebrafish, rodents and non-human pri- Sensor-based techniques are able enough to iden-
mates have been employed for the toxicity tify the selective interaction between the sensor
assessment of nanotherapeutics [163]. In vivo entity and the cellular markers and thus eliminate
toxicity assessment-based approaches stress the limitations of the currently followed endpoint
upon the determination of pharmacokinetic as evaluation methods.
well as pharmacodynamic profile of the test Numerous chemical sensors such as ion-­
chemical. A thorough and quantitative pharmaco- selective electrodes and fluorescence sensors
kinetic investigation of the nanostructure, i.e. have been used for detecting Ag ions and thus can
absorption, metabolism, distribution and excre- be explored for assessment of nanotoxicity of
tion, can lead to advances in the nanostructure’s Ag-based nanomedicines [169]. Although
design for therapeutic and diagnostic applica- researchers used atomic absorption spectroscopy
tions. For instance, Baati et al. explored the phar- (AAS), plasma emission spectroscopy and anodic
macokinetic, biodistribution and toxicity profile stripping voltametric techniques for Ag ion detec-
of titanate nanotubes in mice [164]. The toxicity tion, these techniques rarely have been imple-
338 A. Panghal and S. J. S. Flora

mented for evaluation of Ag NPs toxicity [170]. at nanomolar concentrations [177]. Nanosensors
Chatterjee et al. used rhodamine-based fluoro- can be made by coupling nanostructured ligands
genic and chromogenic probes for detection of either with a chemical moiety (nanochemical
Ag ions and NPs in aqueous media [171]. The sensors) or with a biological component (nano-
implementation of chemical sensors for metallic biosensors). Nanobiosensors are more developed
nanomedicines risk assessment is restricted due in comparison to nanochemical sensors in terms
to complicated coordination properties and of their use for monitoring the nanotherapeutics.
poorly defined stereo-chemical profile. Nanochemical sensors have nanostructure which
Biosensors are devices which are deemed to is radio-labelled or tagged with fluorescence
detect and record the physiological changes. moieties which aid in live tracking of the test
They are usually made by coupling bioentities compound in the biological system. In the sce-
(proteins, antibodies, microorganisms, DNA and nario where NPs labelling is difficult, tracking
cells) with the signal transducers such as elec- the NPs by electron microscopy is a promising
trodes, optical transducers and semiconductors approach to monitor the electron-rich nanomedi-
[172]. These devices have profound several cines in the diverse organs.
applications including the field of nanomedicine.
Cell-based biosensors sense live cells’ physio-
logical changes induced by internal or external 19.6.4 Omics Techniques
stimuli [173]. EIS biosensors are able to provide
rapid and real information about the cytotoxicity Omics technologies such as genomics, pro-
with response to nanomaterial exposure. Further, teomics, transcriptomics and metabolomics are
chip-based biosensors are also a promising tool emerging techniques which have promising abil-
for the evaluation of nanomedicines’ toxicity. ity to evaluate the in vitro and in vivo toxicity of
Chip-based approach has been tested by Kim nanomedicines. Omics techniques are superior
et al. where they used lithographic chip coupled over other conventional techniques in terms of
with gold-sensing electrode to evaluate the size-­ less interference with the NPs properties due to
dependent toxicity of SiO2 NPs [174]. Further, which the probability of false-positive and false-­
neural cell chip fabricated on the conductive sur- negative results is low. Notably, omics technol-
faces modified with nanostructured ligands also ogy provides an insight in the molecular
serves as the purpose for risk assessment of nano- interaction and cellular/tissue/organ responses to
materials [175]. Furthermore, FRET technology the nanotherapeutics exposure [178]. Numerous
has ability to monitor the stimuli-triggered haz- pathways such as oxidative stress, inflammation,
ards at cellular level in real-time manner and apoptosis and proliferation have been found to be
therefore might be a promising technique for involved in NPs toxicity by using transcriptomics
assessment of nanomedicines’ toxicity [176]. and proteomics. Risk assessment of NPs by using
The major advantage of nanosensors is that transcriptomics and proteomics is higher as com-
they are highly sensitive due to their small size pared to metabolomics and genomic. Further,
and high surface to volume ratio which enables implementation of omics technology is more
them to interact with the target at multiple sites. prominent in in vitro assessment of NPs as
The ability of nanosensors to monitor cellular ­compared to in vivo assessment. Additionally,
changes induced by nanomaterials has led to Ag, SiO2 and ZnO NPs have been mainly assessed
development of “nano assessing nano” approach by transcriptomics, whereas effects of Au and
for the assessment of nanotherapeutics-induced CNTs have been studied by proteomics as
toxicity [177]. Geng et al. employed Ag NPs-­ reviewed by Frohlich et al. [158]. Transcriptomics
based nanosensors coupled with the green fluo- has been successfully implicated for the evalua-
rescent protein to perceive the early cellular tion of nanotoxicity of CuO, TiO2, ZnO and SiO2
changes induced by low exposure to Ag NPs and NPs and cadmium quantum dots [179–181].
reported the morphological changes in cells even Further, genomics approach has been exploited
19 Toxicity Evaluation of Nanomedicine 339

for the risk assessment of CNTs in invertebrates toxicity and side effects. Int J Nanomedicine.
2017;12:6107–29.
[182, 183]. The evolution in the technological 6. Meng H, Leong W, Leong KW, Chen C, Zhao
field has influenced the implication of proteomics Y. Walking the line: the fate of nanomaterials at bio-
in nanotoxicity evaluation. Ability of proteomics logical barriers. Biomaterials. 2018;174:41–53.
to evaluate composition, charge and size of pro- 7. Hamilton RF, Wu N, Xiang C, et al. Synthesis,
characterization, and bioactivity of carboxylic acid-­
teins renders it more applicable for evaluation of functionalized titanium dioxide nanobelts. Part Fibre
toxicity of NPs of varying sizes [184, 185]. Toxicol. 2014;11:43.
Metabolic profiling by mass spectroscopy is 8. Jeevanandam J, Barhoum A, Chan YS, Dufresne
another efficient approach for the nanotoxicity A, Danquah MK. Review on nanoparticles and
nanostructured materials: history, sources, tox-
evaluation and provides evidences of alterations icity and regulations. Beilstein J Nanotechnol.
in the cellular signalling with the exposure to 2018;9:1050–74.
nanomaterials, as in the case of SiO2 NPs [179, 9. Ai J, Biazar E, Jafarpour M, et al. Nanotoxicology
186]. and nanoparticle safety in biomedical designs. Int J
Nanomedicine. 2011;6:1117–27.
10. Attarilar S, Yang J, Ebrahimi M, et al. The toxicity
phenomenon and the related occurrence in metal and
19.7 Conclusion metal oxide nanoparticles: a brief review from the
biomedical perspective. Front Bioeng Biotechnol.
2020;8:822.
Nanomedicines differ from conventional medi- 11. Garcés M, Cáceres L, Chiappetta D, Magnani N,
cines in their physiochemical properties such Evelson P. Current understanding of nanoparticle
as size, and surface to volume ratio. toxicity mechanisms and interactions with biologi-
Nanomedicines contribute to drug targeting as cal systems. New J Chem. 2021;45:14328–44.
12. Saiyed M, Patel R, Patel S. Toxicology perspec-
well as efficient drug delivery; and this tive of nanopharmaceuticals: a critical review. Int J
affords them some edge over conventional medi- Pharm Sci Nanotechnol. 2011;4:1287–95.
cines. However, these properties also have been 13. Bahadar H, Maqbool F, Niaz K, Abdollahi
associated with potential toxicity of nanomedi- M. Toxicity of nanoparticles and an overview
of current experimental models. Iran Biomed J.
cines. Early recognition and in-depth understand- 2016;20:1–11.
ing of the molecular mechanisms involved in 14. Bawa R, Barenholz Y, Owen A. The challenge
nanomedicines-induced toxicity are critical for of regulating nanomedicine: key issues. In:
safe use of nanomedicines in public health. Nanomedicines: design, delivery and detection, RSC
drug discovery series. Cambridge: Royal Society of
Several in vitro, in vivo, sensors- and omics-­ Chemistry; 2016. p. 290–314.
based approaches are currently being used for the 15. Kumar V, Sharma N, Maitra SS. In vitro and in vivo
risk assessment of nanomedicines. toxicity assessment of nanoparticles. Int Nano Lett.
2017;7:243–56.
16. Rasmussen K, Rauscher H, Kearns P, González M,
Riego Sintes J. Developing OECD test guidelines for
References regulatory testing of nanomaterials to ensure mutual
acceptance of test data. Regul Toxicol Pharmacol.
1. Farjadian F, Ghasemi A, Gohari O, Roointan A, 2019;104:74–83.
Karimi M, Hamblin MR. Nanopharmaceuticals 17. Tirumala MG, Anchi P, Raja S, Rachamalla M,
and nanomedicines currently on the market: chal- Godugu C. Novel methods and approaches for safety
lenges and opportunities. Nanomedicine (Lond). evaluation of nanoparticle formulations: a focus
2019;14:93–126. towards in vitro models and adverse outcome path-
2. Ramos AP, Cruz MAE, Tovani CB, Ciancaglini ways. Front Pharmacol. 2021;12:612659.
P. Biomedical applications of nanotechnology. 18. Ventola CL. Progress in nanomedicine: approved
Biophys Rev. 2017;9:79–89. and investigational nanodrugs. P T. 2017;42:742–55.
3. Lu W, Yao J, Zhu X, Qi Y. Nanomedicines: rede- 19. Anselmo AC, Mitragotri S. Nanoparticles in the
fining traditional medicine. Biomed Pharmacother. clinic: an update. Bioeng Transl Med. 2019;4:e10143.
2021;134:111103. 20. Adams D, Gonzalez-Duarte A, O’Riordan WD,
4. Bawa R. Nanopharmaceuticals: nanopharmaceuti- et al. Patisiran, an RNAi therapeutic, for heredi-
cals. Eur J Nanomed. 2010;3:34–40. tary transthyretin amyloidosis. N Engl J Med.
5. Brand W, Noorlander CW, Giannakou C, et al. 2018;379:11–21.
Nanomedicinal products: a survey on specific
340 A. Panghal and S. J. S. Flora

21. Nederberg F, Zhang Y, Tan JP, et al. Biodegradable nium dioxide in male rats. Gastroenterol Hepatol
nanostructures with selective lysis of microbial Bed Bench. 2018;11:159–68.
membranes. Nat Chem. 2011;3:409–14. 39. Bartneck M, Ritz T, Keul HA, et al. Peptide-­
22. Li S, Jiang Q, Liu S, et al. A DNA nanorobot func- functionalized gold nanorods increase liver injury in
tions as a cancer therapeutic in response to a molecu- hepatitis. ACS Nano. 2012;6:8767–77.
lar trigger in vivo. Nat Biotechnol. 2018;36:258–64. 40. Li Y, Zeng X, Wang S, et al. Inhibition of autophagy
23. Germain M, Caputo F, Metcalfe S, et al. Delivering protects against PAMAM dendrimers-induced hepa-
the power of nanomedicine to patients today. J totoxicity. Nanotoxicology. 2015;9:344–55.
Control Release. 2020;326:164–71. 41. Souza MR, Mazaro-Costa R, Rocha TL. Can nano-
24. Mitchell MJ, Billingsley MM, Haley RM, Wechsler materials induce reproductive toxicity in male mam-
ME, Peppas NA, Langer R. Engineering preci- mals? A historical and critical review. Sci Total
sion nanoparticles for drug delivery. Nat Rev Drug Environ. 2021;769:144354.
Discov. 2021;20:101–24. 42. Falchi L, Khalil WA, Hassan M, Marei
25. Bulbake U, Doppalapudi S, Kommineni N, Khan WFA. Perspectives of nanotechnology in male
W. Liposomal formulations in clinical use: an fertility and sperm function. Int J Vet Sci Med.
updated review. Pharmaceutics. 2017;9:12. 2018;6:265–9.
26. Ventola CL. The nanomedicine revolution: part 1: 43. Han JW, Jeong JK, Gurunathan S, et al. Male- and
emerging concepts. P T. 2012;37:512–25. female-derived somatic and germ cell-specific toxic-
27. Zhang Y, Yang W-X. Tight junction between endo- ity of silver nanoparticles in mouse. Nanotoxicology.
thelial cells: the interaction between nanopar- 2016;10:361–73.
ticles and blood vessels. Beilstein J Nanotechnol. 44. Hong F, Si W, Zhao X, et al. TiO2 nanoparticle
2016;7:675–84. exposure decreases spermatogenesis via biochemi-
28. Naqvi S, Panghal A, Flora S. Nanotechnology: a cal dysfunctions in the testis of male mice. J Agric
promising approach for delivery of neuroprotective Food Chem. 2015;63:7084–92.
drugs. Front Neurosci. 2020;14:494. 45. Sun J, Zhang Q, Wang Z, Yan B. Effects of nanotox-
29. Szebeni J, Simberg D, González-Fernández Á, icity on female reproductivity and fetal development
Barenholz Y, Dobrovolskaia MA. Roadmap and in animal models. Int J Mol Sci. 2013;14:9319–37.
strategy for overcoming infusion reactions to nano- 46. Habas K, Demir E, Guo C, Brinkworth MH,
medicines. Nat Nanotechnol. 2018;13:1100–8. Anderson D. Toxicity mechanisms of nanoparticles
30. Hoshyar N, Gray S, Han H, Bao G. The effect of in the male reproductive system. Drug Metab Rev.
nanoparticle size on in vivo pharmacokinetics 2021;53:604–17.
and cellular interaction. Nanomedicine (Lond). 47. Gao G, Ze Y, Li B, et al. Ovarian dysfunction and
2016;11:673–92. gene-expressed characteristics of female mice
31. Fu PP, Xia Q, Hwang HM, Ray PC, Yu H. Mechanisms caused by long-term exposure to titanium dioxide
of nanotoxicity: generation of reactive oxygen spe- nanoparticles. J Hazard Mater. 2012;243:19–27.
cies. J Food Drug Anal. 2014;22:64–75. 48. Tang Y, Chen B, Hong W, et al. ZnO nanoparticles
32. Böttcher JP, Knolle PA, Stabenow D. Mechanisms induced male reproductive toxicity based on the
balancing tolerance and immunity in the liver. Dig effects on the endoplasmic reticulum stress signal-
Dis. 2011;29:384–90. ing pathway. Int J Nanomedicine. 2019;14:9563–76.
33. Tavares AJ, Poon W, Zhang YN, et al. Effect of 49. Garcia TX, Costa GM, França LR, Hofmann
removing Kupffer cells on nanoparticle tumor deliv- MC. Sub-acute intravenous administration of sil-
ery. Proc Natl Acad Sci U S A. 2017;114:E10871–80. ver nanoparticles in male mice alters Leydig cell
34. Modrzynska J, Berthing T, Ravn-Haren G, et al. function and testosterone levels. Reprod Toxicol.
Primary genotoxicity in the liver following pulmo- 2014;45:59–70.
nary exposure to carbon black nanoparticles in mice. 50. Mathias FT, Romano RM, Kizys MM, et al. Daily
Part Fibre Toxicol. 2018;15:2. exposure to silver nanoparticles during prepubertal
35. Liu F, Chang X, Tian M, et al. Nano nio induced liver development decreases adult sperm and reproductive
toxicity via activating the Nf-Κb signaling pathway parameters. Nanotoxicology. 2015;9:64–70.
in rats. Toxicol Res (Camb). 2017;6:242–50. 51. Wang Z, Wang Z. Nanoparticles induced embryo-­
36. Sha B, Gao W, Wang S, et al. Oxidative stress fetal toxicity. Toxicol Ind Health. 2020;36:181–213.
increased hepatotoxicity induced by nano-titanium 52. Buerki-Thurnherr T, Schaepper K, Aengenheister L,
dioxide in Brl-3a cells and Sprague-Dawley rats. J Wick P. Developmental toxicity of nanomaterials:
Appl Toxicol. 2014;34:345–56. need for a better understanding of indirect effects.
37. Recordati C, De Maglie M, Bianchessi S, et al. Chem Res Toxicol. 2018;31:641–2.
Tissue distribution and acute toxicity of silver after 53. Dugershaw BB, Aengenheister L, Hansen SSK,
single intravenous administration in mice: nano-­ Hougaard KS, Buerki-Thurnherr T. Recent insights
specific and size-dependent effects. Part Fibre on indirect mechanisms in developmental toxicity of
Toxicol. 2016;13:12. nanomaterials. Part Fibre Toxicol. 2020;17:31.
38. Suker DK, Jasim FA. Liver histopathological altera- 54. Pietroiusti A, Vecchione L, Malvindi MA, et al.
tion after repeated intra-tracheal instillation of tita- Relevance to investigate different stages of preg-
19 Toxicity Evaluation of Nanomedicine 341

nancy to highlight toxic effects of nanoparticles: 68. Zhao H, Li L, Zhan H, Chu Y, Sun B. Mechanistic
the example of silica. Toxicol Appl Pharmacol. understanding of the engineered nanomaterial-­
2018;342:60–8. induced toxicity on kidney. J Nanomater.
55. Teng C, Jia J, Wang Z, Sharma VK, Yan B. Size-­ 2019;2019:2954853.
dependent maternal-fetal transfer and fetal devel- 69. Reddy AR, Reddy YN, Krishna DR, Himabindu
opmental toxicity of ZnO nanoparticles after oral V. Multi wall carbon nanotubes induce oxidative
exposures in pregnant mice. Ecotoxicol Environ Saf. stress and cytotoxicity in human embryonic kidney
2019;182:109439. (HEK293) cells. Toxicology. 2010;272:11–6.
56. Vidanapathirana AK, Thompson LC, Odom J, et al. 70. Blazer-Yost BL, Banga A, Amos A, et al. Effect
Vascular tissue contractility changes following late of carbon nanoparticles on renal epithelial cell
gestational exposure to multi-walled carbon nano- structure, barrier function, and protein expression.
tubes or their dispersing vehicle in Sprague Dawley Nanotoxicology. 2011;5:354–71.
rats. J Nanomed Nanotechnol. 2014;5:201. 71. Tiwari R, Singh RD, Khan H, et al. Oral subchronic
57. Stapleton PA, McBride CR, Yi J, Abukabda AB, exposure to silver nanoparticles causes renal dam-
Nurkiewicz TR. Estrous cycle-dependent modula- age through apoptotic impairment and necrotic cell
tion of in vivo microvascular dysfunction after nano- death. Nanotoxicology. 2017;11:671–86.
material inhalation. Reprod Toxicol. 2018;78:20–8. 72. Xu J, Li Z, Xu P, Xiao L, Yang Z. Nanosized copper
58. Blum JL, Xiong JQ, Hoffman C, Zelikoff oxide induces apoptosis through oxidative stress in
JT. Cadmium associated with inhaled cadmium podocytes. Arch Toxicol. 2013;87:1067–73.
oxide nanoparticles impacts fetal and neonatal devel- 73. Lin YF, Chiu IJ, Cheng FY, et al. The role
opment and growth. Toxicol Sci. 2012;126:478–86. of hypoxia-inducible factor-1α in zinc oxide
59. Hougaard KS, Campagnolo L, Chavatte-Palmer nanoparticle-­induced nephrotoxicity in vitro and
P, et al. A perspective on the developmental tox- in vivo. Part Fibre Toxicol. 2016;13:52.
icity of inhaled nanoparticles. Reprod Toxicol. 74. Walsh TJ, Goodman JL, Pappas P, et al. Safety, toler-
2015;56:118–40. ance, and pharmacokinetics of high-dose liposomal
60. Gurumurthy R. Assessing nanoparticle risks to amphotericin B (AmBisome) in patients infected
human health. Elsevier; 2016. with Aspergillus species and other filamentous
61. Lam CW, James JT, McCluskey R, Hunter fungi: maximum tolerated dose study. Antimicrob
RL. Pulmonary toxicity of single-wall carbon nano- Agents Chemother. 2001;45:3487–96.
tubes in mice 7 and 90 days after intratracheal instil- 75. Balas M, Popescu Din IM, Hermenean A, Cinteza
lation. Toxicol Sci. 2004;77:126–34. LO, Dinischiotu A. Exposure to iron oxide nanopar-
62. Rice KM, Nalabotu SK, Manne ND, et al. Exposure ticles coated with phospholipid-based polymeric
to cerium oxide nanoparticles is associated with acti- micelles induces renal transitory biochemical
vation of mitogen-activated protein kinases signal- and histopathological changes in mice. Materials
ing and apoptosis in rat lungs. J Prev Med Public (Basel). 2021;14:2605.
Health. 2015;48:132–41. 76. Yousef MI, Mutar TF, Kamel MAE. Hepato-renal
63. Dong J. Signaling pathways implicated in car- toxicity of oral sub-chronic exposure to aluminum
bon nanotube-induced lung inflammation. Front oxide and/or zinc oxide nanoparticles in rats. Toxicol
Immunol. 2020;11:552613. Rep. 2019;6:336–46.
64. Dokka S, Toledo D, Shi X, Castranova V, 77. Kreyling WG. Discovery of unique and ENM- spe-
Rojanasakul Y. Oxygen radical-mediated pulmonary cific pathophysiologic pathways: comparison of
toxicity induced by some cationic liposomes. Pharm the translocation of inhaled iridium nanoparticles
Res. 2000;17:521–5. from nasal epithelium versus alveolar epithelium
65. Li C, Liu H, Sun Y, et al. Pamam nanoparticles pro- towards the brain of rats. Toxicol Appl Pharmacol.
mote acute lung injury by inducing autophagic cell 2016;299:41–6.
death through the Akt-Tsc2-Mtor signaling pathway. 78. Zhang L, Bai R, Li B, et al. Rutile TiO2 particles
J Mol Cell Biol. 2009;1:37–45. exert size and surface coating dependent r­etention
66. Hozayen WG, Mahmoud AM, Desouky EM, and lesions on the murine brain. Toxicol Lett.
El-Nahass ES, Soliman HA, Farghali AA. Cardiac 2011;207:73–81.
and pulmonary toxicity of mesoporous silica 79. Wang Y, Xiong L, Tang M. Toxicity of inhaled
nanoparticles is associated with excessive ROS pro- particulate matter on the central nervous system:
duction and redox imbalance in Wistar rats. Biomed neuroinflammation, neuropsychological effects
Pharmacother. 2019;109:2527–38. and neurodegenerative disease. J Appl Toxicol.
67. Johnston HJ, Hutchison G, Christensen FM, Peters 2017;37:644–67.
S, Hankin S, Stone V. A review of the in vivo and 80. Knudsen KB, Northeved H, Ek PK, et al.
in vitro toxicity of silver and gold particulates: par- Differential toxicological response to positively and
ticle attributes and biological mechanisms respon- negatively charged nanoparticles in the rat brain.
sible for the observed toxicity. Crit Rev Toxicol. Nanotoxicology. 2014;8:764–74.
2010;40:328–46. 81. Li J, Martin F. Current perspective on nanomaterial-­
induced adverse effects: neurotoxicity as a case
342 A. Panghal and S. J. S. Flora

