Pharmaco Kinetics
Pharmaco Kinetics
Pharmacokinetics (from Ancient Greek pharmakon "drug" and kinetikos "moving, putting in motion"; see
chemical kinetics), sometimes abbreviated as PK, is a branch of pharmacology dedicated to determine the fate
of substances administered to a living organism. The substances of interest include any chemical xenobiotic
such as: pharmaceutical drugs, pesticides, food additives, cosmetics, etc. It attempts to analyze chemical
metabolism and to discover the fate of a chemical from the moment that it is administered up to the point at
which it is completely eliminated from the body. Pharmacokinetics is the study of how an organism affects a
drug, whereas pharmacodynamics (PD) is the study of how the drug affects the organism. Both together
influence dosing, benefit, and adverse effects, as seen in PK/PD models.
IUPAC definition
Contents Pharmacokinetics:
Overview
Pharmacokinetics describes how the body affects a specific xenobiotic/chemical after administration through
the mechanisms of absorption and distribution, as well as the metabolic changes of the substance in the body
(e.g. by metabolic enzymes such as cytochrome P450 or glucuronosyltransferase enzymes), and the effects and
routes of excretion of the metabolites of the drug.[2] Pharmacokinetic properties of chemicals are affected by
the route of administration and the dose of administered drug. These may affect the absorption rate.[3]
Models have been developed to simplify conceptualization of the
many processes that take place in the interaction between an organism
and a chemical substance. One of these, the multi-compartmental
model, is the most commonly used approximations to reality;
however, the complexity involved in adding parameters with that
modelling approach means that monocompartmental models and
above all two compartmental models are the most-frequently used.
The various compartments that the model is divided into are
commonly referred to as the ADME scheme (also referred to as
LADME if liberation is included as a separate step from absorption):
The two phases of metabolism and excretion can also be grouped together under the title elimination. The
study of these distinct phases involves the use and manipulation of basic concepts in order to understand the
process dynamics. For this reason, in order to fully comprehend the kinetics of a drug it is necessary to have
detailed knowledge of a number of factors such as: the properties of the substances that act as excipients, the
characteristics of the appropriate biological membranes and the way that substances can cross them, or the
characteristics of the enzyme reactions that inactivate the drug.
All these concepts can be represented through mathematical formulas that have a corresponding graphical
representation. The use of these models allows an understanding of the characteristics of a molecule, as well as
how a particular drug will behave given information regarding some of its basic characteristics such as its acid
dissociation constant (pKa), bioavailability and solubility, absorption capacity and distribution in the organism.
The model outputs for a drug can be used in industry (for example, in calculating bioequivalence when
designing generic drugs) or in the clinical application of pharmacokinetic concepts. Clinical pharmacokinetics
provides many performance guidelines for effective and efficient use of drugs for human-health professionals
and in veterinary medicine.
Metrics
The following are the most commonly measured pharmacokinetic metrics:[6] The units of the dose in the table
are expressed in moles (mol) and molar (M). To express the metrics of the table in units of mass, instead of
Amount of substance, simply replace 'mol' with 'g' and 'M' with 'g/dm3'. Similarly, other units in the table may
be expressed in units of an equivalent dimension by scaling.
Pharmacokinetic metrics
Worked
Characteristic Description Symbol Unit Formula example
value
Amount of drug
Dose Design parameter 500 mmol
administered.
Time between drug
Dosing interval Design parameter 24 h
dose administrations.
The peak plasma
Cmax concentration of a drug Direct measurement 60.9 mmol/L
after administration.
tmax Time to reach Cmax . Direct measurement 3.9 h
Rate of infusion
Infusion rate required to balance 50 mmol/h
elimination.
In pharmacokinetics, steady state refers to the situation where the overall intake of a drug is fairly in dynamic
equilibrium with its elimination. In practice, it is generally considered that steady state is reached when a time
of 3 to 5 times the half-life for a drug after regular dosing is started.
Pharmacokinetic models
Pharmacokinetic modelling is performed
by noncompartmental or compartmental
methods. Noncompartmental methods
estimate the exposure to a drug by
estimating the area under the curve of a
concentration-time graph. Compartmental
methods estimate the concentration-time
graph using kinetic models.
Noncompartmental methods are often
more versatile in that they do not assume
any specific compartmental model and
produce accurate results also acceptable
for bioequivalence studies. The final
outcome of the transformations that a
drug undergoes in an organism and the
rules that determine this fate depend on a The time course of drug plasma concentrations over 96 hours
number of interrelated factors. A number following oral administrations every 24 hours. Note that in steady
of functional models have been state and in linear pharmacokinetics AUCτ=AUC∞.[7] Steady state
developed in order to simplify the study is reached after about 5 × 12 = 60 hours. The graph depicts a
of pharmacokinetics. These models are typical time course of drug plasma concentration and illustrates
based on a consideration of an organism main pharmacokinetic metrics
as a number of related compartments.