example. In: Neurotoxicity of nanomaterials and 95. Bahadori F, Kocyigit A, Onyuksel H, Dag A, Topcu
nanomedicine. Elsevier; 2017. p. 75–98. G. Cytotoxic, apoptotic and genotoxic effects of
82. Yuan ZY, Hu YL, Gao JQ. Brain localization and lipid-based and polymeric nano micelles, an in vitro
neurotoxicity evaluation of polysorbate 80-­modified evaluation. Toxics. 2018;6:7.
chitosan nanoparticles in rats. PLoS One. 96. Knudsen KB, Northeved H, Kumar PE, et al. In
2015;10:e0134722. vivo toxicity of cationic micelles and liposomes.
83. Huo T, Barth RF, Yang W, et al. Preparation, biodis- Nanomedicine. 2015;11:467–77.
tribution and neurotoxicity of liposomal cisplatin fol- 97. Yu Z, Li Q, Wang J, et al. Reactive oxygen species-­
lowing convection enhanced delivery in normal and related nanoparticle toxicity in the biomedical field.
F98 glioma bearing rats. PLoS One. 2012;7:e48752. Nanoscale Res Lett. 2020;15:115.
84. Zeng Y, Kurokawa Y, Win-Shwe TT, et al. Effects of 98. Manke A, Wang L, Rojanasakul Y. Mechanisms of
PAMAM dendrimers with various surface functional nanoparticle-induced oxidative stress and toxicity.
groups and multiple generations on cytotoxicity and Biomed Res Int. 2013;2013:942916.
neuronal differentiation using human neural progen- 99. Soni D, Gandhi D, Tarale P, Bafana A, Pandey RA,
itor cells. J Toxicol Sci. 2016;41:351–70. Sivanesan S. Oxidative stress and genotoxicity of
85. You R, Ho YS, Hung CH, et al. Silica nanoparticles zinc oxide nanoparticles to pseudomonas species,
induce neurodegeneration-like changes in behavior, human promyelocytic leukemic (Hl-60), and blood
neuropathology, and affect synapse through MAPK cells. Biol Trace Elem Res. 2017;178:218–27.
activation. Part Fibre Toxicol. 2018;15:28. 100. Abdelazeim SA, Shehata NI, Aly HF, Shams
86. Li N, Georas S, Alexis N, et al. A work group SGE. Amelioration of oxidative stress-mediated
report on ultrafine particles (American Academy apoptosis in copper oxide nanoparticles-induced
of Allergy, Asthma & Immunology): why ambi- liver injury in rats by potent antioxidants. Sci Rep.
ent ultrafine and engineered nanoparticles should 2020;10:10812.
receive special attention for possible adverse 101. Liu W, Hu T, Zhou L, et al. Nrf2 protects against
health outcomes in human subjects. J Allergy Clin oxidative stress induced by SiO(2) nanoparticles.
Immunol. 2016;138:386–96. Nanomedicine (Lond). 2017;12:2303–18.
87. Kan H, Pan D, Castranova V. Engineered nanopar- 102. Pujalté I, Passagne I, Brouillaud B, et al. Cytotoxicity
ticle exposure and cardiovascular effects: the role and oxidative stress induced by different metal-
of a neuronal-regulated pathway. Inhal Toxicol. lic nanoparticles on human kidney cells. Part Fibre
2018;30:335–42. Toxicol. 2011;8:10.
88. Kunovac A, Hathaway QA, Pinti MV, et al. Ros 103. Terada T, Kulkarni JA, Huynh A, Tam YYC, Cullis
promote epigenetic remodeling and cardiac dys- P. Protective effect of edaravone against cationic
function in offspring following maternal engineered lipid-mediated oxidative stress and apoptosis. Biol
nanomaterial (ENM) exposure. Part Fibre Toxicol. Pharm Bull. 2021;44:144–9.
2019;16:24. 104. Li Y, Zhu H, Wang S, et al. Interplay of oxidative
89. Yu X, Hong F, Zhang YQ. Cardiac inflammation stress and autophagy in PAMAM dendrimers-­
involving in PKCε or ERK1/2-activated NF-κB induced neuronal cell death. Theranostics.
signalling pathway in mice following exposure to 2015;5:1363–77.
titanium dioxide nanoparticles. J Hazard Mater. 105. Mukherjee SP, Byrne HJ. Polyamidoamine den-
2016;313:68–77. drimer nanoparticle cytotoxicity, oxidative stress,
90. Feng L, Ning R, Liu J, et al. Silica nanoparticles caspase activation and inflammatory response:
induce JNK-mediated inflammation and myo- experimental observation and numerical simulation.
cardial contractile dysfunction. J Hazard Mater. Nanomedicine. 2013;9:202–11.
2020;391:122206. 106. Rasras S, Kalantari H, Rezaei M, et al. Single-walled
91. Thompson LC, Sheehan NL, Walters DM, Lust RM, and multiwalled carbon nanotubes induce oxidative
Brown JM, Wingard CJ. Airway exposure to modi- stress in isolated rat brain mitochondria. Toxicol Ind
fied multi-walled carbon nanotubes perturbs cardio- Health. 2019;35:497–506.
vascular adenosinergic signaling in mice. Cardiovasc 107. Rouse JG, Yang J, Barron AR, Monteiro-Riviere
Toxicol. 2019;19:168–77. NA. Fullerene-based amino acid nanoparticle inter-
92. Shah V, Taratula O, Garbuzenko OB, et al. actions with human epidermal keratinocytes. Toxicol
Genotoxicity of different nanocarriers: possible In Vitro. 2006;20:1313–20.
modifications for the delivery of nucleic acids. Curr 108. Turabekova M, Rasulev B, Theodore M, Jackman
Drug Discov Technol. 2013;10:8–15. J, Leszczynska D, Leszczynski J. Immunotoxicity
93. Singh N, Manshian B, Jenkins GJ, et al. of nanoparticles: a computational study suggests
Nanogenotoxicology: the DNA damaging poten- that CNTs and C60 fullerenes might be recognized
tial of engineered nanomaterials. Biomaterials. as pathogens by Toll-like receptors. Nanoscale.
2009;30:3891–914. 2014;6:3488–95.
94. Ng CT, Li JJ, Bay BH, Yung LY. Current studies into 109. Szebeni J, Alving CR, Rosivall L, et al. Animal
the genotoxic effects of nanomaterials. J Nucleic models of complement-mediated hypersensitiv-
Acids. 2010;2010:947859.
19 Toxicity Evaluation of Nanomedicine 343

ity reactions to liposomes and other lipid-based earlier biomarker for nanotoxicological evaluation.
nanoparticles. J Liposome Res. 2007;17:107–17. ACS Nano. 2014;8:2562–74.
110. Hu X, Dong M, Liang X, Liu Z, Li Q. Reactive 124. Hou CC, Tsai TL, Su WP, et al. Pronounced induction
oxygen species-mediated inflammation and apop- of endoplasmic reticulum stress and tumor suppres-
tosis in hand-foot syndrome induced by PEGylated sion by surfactant-free poly(lactic-co-glycolic acid)
liposomal doxorubicin. Int J Nanomedicine. nanoparticles via modulation of the PI3K signaling
2021;16:471–80. pathway. Int J Nanomedicine. 2013;8:2689–707.
111. Khanna P, Ong C, Bay BH, Baeg GH. Nanotoxicity: 125. Tsai YY, Huang YH, Chao YL, et al. Identification of
an interplay of oxidative stress, inflammation and the nanogold particle-induced endoplasmic reticu-
cell death. Nanomaterials (Basel). 2015;5:1163–80. lum stress by omic techniques and systems biology
112. Wu W, Samet JM, Peden DB, Bromberg analysis. ACS Nano. 2011;5:9354–69.
PA. Phosphorylation of P65 is required for zinc 126. Stern ST, Adiseshaiah PP, Crist RM. Autophagy and
oxide nanoparticle-induced interleukin 8 expression lysosomal dysfunction as emerging mechanisms of
in human bronchial epithelial cells. Environ Health nanomaterial toxicity. Part Fibre Toxicol. 2012;9:20.
Perspect. 2010;118:982–7. 127. Sohaebuddin SK, Thevenot PT, Baker D, Eaton JW,
113. Chen J, Zhang J, Cao J, Xia Z, Gan J. Inflammatory Tang L. Nanomaterial cytotoxicity is composition,
MAPK and NF-κB signaling pathways differenti- size, and cell type dependent. Part Fibre Toxicol.
ated hepatitis potential of two agglomerated titanium 2010;7:22.
dioxide particles. J Hazard Mater. 2016;304:370–8. 128. Xia T, Kovochich M, Liong M, Zink JI, Nel
114. Garapaty A, Champion JA. Shape of ligand immo- AE. Cationic polystyrene nanosphere toxicity
bilized particles dominates and amplifies the mac- depends on cell-specific endocytic and mitochon-
rophage cytokine response to ligands. PLoS One. drial injury pathways. ACS Nano. 2008;2:85–96.
2019;14:e0217022. 129. Thomas TP, Majoros I, Kotlyar A, Mullen D, Holl
115. Mohammadinejad R, Moosavi MA, Tavakol S, et al. MM, Baker JR Jr. Cationic poly(amidoamine)
Necrotic, apoptotic and autophagic cell fates trig- dendrimer induces lysosomal apoptotic path-
gered by nanoparticles. Autophagy. 2019;15:4–33. way at therapeutically relevant concentrations.
116. Hsin YH, Chen CF, Huang S, Shih TS, Lai PS, Biomacromolecules. 2009;10:3207–14.
Chueh PJ. The apoptotic effect of nanosilver is 130. Cho WS, Duffin R, Howie SE, et al. Progressive
mediated by a ROS- and JNK-dependent mecha- severe lung injury by zinc oxide nanoparticles; the
nism involving the mitochondrial pathway in role of Zn2+ dissolution inside lysosomes. Part Fibre
NIH3T3 cells. Toxicol Lett. 2008;179:130–9. Toxicol. 2011;8:27.
117. Kim S, Ryu DY. Silver nanoparticle-induced oxi- 131. Feng X, Zhang Y, Zhang C, et al. Nanomaterial-­
dative stress, genotoxicity and apoptosis in cul- mediated autophagy: coexisting hazard and
tured cells and animal tissues. J Appl Toxicol. health benefits in biomedicine. Part Fibre Toxicol.
2013;33:78–89. 2020;17:53.
118. Siddiqui MA, Alhadlaq HA, Ahmad J, Al-Khedhairy 132. Zhu S, Zhang J, Zhang L, et al. Inhibition of Kupffer
AA, Musarrat J, Ahamed M. Copper oxide nanopar- cell autophagy abrogates nanoparticle-induced liver
ticles induced mitochondria mediated apopto- injury. Adv Healthc Mater. 2017;6:1601252.
sis in human hepatocarcinoma cells. PLoS One. 133. Jiang X, Tang Q, Zhang J, et al. Autophagy-­
2013;8:e69534. dependent release of zinc ions is critical for acute
119. Ryu W-I, Park Y-H, Bae HC, et al. ZnO nanoparticle lung injury triggered by zinc oxide nanoparticles.
induces apoptosis by ROS triggered mitochondrial Nanotoxicology. 2018;12:1068–91.
pathway in human keratinocytes. Mol Cell Toxicol. 134. Zhang X, Zhang H, Liang X, et al. Iron oxide
2014;10:387–91. nanoparticles induce autophagosome accumulation
120. Gopinath P, Gogoi SK, Sanpui P, Paul A, through multiple mechanisms: lysosome impair-
Chattopadhyay A, Ghosh SS. Signaling gene cas- ment, mitochondrial damage, and ER stress. Mol
cade in silver nanoparticle induced apoptosis. Pharm. 2016;13:2578–87.
Colloids Surf B Biointerfaces. 2010;77:240–5. 135. Hawkins SJ, Crompton LA, Sood A, et al.
121. Hanna DH, Saad GR. Induction of mitochondria Nanoparticle-induced neuronal toxicity across
mediated apoptosis in human ovarian cancer cells by placental barriers is mediated by autophagy
folic acid coated tin oxide nanoparticles. PLoS One. and dependent on astrocytes. Nat Nanotechnol.
2021;16:e0258115. 2018;13:427–33.
122. Christen V, Camenzind M, Fent K. Silica nanopar- 136. Chen HH, Yu C, Ueng TH, et al. Acute and subacute
ticles induce endoplasmic reticulum stress response, toxicity study of water-soluble polyalkylsulfonated
oxidative stress and activate the mitogen-activated C60 in rats. Toxicol Pathol. 1998;26:143–51.
protein kinase (MAPK) signaling pathway. Toxicol 137. De Stefano D, Carnuccio R, Maiuri
Rep. 2014;1:1143–51. MC. Nanomaterials toxicity and cell death modali-
123. Chen R, Huo L, Shi X, et al. Endoplasmic reticu- ties. J Drug Deliv. 2012;2012:167896.
lum stress induced by zinc oxide nanoparticles is an 138. Braydich-Stolle LK, Schaeublin NM, Murdock
RC, et al. Crystal structure mediates mode of
344 A. Panghal and S. J. S. Flora

cell death in TiO2 nanotoxicity. J Nanopart Res. 153. Kaufmann SH, Lee SH, Meng XW, et al. Apoptosis-­
2009;11:1361–74. associated caspase activation assays. Methods.
139. Schaeublin NM, Braydich-Stolle LK, Schrand AM, 2008;44:262–72.
et al. Surface charge of gold nanoparticles mediates 154. Drasler B, Sayre P, Steinhäuser KG, Petri-Fink
mechanism of toxicity. Nanoscale. 2011;3:410–20. A, Rothen-Rutishauser B. In vitro approaches to
140. Vergallo C, Panzarini E, Carata E, et al. Cytotoxicity assess the hazard of nanomaterials. NanoImpact.
of Β-D-glucose/sucrose-coated silver nanoparticles 2017;8:99–116.
depends on cell type, nanoparticles concentration 155. Mangini M, Verde A, Boraschi D, Puntes VF, Italiani
and time of incubation. In: AIP conference pro- P, De Luca AC. Interaction of nanoparticles with
ceedings, vol. 1749. AIP Publishing LLC; 2016. endotoxin importance in nanosafety testing and
p. 020012. exploitation for endotoxin binding. Nanotoxicology.
141. Panzarini E, Mariano S, Dini L. Investigations of the 2021;15:558–76.
toxic effects of glycans-based silver nanoparticles on 156. Smulders S, Kaiser JP, Zuin S, et al. Contamination
different types of human cells. In: AIP conference of nanoparticles by endotoxin: evaluation of differ-
proceedings, vol. 1873. AIP Publishing LLC; 2017. ent test methods. Part Fibre Toxicol. 2012;9:41.
p. 020012. 157. Sadik OA, Zhou AL, Kikandi S, Du N, Wang Q,
142. Bauer AT, Strozyk EA, Gorzelanny C, et al. Varner K. Sensors as tools for quantitation, nanotox-
Cytotoxicity of silica nanoparticles through exocy- icity and nanomonitoring assessment of engineered
tosis of Von Willebrand factor and necrotic cell death nanomaterials. J Environ Monit. 2009;11:1782–800.
in primary human endothelial cells. Biomaterials. 158. Fröhlich E. Role of omics techniques in the tox-
2011;32:8385–93. icity testing of nanoparticles. J Nanobiotechnol.
143. Wei X, Shao B, He Z, et al. Cationic nanocarriers 2017;15:84.
induce cell necrosis through impairment of Na(+)/ 159. Huang HJ, Lee YH, Hsu YH, Liao CT, Lin YF, Chiu
K(+)-ATPase and cause subsequent inflammatory HW. Current strategies in assessment of nanotoxic-
response. Cell Res. 2015;25:237–53. ity: alternatives to in vivo animal testing. Int J Mol
144. Lai L, Jin JC, Xu ZQ, Mei P, Jiang FL, Liu Y. Necrotic Sci. 2021;22:4216.
cell death induced by the protein-mediated intercel- 160. Barenholz Y. Doxil®--the first FDA-approved
lular uptake of CdTe quantum dots. Chemosphere. nano-drug: lessons learned. J Control Release.
2015;135:240–9. 2012;160:117–34.
145. Shukla RK, Badiye A, Vajpayee K, Kapoor 161. Henney JE. Toxicity testing: the FDA perspective.
N. Genotoxic potential of nanoparticles: structural Ann N Y Acad Sci. 2000;919:75–8.
and functional modifications in DNA. Front Genet. 162. Arvidson KB. FDA toxicity databases and real-time
2021;12:728250. data entry. Toxicol Appl Pharmacol. 2008;233:17–9.
146. Demir E, Creus A, Marcos R. Genotoxicity and 163. Lama S, Merlin-Zhang O, Yang C. In vitro and in vivo
DNA repair processes of zinc oxide nanoparticles. J models for evaluating the oral toxicity of nanomedi-
Toxicol Environ Health A. 2014;77:1292–303. cines. Nanomaterials (Basel). 2020;10:2177.
147. Kruszewski M, Grądzka I, Bartłomiejczyk T, et al. 164. Baati T, Njim L, Jaafoura S, et al. Assessment of
Oxidative DNA damage corresponds to the long pharmacokinetics, toxicity, and biodistribution of a
term survival of human cells treated with silver high dose of Titanate nanotubes following intrave-
nanoparticles. Toxicol Lett. 2013;219:151–9. nous injection in mice: a promising nanosystem of
148. Choudhury SR, Ordaz J, Lo CL, Damayanti NP, medical interest. ACS Omega. 2021;6:21872–83.
Zhou F, Irudayaraj J. From the cover: zinc oxide 165. Fonseca-Gomes J, Loureiro JA, Tanqueiro SR, et al.
nanoparticles-induced reactive oxygen species pro- In vivo bio-distribution and toxicity evaluation of
motes multimodal cyto- and epigenetic toxicity. polymeric and lipid-based nanoparticles: a poten-
Toxicol Sci. 2017;156:261–74. tial approach for chronic diseases treatment. Int J
149. Pogribna M, Hammons G. Epigenetic effects of Nanomedicine. 2020;15:8609–21.
nanomaterials and nanoparticles. J Nanobiotechnol. 166. Zou W, Li L, Chen Y, et al. In vivo toxicity evalu-
2021;19:2. ation of PEGylated CuIns2/ZnS quantum dots in
150. Kopp B, Dario M, Zalko D, Audebert M. Assessment BALB/c mice. Front Pharmacol. 2019;10:437.
of a panel of cellular biomarkers and the kinetics 167. Højelse F. Preclinical safety assessment: in vitro --
of their induction in comparing genotoxic modes in vivo testing. Pharmacol Toxicol. 2000;86(Suppl
of action in Hepg2 cells. Environ Mol Mutagen. 1):6–7.
2018;59:516–28. 168. Ibrahim KE, Al-Mutary MG, Bakhiet AO, Khan
151. Seidel C, Kirsch A, Fontana C, et al. Epigenetic HA. Histopathology of the liver, kidney, and spleen
changes in the early stage of silica-induced cell of mice exposed to gold nanoparticles. Molecules.
transformation. Nanotoxicology. 2017;11:923–35. 2018;23:1848.
152. Savage DT, Hilt JZ, Dziubla TD. In vitro methods 169. Lai C-Z, Fierke MA, da Costa RC, Gladysz JA, Stein
for assessing nanoparticle toxicity. Methods Mol A, Bühlmann P. Highly selective detection of silver
Biol. 1894;2019:1–29. in the low ppt range with ion-selective electrodes
19 Toxicity Evaluation of Nanomedicine 345

based on ionophore-doped fluorous membranes. 179. Shin TH, Lee DY, Lee HS, et al. Integration of
Anal Chem. 2010;82:7634–40. metabolomics and transcriptomics in nanotoxicity
170. Xu K, Pérez-Ràfols C, Cuartero M, Crespo studies. BMB Rep. 2018;51:14–20.
GA. Electrochemical detection of trace silver. 180. Alsagaby SA, Vijayakumar R, Premanathan M, et al.
Electrochim Acta. 2021;374:137929. Transcriptomics-based characterization of the toxic-
171. Chatterjee A, Santra M, Won N, et al. Selective fluo- ity of ZnO nanoparticles against chronic myeloid leu-
rogenic and chromogenic probe for detection of sil- kemia cells. Int J Nanomedicine. 2020;15:7901–21.
ver ions and silver nanoparticles in aqueous media. J 181. Chen Z, Gao SH, Jin M, et al. Physiological and
Am Chem Soc. 2009;131:2040–1. transcriptomic analyses reveal CuO nanopar-
172. Hondroulis E, Shah P, Zhu X, Li C-Z. Biosensing ticle inhibition of anabolic and catabolic activi-
devices for toxicity assessment of nanomaterials. In: ties of sulfate-reducing bacterium. Environ Int.
Biointeractions of nanomaterials. CRC Press; 2014. 2019;125:65–74.
p. 117–30. 182. Zhao L, Wan H, Liu Q, Wang D. Multi-walled car-
173. Asphahani F, Zhang M. Cellular impedance biosen- bon nanotubes-induced alterations in microRNA
sors for drug screening and toxin detection. Analyst. let-7 and its targets activate a protection mechanism
2007;132:835–41. by conferring a developmental timing control. Part
174. Kim T-H, Kang S-R, Oh B-K, Choi J-W. Cell chip Fibre Toxicol. 2017;14:27.
for detection of silica nanoparticle-induced cytotox- 183. Zhao Y, Wu Q, Li Y, Nouara A, Jia R, Wang D. In
icity. Sens Lett. 2011;9:861–5. vivo translocation and toxicity of multi-walled
175. Kafi M, Cho H-Y, Choi JW. Neural cell chip carbon nanotubes are regulated by microRNAs.
based electrochemical detection of nanotoxicity. Nanoscale. 2014;6:4275–84.
Nanomaterials. 2015;5:1181–99. 184. Nienhaus K, Nienhaus GU. Towards a molecular-­
176. Liu L, He F, Yu Y, Wang Y. Application of FRET level understanding of the protein corona around
biosensors in mechanobiology and mechanophar- nanoparticles–recent advances and persisting chal-
macological screening. Front Bioeng Biotechnol. lenges. Curr Opin Biomed Eng. 2019;10:11–22.
2020;8:595497. 185. Qin M, Zhang J, Li M, et al. Proteomic analysis of
177. Geng Y, Chattopadhyay AN, Zhang X, et al. Nano intracellular protein corona of nanoparticles eluci-
assessing nano: nanosensor-enabled detection of cell dates nano-trafficking network and nano-bio interac-
phenotypic changes identifies nanoparticle toxico- tions. Theranostics. 2020;10:1213–29.
logical effects at ultra-low exposure levels. Small. 186. Shim W, Paik MJ, Nguyen D-T, et al. Analysis of
2020;16:e2002084. changes in gene expression and metabolic profiles
178. Fan TW, Higashi RM, Lane AN. Integrating metabo- induced by silica-coated magnetic nanoparticles.
lomics and transcriptomics for probing SE antican- ACS Nano. 2012;6:7665–80.
cer mechanisms. Drug Metab Rev. 2006;38:707–32.
Biochemical Indices of Drug
Toxicity
20
Emmanuel Kwaku Ofori

Abstract Keywords

Generally, drugs or chemicals have the poten- Biomarker · Electrolytes · Enzymes ·


tial to trigger unpleasant responses when taken Hepatocytes · Toxicity
in excess. Changes in biochemical markers
provide information on the mechanism of tox-
icity, the functional state of important organ 20.1 Introduction
systems, and the identification of target tissues
(liver, kidney, hematopoietic and immune sys- The use of drugs and the adverse or toxic conse-
tems, etc.). This is because variations in the quences that arise is a public health problem. The
levels of enzymes and non-enzymes in the pharmaceutical industry’s primary focus contin-
serum signal tissue or cellular damage, which ues to be on reducing toxicity of drugs to the
results in an abnormal release of intracellular greatest extent possible. It has been proposed that
components into circulation. It is, thus, impor- a variety of biochemical markers be tracked to
tant to understand the biochemical indices investigate the potentially detrimental effects of
involved in drug toxicity to enable rapid chemical compounds, including plant extracts.
response, which may include withdrawal of The term “biochemical marker” refers to a mea-
toxic agent. The biochemical indicators of surable biological observation that may be used
drug toxicity, as well as the basis behind the to substitute for and, in some cases, anticipate a
measurement of enzymes or proteins impli- clinically meaningful endpoint that is more diffi-
cated in drug toxicity, are covered in this chap- cult to detect [1]. It is important to note that bio-
ter. This chapter will detail the kidney, liver, chemical markers are used in toxicology for three
and cardiac function tests used, as well as the primary purposes: they can be used to confirm
interpretation of results. We’ll also go through the presence of a harmful agent, they can be used
what we know about novel toxicity biomark- to monitor individual susceptibility to a toxicant,
ers and how they are being applied to human and they can be used to quantitatively assess the
populations after being studied in deleterious effects of a toxicant on an organism
animal models. or an individual [1, 2]. These biochemical mark-
ers are also used for a variety of other purposes,
E. K. Ofori (*) including the prediction and treatment of adverse
Department of Chemical Pathology, University of medication responses, the identification of cell
Ghana Medical School, Accra, Ghana types, pharmacodynamics, and dose-response
e-mail: ekofori1@ug.edu.gh

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 347
S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_20
348 E. K. Ofori

studies [3]. These techniques may also be applied creatine kinase (CK) are the most routinely
in the context of health and disease states for assessed biochemical markers in drug toxicity.
chemical screening during drug discovery, diag- Among the non-enzymatic parameters that are
nosis, and characterization, as well as for moni- commonly investigated in toxicological studies
toring prognostic markers and designing tailored are the measurements of bilirubin, creatinine,
treatments [4, 5]. blood urea nitrogen (BUN), malondialdehyde
Ideally, good biochemical markers should be (MDA), glucose (including fasting levels), total
measurable with little or no systematic variability, protein (including albumin and globulin levels),
should improve when the condition improves and B-type natriuretic protein (BNP), troponins, cho-
vice versa, and should be able to distinguish clini- lesterol, and triglyceride levels. In medication
cally significant changes over time from back- toxicity research, certain common electrolytes
ground values such as biological fluctuations and and minerals are being studied extensively. These
measurement errors [6–8]. The measurement of include sodium, potassium, phosphate, magne-
enzyme and non-enzyme activity is extremely sium, calcium, and chloride, to name a few. It has
important in detecting metabolic changes early in been possible to test these parameters using stan-
the course of a disease as well as in detecting dard biochemical procedures, automated clinical
organ-specific consequences of a medication’s chemistry analyzer techniques, and commercial
action, determining toxic processes, and establish- kits [10, 11]. It is the purpose of this chapter to
ing pharmacological effects. To use enzyme levels offer a concise summary of various conventional
as a marker of cellular or tissue damage, the basic and novel biochemical indicators that are associ-
principle relies on a comparison of the changes in ated with organ damage and medication toxicity.
activity in serum or plasma enzymes, which are
primarily present in the intracellular environment
and are only released into the serum in very low 20.2 Kidneys
concentrations [1, 4, 9]. It follows that fluctuation
of these enzymes in serum is indicative of tissue or The kidney is one of the organs that is commonly
cellular injury, which results in an aberrant release evaluated in the pharmaceutical business because
of intracellular components into the bloodstream. it plays a critical role in drug excretion and detox-
Blood serum or plasma is a particularly useful pro- ification [12, 13]. The glomerulus is responsible
tein pool for repeated biomarker analysis through- for the filtration of the blood in the nephron,
out time because it contains a large number of which is the beginning of renal excretion. After
proteins. It is also highly significant in toxicologi- passing through this filtering barrier, the primary
cal investigations to analyze urine for the presence urine enters the tubular system for processing. It
of kidney involvement, which might be difficult to then passes via the proximal tubule, where essen-
detect without it. Urinalysis can provide an early tial components are reabsorbed, and onto the
indication of whether or not the kidney is working loop of Henle, where urine concentration is
normally or whether or not its functionality has determined, the distal tubule, and lastly the col-
been impaired. The pH, specific gravity, glucose, lecting duct, where the final urinary concentra-
ketones, protein, nitrites (including bilirubin), uro- tion is adjusted and the urine is discharged [13,
bilinogen, and cellular microscopy of urine are all 14] In addition, the kidneys receive about 25% of
measured in addition to the color and appearance. the cardiac output and play a substantial role in
Table 20.1 contains a collection of biochemi- the biotransformation of medicines into hazard-
cal indicators that are commonly used in drug ous metabolites [15, 16], making them particu-
and organ toxicity investigations. The enzymes larly vulnerable to tubular epithelial cell injury
aspartate aminotransferase (AST), alanine ami- [17].
notransferase (ALT), lactate dehydrogenase Waste materials and extra fluid can accumu-
(LDH), alkaline phosphatase (ALP), gamma-­ late in the body if the ability of the kidneys to
glutamyltransferase (GGT), 5′ nucleotidase, and filter blood is significantly diminished. It has
20 Biochemical Indices of Drug Toxicity 349

Table 20.1 Biochemical markers of drug toxicity and organ damage


Enzymes Non-enzymes Electrolytes
GGT (gamma-glutamyltransferase) Total bilirubin Sodium
ALT (alanine aminotransferase) Total cholesterol Potassium
AP (alkaline phosphatase) Triglycerides Chloride
AST (aspartate aminotransferase) Total protein Calcium
LDH (lactate dehydrogenase) Albumin Phosphorus
CK (creatine kinase) Glucose Magnesium
Carbonyl transferase Bile acids Iron
Malate dehydrogenase Creatinine Copper
Sorbitol dehydrogenase BUN (blood urea nitrogen)
Paraoxonase Glutathione
5′ nucleotidase Troponins
Lipase Malondialdehyde
Amylase BNP (B-type natriuretic protein)

been shown that a loss in the kidneys’ ability to diagnosis of kidney disease. The levels of serum
reabsorb endogenous components such as small creatinine and BUN are not completely predic-
proteins, glucose, or metabolites following a tive of GFR. They are generic and insensitive
renal injury might increase the amount of these when it comes to distinguishing between various
components excreted in the urine. Injury repair phases of cellular damage or the consequences of
and remission rely on the severity of the injury cellular injury [22, 23]. These significant disad-
and entail the migration, proliferation, and differ- vantages have resulted in clinical outcomes that
entiation of epithelial cells, which help the body have been delayed. As a result, the discovery and
restore its structure and function after it has been validation of biomarkers for the early identifica-
damaged. Biomarkers with improved sensitivity tion of AKI are an absolute requirement. This
and specificity are required in drug development article provides a concise summary of various
risk assessment studies as well as in predicted classic and developing biomarkers for the early
toxicological screenings for chemical substances diagnosis of kidney disease, as well as potential
and the evaluation of therapeutic agents for the clinical applications.
activation of kidney regeneration [18].
It has been shown that acute kidney injury
(AKI), also known as “acute renal failure,” is 20.2.1 Serum Creatinine and Urea
linked with a significant mortality rate of up to
80% in some studies [19, 20]. Blood and/or pro- The serum creatinine and blood urea nitrogen
tein in the urine, high blood pressure, frequent (BUN) levels are the most routinely utilized
and/or painful urination, puffiness around the markers for the detection and monitoring of renal
eyes, and edema of the hands and feet are some function. Muscle catabolism results in the pro-
of the traditional warning symptoms of kidney duction of creatinine, which is a chemical waste
disease [8–21]. Irreversible damage to the kidney molecule [24, 25]. Phosphocreatine, a molecule
results in a decrease in the glomerular filtration engaged in energy generation in muscles, under-
rate (GFR), which compromises kidney function goes spontaneous cyclization, which results in
and ultimately leads to renal failure and mortal- the formation of creatine and inorganic phospho-
ity. GFR decreases as a result of irreversible kid- rus and is catalyzed by creatine kinase (CK) [9,
ney injury. Patient monitoring for renal damage 26]. Phosphocreatine is a molecule involved in
has traditionally relied on serum creatinine, blood energy production in muscles. Further spontane-
urea nitrogen (BUN), other waste products, and ous breakdown of creatine into creatinine
the detection of urine components, among other ­happens at a very regular and uniform pace dur-
things. Nonetheless, there are certain limits to ing each day. The level of creatinine in the blood
their diagnostic capacities when it comes to the is a reasonably good predictor of intrinsic kidney
350 E. K. Ofori

function. The glomerulus filters creatinine freely, the glomerular filtrate in the same concentration
and the clearance of creatinine from the plasma as it does in the bloodstream. Urea concentra-
to the urine may be used to estimate the glomeru- tions in the blood rise as a result of decreased glo-
lar filtration rate (GFR) to a reasonable degree of merular filtration [13, 28]. The concentration of
accuracy [27]. A little quantity of creatinine is serum urea may also be determined as urea nitro-
released by the proximal tubules of the kidney; gen, and this test is referred to as blood urea
however, unlike urea, no creatinine is resorbed by nitrogen (BUN). Uremia (increased BUN levels)
the tubules in the same way. Additional studies can develop in patients with renal failure (both
have shown that the use of creatinine clearance acute and chronic), according to the literature
might result in artificially high GFR values, par- [40].
ticularly in persons with chronic renal failure [28, A variety of disorders can harm the kidneys,
29]. resulting in inefficient urine production and
If the kidneys become compromised for what- excretion. Congestive heart failure can result in
ever reason, the quantity of creatinine in the low blood pressure, elevated urea levels, and a
blood will rise as a result of inefficient or resulting drop in glomerular filtration rate (GFR)
decreased clearance of creatinine by the kidneys, [41, 42]. Urinary tract infections, on the other
which will cause the blood to become acidic. hand, might result in elevated urea levels [39].
High creatinine levels are indicative of compro- When there is an increase in protein catabolism,
mised renal function or kidney disease. When such as during gastrointestinal bleeding, clinical
comparing blood creatinine concentration to urea dehydration, hypovolemia, shock, congestive
concentration, it has been shown that the former heart failure, pre-eclampsia, or rhabdomyolysis,
is impacted by muscle mass while the latter is urea concentrations might rise or fall [13, 43].
essentially unaffected by dietary impacts and Hemodialysis is done in severe instances to elim-
protein catabolism [30, 31]. The blood creatinine inate soluble urea and other waste products from
level is also affected by factors such as age, gen- the bloodstream. The measurements of urea and
der, and weight [32, 33]. The presence of gastro- creatinine are used in combination to determine
intestinal bleeding has also been observed to renal function. They are, on the other hand, rela-
raise serum creatinine concentrations, with no tively impervious to even little renal damage due
apparent adverse effect on the kidney [34, 35]. to infection. Changes in creatinine concentra-
In humans, urea is the most important end tions in the blood tend to correspond to changes
product of nitrogen metabolism. It is produced by in urea concentrations in the blood when there is
the liver’s cells from ammonia produced by the renal toxicity. However, as compared to urea, the
breakdown of amino acids derived either from concentration of creatinine varies more slowly
exogenous protein digestion or from endogenous over time. Even though it is not conclusive, the
tissue proteins [36]. Ammonia is generated by the urea to creatinine ratio can be useful in making a
breakdown of amino acids derived either from differential diagnosis of azotemia.
exogenous protein digestion or from endogenous
tissue proteins [37]. Urea is excreted by several
organs, including the kidneys, the gut, the saliva, 20.2.2 The Glomerular Filtration
and the perspiration. Rate (GFR) and Clearance
Because the kidney is the most significant
route of urea elimination, urea has long been To represent the flow rate of filtered fluid via the
employed as a marker of renal function. Like cre- kidney, a computed value is used [44, 45]. Renal
atinine, urine is freely filtered by the glomerulus; blood flow and pressure are both influenced by
however, minor quantities are reabsorbed by the the clearance concept, and GFR is intimately
renal tubules and released by the proximal tubules connected to both [46]. It is possible to define
throughout the filtration process [38, 39]. When clearance as the amount of blood that can be
the body is functioning normally, urea arrives in cleared of the drug under consideration in a par-
20 Biochemical Indices of Drug Toxicity 351

ticular length of time. It is the assessment of how renal dysfunction [50–52]. Cystatin C levels in
much a certain item may be removed from a urine can be used to detect drug-induced renal
given volume of blood in a specified amount of damage [53] and are related to a reduction in
time by the kidneys. If the item being examined tubular reabsorption as a result of the injury.
is to be relevant as a biomarker for clearance Furthermore, biological parameters such as gen-
investigations, it must be eliminated only by pas- der, age, ethnicity, and muscle mass have no sig-
sive filtration at the glomerulus. For the tubules to nificant effect on cystatin C levels in the blood.
function properly, there must be almost no re-­ Cystatin C appears to be a more sensitive bio-
absorption or additional secretion. The substance marker of reduced GFR than creatinine, although
must also exist in the same biochemical form in it has not been well tested to the point where it
both the blood and the urine, which necessitates can be recommended for frequent usage.
the absence of any metabolic change before uri-
nary excretion may take place. Inulin (a plant
polysaccharide) has been shown to satisfy this 20.2.4 Fibrinogen
need in experiments; but, in reality, creatinine is
utilized since it is naturally available in plasma Fibrinogen is a protein that is essential to the
and urine as a byproduct of muscle metabolism clotting process and is found in large quantities in
and does not need to be injected into the body. the blood [54]. As an acute response protein, it
Thus, creatinine clearance is defined as the vol- has been shown to have an important function in
ume of plasma that is cleared of creatinine in a inflammatory illnesses [55, 56]. When there is
certain amount of time per unit of time. Several AKI, the fibrinogen protein gene is activated in
blood samples are obtained together with a timed the kidneys, and the fibrinogen protein is expelled
urine collection to evaluate this parameter (usu- more in the urine [57]. In patients with and with-
ally a 24 h collection). The data can be used to out AKI, urinary fibrinogen may be used to suc-
determine how well the kidneys are doing in cessfully discriminate between the two groups,
terms of excretory function. As a whole, these with the excreted protein reaching an AUC-ROC
computed numbers can be used to track the evo- of 0.98, which is comparable to other recognized
lution of kidney damage that has already biomarkers of renal damage [18, 57].
occurred. Early identification of a growing injury, Consequently, fibrinogen is developing as a novel
on the other hand, is not achievable [47, 48]. As a translational biomarker for the diagnosis of AKI,
result, there is an urgent need for novel biomark- and it has the potential to be examined further as
ers that are better, cheaper, and more efficient a therapeutic target to treat AKI.
than the ones now available.

20.2.5 Kidney Injury Molecule-1


20.2.3 Blood Cystatin C
Kidney injury molecule-1 (KIM-1) is a type 1
It has been discovered that cystatin C in the cell membrane glycoprotein of 104 kDa that con-
bloodstream is a 13-kDa peptide of low molecu- tains immunoglobulin-like domains and is not
lar weight that is widely dispersed in nucleated detectable in normal kidney tissue or urine [58].
cells of the human body [49]. Cystatin C is a pro- Because KIM-1 protein is produced by tubule
tein that is readily filtered by the glomerulus, epithelial cells in response to damage, it is an
completely reabsorbed by the kidneys, and not excellent biomarker for renal tubular function
released by the tubules, making it a promising [59]. In addition, KIM-1 is very sensitive to inter-
marker for glomerular function (GFR). ferences from unrelated kidney problems [60–
Additionally, the severity of acute and end-stage 62], as well as precise and specific. It is also
renal damage, diabetes, and cardiovascular risk is extremely stable in urine and is unaffected by
all associated with this endogenous marker of interferences from other kidney disorders. Kim-1
352 E. K. Ofori

has superior diagnostic capabilities than the usual serum creatinine and BUN levels are observed
indicators of renal injury (serum urea and creati- [73]. Because NAG excretion is increased in
nine), and it is capable of detecting subtle types other glomerular disorders, such as nephrotic
of tubular damage [63]. syndrome [19, 74], the use of NAG is still limited
in clinical practice.

20.2.6 Neutrophil Gelatinase-­
Associated Lipocalin 20.2.8 Beta-2-Microglobulin

A tiny 25 kDa secreted protein of the lipocalin Circulating β2-microglobulin in circulation is a


superfamily, neutrophil gelatinase-associated single polypeptide chain with a molecular weight
lipocalin (NGAL) has been identified as a poten- of 12 kDa, which is easily filtered by the glom-
tial marker of renal tubular damage [64]. NGAL eruli and virtually fully reabsorbed and degraded
was first discovered in innately activated neutro- by the renal tubules [75]. The production of
phils and has now been identified in other cell β2-microglobulin in persons who appear to be in
types [65]. As a result of acute kidney injury good health (normal) is rather consistent [76].
(AKI), NGAL expression is significantly Because of a decrease in their absorption in the
expressed in the renal tubules, ultimately result- renal tubules, there is an increase in the concen-
ing in elevated plasma and urine NGAL concen- tration of β2-microglobulin in urine [77].
trations [64, 66]. This means that NGAL can be Treatment with some medicines, such as antiret-
used as a valuable early indication of AKI in both roviral treatments in HIV patients [78] and cis-
the serum and urine. In humans, the protein platin therapies [79], has been shown to
expression levels of NGAL are increased rapidly significantly reduce tubular reabsorption of
in the epithelial cells of the lungs, colon, and liver β2-microglobulin.
within a few hours of the onset of cellular injury
[67, 68]. In addition, the protein expression levels
of NGAL are increased rapidly in the epithelial 20.2.9 Electrolytes
cells of the intestine [67, 68]. NGAL levels in
urine 1 day after kidney transplantation were Electrolytes are ions in solution that are both pos-
found to be predictive of recipients who would itively and negatively charged, and they may be
have delayed graft function and those who would found in all bodily fluids. These ions can conduct
require dialysis [69, 70]. electrical currents. When evaluating the safety of
medicine and, to a significant degree, the func-
tion of the kidneys, one of the most widely
20.2.7 N-Acetyl-beta- employed biochemical markers is serum electro-
glucosaminidase lyte concentration. Sodium, potassium, and chlo-
ride are among the electrolytes that are most
The protein N-acetyl-glucosaminidase (NAG) is frequently monitored in toxicology studies. Iron,
a lysosomal enzyme that is mostly present in magnesium, copper, calcium, and phosphate are
proximal tubule cells [71]. In addition, because all checked to a certain extent as well. Electrolyte
NAG is a big protein that is difficult to filter by imbalances can arise as a result of vomiting or
the glomerulus, its excretion into urine is associ- diarrhea in the gastrointestinal system [80]. Aside
ated with greater tubular cell damage [19, 72]. As from electrolyte loss or gain, acid-base disorders,
a result, it is a sensitive and specific urinary intrinsic kidney disease, and stimulants in the
marker of renal injury. Increases in blood NAG respiratory center can all cause electrolyte loss or
levels are frequently reported before increases in gain [81, 82].
20 Biochemical Indices of Drug Toxicity 353

20.2.10 Sodium with the remaining 2% found in the extracellular


fluid (ECF) being distributed across the intersti-
Sodium is the most prevalent cation in the body tial and plasma compartments [93, 94].
and the most abundant cation in the extracellular Maintaining the intracellular potassium gradient
fluid. Ionized sodium (Na+) is necessary for the is accomplished by an ATP-structured energetic
maintenance of normal osmotic pressure and pumping of sodium, which is balanced by the
fluid volume, as well as the excitability of neuro- calcium-mediated intracellular pumping of
muscular fibers [83, 84]. At the glomerulus, potassium and hydrogen ions into the cells [83,
plasma sodium (Na+) is loosely filtrated and reab- 95]. Potassium may also play important roles in
sorbed in both the proximal tubules and the loop the regulation of intracellular volume, clinical
of Henle, where it is combined with chloride ions enzymology, protein synthesis, and glucose
and water. Aldosterone controls sodium reab- metabolism, among other functions [95]. The
sorption in the distal convoluted tubules, and amount of potassium in the blood does not alter
sodium ions are exchanged at the cost of a univa- appreciably in response to water loss or retention.
lent cation (e.g., potassium and hydrogen ions) Factors that produce even a slight or unexpected
[85, 86]. It is possible to discriminate between alteration in intracellular K+ concentrations will
pre-renal and intrinsic acute renal failure with result in a significant shift in extracellular K+
high accuracy by utilizing fractional sodium concentrations. Insulin stimulates the uptake of
excretion [87]. Changes in body water and potassium by the cells. When the pH of a solution
plasma volume can have an impact on serum is acidic, hydrogen ions (H+) flow into cells (to
sodium concentrations, either directly or indi- be buffered) in exchange for potassium ions (K+).
rectly. The increase in sodium causes a decrease
in water retention, which increases the extracel-
lular fluid (ECF) volume [88, 89]. The loss of 20.2.12 Chloride
sodium results in the loss of water and the shrink-
ing of the ECF volume. The extracellular fluid With sodium, chloride is a major anion in extra-
compartment’s volume is therefore controlled by cellular fluid, and the two of them work together
the sodium concentration of ECF [90]. A wide to maintain electro-neutrality and osmolality in
range of xenobiotics affects sodium metabolism, the body. In the ECF, it is the most abundant
causing it to be retained or depleted [90]. anion linked with sodium [96, 97]. After passing
Controlling bodily water is performed by the through the glomerulus, chloride is passively
measurement of plasma osmolality, blood vol- reabsorbed in the proximal convoluted tubules
ume, and sodium concentrations, which are all and actively reabsorbed with sodium in the loop
interconnected. Osmo- and baro-regulations are of Henle and the distal tubules after being con-
used to accomplish this. centrated in the kidney. The ability of the kidneys
to vary daily chloride excretion allows total body
chloride levels to remain relatively constant and
20.2.11 Potassium serum chloride concentrations to remain within a
restricted range despite significant fluctuations in
In the intracellular environment, potassium is the daily intake. The measurement of serum Cl− con-
most abundant intracellular cation and is respon- centration seldom provides additional informa-
sible for the maintenance of electrochemical gra- tion to that acquired from the measurement of
dients and the transmission of impulses [91, 92]. Na+ concentration. Patients who vomit or have
The resting potential of excitable cells such as unusual chloride-losing episodes may benefit
neurons and muscle, including the myocardium, from measuring serum Cl−, which may be used to
is determined by potassium (K+), even though it calculate the “anion gap” and, as a result, may be
is present in low amounts in ECF. Almost all of able to be diagnosed with various acid-base
the total body K+ (98%) is found within cells, disorders.
354 E. K. Ofori

20.2.13 Calcium ized calcium concentrations as a result of the


effects of pH on the extracellular fluid (ECF) or
When it comes to minerals, calcium is the most protein binding. Under the influence of vitamin
prevalent, accounting for around 1.5% of total D, calcium and phosphate are absorbed in the gut
body mass [97]. The bones and teeth include together with other nutrients. Calcium is stored in
hydroxyapatite [Ca10 (PO4)6(OH)2], which the bones and excreted through the kidneys by
accounts for about 99% of the body’s calcium. the body as a waste product. The hormone para-
The remainder is primarily concentrated in the thyroid hormone, vitamin D, and the hormone
ECF compartment, with around 9 mmol in the calcitonin all affect renal excretion of calcium
plasma. Calcium is essential in the physiology [102].
and biochemistry of animals and cells, as well as
in the biochemistry of cells.
Calcium has a role in the release of neu- 20.2.14 Magnesium
rotransmitters, the contraction of all muscle cell
types, coagulation, cell growth, membrane trans- Magnesium is the second most important intra-
port processes, fertilization, and signal transduc- cellular cation after sodium. Magnesium is a
tion pathways, where it serves as a second mineral nutrient that is found in every cell type in
messenger [98]. This cation is also involved in every organism. An adult weighing 70 kg pos-
the generation of cardiac action potentials and sesses around 25 g magnesium (1000 mmol). The
the functioning of pacemakers, as well as the bones and teeth contain half of this total quantity.
contraction of cardiac, skeletal, and smooth mus- Of the remaining half, 98% is found in cells
cle, which has implications for myocardial where magnesium is the second most abundant
infarction and pharmaceutical treatment [98]. intracellular cation after potassium. The remain-
Coagulation experiments have shown that cal- der (1–2%) is found in the extracellular fluid
cium ions are necessary as a cofactor [99]. (ECF), with plasma magnesium concentrations
Total serum calcium is composed of three averaging around 0.7 mmol/L. When numerous
major forms: ionized calcium (which accounts enzyme systems, particularly those involved in
for approximately 50% of total), protein-bound energy metabolism, are activated, magnesium is
calcium (which accounts for 40% of total), and required to ensure that they function properly. As
the remainder, which is complexed with anions previously stated, it is required for the binding of
such as bicarbonate, citrate, lactate, and phos- macromolecules to organelles (e.g., the binding
phate (which accounts for approximately 10% of of mRNA to ribosomes). Magnesium plays a
total) [100]. The majority of the calcium that is vital role in the regulation of calcium entrance
bound to proteins is linked to albumin. Calcium into cells as well as calcium activity inside cells
in the form of ions, or free calcium, is the meta- [103, 104].
bolically active form of the mineral. Two vari- Because magnesium is a component of chlo-
ables have a significant impact on plasma calcium rophyll, green vegetables, cereals, and animal
levels: albumin levels and pH [101]. Changes in meat are all excellent suppliers of the mineral.
plasma albumin affect total calcium, regardless The amount of magnesium reabsorption in the
of whether or not ionized fractions are present. tubules is enhanced by parathyroid hormone
Laboratory technicians compute “adjusted” or (PTH). Inhibition of Mg2+ tubular reabsorption is
“corrected” calcium when plasma albumin is observed when aldosterone activity is elevated,
much higher or lower than normal to correct for but Mg2+ cellular uptake appears to be promoted
this. When examining calcium, it is critical to when thyroxine is administered. One may distin-
consider the relationship between ionized cal- guish between three different fractions of serum
cium and the acid-base state. Acidosis produces a magnesium: protein-bound, ionized (the physio-
rise in plasma ionized calcium concentrations, logically active form), and complexed with
whereas alkalosis causes a drop in plasma ion- anions such as phosphate, bicarbonate, and citrate
20 Biochemical Indices of Drug Toxicity 355

[105, 106]. Magnesium homeostasis is deter- the most important organ in the regulation of
mined by the balance between the absorption of phosphate homeostasis. A considerable change in
magnesium by the small intestine and the excre- phosphate levels is therefore thought to be the
tion of magnesium by the renal system. outcome of renal disease.

20.2.15 Phosphates 20.2.16 Iron

Phosphates, a widely distributed element, are a Iron, a trace element, in addition to DNA synthe-
key intracellular anion in mammals and serve a sis and oxygen transport, is necessary for cellular
variety of activities [107]. Phosphate may be development and defense as well as the energy
present in a variety of tissues and fluids through- generation [111]. Iron is an essential trace ele-
out the body, including plasma, extracellular ment for all of life’s main cell functions. Iron is
fluid, cell membrane structures, intracellular essential to life [112] because of its extraordinary
fluid, collagen, and bone [108]. Organic and inor- flexibility in functioning as both an electron
ganic phosphates account for approximately donor and an electron acceptor. Ferritin is formed
700 g of phosphorus per 70 kg of body weight in in the intestinal mucosa when the iron is absorbed
an adult. The majority of the phosphate in the into apoferritin and stored by the mucosal cells.
body (80–85%) is found in the bones and teeth as Alternatively, iron is transferred through the
the mineral hydroxyapatite [Ca10 (PO4)6(OH)2]. intestinal mucosa to the circulation, where it
The remainder (15–20%) is primarily found binds with transferrin. The liver contains the
within cells as organic phosphate compounds, majority of the iron reserves, with a small amount
which are toxic to cells (AMP, ADP, ATP). also present in the bone marrow and spleen [113,
Organic and inorganic phosphates are both pres- 114]. Iron concentrations are tested in serum or
ent in serum phosphate levels. Phospholipids, plasma, and it is most typically employed as a
phosphate esters, phosphoproteins, nucleic acids, marker of iron status (deficiency or excess) and
and other organic compounds contain organic inflammation. Iron has the potential to be poison-
phosphate. In the environment outside of cells, ous as well. It can accelerate the transformation
phosphate is primarily inorganic, occurring as a of hydrogen peroxide into free radicals or the for-
combination of HPO42− (80%) and H2PO4− (20%) mation of insoluble salts. A broad range of cel-
at physiological pH levels. lular structures can be damaged by free radicals,
Phosphate is a significant component of phos- which can eventually lead to the death of the cell
pholipid membranes, RNAs, nicotinamide [115, 116].
diphosphate (an enzyme cofactor), cyclic ade-
nine, guanine nucleotides (second messengers),
and phosphoproteins, and it is essential for the 20.3 Liver
intracellular metabolism of proteins, lipids, and
carbohydrates. All of the variables that promote The liver, which is positioned in the upper right
glucose absorption in the body including glucose region of the abdominal cavity behind the dia-
and fructose, alkalosis, insulin, adrenergic stimu- phragm and is responsible for the metabolism of
lation, and anabolism work together to promote almost every foreign material, is a vital organ in
glucose uptake. Many of the variables that influ- the body’s defense against infection. The most
ence blood calcium concentrations also have an prevalent symptom of drug toxicity is liver dam-
impact on serum phosphate concentrations, either age induced by drugs or chemicals [117, 118],
directly or through indirect means. As a result, which is responsible for more than half of all
laboratory readings for calcium and phosphate cases of acute liver failure [119] and is the most
should be evaluated in conjunction with one common cause of death from drug overdose
another [109, 110]. The kidney continues to be [120].
356 E. K. Ofori

Chronic hepatic damage is the major hurdle to enzyme abnormalities is interpreted in the con-
drug research, and it is also the most prevalent text of the patient’s features, it may be possible to
reason for medication recalls from the market- influence the course of subsequent diagnostic
place [120]. Drugs become more hydrophilic as a investigation.
result of biochemical activity in the hepatocyte,
which results in water-soluble molecules that are
excreted in the urine or bile [121]. Activation of 20.3.1 Aminotransferases
oxidative pathways in the liver, particularly the
cytochrome P-450 enzyme system, is required Drug-induced liver damage can be detected by
for biotransformation [122]. In the presence of elevated levels of the enzymes alanine amino-
transport proteins on the hepatocyte membrane, transferase (ALT) and aspartate aminotransferase
the hydrophilic product is exported into the (AST) [125]. Indications of hepatocellular dam-
bloodstream or bile, and it is then expelled from age include the presence of aminotransferases
the body via the kidney or the gastrointestinal (also known as transaminases), which are the
tract after undergoing a series of metabolic steps, most prevalent and specific. AST and ALT, which
which typically include conjugation to an amino were originally called serum glutamate oxaloac-
acid, a glucuronide, a sulfate, or glutathione. etate transaminase (SGOT) and serum glutamate
Assessing liver damage in basic toxicological pyruvate transaminase (SGPT), respectively,
research and toxicity testing is often done using transfer aspartate and alanine to the keto group of
serum biochemical measures, which are then ketoglutaric acids [126, 127]. The quantity of this
confirmed by histopathology after the first enzyme present in the blood is determined by an
assessment. AST test. The presence of AST in the blood is
The most common clinical patterns of liver generally insignificant. When bodily tissues or
injury in humans are intrinsic hepatocellular organs such as the heart or liver become ill or
(affecting hepatocytes), cholestatic (affecting the injured, more AST is released into the blood-
biliary system), and mixed hepatocellular/choles- stream to compensate. According to the intensity
tatic (affecting both hepatocytes and the biliary of tissue injury, the amount of AST in the blood
system) [123, 124]. Intrinsic hepatocellular is proportional to that severity. The AST test and
(affecting hepatocytes) and cholestatic (affecting the ALT test can both be conducted at the same
the biliary system) are the most common clinical time in the same lab. According to [128], the
patterns of liver injury in humans. Examples of AST/ALT ratio can indicate if the liver or another
liver injury indicators include a variety of organ has been injured. Both enzymes are highly
enzymes and peripheral proteins that are pro- active in tissues, with the liver, heart, and mus-
duced in reaction to cellular damage, as well as cles being among the most active sites. Any dam-
proteins that have undergone significant altera- age or injury to the cells of these tissues may
tion within the liver. Because many severe liver result in the release of these enzymes into the
ailments are accompanied by normal levels and circulation, boosting the activity of these enzymes
because abnormal levels can be detected in in the serum. If you are trying to figure out
asymptomatic healthy persons, a single liver whether your liver or another organ has been
function test is of limited value in the screening harmed, the AST to ALT ratio might be helpful.
for liver disease and other chronic diseases. The The significance of elevations in serum AST and
use of a battery of liver function tests, on the ALT levels is often proportional to the number of
other hand, is a very sensitive procedure. The hepatocytes that have been injured.
amount of false negatives is reduced as a result of Even though ALT is commonly located in the
this method. The use of a battery of liver tests is liver, AST may be found in a variety of other
also associated with a high level of specificity, organs as well. In preclinical research and clini-
which is especially important when a large num- cal surveillance of adverse effects, these blood
ber of tests are abnormal. When the pattern of indicators of hepatocyte damage have been used
20 Biochemical Indices of Drug Toxicity 357

for several decades. When it comes to liver injury forms of AP are found in the liver, bone, stomach,
or illness, ALT is a valid screening test, whereas placenta, and kidney, among other tissues [132].
AST is found mostly in skeletal muscle and the Predominant forms can also be found in bone tis-
heart and is most often associated with damage to sues, where they stimulate the activity of osteo-
these organs. Even though AST is not highly spe- blasts, as can be seen in broken bones. Low levels
cific, elevated levels indicate liver cell destruc- of alkaline phosphatase are caused by a variety of
tion. The levels of AST and ALT are raised in conditions including hypothyroidism, pernicious
almost all liver illnesses to some degree or anemia, and zinc deficiency [129, 134].
another. Extreme viral hepatitis, drug- or toxin-­
induced liver necrosis, and circulatory shock are
the conditions with the highest levels of amino- 20.3.4 Gamma-Glutamyl
transaminases [129, 130]. Even though enzyme Transpeptidase
levels might indicate the degree of hepatocellular
damage, they do not necessarily correspond to Gamma-glutamyl transpeptidase (GGT), also
the outcome of the test. Falling AST and ALT known as gamma-glutamyltransferase, is a pro-
values in patients with fulminant hepatic failure tein present in the cell membranes of several
may indicate either a favorable prognosis or a organs, including the kidney, bile duct, pancreas,
bleak outlook [131]. ALT grows more than AST gallbladder, spleen, heart, brain, and seminal ves-
when there is liver damage or illness, and the icles [135, 136]. Gamma-glutamyl transpepti-
quantity of ALT increase is larger than the amount dase (GGT) is a hepatobiliary damage biomarker
of AST. characterized by cholestasis and biliary repercus-
sions. Birth weight and baby GGT levels are high
during the first year of life and continue to climb
20.3.2 Cholestatic Enzymes for the next 60 years [137]. Men have greater
GGT concentrations in their serum than women
When the biliary system is physically or func- [138]. When a person has liver disease, GGT
tionally blocked, either within or outside of the activity and alkaline phosphatase levels are sig-
liver, it is referred to as cholestasis (or bile flow nificantly connected. Pharmacological agents
blockage). Cholestasis is characterized by an such as phenobarbitone, phenytoin, paracetamol,
increase in the levels of alkaline phosphatase, and tricyclic antidepressants can cause GGT to
gamma-glutamyl transferase, and bilirubin in the be elevated. Anticonvulsant medicines have been
blood. When compared to only measuring blood shown to boost GGT and AP activities in people
bilirubin levels, ALP and GGT have higher sensi- even when there is no evidence of a liver injury
tivity for identifying this abnormality in the [139]. Several illnesses, including diabetes mel-
bloodstream. However, because ALP activity is litus, acute pancreatitis, and myocardial infarc-
influenced by a wide range of diverse factors, it is tion, are associated with elevated GGT levels
not specifically designed for this use. [140, 141]. Because GGT is not found in bone,
the majority of its diagnostic applications are
limited to the exclusion of bone disease.
20.3.3 Alkaline Phosphatase

Alkaline phosphatase (AP) is a multi-isoenzyme 20.3.5 Blood Bilirubin


complex that hydrolyzes organic phosphate ester
links, releasing an organic radical and inorganic In normal red blood cell hemoglobin disintegra-
phosphate [132]. It is produced by the hydrolysis tion, bilirubin is a yellow breakdown product that
of organic phosphate ester linkages. Cholestasis is used to identify liver impairment [142, 143].
and hepatobiliary damage are the most common Heme oxygenase is a catalytic enzyme that con-
markers [133] for this condition. Different iso- verts heme to biliverdin. Biliverdin reductase
358 E. K. Ofori

then converts biliverdin to unconjugated bilirubin 20.3.6 Glutamate Dehydrogenase


(UCB), which is excreted in the urine. This sort
of bilirubin is referred to as indirect bilirubin, In the mitochondria, the enzyme glutamate dehy-
which is a phrase used to characterize it. Nonpolar drogenase (GDH) is responsible for catalyzing
and somewhat insoluble in water, UCB is a mol- the conversion of glutamate to ketoglutarate
ecule that binds to albumin and is transported to [146]. GDH is found in high concentrations in
the liver, where it is conjugated with glucuronic the liver tissue of humans and most mammalian
acid by the action of the enzyme glucuronyl species, and it is a sensitive and specific marker
transferase. The conjugation phase makes biliru- of liver disease in these animals [147]. Besides
bin more water-soluble, which makes it simpler the kidney and stomach, it may be present in a
to eliminate from the body. A substance known as variety of other human tissues including muscle
conjugated bilirubin (CB) is produced in the bile, and the salivary gland [148]. Given the fact that
which travels to the duodenum. The activity of this enzyme is situated in the mitochondria of
bacteria in the gut causes it to be converted into cells, it must be disturbed before it can be released
urobilinogen in the intestine. Urobilinogen is in significant quantities into the bloodstream. As
reabsorbed and circulated through the enterohe- a result, any significant increase in GDH levels in
patic system before being ejected by the kidneys the serum is considered an indication of hepatic
in a proportionately large amount. necrosis.
Detectable quantities of conjugated bilirubin
in the blood are only seen in patients suffering
from hepatobiliary disease [129]. Because light 20.3.7 Sorbitol Dehydrogenase
can induce changes in bilirubin levels, serum
and plasma samples must be stored in the dark Sorbitol dehydrogenase (SD) has been discov-
before being tested. However, whereas high ered in several different human and animal tis-
total bilirubin levels in the blood are an excel- sues over time. Hepatic, renal, and seminal
lent early diagnostic of cholestasis, they may vesicle mitochondria are likely to be the most
not be a particularly sensitive indicator of liver common locations for this enzyme [128]. A sen-
malfunction or illness prognosis in the long sitive enzyme marker for liver necrosis (levels are
term. As a result of hepatotoxicity, the amount generally low, but spike following acute bouts of
of urobilinogen in the urine may increase [144]. liver damage), however, it should be used in com-
Following alcoholic liver damage and hemoly- bination with other enzyme measurements such
sis, it has also been discovered that urinary uro- as ALT or other hepatic enzymes.
bilinogen levels increase. Hyperbilirubinemia
can be caused by a variety of factors, including
increased bilirubin synthesis, reduced liver 20.3.8 Plasma Proteins
absorption or conjugation, and impaired biliary
elimination [145]. The presence of urobilinogen Total proteins are made up of all of the protein
in the urine is a sensitive indicator of hepatocel- species that have been measured combined. The
lular failure. If you have this symptom, you glomerulus does not filter big or highly charged
most likely have alcoholic liver damage, cirrho- proteins, but small proteins pass readily across
sis, or malignant liver disease. When a person the glomerular barrier and are reabsorbed by the
has cholestatic jaundice, the protein urobilino- proximal tubules after passing through the glo-
gen vanishes from their urine. Ehrlich’s alde- merular barrier. Proteins are excreted in the urine
hyde reagent becomes purple when it comes as a result of damaged nephrons [149]. Among
into contact with urobilinogen. This reagent is other things, proteinuria is characterized by an
packaged in a dipstick, which allows for fast abnormally high protein excretion in the urine,
semi-qualitative testing using freshly voided which is a prognostic sign of moderate to severe
urine in a short period. kidney damage and a reliable predictor of
20 Biochemical Indices of Drug Toxicity 359

p­ rogressive renal function loss in many clinical acute and chronic liver diseases increase the
situations [150, 151]. However, the liver does not activity of these enzymes. The fact that OTC is
manufacture globulins, but it does produce the categorized as a “liver-specific” enzyme is owing
proteins as well as albumin and blood-clotting to its high concentration in hepatic tissue when
factors [152, 153]. The total protein content in compared to other tissues; nonetheless, the tech-
the blood is generally proportional to the concen- nical requirements for its testing frequently pre-
tration of albumin in the blood unless otherwise vent it from being used to its full potential.
stated. Total protein changes are often associated Malondialdehyde (MDA) is a three-carbon
with decreased synthesis (liver) or increased loss dialdehyde that is produced as a byproduct of the
(muscle) (kidney). metabolism of fatty acids [160]. MDA is a bio-
Albumin is the most abundant protein in marker for oxidative stress and lipid peroxidation
plasma produced by the liver in terms of amount, in general [161], as well as for lipid peroxidation
and it may be used as a biomarker of hepatic in particular. It has been shown that lipid peroxi-
function to monitor liver function. Albumin in dation plays a role in the pathophysiology of sev-
serum has a half-life of around 21 days, which is eral different types of tissue injuries, including
rather short. Patients suffering from cirrhosis and tissue damage produced by a range of toxic sub-
ascites frequently have reduced serum albumin stances. Because MDA is produced as a result of
levels [154]. Because albumin detects both glo- lipid peroxidation, it can be used as a biomarker
merular and tubular damage, it is the protein that to detect cell membrane damage. It has been
is most likely to be detected at high levels early in demonstrated that there is a possible relationship
a variety of renal diseases. In addition to nutri- between kidney lipid peroxidation levels and
tional conditions, hormonal balance, and osmotic renal damage [162].
pressure [155], albumin production is controlled
by a range of other variables as well.
Ceruloplasmin, an acute-phase protein produced 20.3.10 Other Non-enzymes
by the liver, is another product of the liver. It has of Toxicological Relevance
been shown that infections, rheumatoid arthritis,
pregnancy, and obstructive jaundice can result in 20.3.10.1 Triglycerides
a rise in plasma concentration [156]. A low Triglycerides (TGs), also known as neutral fats,
plasma level of ceruloplasmin is usually observed triacylglycerols, or triacylglycerides, are lipids
in patients with Wilson’s sickness, and this is a that are composed of three long-chain fatty acids
critical diagnostic indicator [157]. Low levels of esterified to a glycerol. Triglycerides (TGs) are a
ceruloplasmin can be caused by a variety of con- type of lipid that is found in a variety of foods.
ditions including neonates, kwashiorkor, maras- Exogenous (chylomicrons) and endogenous (pre-­
mus, protein-losing enteropathy, copper lipoproteins) transport of these substances to tar-
deficiency, and aceruloplasminemia [129, 158]. get cells has been demonstrated [163]. While
endogenous triglycerides are synthesized and
stored in the liver, exogenous triglycerides are
20.3.9 Other Enzymes obtained from the food [164]. TGs are the body’s
of Toxicological Relevance principal source of energy and also serve as the
human body’s primary and most dependable
In addition to the enzymes indicated above, energy reserves, according to the World Health
numerous more enzymes are used as markers to Organization. It has been shown that TG plays a
identify drug-induced damage in addition to the role in metabolic pathways that regulate the rate
enzymes stated above. Among the enzymes of fatty acid oxidation as well as the fate of lipo-
involved in the urea cycle, ornithine transcarba- proteins in the body [165]. When TG is stored in
mylase (OTC) is present almost exclusively in the cytoplasm of a cell (e.g., muscle), it is
the mitochondria of animal liver cells [159]. Both ­surrounded by a monolayer of phospholipids and
360 E. K. Ofori

hydrophobic proteins, which is known as the TG vated because they are engaged in a wide number
storage complex [166]. of signaling pathways. Cholesterol catabolism
Only trace levels of triglycerides are normally and elimination, as well as regulation of pancre-
allowed in the blood. To establish whether or not atic secretions and the release of gastrointestinal
there are high triglyceride levels in the blood, the peptides [176], are all functions of bile acids. The
serum triglyceride concentration is determined small intestine is also aided in the digestion and
[167]. Increased serum TG levels produce an absorption of dietary fat (and, indirectly, fat-­
increase in blood viscosity as well as platelet soluble vitamins). In this way, bile acid concen-
aggregation, which results in a reduction in vas- trations can be employed as a valid biomarker of
cular flow. An increase in total lipid (TG) levels hepatobiliary function in the laboratory setting.
in the blood is usually coupled with a decrease in
HDL cholesterol levels in diabetes patients, and 20.3.10.4 Glucose
both are risk factors for cardiovascular disease Glucose is the most important simple sugar
(CAD [168]). According to toxicity studies [169], (monosaccharide) in mammalian metabolism
there may be a variety of associated lipoprotein because it is the most readily available. It is a car-
metabolism abnormalities. bohydrate molecule with six carbon atoms.
Glucose is a critical component of cellular respi-
20.3.10.2 Cholesterol ration and one of the primary products of photo-
Cholesterol, a steroid oil hydrocarbon with a synthesis, and it is produced in large quantities by
four-link structure, is a substance that plays an plants. Cells employ glucose as an energy source
essential role in the function of membranes and and as a metabolic intermediate supply material
the metabolism of lipids [170]. In addition to for a wide range of biosynthetic actions [177].
being a precursor to steroid hormone biosynthe- There are several pathways involved in the
sis (including glucocorticoids, estrogens, proges- metabolism of glucose in the body. These include
terone, androgens, and mineralocorticoids), glycolysis, gluconeogenesis, glycogenolysis,
cholesterol is also a bile acid and vitamin D pro- glycogenesis, the pentose-phosphate pathway,
ducer [171]. Aside from these functions, choles- and the citric acid cycle [178, 179]. The primary
terol is also involved in signaling and sperm purpose of testing blood glucose levels is to
production [172]. Lipoproteins, which are pro- determine how well the body is handling carbo-
tein bundles, are responsible for transporting hydrates. Normal plasma glucose levels can only
cholesterol through the body. A lipid profile be maintained by maintaining a perfect match
includes measurements of total cholesterol, LDL between glucose consumption and endogenous
cholesterol, HDL cholesterol, and triglycerides, glucose production or dietary glucose supply (or
among other things. Chronic hypercholesterol- both).
emia (hyperlipidemia) is a primary cause of coro-
nary artery disease and stroke [173]. 20.3.10.5 Cytokines
The use of blood cytokines as toxicity markers
20.3.10.3 Bile Acids has lately gained prominence [180, 181, 183].
Bile acids are the byproducts of cholesterol The use of cytokines as biomarkers, on the other
metabolism in animals, and their principal func- hand, is problematic because of their short circu-
tions in the intestines are to act as powerful deter- lation half-life, low baseline levels, and lack of
gents or emulsifying agents to aid in the digestion tissue-specific expressions. Interleukin-18 (IL-­
and absorption of dietary fats [174, 175]. They 18) is a pro-inflammatory cytokine that is gener-
are produced by the liver, and they are stored in ated by macrophages and other cells and belongs
the gallbladder. Feeding causes the gallbladder to to the IL-1 superfamily, according to the National
contract, allowing bile acids to pass into the gut Institutes of Health [182]. Plasma IL-18 levels
through the digestive system. When there is liver are elevated in patients suffering from
damage or dysfunction, total bile acids are ele- ­inflammatory arthritis, inflammatory bowel dis-
20 Biochemical Indices of Drug Toxicity 361

ease (IBD), systemic lupus erythematosus (SLE), creatine kinase (CK), an enzyme that leaks into
psoriasis, hepatitis, and multiple sclerosis (MS) the bloodstream. Lactate dehydrogenase (LDH)
[183–185]. Furthermore, in individuals with is a tetrameric protein that catalyzes the revers-
acute renal damage, urine interleukin-18 (IL-18) ible conversion of pyruvate to lactate in the pres-
levels are significantly raised before increases in ence of oxygen [192]. LDH is an intracellular
serum creatinine levels [186, 187]. Although this enzyme that is widely distributed throughout the
IL-18 appears to be a promising potential bio- body and is particularly abundant in tissues that
marker in the context of AKI, its pro-­inflammatory utilize glucose for energy [193] such as the skel-
properties and upregulation machinery under etal and brain muscles, cardiac muscle, kidneys,
inflammatory settings may limit its sensitivity and liver [194]. As a result of this distribution, a
and specificity when used in clinical rise in LDH can cause damage to a wide range of
applications. tissues as a result of its elevation. The majority of
Inflammatory peptide C-reactive peptide LDH isoenzymes are distributed in a tissue-­
(CRP) is an acute protein that has been proposed specific manner.
as a biochemical biomarker for the risk of coro- Troponins are the protein filaments that are
nary heart disease. CRP levels in the blood are responsible for contractile muscle contraction in
used to demonstrate the presence of active inflam- the heart and skeletal muscles [194]. Troponin
matory cells in plaques [188, 189]. T’s role is to provide a connection between the
troponin complex and the tropomyosin strand of
the actin thin filament. Troponin I suppresses the
20.4 Muscle activity of the actinomycin ATPase enzyme.
Troponin I is found in a variety of plants.
It is becoming increasingly usual to encounter Troponin C regulates contraction by forming a
muscle toxicity as a concern in drug develop- complex with four calcium ions. Cardiomyocyte
ment, and research into methods to predict skel- troponins I and T have amino acid sequences that
etal muscle injury is becoming more prevalent. differ from those found in skeletal muscle, and
Toxicologically significant are traditional muscle these differences can be used to distinguish
injury signs such as elevations in aminotransfer- between the two tissue types [195, 196]. An
ase (AST), creatine kinase (CK), and lactate increase in cardiac troponin levels in the blood
dehydrogenase (LDH) [190]. These conventional might indicate the presence of heart illness, the
identifiers are not without their downsides, most frequent of which is myocardial infarction
though (specificity and sensitivity). Several pro- [196]. As a result of the release of the enzyme
teins, including skeletal troponin I and T, creatine into the circulation when the heart is wounded,
kinase protein M, myosin light chain 3, fatty increased troponin levels improve diagnostic sen-
acid-binding protein 3, aldolase A, and myoglo- sitivity and specificity in identifying cardiac
bin, are sensitive and specific indicators of drug-­ muscle cell death in the patient.
induced skeletal muscle injury [190, 191] and During times of ventricular stress, cardiomyo-
have shown promise as sensitive and specific cytes secrete the peptide hormone B-type natri-
indicators of drug-induced skeletal muscle injury. uretic protein (BNP), which has anti-arrhythmic
properties [197]. BNP (brain-derived neuro-
trophic factor) is a cardiac-specific hormone that
20.5 Cardiac detects mechanical stress inside the myocardium
[198]. It has been proposed as a biomarker to
The list of indicators used to measure cardiotox- detect cardiac pressure and volume overload
icity includes markers for structural and func- when pathological symptoms are present. BNP is
tional alterations, as well as markers for oxidative elevated when these symptoms are present. It is a
stress. Inflammatory or recent muscular injury or powerful “negative predictor,” since a low result
damage is indicated by elevated blood levels of implies that the volume parameters are within
362 E. K. Ofori

normal limits. Increased BNP levels are an inde- spectrometry (MS) are powerful analytical meth-
pendent predictor of death in patients suffering ods for generating multivariate metabolic data.
from acute coronary syndromes [199, 200]. Because it is non-destructive and acceptable for
intact biomaterials, nuclear magnetic resonance
(NMR) has the benefit of providing intrinsically
20.6 Recent Developments more information-rich results in terms of molec-
and Future Perspectives ular structure identification. Even though MS is
more analytically sensitive than NMR, it requires
Currently, available early toxicity indicators are the use of extraction and derivatization processes
ineffective in effectively addressing pharmaceu- before the experiment. The successful applica-
tical toxicity concerns. Molecular epidemiology tion of NMR- and MS-based metabonomics in
and genomic studies have received a great deal toxicological and clinical research will aid us in
of attention in recent decades to identify specific our quest to get a better understanding of phar-
biomarkers for nephrotoxicity and hepatotoxic- maceutical toxicity.
ity, as well as for learning more about the under-
lying mechanisms of various kidney and liver
insults [201]. It will be easier to gain a compre- 20.7 Conclusion
hensive understanding of how toxic insults
affect biological systems with the introduction Variations in serum biochemical parameters that
of these “omic” technologies (genomics, pro- occur during toxicity have been discussed. The
teomics, metabolomics, and cytomics) in the working state of important organ systems such as
future, which will lead to more accurate toxicity the liver, renal, cardiac, hematopoietic, and
prediction models that can be used in drug immunological systems may be determined using
development, as well as more effective drug biochemical markers. In toxicological research,
development. When it comes to biomarker dis- enzymes are frequently used to identify and eval-
covery and characterization, one of the more uate cell damage, and they are also used to assess
recent breakthroughs in the quest for biomark- the severity of the damage. The use of biomarkers
ers has been the detection of plasma microR- in drug development, illness, and monitoring of
NAs (miRNAs), which are small RNAs that are favorable effects of therapeutic interventions
found in the plasma of healthy people. is beneficial in a number of ways. It is also vital
Endogenous RNA molecules of 22 nucleotides to conduct research and discover early and sensi-
in length, microRNAs are produced in the tive biomarkers that will enable the creation of
nucleus and processed in the cytoplasm before prompt detection of toxicity. Several emerging
being recruited to silencing complexes, where biomarkers, including urine protein biomarkers,
they block the translation of certain target microRNAs, proteomics, metabolomics, and tar-
mRNA transcripts. DNA microarrays allow for geted mass spectrometry assays, have the poten-
the simultaneous monitoring of the expression tial to exceed traditional indicators in mammalian
of hundreds or thousands of genes using a single models in terms of sensitivity and predictive
sample of DNA. A great deal of work has been capacity.
made in understanding the beneficial effects of
microRNAs in cancer, cardiovascular disease,
and other organ damage illnesses. References
Analytical techniques such as nuclear mag-
netic resonance (NMR), gas chromatography, 1. Aronson JK, Ferner RE. Biomarkers—a
general review. Curr Protoc Pharmacol.
high-performance liquid chromatography 2017;76(1):9.23.1–9.23.17.
(HPLC), and mass spectrometry (MS) can be 2. Gil F, Hernández AF. Significance of biochemical
employed to investigate metabolite expression markers in applied toxicology. In: General, applied
patterns in biological matrices. NMR and mass and systems toxicology. Wiley; 2009.
20 Biochemical Indices of Drug Toxicity 363

3. Zhao X, et al. Biomarkers in pharmaceutical 22. Kashani K, Cheungpasitporn W, Ronco


research. Clin Chem. 2015;61(11):1343–53. C. Biomarkers of acute kidney injury: the pathway
4. Califf RM. Biomarker definitions, and their applica- from discovery to clinical adoption. Clin Chem Lab
tions. Exp Biol Med. 2018;243(3):213–21. Med. 2017;55(8):1074–89.
5. Davis KD, et al. Discovery and validation of bio- 23. Lin C-H, Chang Y-C, Chuang L-M. Early detec-
markers to aid the development of safe and effective tion of diabetic kidney disease: present limita-
pain therapeutics: challenges and opportunities. Nat tions and future perspectives. World J Diabetes.
Rev Neurol. 2020;16(7):381–400. 2016;7(14):290.
6. Strimbu K, Tavel JA. What are biomarkers? Curr 24. Andrews R, et al. The effect of dietary creatine sup-
Opin HIV AIDS. 2010;5(6):463. plementation on skeletal muscle metabolism in con-
7. Issa NT, et al. Drug metabolism in preclinical drug gestive heart failure. Eur Heart J. 1998;19(4):617–22.
development: a survey of the discovery process, tox- 25. Thongprayoon C, Cheungpasitporn W, Kashani
icology, and computational tools. Curr Drug Metab. K. Serum creatinine level, a surrogate of muscle
2017;18(6):556–65. mass, predicts mortality in critically ill patients. J
8. Zhang Z, Tang W. Drug metabolism in drug dis- Thorac Dis. 2016;8(5):E305.
covery and development. Acta Pharm Sin B. 26. Wyss M, Kaddurah-Daouk R. Creatine and creatinine
2018;8(5):721–32. metabolism. Physiol Rev. 2000;80(3):1107–213.
9. Dzoyem JP, Kuete V, Eloff JN. Biochemical param- 27. Kampmann J, Hansen JM. Glomerular filtration
eters in toxicological studies in Africa: significance, rate and creatinine clearance. Br J Clin Pharmacol.
the principle of methods, data interpretation, and 1981;12(1):7.
use in plant screenings. In: Toxicological sur- 28. Schwartz GJ, Work DF. Measurement and estima-
vey of African medicinal plants. Elsevier; 2014. tion of GFR in children and adolescents. Clin J Am
p. 659–715. Soc Nephrol. 2009;4(11):1832–43.
10. Ebubekir B, Nurinnisa O, Nurcan K-B. Automation 29. Samra M, Abcar AC. False estimates of elevated cre-
in the clinical laboratory: integration of several ana- atinine. Perm J. 2012;16(2):51.
lytical and interlaboratory pre-and post-analytical 30. Odden MC, Shlipak MG, Tager IB. Serum creati-
systems. Turk J Biochem. 2017;42(1):1–13. nine and functional limitation in elderly persons. J
11. Hawker CD. Laboratory automation: total and sub- Gerontol A Biomed Sci Med Sci. 2009;64(3):370–6.
total. Clin Lab Med. 2007;27(4):749–70. 31. Kim S-W, et al. A new equation to estimate muscle
12. Bajaj P, et al. Emerging kidney models to investigate mass from creatinine and cystatin C. PLoS One.
metabolism, transport, and toxicity of drugs and xeno- 2016;11(2):e0148495.
biotics. Drug Metab Dispos. 2018;46(11):1692–702. 32. Knight EL, et al. Factors influencing serum cys-
13. Fuchs TC, Hewitt P. Biomarkers for drug-induced tatin C levels other than renal function and the
renal damage and nephrotoxicity—an overview for impact on renal function measurement. Kidney Int.
applied toxicology. AAPS J. 2011;13(4):615–31. 2004;65(4):1416–21.
14. Braun J-P, Lefebvre HP. Kidney function and dam- 33. Cirillo M, Anastasio P, De Santo NG. Relationship
age. In: Clinical biochemistry of domestic animals. of gender, age, and body mass index to errors in
6th ed. Amsterdam: Elsevier; 2008. p. 485–528. predicted kidney function. Nephrol Dial Transplant.
15. Perazella MA. Pharmacology behind common 2005;20(9):1791–8.
drug nephrotoxicities. Clin J Am Soc Nephrol. 34. Cakmak U, et al. Effects of acute kidney injury on
2018;13(12):1897–908. clinical outcomes in patients with upper gastrointes-
16. Schnellmann RG. Toxic responses of the kidney. In: tinal bleeding. Ren Fail. 2016;38(2):176–84.
Casarett and Doull’s toxicology: the basic science 35. Rockey DC. Gastrointestinal bleeding. Gastroenterol
of poisons, vol. 1. New York: McGraw-Hill; 2008. Clin. 2005;34(4):581–8.
p. 583–7. 36. Weiner ID, Mitch WE, Sands JM. Urea and ammonia
17. Pazhayattil GS, Shirali AC. Drug-induced impair- metabolism and the control of renal nitrogen excre-
ment of renal function. Int J Nephrol Renov Dis. tion. Clin J Am Soc Nephrol. 2015;10(8):1444–58.
2014;7:457. 37. Stewart GS, Smith CP. Urea nitrogen sal-
18. Campion S, et al. The current status of biomarkers vage mechanisms and their relevance to rumi-
for predicting toxicity. Expert Opin Drug Metab nants, non-­ ruminants, and man. Nutr Res Rev.
Toxicol. 2013;9(11):1391–408. 2005;18(1):49–62.
19. Han W, et al. Urinary biomarkers in the early 38. Duarte CG, Preuss HG. Assessment of renal func-
diagnosis of acute kidney injury. Kidney Int. tion—glomerular and tubular. Clin Lab Med.
2008;73(7):863–9. 1993;13(1):33–52.
20. Bonventre JV, Yang L. Cellular pathophysiol- 39. Lyman JL. Blood urea nitrogen and creatinine.
ogy of ischemic acute kidney injury. J Clin Invest. Emerg Med Clin North Am. 1986;4(2):223–33.
2011;121(11):4210–21. 40. Meyer TW, Hostetter TH. Uremia. N Engl J Med.
21. Abdulkader RC, et al. Aging and decreased glo- 2007;357(13):1316–25.
merular filtration rate: an elderly population-based 41. Ljungman S, Laragh JH, Cody RJ. Role of the kidney
study. PLoS One. 2017;12(12):e0189935. in congestive heart failure. Drugs. 1990;39(4):10–21.
364 E. K. Ofori

42. Parrinello G, et al. Blood urea nitrogen to creatinine 61. Moresco RN, et al. Urinary kidney injury molecule-
ratio is associated with congestion and mortality in ­1 in renal disease. Clin Chim Acta. 2018;487:15–21.
heart failure patients with renal dysfunction. Intern 62. Bonventre JV. Kidney injury molecule-1: a trans-
Emerg Med. 2015;10(8):965–72. lational journey. Trans Am Clin Climatol Assoc.
43. Muzykovsky K, Sarosky KM. Acute kidney injury. 2014;125:293.
In: Primary care: an interprofessional perspective. 63. Vaidya VS, et al. Kidney injury molecule-1 out-
Springer; 2014. p. 413. performs traditional biomarkers of kidney injury
44. Kaufman DP, Basit H, Knohl SJ. Physiology, glomer- in preclinical biomarker qualification studies. Nat
ular filtration rate (GFR). In: StatPearls [Internet]. Biotechnol. 2010;28(5):478–85.
Treasure Island: StatPearls Publishing; 2018. 64. Singer E, et al. Neutrophil gelatinase-associated
45. Gaspari F, Perico N, Remuzzi G. Measurement lipocalin: pathophysiology and clinical applications.
of glomerular filtration rate. Kidney Int Suppl. Acta Physiol. 2013;207(4):663–72.
1997;63:S151–4. 65. Polignano D, et al. Neutrophil gelatinase-associated
46. Perrone RD, Madias NE, Levey AS. Serum creati- lipocalin (NGAL) as a marker of kidney damage.
nine as an index of renal function: new insights into Am J Kidney Dis. 2008;52(3):595–605.
old concepts. Clin Chem. 1992;38(10):1933–53. 66. Soni SS, et al. NGAL: a biomarker of acute kid-
47. Pickering JW, Endre ZH. Challenges facing early ney injury and other systemic conditions. Int Urol
detection of acute kidney injury in the critically ill. Nephrol. 2010;42(1):141–50.
World J Crit Care Med. 2012;1(3):61. 67. Devarajan P. Neutrophil gelatinase-associated lipo-
48. Sandilands EA, et al. Measurement of renal function calin: a promising biomarker for human acute kid-
in patients with chronic kidney disease. Br J Clin ney injury. Biomark Med. 2010;4(2):265–80.
Pharmacol. 2013;76(4):504–15. 68. Cruz DN, Ronco C, Katz N. Neutrophil
49. Onopiuk A, Tokarzewicz A, Gorodkiewicz gelatinase-­associated lipocalin: a promising bio-
E. Cystatin C: a kidney function biomarker. Adv marker for detecting cardiac surgery-associated
Clin Chem. 2015;68:57–69. acute kidney injury. J Thorac Cardiovasc Surg.
50. Inker LA, et al. Estimating glomerular filtration rate 2010;139(5):1101–6.
from serum creatinine and cystatin C. N Engl J Med. 69. Hall IE, et al. IL-18, and urinary NGAL predict dial-
2012;367(1):20–9. ysis and graft recovery after kidney transplantation.
51. Taglieri N, Koenig W, Kaski JC. Cystatin C and car- J Am Soc Nephrol. 2010;21(1):189–97.
diovascular risk. Clin Chem. 2009;55(11):1932–43. 70. Parikh C, et al. Urine NGAL, and IL-18 are pre-
52. Krolewski AS, et al. Serum concentration of cystatin dictive biomarkers for delayed graft function fol-
C and risk of end-stage renal disease in diabetes. lowing kidney transplantation. Am J Transplant.
Diabetes Care. 2012;35(11):2311–6. 2006;6(7):1639–45.
53. Dieterle F, et al. Urinary clusterin, cystatin C, 71. Ali RJ, Al-Obaidi FH, Arif HS. The role of urinary
β2-microglobulin and total protein as markers to N-acetyl beta-D-glucosaminidase in children with
detect drug-induced kidney injury. Nat Biotechnol. urological problems. Oman Med J. 2014;29(4):285.
2010;28(5):463–9. 72. Liangos O, et al. Urinary N-acetyl-β-(D)-
54. Budzynski AZ, Shainoff JR. Fibrinogen and fibrin: glucosaminidase activity and kidney injury
biochemistry and pathophysiology. Crit Rev Oncol molecule-1 level are associated with adverse out-
Hematol. 1986;6(2):97–146. comes in acute renal failure. J Am Soc Nephrol.
55. Charlie-Silva I, et al. Acute-phase proteins during 2007;18(3):904–12.
the inflammatory reaction by bacterial infection: 73. Obermüller N, et al. Current developments in early
fish-model. Sci Rep. 2019;9(1):1–13. diagnosis of acute kidney injury. Int Urol Nephrol.
56. Luyendyk JP, Schoenecker JG, Flick MJ. The multi- 2014;46(1):1–7.
faceted role of fibrinogen in tissue injury and inflam- 74. Vaidya VS, Ferguson MA, Bonventre JV. Biomarkers
mation. Blood. 2019;133(6):511–20. of acute kidney injury. Annu Rev Pharmacol Toxicol.
57. Hoffmann D, et al. Fibrinogen excretion in the urine 2008;48:463–93.
and immunoreactivity in the kidney serves as a 75. Nomura T, et al. β2-Microglobulin-mediated signal-
translational biomarker for acute kidney injury. Am ing as a target for cancer therapy. Anticancer Agents
J Pathol. 2012;181(3):818–28. Med Chem. 2014;14(3):343–52.
58. Bonventre JV. Kidney injury molecule-1 (KIM-­ 76. Karlsson F, Wibell L, Evrin P. beta 2-Microglobulin
1): a urinary biomarker and much more. Oxford in clinical medicine. Scand J Clin Lab Invest Suppl.
University Press; 2009. p. 3265–8. 1980;154:27–37.
59. Ichimura T, et al. Kidney injury molecule-1 (KIM-­ 77. Bethea M, Forman D. Beta 2-microglobulin: its sig-
1), a putative epithelial cell adhesion molecule nificance and clinical usefulness. Ann Clin Lab Sci.
containing a novel immunoglobulin domain, is up-­ 1990;20(3):163–8.
regulated in renal cells after injury. J Biol Chem. 78. Gatanaga H, et al. Urinary β 2-microglobulin as a
1998;273(7):4135–42. possible sensitive marker for renal injury caused
60. Yin C, Wang N. Kidney injury molecule-1 in kidney by tenofovir disoproxil fumarate. AIDS Res Hum
disease. Ren Fail. 2016;38(10):1567–73. Retroviruses. 2006;22(8):744–8.
20 Biochemical Indices of Drug Toxicity 365

79. George B, Joy MS, Aleksunes LM. Urinary pro- 100. Shaker JL, Deftos L. Calcium and phosphate homeo-
tein biomarkers of kidney injury in patients stasis. In: Endotext [Internet]. South Dartmouth:
receiving cisplatin chemotherapy. Exp Biol Med. MDText.com, Inc; 2018.
2018;243(3):272–82. 101. Agnes F, et al. Ionized calcium in calf serum: relation
80. Pourfridoni M, et al. Fluid and electrolyte dis- to total serum calcium, albumin, total protein, and
turbances in COVID-19 and their complications. pH. J Veterinary Med Ser A. 1993;40(1–10):605–8.
Biomed Res Int. 2021;2021:6667047. 102. Jeon US. Kidney and calcium homeostasis.
81. Langston C. Managing fluid and electrolyte disor- Electrolyte Blood Press. 2008;6(2):68–76.
ders in kidney disease. Vet Clin North Am Small 103. Pasternak K, Kocot J, Horecka A. Biochemistry of
Anim Pract. 2017;47(2):471–90. magnesium. J Elem. 2010;15(3):601–16.
82. Dhondup T, Qian Q. Acid-base and electrolyte 104. Blaine J, Chonchol M, Levi M. Renal control of cal-
disorders in patients with and without chronic cium, phosphate, and magnesium homeostasis. Clin
kidney disease: an update. Kidney Dis (Basel). J Am Soc Nephrol. 2015;10(7):1257–72.
2017;3(4):136–48. 105. Touyz RM. Magnesium in clinical medicine. Front
83. Pohl HR, Wheeler JS, Murray HE. Sodium and Biosci. 2004;9(1–3):1278–93.
potassium in health and disease. In: Interrelations 106. Jahnen-Dechent W, Ketteler M. Magnesium basics.
between essential metal ions and human diseases. Clin Kidney J. 2012;5(Suppl_1):i3–i14.
Dordrecht: Springer Netherlands; 2013. p. 29–47. 107. Bansal VK. Serum inorganic phosphorus. In:
84. Strazzullo P, Leclercq C. Sodium. Adv Nutr. Clinical methods: the history, physical, and labora-
2014;5(2):188–90. tory examinations. 3rd ed. Boston: Butterworths;
85. Subramanya AR, Ellison DH. Distal convoluted 1990.
tubule. Clin J Am Soc Nephrol. 2014;9(12):2147–63. 108. Anderson JJ, Garner SC. Calcium and phosphorus in
86. McCormick JA, Ellison DH. The distal convoluted health and disease, vol. 10. CRC Press; 1995.
tubule. Compr Physiol. 2015;5(1):45. 109. Lederer E. Regulation of serum phosphate. J Physiol.
87. Balogun RA, Okusa MD. Fractional excretion of 2014;592(18):3985–95.
sodium, urea, and other molecules in acute kidney 110. Penido MGM, Alon US. Phosphate homeosta-
injury. In: UpToDate. Wolters Kluwer Health; 2020. sis and its role in bone health. Pediatr Nephrol.
88. Ackerman GL. Serum sodium. In: Clinical methods: 2012;27(11):2039–48.
the history, physical, and laboratory examinations. 111. Abbaspour N, Hurrell R, Kelishadi R. Review on
3rd ed. Boston: Butterworths; 1990. iron and its importance for human health. J Res Med
89. van den Bosch JJ, et al. Plasma sodium, extracellu- Sci. 2014;19(2):164.
lar fluid volume, and blood pressure in healthy men. 112. Oliveira F, Rocha S, Fernandes R. Iron metabo-
Physiol Rep. 2021;9(24):e15103. lism: from health to disease. J Clin Lab Anal.
90. George B, et al. Xenobiotic transporters, and kidney 2014;28(3):210–8.
injury. Adv Drug Deliv Rev. 2017;116:73–91. 113. Saito H. Metabolism of iron stores. Nagoya J Med
91. Kowey PR. The role of potassium. In: Women’s Sci. 2014;76(3–4):235.
health and menopause. Springer; 2002. p. 151–7. 114. Muñoz M, García-Erce JA, Remacha ÁF. Disorders
92. Udensi UK, Tchounwou PB. Potassium homeosta- of iron metabolism. Part II: iron deficiency and iron
sis, oxidative stress, and human disease. Int J Clin overload. J Clin Pathol. 2011;64(4):287–96.
Exp Physiol. 2017;4(3):111. 115. Valko M, et al. Free radicals and antioxidants in nor-
93. Bhave G, Neilson EG. Body fluid dynamics: back to mal physiological functions and human disease. Int
the future. J Am Soc Nephrol. 2011;22(12):2166–81. J Biochem Cell Biol. 2007;39(1):44–84.
94. Tobias A, Ballard BD, Mohiuddin SS. Physiology, 116. Bhattacharya S. Reactive oxygen species and cellu-
water balance. In: StatPearls [Internet]. Treasure lar defense system. In: Free radicals in human health
Island: StatPearls Publishing; 2019. and disease. Springer; 2015. p. 17–29.
95. Pirahanchi Y, Aeddula NR. Physiology, sodium-­ 117. Suk KT, Kim DJ. Drug-induced liver injury: present
potassium pump (Na+ K+ pump). In: StatPearls and future. Clin Mol Hepatol. 2012;18(3):249.
[Internet]. Treasure Island: StatPearls Publishing; 118. Kaplowitz N. Drug-induced liver injury. Clin Infect
2019. Dis. 2004;38(Supplement_2):S44–8.
96. Morrison G. Serum chloride. In: Clinical methods: 119. Lee WM. Drug-induced hepatotoxicity. N Engl J
the history, physical, and laboratory examinations. Med. 2003;349(5):474–85.
3rd ed. Boston: Butterworths; 1990. 120. Regev A. Drug-induced liver injury and drug devel-
97. Beto JA. The role of calcium in human aging. Clin opment: an industry perspective. In: Seminars in
Nutr Res. 2015;4(1):1–8. liver disease. Thieme Medical Publishers; 2014.
98. Brini M, et al. Calcium in health and disease. 121. Pandit A, Sachdeva T, Bafna P. Drug-induced
In: Interrelations between essential metal ions hepatotoxicity: a review. J Appl Pharm Sci.
and human diseases. Dordrecht: Springer; 2013. 2012;2(5):233–43.
p. 81–137. 122. Buhler DR, Williams DE. The role of biotransfor-
99. Minisola S, et al. The diagnosis and management of mation in the toxicity of chemicals. Aquat Toxicol.
hypercalcemia. BMJ. 2015;350:h2723. 1988;11(1–2):19–28.
366 E. K. Ofori

123. Francis P, Navarro VJ. Drug-induced hepatotoxicity. 144. Gulati K, et al. Hepatotoxicity: Its mechanisms,
In: StatPearls [Internet]. Treasure Island: StatPearls experimental evaluation, and protective strategies.
Publishing; 2021. Am J Pharmacol. 2018;1(1):1004.
124. Bashir A, et al. Liver toxicity. In: StatPearls 145. Fevery J. Bilirubin in clinical practice: a review.
[Internet]. Treasure Island: StatPearls Publishing; Liver Int. 2008;28(5):592–605.
2021. 146. Miñambres B, et al. A new class of gluta-
125. Shi Q, et al. Biomarkers for drug-induced liver mate dehydrogenases (GDH). J Biol Chem.
injury. Expert Rev Gastroenterol Hepatol. 2000;275(50):39529–42.
2010;4(2):225–34. 147. Jaeschke H, McGill MR. Serum glutamate dehydro-
126. McGill MR. The past and present of serum amino- genase—a biomarker for liver cell death or mitochon-
transferases and the future of liver injury biomark- drial dysfunction? Toxicol Sci. 2013;134(1):221–2.
ers. EXCLI J. 2016;15:817. 148. Plaitakis A, et al. The glutamate dehydrogenase
127. Hannig C, Spitzmüller B, Hannig M. Transaminases pathway and its roles in cell and tissue biology in
in the acquired pellicle. Arch Oral Biol. health and disease. Biology. 2017;6(1):11.
2009;54(5):445–8. 149. Gorriz JL, Martinez-Castelao A. Proteinuria: detec-
128. Ozer J, et al. The current state of serum biomarkers of tion and role in native renal disease progression.
hepatotoxicity. Toxicology. 2008;245(3):194–205. Transplant Rev. 2012;26(1):3–13.
129. Thapa B, Walia A. Liver function tests and their 150. Cravedi P, Remuzzi G. Pathophysiology of pro-
interpretation. Indian J Pediatr. 2007;74(7):663–71. teinuria and its value as an outcome measure in
130. Losser M-R, Payen D. Mechanisms of liver dam- chronic kidney disease. Br J Clin Pharmacol.
age. In: Seminars in liver disease. Thieme Medical 2013;76(4):516–23.
Publishers, Inc; 1996. 151. Cravedi P, Ruggenenti P, Remuzzi G. Proteinuria
131. Sunheimer R, et al. Serum analyte pattern character- should be used as a surrogate in CKD. Nat Rev
istic of fulminant hepatic failure. Ann Clin Lab Sci. Nephrol. 2012;8(5):301–6.
1994;24(2):101–9. 152. Ozougwu JC. Physiology of the liver. Int J Res
132. Sharma U, Pal D, Prasad R. Alkaline phos- Pharm Biosci. 2017;4(8):13–24.
phatase: an overview. Indian J Clin Biochem. 153. Kalra A, et al. Physiology, liver. In: StatPearls
2014;29(3):269–78. [Internet]. Treasure Island: StatPearls Publishing;
133. Woreta TA, Alqahtani SA. Evaluation of abnormal 2018.
liver tests. Med Clin North Am. 2014;98(1):1–16. 154. Bernardi M, Maggioli C, Zaccherini G. Human albu-
134. Lowe D, Sanvictores T, John S. Alkaline phos- min in the management of complications of liver cir-
phatase. In: StatPearls [Internet]. Treasure Island: rhosis. In: Ann Update Intensive Care Emerg Med,
StatPearls Publishing; 2017. vol. 2012; 2012. p. 421–30.
135. Center SA. Interpretation of liver enzymes. Vet Clin 155. Runström G, Mann A, Tighe B. The fall and rise of
N Am Small Anim Pract. 2007;37(2):297–333. tear albumin levels: a multifactorial phenomenon.
136. Dillon JF, Miller MH. Gamma-glutamyl transferase Ocul Surf. 2013;11(3):165–80.
“To be or not to be” liver function test? London: 156. Gowda S, et al. A review on laboratory liver function
SAGE Publications; 2016. p. 629–31. tests. Pan Afr Med J. 2009;3:17.
137. Loh TP, Metz MP. Trends and physiology of com- 157. Członkowska A, et al. Wilson disease. Nat Rev Dis
mon serum biochemistries in children aged 0–18 Primers. 2018;4(1):1–20.
years. Pathology. 2015;47(5):452–61. 158. Matreja PS, Singh V. An overview on liver function
138. Ha KH, et al. Gender differences in the association test. Academica. 2021;11(10):2199–204.
between serum γ-glutamyltransferase and blood pres- 159. Haskins N, et al. Mitochondrial enzymes of the urea
sure change: a prospective community-based cohort cycle cluster at the inner mitochondrial membrane.
study. J Korean Med Sci. 2014;29(10):1379–84. Front Physiol. 2021;11:1753.
139. Ahmed SN, Siddiqi ZA. Antiepileptic drugs and 160. Valenzuela A. The biological significance of malo-
liver disease. Seizure. 2006;15(3):156–64. ndialdehyde determination in the assessment of tis-
140. Kim JG, et al. Serum gamma-glutamyltransferase is sue oxidative stress. Life Sci. 1991;48(4):301–9.
a predictor of mortality in patients with acute myo- 161. Lykkesfeldt J. Malondialdehyde as a biomarker of
cardial infarction. Medicine. 2018;97(29):e11393. oxidative damage to lipids caused by smoking. Clin
141. Jiang S, Jiang D, Tao Y. Role of gamma-­ Chim Acta. 2007;380(1–2):50–8.
glutamyltransferase in cardiovascular diseases. Exp 162. Richard M, et al. Trace elements, and lipid peroxi-
Clin Cardiol. 2013;18(1):53. dation abnormalities in patients with chronic renal
142. Creeden JF, et al. Bilirubin as a metabolic failure. Nephron. 1991;57(1):10–5.
hormone: the physiological relevance of 163. Feingold KR, Grunfeld C. Introduction to lipids
low levels. Am J Physiol Endocrinol Metab. and lipoproteins. In: Endotext [Internet]. South
2021;320(2):E191–207. Dartmouth: MDText.com, Inc.; 2015.
143. Kalakonda A, Jenkins BA, John S. Physiology, 164. Salih KJ. The major pathways of lipids (triglyceride
bilirubin. In: StatPearls [Internet]. Treasure Island: and cholesterol) and lipoprotein metabolism. Zanco
StatPearls Publishing; 2017. J Pure Appl Sci. 2021;33(4):61–72.
20 Biochemical Indices of Drug Toxicity 367

165. Fielding BA, Frayn KN. Lipoprotein lipase and 185. Favilli F, et al. IL-18 activity in systemic lupus ery-
the disposition of dietary fatty acids. Br J Nutr. thematosus. Ann N Y Acad Sci. 2009;1173(1):301–9.
1998;80(6):495–502. 186. Parikh CR, et al. Urine IL-18 is an early diagnostic
166. Onal G, et al. Lipid droplets in health and disease. marker for acute kidney injury and predicts mortal-
Lipids Health Dis. 2017;16(1):1–15. ity in the intensive care unit. J Am Soc Nephrol.
167. Rosenson RS, Lowe GD. Effects of lipids 2005;16(10):3046–52.
and lipoproteins on thrombosis and rheology. 187. Lonnemann G, et al. Interleukin-18, interleu-
Atherosclerosis. 1998;140(2):271–80. kin-18 binding protein and impaired production
168. Reiner Ž. Hypertriglyceridaemia, and risk of interferon-­gamma in chronic renal failure. Clin
of coronary artery disease. Nat Rev Cardiol. Nephrol. 2003;60(5):327–34.
2017;14(7):401–11. 188. Auer J, et al. C-reactive protein, and coronary artery
169. Goldberg IJ, Trent CM, Schulze PC. Lipid disease. Jpn Heart J. 2002;43(6):607–19.
metabolism and toxicity in the heart. Cell Metab. 189. Spagnoli LG, et al. Role of inflammation in athero-
2012;15(6):805–12. sclerosis. J Nucl Med. 2007;48(11):1800–15.
170. Craig M, Yarrarapu SNS, Dimri M. Biochemistry, 190. Brancaccio P, Lippi G, Maffulli N. Biochemical
cholesterol. In: StatPearls [Internet]. Treasure markers of muscular damage. Clin Chem Lab Med.
Island: StatPearls Publishing; 2018. 2010;48(6):757–67.
171. Mouritsen OG, Zuckermann MJ. What’s so special 191. Padrão AI, et al. Uncovering the exercise-related
about cholesterol? Lipids. 2004;39(11):1101–13. proteome signature in skeletal muscle. Proteomics.
172. Sèdes L, et al. Cholesterol: a gatekeeper of male fer- 2016;16(5):816–30.
tility? Front Endocrinol. 2018;9:369. 192. Farhana A, Lappin SL. Biochemistry, lactate dehy-
173. Ma H, Shieh K-J. Cholesterol and human health. J drogenase. In: StatPearls [Internet]. Treasure Island:
Am Sci. 2006;2(1):46–50. StatPearls Publishing; 2021.
174. Kumar M, et al. Cholesterol-lowering probiotics as 193. Klein R, et al. Clinical and diagnostic significance
potential biotherapeutics for metabolic diseases. Exp of lactate dehydrogenase and its isoenzymes in ani-
Diabetes Res. 2012;2012:902917. mals. Vet Med Int. 2020;2020:5346483.
175. Staels B, Fonseca VA. Bile acids and metabolic 194. Gomes AV, Potter JD, Szczesna-Cordary D. The role
regulation: mechanisms and clinical responses of troponins in muscle contraction. IUBMB Life.
to bile acid sequestration. Diabetes Care. 2002;54(6):323–33.
2009;32(suppl_2):S237–45. 195. Al-Otaiby MA, Al-Amri HS, Al-Moghairi AM. The
176. Alrefai WA, Gill RK. Bile acid transporters: struc- clinical significance of cardiac troponins in medical
ture, function, regulation, and pathophysiological practice. J Saudi Heart Assoc. 2011;23(1):3–11.
implications. Pharm Res. 2007;24(10):1803–23. 196. Babuin L, Jaffe AS. Troponin: the biomarker of
177. Rolland F, Winderickx J, Thevelein JM. Glucose-­ choice for the detection of cardiac injury. CMAJ.
sensing mechanisms in eukaryotic cells. Trends 2005;173(10):1191–202.
Biochem Sci. 2001;26(5):310–7. 197. D’Souza S, Baxter G. B Type natriuretic peptide:
178. Dashty M. A quick look at biochemistry: a good omen in myocardial ischemia? Heart. BMJ
carbohydrate metabolism. Clin Biochem. Publishing Group Ltd. 2003;89(7):707–9.
2013;46(15):1339–52. 198. Weber M, Hamm C. Role of B-type natriuretic
179. Rui L. Energy metabolism in the liver. Compr peptide (BNP) and NT-proBNP in clinical routine.
Physiol. 2014;4(1):177. Heart. 2006;92(6):843–9.
180. Tarrant JM. Blood cytokines as biomarkers of 199. Sabatine MS, et al. Multimarker approach to risk
in vivo toxicity in preclinical safety assess- stratification in non-ST elevation acute coronary
ment: considerations for their use. Toxicol Sci. syndromes: simultaneous assessment of troponin I,
2010;117(1):4–16. C-reactive protein, and B-type natriuretic peptide.
181. Yang X, Salminen WF, Schnackenberg LK. Current Circulation. 2002;105(15):1760–3.
and emerging biomarkers of hepatotoxicity. Curr 200. Glaser R, et al. Placental growth factor and B-type
Biomark Find. 2012;2:43. natriuretic peptide as independent predictors of risk
182. Wang M, Markel TA, Meldrum DR. Interleukin from a multi-biomarker panel in suspected acute cor-
18 in the heart. Shock. 2008;30(1):3–10. onary syndrome (Acute Risk and Related Outcomes
183. Dinarello C, et al. Interleukin-18 and IL-18 binding Assessed With Cardiac Biomarkers [ARROW])
protein. Front Immunol. 2013;4:289. study. Am J Cardiol. 2011;107(6):821–6.
184. Novick D, et al. Interleukin-18, more than a Th1 201. Cui Y, Paules RS. Use of transcriptomics in under-
cytokine. In: Seminars in immunology. Elsevier; standing mechanisms of drug-induced toxicity.
2013. Pharmacogenomics. 2010;11(4):573–85.
Therapeutic Drug Monitoring
and Clinical Toxicology: Challenges
21
and Future Directions

Seth Kwabena Amponsah and Yashwant V. Pathak

Abstract Some of the assays used in TDM and clinical


toxicology lack sensitivity. Taking samples at
Therapeutic drug monitoring (TDM) is the the right time is another challenge associated
clinical practice of measuring levels of drugs with TDM and clinical toxicology.
in the plasma, serum, or blood at predeter- Inaccuracies from sampling site and handling
mined times or intervals in an effort to main- of samples prior to analysis can affect results.
tain blood concentration of a drug within Some drugs also have active metabolites that
optimum range. Drugs that are usually moni- might not be detected by TDM and clinical
tored are those with low therapeutic indices, toxicological assays. Additionally, TDM and
drugs that have irreversible adverse effects, as clinical toxicology are expensive to under-
well as drugs used in the treatment of diseases take. Indeed, the significance of TDM and
whose symptoms are similar to the toxic clinical toxicology in clinical practice cannot
effects of the drug. TDM is useful in detecting be overemphasized. Nonetheless, more
compliance and non-compliance to drug ther- research can be done on alternate sampling
apy in patients. It also provides a means of matrices such as saliva and dried blood spot.
detecting treatment failure. Clinical toxicol- These matrices would make TDM and clini-
ogy is the study of the physiological effects of cal toxicology more convenient and easy to
toxic agents, their mechanism of action, and do. There is the need for better interpretation
ways of managing these effects. Furthermore, of results obtained from TDM and clinical
clinical toxicology helps in the identification toxicological assays. Hence, health profes-
of chemicals, drugs, or toxins that may affect sionals need to be trained and re-trained on
patients. appropriate interpretation of TDM and clini-
Even though TDM and clinical toxicology cal toxicological results so patients are man-
are useful, they come with some limitations. aged appropriately.

Keywords
S. K. Amponsah (*)
Department of Medical Pharmacology, University of
Ghana Medical School, Accra, Ghana Blood concentration · Challenges · Clinical
e-mail: skamponsah@ug.edu.gh interpretation · Sampling · Sensitivity
Y. V. Pathak
USF Health Taneja College of Pharmacy, University
of South Florida, Tampa, FL, USA
e-mail: ypathak1@usf.edu

© The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 369
S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6_21
370 S. K. Amponsah and Y. V. Pathak

21.1 Introduction istration of therapeutic agents, TDM can be used


to determine the level of toxicity by quantifying
Therapeutic drug monitoring (TDM) is a means the amount of the toxic substance in the blood.
of ensuring safety and efficacy of drug therapy However, clinical toxicology would seek to
by regular and consistent monitoring of blood identify and manage toxicity. Therefore, TDM
concentration of the drug. It is a means of ensur- and clinical toxicology can be complementary.
ing that blood concentrations of certain drugs
remain within a given range in order to maxi-
mize the effectiveness of drugs as well as mini- 21.2 Reasons for TDM
mize side effects. The initial focus of therapeutic
drug monitoring was mainly on adverse drug TDM is recommended or requested for only
reactions [1]. As a result, the drugs that were when absolutely necessary. This is because it
mainly monitored were drugs of narrow thera- can increase healthcare cost for patients. Drugs
peutic indices such as digoxin, phenytoin, lith- with low therapeutic indices and drugs whose
ium, theophylline, and some of the toxicity causes irreversible organ damage or
aminoglycoside antibiotics [2]. Later on, clinical death are usually monitored. Drugs that do not
pharmacokinetic monitoring was introduced in produce enough clinical efficacy to determine
therapeutic drug monitoring. Here, the concen- whether there are therapeutic or toxic effects
tration of the drug in the blood is related to the are also monitored [2]. Patient factors such as
responses that the drug produces. Indeed, TDM poor response to treatment and suspected non-­
is not merely the measurement of drug levels in compliance to treatment can also be a reason
blood sample. TDM involves expert clinical for the requisition of TDM [6]. TDM would
interpretation of drug levels which would ulti- also be requested if the manifestations of the
mately help to optimize a patient’s treatment. disease condition and the toxic effects of a drug
Therefore, drugs that do not offer any meaning- are similar [7]. Drugs such as digoxin have
ful clinical information when levels are assayed toxic symptoms that are very similar to symp-
in blood often do not require TDM. toms of the heart disease for which it is used.
The concept of TDM is based on two Aronson and Hardman report that monitoring
assumptions; there is a relationship between the plasma levels of digoxin helps to point out
the administered dose and the concentration of the actual source of the patient’s symptoms [8].
the drug in the blood. This is also affected by A similar phenomenon is observed with amino-
the time of measuring. The second is there is a glycoside antibiotics. Additionally, TDM aids
relationship between the plasma drug concen- in monitoring compliance to drugs used for
tration and therapeutic or physiological effects prophylaxis. A typical example is the use of
[3, 4]. In the population, there may be individ- phenytoin to prevent episodes of seizures. In
uals who may possess unique pharmacokinetic most cases, patients who take drugs for prophy-
and pharmacodynamic characteristics which laxis lack the awareness of the need to comply
may affect drug concentration at steady state. with treatment and, hence, are likely to be non-
Hence, TDM is very important in individual- adherent. In some cases, a patient may not be
izing drug dose, which invariably would aid in showing significant response to treatment
the attainment of optimal plasma drug concen- despite therapeutic concentrations of the drug
tration needed to maximize efficacy and reduce (at steady state) in the blood. This may likely be
toxicity. as a result of treatment failure. TDM therefore
Clinical toxicology is the study of the physi- offers clinicians any easy way to detect treat-
ological effects of toxic agents, their mechanism ment failure. According to Molden, TDM can
of action, and ways of managing their adverse be used in assessing different measures of treat-
effects [5]. In cases of toxicity during the admin- ment failure [9].
21 Therapeutic Drug Monitoring and Clinical Toxicology: Challenges and Future Directions 371

21.3 Challenges with TDM this may be as a result of cross-reactivity of assay


material and many inactive metabolites of the
21.3.1 Drug in Circulation May Not drug [13]. More often, there exists high variation
Correspond to Amount at Site between results obtained from different sample
of Action batches and sometimes within the same run.
Chromatographic methods are more accurate in
A limited number of drugs can be monitored; and quantifying plasma drug concentration. However,
subsequent relevant clinical interpretations are these methods are expensive and technically
made. Aside from concentration of a drug in cir- demanding; hence, immunoassays are still used
culation, the amount of drug present at the site of in the analysis of several drugs such as theophyl-
action is often difficult to determine. Pichini has line, methotrexate, cyclosporine, gentamicin, and
suggested the analysis of drugs in areas such as amikacin. Chromatographic and immunoassay
bronchial secretions, peritoneal fluid, interstitial methods normally measure total concentration of
fluid, tears, and nails [10]. This is because these the drug in the blood. This includes both protein-­
areas may help show the concentrations of the bound drug and free drug. For drugs that are
drug at the site of action. Even with this, it may highly protein-bound (for instance, phenytoin
be difficult to measure the amount of drug at the which is about 90% protein bound), total concen-
actual site of action which is the receptor site. tration obtained from the assay methods may not
This is partly because receptors of a drug may not always correlate with what pertains in systemic
be localized in one part of the body. As a result, circulation [14]. It is well documented that some
the most convenient physiological fluid used in endogenous substances cross-react and interfere
therapeutic drug monitoring is the blood (plasma). with serum concentration of drugs measured. In
For TDM of a drug to be beneficial, the desired the immunoassay of digoxin, digoxin-like immu-
effect and adverse effect of the drug should have noreactive substances (DLIS) alter serum con-
a correlation with the plasma concentration [11]. centration measurement by cross-reacting with
A limited number of drugs have this correlation; the antidigoxin antibodies used in the assay [15].
and these include amikacin, gentamicin, phenyt- This may lead to false low digoxin levels mea-
oin, lithium, vancomycin, methotrexate, cyclo- sured. DLIS levels are normally clinically insig-
sporine, digoxin, phenobarbital, carbamazepine, nificant in healthy persons, but levels may rise
and valproate [12]. significantly in conditions such as hypertension,
liver disease, uremia, and congestive heart fail-
ure. This can be dangerous since it might lead to
21.3.2 Low Sensitivity of Some the tendency for an increased dose adjustment of
of the Assays Used in TDM digoxin. Given the narrow therapeutic range of
digoxin, this might be detrimental to the patient.
Lack of sensitivity of some drug assay methods is
a limitation of TDM. Analytical methods that can
be used to assay drugs in biological samples 21.3.3 Difficulties in Sampling
could be gas chromatography, high performance at the Right Time
liquid chromatography, or ultra-performance liq-
uid chromatography coupled to ultraviolet, mass, Another challenge associated with TDM is
or fluorescence spectrophotometry. Other meth- selecting the right sampling time. A key to obtain-
ods, which are non-specific, include radioimmu- ing useful and accurate measurement of blood
noassay (RIA), enzyme-linked immunosorbent concentration is taking samples at the right time.
assay (ELISA), or enzyme-linked turbidimetric Determining the right time to take a biological
inhibition assay (PETINIA). The non-specific sample can be quite challenging. The pharmaco-
assays, for example immunoassays, could over- kinetic properties of drugs play an important role
estimate plasma concentrations of a drug, and in determining the sampling times. Others factors
372 S. K. Amponsah and Y. V. Pathak

such as age, liver and renal function, drug inter- Table 21.1 Sampling times for some common drugs that
require monitoring
actions, and some genetic factors may also affect
drug levels and the time it takes to reach steady Drug Appropriate sampling time
state. Therefore all these factors should be con- Gentamicin At steady state; after at least 4
half-lives (half-life = 2–3 hours)
sidered in sampling times for TDM. TDM is Trough samples; 30 minutes before
expensive; hence, it is imperative to determine next dose
appropriate sampling times in order to obtain Peak samples; 30 minutes after IV
clinically relevant data. In normal practice, sam- infusion or IV bolus
Digoxin At steady state; 8 days
pling usually takes place just before the next dose Trough sample; before next dose
(trough concentration of the drug). In other cases, Peak sample; at least 6 hours after last
the sample is taken immediately after drug dose
administration (peak plasma concentration). Carbamazepine At steady state; 2–4 weeks after
initiation
Knowledge of the sampling time is of clinical
Trough sample; within 2 hours before
importance in the interpretation of information next dose
obtained from drug monitoring. For some drugs Phenobarbital At steady state; 2–3 weeks after
that require TDM, there exist possible sampling initiation
times; and these are presented in Table 21.1. Trough sample; within 2 hours before
next dose
Aside from these times, sampling can be done Peak sample; at least 3 hours after last
anytime toxicity is suspected or when the patient dose
is showing no response to ongoing treatment Phenytoin At steady state; 5 to 10 days after
[16]. initiation
Trough sample; within 2 hours before
next dose
Valproic acid At steady state; 2–4 days after
21.3.4 Inability to Assay Some Active initiation
Metabolites of Drug Trough sample; within 2 hours before
next dose
The presence of active metabolites of a drug in
circulation may also contribute to the therapeutic
effect of the drug. Some of the drugs that are rou- 21.3.5 Inaccuracies in Detecting
tinely monitored may have some active metabo- Medication Compliance
lites which are often or may not usually be
measured [6]. These metabolites are sometimes TDM has been used over the years in assessing
equally as active or more active than the parent drug adherence (compliance) in patients.
drug. For instance, the active metabolite of clo- However, TDM is only valuable in detecting
zapine, norclozapine, has been shown to be as short-term compliance or non-compliance. TDM
active as the parent drug [17]. has also shown very little benefit in improving
In the determination of plasma concentration compliance to drug in cases of prophylaxis. This
of a drug, the measurement obtained is usually a is especially true in non-hospitalized patients or
total of both the parent drug and the active metab- outpatients. Patients are likely to take a dose of
olite. Other assay methods are also able to mea- their treatment prior to blood sampling for analy-
sure the parent drug but not the active metabolites. sis or before medical appointment [19]. Drug
Some reports suggest that desethylamiodarone plasma levels alone may not be entirely accurate
which is an active metabolite of amiodarone is in detecting compliance. Additionally, individu-
not measured and accounted for in the therapeu- als who are fast metabolizers may also record
tic range of amiodarone during TDM [18]. Since low plasma drug levels on certain occasions [11].
the metabolite is active and accounts for part of This may be falsely attributed to medication non-­
the therapeutic activity, the practice of measuring compliance. The concomitant administration of
the parent compound only may be inaccurate. drugs that are enzyme inducers or inhibitors may
21 Therapeutic Drug Monitoring and Clinical Toxicology: Challenges and Future Directions 373

also affect plasma drug concentration [20, 21]. local anesthetics like bupivacaine, blood samples
For instance, it has been shown that the concomi- from central veins and arteries have been shown
tant administration of clozapine and carbamaze- to be more reliable than peripheral venous sam-
pine significantly decreases serum clozapine ples [27]. Cyclosporine administered intrave-
concentrations [22]. This can be attributed to nously is known to have high concentrations in
potent liver enzyme-inducing ability of samples from central venous catheter than
carbamazepine. peripheral venous samples [28]. After blood sam-
ples are collected, they are normally stored in
tubes before analysis. The purpose of these col-
21.3.6 TDM Is Expensive lection tubes is to preserve the blood sample with
its content of drug and metabolite(s) till assay
One of the shortcomings of TDM is the cost [29–31]. These collection tubes may contain anti-
involved in analyzing samples. TDM can be coagulants such as ethylenediamine tetraacetic
expensive since it involves repeated testing. Two acid (EDTA), heparin, and trisodium citrate
main strategies for TDM exist: reactive TDM (TSC) and sometimes no anticoagulants at all
where monitoring is done if toxicity or treatment [32]. These anticoagulants and barrier gels found
failure is suspected, and the other is proactive in the collection tubes can affect drug concentra-
TDM, which is routine repeated testing done at tion in the blood (plasma or serum) that will be
specific times. Even though it can be argued that measured. Lithium heparin tubes have been
TDM makes overall healthcare cost-effective, reported to be ideal for TDM [33, 34]. One the
there is the likelihood that regular TDM could be contrary, for monitoring serum lithium levels,
additional cost for patient. A study conducted by lithium heparin tubes have been found to inter-
Campbell et al. showed that out-of-pocket costs fere with test results. This is because the lithium
lead to a decrease in the willingness of patients to in the tube can react with porphyrin compound in
participate in TDM [23]. This means TDM may the blood to cause a falsely elevated serum lith-
not be affordable to patients especially if there is ium concentration [35]. There are a number of
no external support. Some analytical procedures systematic reviews and studies that have reported
such as liquid chromatography-mass spectrome- cases of false elevated lithium levels as a result of
try are costly; hence if these methods are the use of lithium heparin tubes [35].
employed in TDM, patients might find this pro-
cedure expensive [24]. One of the main benefits
of TDM is dosage adjustment. Some centers do 21.4 Future Directions
their adjustments using “trial and error” method
of different dosages till an optimal serum concen- 21.4.1 Alternate Sampling Matrices
tration is achieved. This approach in TDM has
been shown to lead to unnecessary healthcare In analysis of biological samples during TDM,
cost for the patient [25]. the most common body fluid used is venous
blood. The traditional invasive venous blood
sampling comes with its challenges. It presents a
21.3.7 Challenges with Sampling lot of discomfort especially in pediatric patients.
and Sample Handling Before Alternate matrices with corresponding highly
Analysis sensitive assays for TDM are recommended.
Saliva has been utilized in monitoring drugs such
The process of drawing samples and handling of as carbamazepine, primidone, phenytoin, and
specimen before analysis are critical in TDM ethosuximide [36]. Saliva is a suitable matrix for
and, hence, have to be done properly. It requires analyzing these drugs because the concentrations
taking extra precautions at the site of sampling to of these drugs in saliva are directly proportional
avoid contamination [26]. In the analysis of some or can be used to extrapolate concentrations of
374 S. K. Amponsah and Y. V. Pathak

the drug in serum [37]. Even though there are a compared to therapeutic reference ranges. The
number of drugs, especially anticonvulsants, that therapeutic reference ranges are serum/plasma
can be assayed in saliva, some drugs such as val- drug concentrations that are expected to produce
proic acid cannot be analyzed using saliva [36]. desired therapeutic effect [40]. These reference
Saliva may be advantageous because, comparing ranges may vary from one laboratory to the labo-
this with the traditional venous blood sampling, it ratory. They may also not be universally favor-
is noninvasive, has high patient preference able to all patients. The purpose of TDM is to
(because of the ease of taking samples especially manage the disease of a patient appropriately and
in children), there is no need for a professional not merely obtaining and recording values. A lot
phlebotomist in taking samples, and its cost-­ of factors can alter effective therapeutic concen-
effectiveness [37, 38]. trations of different drugs in different patients.
Sara Capiau and colleagues report in a study These include, but are not limited to, genetic vari-
that an ideal sample collection method should ables, concomitant administration of drugs, age,
have the following properties: noninvasive, and pharmacokinetic characteristics of patient.
allows for self-sampling, requires little sample Some patients may achieve maximum benefits of
volume, applicable to everyone, robust, and eco- the drug at lower concentrations than the sug-
nomic [25]. Dried blood spot sampling is a gested optimal therapeutic ranges, while others
method where capillary blood is collected from a may experience adverse effect within the sug-
finger or heel prick onto a filter paper [39, 40]. gested ranges [40]. In the management of sei-
This method meets the criteria specified by Sara zures with phenytoin, one reference range for all
Capiau et al. [25]. Dried blood spot sampling is patients may not be applicable [41]. The seizure
minimally invasive and requires a very small type, severity of the illness, and genetic abnor-
amount of blood (5–35 μL) [25]. This method malities may affect optimal drug concentration
facilitates home sample collection, especially for needed by the patient. Therefore, establishing an
children and patients with psychological disor- individualized therapeutic concentration range
ders. It also helps in dealing with one of the major for each patient is the most appropriate approach
challenges in TDM, which is sampling at the in TDM.
right time. In most cases, trough level samples
can be taken immediately before the next dose.
This is often done early morning or late evening 21.4.3 Appropriate Clinical
when the clinics and laboratories may not be Interpretation of TDM Results
working. Dried blood spot sampling will make
sampling possible at these times for a patient who TDM is not only concerned with mere measure-
will be at home. Dried blood spot sampling has ment of drug concentration in the blood [12] but
also been shown to be more stable than frozen also making appropriate clinical interpretation of
blood samples and hence makes it more conve- the data. Dosage adjustment normally follows
nient for storage and later transport [39]. Bio-­ analysis of drug levels. Before dose adjustment is
analytical methods for TDM could be validated done, expert clinical interpretation has to be
to utilize dried blood spot samples. made in order to derive meaningful results from
the procedure [42]. Most centers, especially in
developing countries, have TDM limited to assay
21.4.2 Individualized Therapeutic only, instead of both assay and clinical interpreta-
Concentration tion [42]. Ideally, there should be well-­established
clinical pharmacology and toxicology units in
Individualized therapeutic concentration in TDM hospitals that will make recommendations based
is a principle that when used would bring great on TDM data. The need to improve understand-
improvement and benefit to patients. Usually in ing of pharmacokinetic principles and related
TDM, serum and plasma drug concentrations are TDM data among health professionals cannot be
21 Therapeutic Drug Monitoring and Clinical Toxicology: Challenges and Future Directions 375

overemphasized [43]. Current health profession- dose to be administered. The Bayesian algorithm
als should therefore be trained in appropriate is considered the gold standard for dosage adjust-
clinical interpretation of TDM data obtained ment calculations. Timing of when samples
from laboratories. should be taken is often challenging. The
Bayesian model helps to solve this problem by
comparing a random concentration to a popula-
21.4.4 Merging Target Concentration tion average concentration-time curve [46, 47].
Intervention (TCI) with TDM Pharmacokinetic models can be applied to pre-
dict specific individualized pharmacokinetic pro-
One area that can be explored is merging target files in order to make room for inter-patient
concentration intervention (TCI) with TDM [21]. variability [48]. More research and validation
TCI is a method that makes use of pharmacoki- need to be done on the computer-assisted method
netic and pharmacodynamic principles to esti- of TDM as this can lead to significant advance-
mate how patients would reach target ment in patient therapy.
concentration when given a specific dose of a
drug. Some of the parameters examined include
clearance (Cl), maximum effect of the drug at 21.5 Conclusion
high concentrations (Emax), and concentration that
produces 50% of Emax (EC50) [44]. It is ideal to There are a number of challenges associated with
use TCI when the effect of a treatment being the TDM and clinical toxicology, and these
administered is difficult to quantify. It is there- include low sensitivity of some of the assays, dif-
fore used to maintain drug concentration of a tar- ficulties in sampling at the right time, and inabil-
get drug which has been shown to be effective in ity to assay some active metabolites of drug,
majority of populations [45]. among others. Despite these challenges, TDM
Another reason for using TCI is when a group-­ and clinical toxicology play important roles in
based dosing system (such as dosing based on patient therapy (care). Nonetheless, the practice
weight) does not reduce variability between dif- can be improved by using alternate sampling
ferent members [45]. Even though TCI and TDM matrices (like saliva and dried blood spot), indi-
are different principles that are practiced sepa- vidualized therapeutic concentrations, appropri-
rately, merging the two may be of more benefit ate clinical interpretation of results, and use of
than practicing them separately. Merging these computer-assisted TDM.
two means TDM is now going to consider phar-
macogenomics. Hence, monitoring of serum/
plasma drug concentrations would not be aimed References
at only dosage adjustment but rather reaching
individual target concentration [44]. 1. Amponsah SK, Boadu JA, Dwamena DK, Opuni
KFM. Bioanalysis of aminoglycosides using high-­
performance liquid chromatography. ADMET
DMPK. 2022;10(1):27–62.
21.4.5 Computer-Assisted TDM 2. Dasgupta A, editor. Therapeutic drug monitoring:
newer drugs and biomarkers. Academic Press; 2012.
3. Kang JS, Lee MH. Overview of therapeutic drug
Finally, computed-assisted TDM is another area monitoring. Korean J Intern Med. 2009;24(1):1.
that can be studied in terms of improving the 4. Persky AM, Pollack GM. Foundations in pharmacoki-
practice of TDM. Computerized clinical decision netics. In: UNC Eshelman School of Pharmacy. North
support systems may have the potential of Carolina: University of North Carolina Chapel Hill;
2013.
improving the benefits of drugs that are normally 5. Ref Sullivan DW, Gad S. Clinical toxicology and clin-
monitored. During dosage adjustment, some ical analytical toxicology. In: Information resources
models are used in the calculation of the right in toxicology. Academic Press; 2020. p. 237–40.
376 S. K. Amponsah and Y. V. Pathak

6. Ghiculescu RA. Therapeutic drug monitoring: which sus guidelines for therapeutic drug monitoring
drugs, why, when and how to do it. Aust Prescr. in psychiatry: update 2011. Pharmacopsychiatry.
2008;31:42–4. 2011;21(06):195–235.
7. Gross AS. Best practice in therapeutic drug monitor- 24. Avent ML, Rogers BA, Cheng AC, Paterson
ing. Br J Clin Pharmacol. 2001;52(S1):5–9. DL. Current use of aminoglycosides: indications,
8. Aronson JK, Hardman M. ABC of monitoring drug pharmacokinetics and monitoring for toxicity. Intern
therapy: measuring plasma drug concentrations. BMJ. Med J. 2011;41(6):441–9.
1992;305:1078–80. 25. Capiau S, Alffenaar JW, Stove CP. Alternative sam-
9. Molden E. 17 Using TDM data to study treatment pling strategies for therapeutic drug monitoring. In:
failure. Pharmacopsychiatry. 2020;53(03) Clinical challenges in therapeutic drug monitoring.
10. Pichini S, Altieri I, Zuccaro P, Pacifici R. Drug moni- Elsevier; 2016. p. 279–336.
toring in nonconventional biological fluids and matri- 26. Kontny NE, Hempel G, Boos J, Boddy AV, Krischke
ces. Clin Pharmacokinet. 1996;30(3):211–28. M. Minimization of the preanalytical error in plasma
11. Opuni KFM, Boadu JA, Amponsah SK, Okai samples for pharmacokinetic analyses and therapeutic
CA. High performance liquid chromatography: a ver- drug monitoring-using doxorubicin as an example.
satile tool for assaying antiepileptic drugs in biologi- Ther Drug Monit. 2011;33(6):766–71.
cal matrices. J Chromatogr B. 2021;1179:122750. 27. Biscoping J, Adams HA, Menges T, Krumholz W,
12. Figueras A. Review of the evidence to include TDM Hempelmann G. The significance of the sampling site
in the essential in vitro diagnostics list and prioritiza- in the determination of plasma levels of local anes-
tion of medicines to be monitored. Fundació Institut thetics using 0.75% bupivacaine as an example. Reg
Català de Farmacologia: Barcelona, Spain; 2019. Anaesth. 1990;13(1):16–20.
13. Steimer W. Performance and specificity of monoclo- 28. Shulman RJ, Ou C, Reed T, Gardner P. Central venous
nal immunoassays for cyclosporine monitoring: how catheters versus peripheral veins for sampling blood
specific is specific? Clin Chem. 1999;45(3):371–81. levels of commonly used drugs. J Parenter Enter Nutr.
14. Warner AM. Pitfalls in monitoring therapeutic drugs. 1998;22(4):234–7.
Lab Med. 1997;28(10):653–7. 29. Amponsah SK, Yeboah S, Kukuia KKE, N’guessan
15. Wu SL, Li W, Wells A, Dasgupta A. Digoxin-like and BB, Adi-Dako O. A pharmacokinetic evaluation of
digitoxin-like immunoreactive substances in elderly a pectin based oral multiparticulate matrix carrier of
people: impact on therapeutic drug monitoring of carbamazepine. Adv Pharmacol Pharm Sci. 2021; ID
digoxin and digitoxin concentrations. Am J Clin 5527452, 7 pages.
Pathol. 2001;115(4):600–4. 30. Amponsah SK, N'guessan BB, Akandawen M, Aning
16. Hamzah A, Fattah AB. Evaluation of blood sampling A, Agboli SY, Danso EA, Opuni KFM, Asiedu-­
times and indications for therapeutic drug monitoring Gyekye IJ, Appiah-Opong R. Effect of Cellgevity®
services. Malays J Pharm Sci. 2008;6(1):1–11. supplement on selected rat liver Cytochrome P450
17. Hendset M, Haslemo T, Rudberg I, Refsum H, enzyme activity and pharmacokinetic parameters of
Molden E. The complexity of active metabolites in carbamazepine. Evid Based Complement Alternat
therapeutic drug monitoring of psychotropic drugs. Med. 2022; ID 7956493, 8 pages.
Pharmacopsychiatry. 2006;39(04):121–7. 31. Amponsah SK, Opuni KFM, Antwi KA, Kunkpeh
18. Campbell TJ, Williams KM. Therapeutic drug moni- VP. Effect of aminophylline on the pharmacokinet-
toring: antiarrhythmic drugs. Br J Clin Pharmacol. ics of amikacin in Sprague-Dawley rats. J Infect Dev
2001;52(S1):21–33. Ctries. 2019;13(3):251–4.
19. Fluckiger P, Aícua-Rapún I, André P, Rossetti AO, 32. Kulkarni P, Karanam A, Gurjar M, Dhoble S, Naik
Decosterd LA, Buclin T, Novy J. Therapeutic drug AB, Vidhun BH, Gota V. Effect of various anticoagu-
monitoring of newer generation antiseizure medi- lants on the bioanalysis of drugs in rat blood: implica-
cations at the point of treatment failure. Seizure. tion for pharmacokinetic studies of anticancer drugs.
2022;94:66–9. Springerplus. 2016;5(1):1–8.
20. Harivenkatesh N, Haribalaji N, David DC, Kumar 33. Steuer C, Huber AR, Bernasconi L. Where clinical
CP. Therapeutic drug monitoring of antiepilep- chemistry meets medicinal chemistry. Systematic
tic drugs in a tertiary care hospital in India. Clin analysis of physico-chemical properties predicts sta-
Neuropharmacol. 2015;38(1):1–5. bility of common used drugs in gel separator serum
21. Advani M, Seetharaman R, Pawar S, Mali S, tubes. Clin Chim Acta. 2016;462:23–7.
Lokhande J. Past, present and future perspectives of 34. Schrapp A, Mory C, Duflot T, Pereira T, Imbert L,
therapeutic drug monitoring in India. Int J Clin Pract. Lamoureux F. The right blood collection tube for
2021;75:e14189. therapeutic drug monitoring and toxicology screening
22. Campbell JP, Burton E, Wymer S, Shaw M, Vaughn procedures: standard tubes, gel or mechanical separa-
BP. Out-of-pocket cost is a barrier to therapeutic drug tor? Clin Chim Acta. 2019;488:196–201.
monitoring in inflammatory bowel disease. Dig Dis 35. Brizzee L, Stone A, Palmer MC. False lithium toxicity
Sci. 2017;62(12):3336–43. secondary to lithium heparin test tube: a case report
23. Hiemke C, Baumann P, Bergemann N, Conca and review. Mental Health Clin. 2020;10(3):90–4.
A, Dietmaier O, Egberts K, et al. AGNP consen-
21 Therapeutic Drug Monitoring and Clinical Toxicology: Challenges and Future Directions 377

36. Patsalos PN, Berry DJ. Therapeutic drug monitor- 43. Samani K. Therapeutic drug monitoring: an e-­learning
ing of antiepileptic drugs by use of saliva. Ther Drug resource. Biosci Horiz. 2009;2(2):113–24.
Monit. 2013;35(1):4–29. 44. Holford NH. Target concentration intervention:
37. van den Elsen SH, Akkerman OW, Jongedijk EM, beyond Y2K. Br J Clin Pharmacol. 1999;48(1):9–13.
Wessels M, Ghimire S, van der Werf TS, et al. 45. Holford N. Pharmacodynamic principles and tar-
Therapeutic drug monitoring using saliva as matrix: get concentration intervention. Transl Clin Pharm.
an opportunity for linezolid, but challenge for moxi- 2018;26(4):150–4.
floxacin. Eur Respir J. 2020;55(5):1901903. 46. Burger DM, Hugen PW, Aarnoutse RE, Hoetelmans
38. Gorodischer R, Burtin P, Hwang P, Levine M, Koren RM, Jambroes M, Nieuwkerk PT, et al. Treatment
G. Saliva versus blood sampling for therapeutic drug failure of nelfinavir-containing triple therapy can
monitoring in children: patient and parental prefer- largely be explained by low nelfinavir plasma concen-
ences and an economic analysis. Ther Drug Monit. trations. Ther Drug Monit. 2003;25(1):73–80.
1994;16(5):437–43. 47. Amponsah SK, Adjei GO, Enweronu-Laryea C,
39. Wilhelm AJ, den Burger JC, Swart EL. Therapeutic Bugyei KA, Hadji-Popovski K, Kurtzhals JA,
drug monitoring by dried blood spot: progress to Kristensen K. Population pharmacokinetic charac-
date and future directions. Clin Pharmacokinet. teristics of amikacin in suspected cases of neonatal
2014;53(11):961–73. sepsis in a low-resource African setting: a prospec-
40. Amponsah SK, Opuni KFM, Donkor AA. Donkor, tive non-randomized single-site study. Curr Ther Res.
animal model investigation suggests betamethasone 2017;84:e1–6.
alters the pharmacokinetics of amikacin. ADMET 48. Goicoechea M, Vidal A, Capparelli E, Rigby A,
DMPK. 2018;6(4):279–83. Kemper C, Diamond C, California Collaborative
41. Wu MF, Lim WH. Phenytoin: a guide to thera- Treatment Group. A computer-based system to aid in
peutic drug monitoring. Proc Singapore Healthc. the interpretation of plasma concentrations of antiret-
2013;22(3):198–202. rovirals for therapeutic drug monitoring. Antivir Ther.
42. Nwobodo N. Therapeutic drug monitoring in a 2007;12(1):55.
developing nation: a clinical guide. JRSM open.
2014;5(8):2054270414531121.
Index

A C
Adverse drug reactions (ADRs), 75–77, 80, 96, Carbon nanotubes (CNTs), 234–237, 242, 243, 247, 249,
107–110, 120, 146, 151, 156, 199, 208, 273, 275, 329, 331–333, 338, 339
315, 370 Challenges, 4, 11, 13, 15, 35, 44, 53, 56, 58, 59, 68–70,
Allograft rejection, 187, 256, 257, 259, 263, 265 74, 75, 77, 80, 81, 99, 100, 106, 107, 109, 161,
Alternative matrices, 3, 104, 110 163, 170, 193, 234, 238–241, 274, 305, 310, 314,
Analysis, 2, 4, 11–15, 25, 29, 44–47, 50, 52–58, 60, 68, 316–318, 370–375
69, 72, 74–81, 96, 98–100, 102–107, 109, Clinical interpretation, 15, 91, 97, 98, 370, 371, 374, 375
117–120, 126–140, 151, 161–163, 218, 219, 234, Clinical toxicology (CT), 10–16, 21, 47, 91, 103,
240, 277, 278, 318, 336, 348, 371–374 117–140, 145, 161–163, 170, 292, 370, 372, 375
Analytical techniques, 10–12, 14, 15, 44, 45, 59, 74, 100, Computational biomarker, 134–138
102, 103, 163, 167, 362 Critically ill patients, 30–31, 144–149, 151, 153,
Antibiotics, 10–14, 22, 32, 35, 45, 52, 81, 90, 97, 155–157
99–101, 127, 145–151, 157, 167, 169, 171, Cyclosporine, 22–24, 89, 97, 182–193, 201, 234, 256,
187, 199–201, 239–241, 262, 304, 307, 310, 257, 260–264, 293, 316, 371, 373
311, 370 Cytochrome P450 (CYP), 27–28, 30, 33–36, 177, 184, 185,
Anticancer drugs, 12, 165, 169–173, 177, 256 188, 198, 201, 205–211, 286, 290, 292, 308, 309
Anticoagulants, 12–14, 28, 29, 52, 55, 145, 155, 157,
162, 200, 201, 218, 242–244, 288, 310, 373
Antifungal, 12, 14, 45, 99, 101, 145, 152–154, 157, 187, D
201, 239 Data, 2, 3, 11, 12, 14, 15, 32, 36, 45, 46, 59, 68–70,
Antimicrobial, 23, 28, 30, 31, 33, 45, 48, 145–157, 200, 72–81, 98, 102, 106–109, 117–124, 127–136,
294, 310 138, 140, 149, 152, 154–156, 161–163, 177, 218,
Antiviral, 12, 14, 48, 145, 153–157, 201, 304, 318 234, 277, 278, 287, 292, 295, 310–313, 316–319,
a priori TDM, 22, 170, 199 325, 328, 337, 351, 362, 372, 374, 375
Artificial intelligence (AI), 68–81, 105–109, 273–282 Deep learning (DL), 68–69, 72, 74, 75, 78–81, 105, 107,
Automated machine learning (AutoML), 277 108, 274, 276–279, 281, 282
Dried blood spots (DBS), 37, 43–60, 99, 109
Drug assays, 25, 89, 92, 98, 166, 371
B Drug design, 68–72, 75, 80, 81
Bioactivation, 34, 35, 205, 215 Drug dosing, 2, 26, 167–169, 198, 285–287, 291–294,
Bioanalysis, 1 298, 313, 314, 316
Biochips, 101 Drug individualization, 286, 296–298, 303–319
Biological matrices, 1–5, 11, 13, 47, 55, 96, 98, Drug-induced liver injury (DILI), 79, 107, 273–282
102–106, 234, 235, 238, 239, 242–244, 247, 362 Drug interactions, 15, 22–24, 28, 75, 92, 172, 189, 198,
Biological samples, 1, 2, 5, 47, 100, 103, 104, 241, 241, 319, 372
371, 373 Drug levels, 1–3, 5, 24, 90, 93, 103, 104, 162, 178, 198,
Biomarkers, 58, 68, 75, 100, 105, 109, 119, 126, 200, 211, 212, 217, 370, 372, 374
132–138, 206, 265, 327, 334, 336, 348, 349, 351, Drug metabolism, 22, 25–27, 33, 36, 193, 202, 204–208,
357, 359–362 211, 214, 292, 307, 313, 316
Blood concentrations, 2, 5, 45, 46, 97, 98, 121, 146, 147, Drug monitoring, 5, 28, 70, 72–74, 91, 96, 167, 199, 202,
183, 193, 198, 315, 370, 371 234, 372

© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature 379
Switzerland AG 2022
S. K. Amponsah, Y. V. Pathak (eds.), Recent Advances in Therapeutic Drug Monitoring and Clinical
Toxicology, https://doi.org/10.1007/978-3-031-12398-6
380 Index

E O
Electrochemical determination, 245 Optimal dosing, 294–295, 297
Electrolytes, 22, 28, 241, 348, 349, 352 Organ toxicity, 256–258, 265, 324, 348
Endpoints, 72, 75, 78–80, 117–119, 126, 128–140, 337,
347
Enzyme polymorphisms, 209 P
Enzymes, 3, 11, 13, 24, 25, 27–28, 30, 33–35, 54, 93, Pharmacodynamics (PD), 2, 3, 10, 26, 36, 45, 88, 93,
100, 103, 146, 151, 172, 177, 178, 184–186, 188, 96–98, 117, 119, 126–127, 129, 145–147, 150, 153,
198, 200, 201, 204–214, 216, 237, 238, 240, 241, 154, 156, 162, 170–172, 176, 188, 208–209, 259,
244, 245, 249, 263–265, 286, 288–290, 292–294, 285–288, 297, 308, 311, 317, 337, 347, 370, 375
305, 307–310, 314, 315, 331, 348, 349, 352, Pharmacogenetics, 27, 199, 201, 202, 218, 259, 286, 290,
354–359, 361, 362, 372 291, 298, 313–314
Pharmacokinetics (PK), 2–4, 10, 26–28, 32, 33, 36, 37,
44, 88, 93, 96–98, 102, 104, 108, 117–140,
F 145–148, 151, 152, 154, 156, 162, 166–172,
Fluorescence, 11, 14, 25, 49, 50, 93, 105, 218, 235–237, 175–178, 182, 184, 188, 193, 198, 199, 204, 208,
242, 244, 246–249, 337, 338, 371 240, 246, 248, 285–288, 293, 298, 306–309, 311,
313, 317, 318, 324, 335, 337, 370, 371, 374, 375
Pharmacovigilance, 76, 77, 81, 107
H Physiologically based pharmacokinetic (PBPK)
Hematocrit (Hct), 44–46, 50–59, 187, 312 modeling, 33, 286, 305, 311–313
Hepatocytes, 33, 34, 36, 264, 276, 327, 356 PK/PD modeling, 311, 317, 318
Plasma concentrations, 3, 4, 10, 22–26, 29, 30, 34–37,
45, 46, 90, 92, 93, 97, 98, 108, 120, 121,
I 124–126, 145, 146, 150, 151, 166, 167, 172–174,
Immunosuppressant, 10, 12, 14, 28, 32, 45, 49, 89, 90, 177, 178, 182, 184, 200, 202, 204, 208, 211, 215,
100, 101, 169, 181–183, 185, 186, 188–193, 201, 233, 234, 239–241, 246, 248, 309, 359, 371, 372
216, 218, 259, 260, 262, 264, 265 Polymorphism, 27, 28, 177, 182, 188, 201, 208–214,
Immunosuppressant toxicity, 186–191, 193 259, 265
Immunosuppression, 101, 190, 256–261, 263–265, 294 Pro-drugs, 201, 204, 207, 212, 214–216
Immunosuppressive drugs, 182, 183, 188, 193, 213,
255–265
Individualized drug therapy, 166, 169, 170, 172 Q
Quality assurance, 161, 162
Quantum dots, 235–238, 242, 246, 249, 337, 338
L
Liquid chromatography coupled with electrospray-­
ionization mass spectrophotometry (LC-ESI-MS), S
103 Safety, 4, 23, 37, 68, 73–77, 79–81, 88, 89, 92, 96, 101,
102, 105, 107, 108, 117, 118, 120, 145, 166, 177,
178, 201–203, 205, 208, 214, 216, 217, 256, 287,
M 313, 324, 327, 352, 370
Machine learning (ML), 68, 70, 73–77, 79–81, 105, 107, Sampling, 2–4, 25, 32, 44, 45, 47–60, 88, 90, 91, 96, 98,
108, 274, 277–280, 282 99, 102, 104, 106, 121, 123, 124, 150, 162, 163,
Matrices, 2, 4, 10, 11, 13–15, 46, 47, 50–54, 56–58, 72, 176, 318, 336, 371–375
99–107, 109, 110, 163, 234, 235, 237, 238, 243, Sensitivity, 11, 13, 14, 55, 79, 80, 97, 99, 101, 102, 144,
245, 249, 276–280, 373, 375 145, 199, 213, 234, 240–245, 247, 248, 290, 304,
Medication overdoses, 295, 296 311, 349, 357, 361, 362, 371, 375
Metabolites, 1–3, 10, 13, 22, 25–27, 33–35, 47, 50, 51, Solid organ transplantation, 216, 255–265
58, 59, 92, 93, 100, 102, 104–107, 120, 126, 167, Steady state, 24, 31, 89, 90, 97, 121, 125–126, 150, 156,
169, 174, 177, 184, 185, 187, 200–209, 211–219, 176, 178, 186, 200, 203, 241, 249, 370, 372
245, 248, 249, 262, 317, 318, 336, 348, 349, 362,
371–373, 375
T
Target concentration intervention (TCI), 375
N Therapeutic drug monitoring (TDM), 2, 3, 10–16, 21–37,
Nanomedicines, 324–335, 337–339 43–59, 68, 88–93, 96–110, 117–140, 145–155,
Nanoparticles, 100, 101, 234–236, 238, 240–249, 325 157, 161–163, 165–168, 170–178, 181–193,
Nanotoxicity, 328, 337–339 197–219, 233–249, 287, 318–319, 370–375
Index 381

Therapeutic indices, 4, 26, 44, 58, 88, 92, 117, 152, 157, 167, 169, 172, 174–176, 182, 183, 185, 188–190,
166, 167, 170–172, 182, 183, 200, 203, 210, 192, 193, 198–200, 202–208, 210–214, 216–219,
213–215, 247, 249, 293, 307, 327, 370 238–241, 245, 246, 257, 258, 261–263, 273–282,
Therapeutic monitoring, 26, 93, 109, 166, 238, 287, 291, 294, 297, 298, 307, 310, 312, 314,
240, 241 324–339, 347–362, 370, 372, 373
Therapy individualization, 215, 285–298, 303–305, 310, Toxicology, 1, 3, 43, 44, 47–50, 78, 101–104, 106,
311, 313 165–178, 181, 192, 197, 198, 347, 352, 374
Thermal desorption-tandem mass spectrometry
(LDTD-MS-MS), 102, 103
Toxic concentrations, 4–5 U
Toxicity, 2, 4, 15, 22–32, 36, 68, 70, 75, 77–81, 88–92, Uridine diphosphate glucuronosyltransferases (UGTs),
96–99, 101, 105, 108, 110, 144–157, 162, 166, 34, 198, 205, 207, 211–213, 293

You might also like