The simplest idea is to think of an
organism as only one homogenous compartment. This monocompartmental model presupposes that blood
plasma concentrations of the drug are a true reflection of the drug's concentration in other fluids or tissues and
that the elimination of the drug is directly proportional to the drug's concentration in the organism (first order
kinetics).
However, these models do not always truly reflect the real situation within an organism. For example, not all
body tissues have the same blood supply, so the distribution of the drug will be slower in these tissues than in
others with a better blood supply. In addition, there are some tissues (such as the brain tissue) that present a real
barrier to the distribution of drugs, that can be breached with greater or lesser ease depending on the drug's
characteristics. If these relative conditions for the different tissue types are considered along with the rate of
elimination, the organism can be considered to be acting like two compartments: one that we can call the
central compartment that has a more rapid distribution, comprising organs and systems with a well-developed
blood supply; and a peripheral compartment made up of organs with a lower blood flow. Other tissues, such
as the brain, can occupy a variable position depending on a drug's ability to cross the barrier that separates the
organ from the blood supply.
This two compartment model will vary depending on which compartment elimination occurs in. The most
common situation is that elimination occurs in the central compartment as the liver and kidneys are organs with
a good blood supply. However, in some situations it may be that elimination occurs in the peripheral
compartment or even in both. This can mean that there are three possible variations in the two compartment
model, which still do not cover all possibilities.[8]
This model may not be applicable in situations where some of the enzymes responsible for metabolizing the
drug become saturated, or where an active elimination mechanism is present that is independent of the drug's
plasma concentration. In the real world each tissue will have its own distribution characteristics and none of
them will be strictly linear. If we label the drug's volume of distribution within the organism VdF and its
volume of distribution in a tissue VdT the former will be described by an equation that takes into account all
the tissues that act in different ways, that is:
This represents the multi-compartment model with a number of curves that express complicated equations in
order to obtain an overall curve. A number of computer programs have been developed to plot these
equations.[8] However complicated and precise this model may be, it still does not truly represent reality
despite the effort involved in obtaining various distribution values for a drug. This is because the concept of
distribution volume is a relative concept that is not a true reflection of reality. The choice of model therefore
comes down to deciding which one offers the lowest margin of error for the drug involved.
Noncompartmental analysis
Single-compartment model
Linear pharmacokinetics is so-called because the graph of the relationship between the various factors
involved (dose, blood plasma concentrations, elimination, etc.) gives a straight line or an approximation to one.
For drugs to be effective they need to be able to move rapidly from blood plasma to other body fluids and
tissues.
Multi-compartmental models
The graph for the non-linear relationship between the various factors is
represented by a curve; the relationships between the factors can then be
found by calculating the dimensions of different areas under the curve.
The models used in non-linear pharmacokinetics are largely based on
Michaelis–Menten kinetics. A reaction's factors of non-linearity include
the following:
Bioavailability
At a practical level, a drug's bioavailability can be defined as the
proportion of the drug that reaches its site of action. From this
perspective the intravenous administration of a drug provides the
greatest possible bioavailability, and this method is considered to yield Plasma drug concentration vs time
a bioavailability of 1 (or 100%). Bioavailability of other delivery after an IV dose
methods is compared with that of intravenous injection (absolute
bioavailability) or to a standard value related to other
delivery methods in a particular study (relative
bioavailability).
Therefore, if a drug has a bioavailability of 0.8 (or 80%) and it is administered in a dose of 100 mg, the
equation will demonstrate the following:
De = 0.8 × 100 mg = 80 mg
That is the 100 mg administered represents a blood plasma concentration of 80 mg that has the capacity to
have a pharmaceutical effect.
This concept depends on a series of factors inherent to each drug, such as:[11]
Pharmaceutical form
Chemical form
Route of administration
Stability
Metabolism
These concepts, which are discussed in detail in their respective titled articles, can be mathematically
quantified and integrated to obtain an overall mathematical equation:
where is the drug's rate of administration and is the rate at which the absorbed drug reaches the
circulatory system.
Finally, using the Henderson-Hasselbalch equation, and knowing the drug's (pH at which there is an
equilibrium between its ionized and non ionized molecules), it is possible to calculate the non ionized
concentration of the drug and therefore the concentration that will be subject to absorption:
When two drugs have the same bioavailability, they are said to be biological equivalents or bioequivalents.
This concept of bioequivalence is important because it is currently used as a yardstick in the authorization of
generic drugs in many countries.
LADME
A number of phases occur once the drug enters into contact with the organism, these are described using the
acronym LADME:
Some textbooks combine the first two phases as the drug is often administered in an active form, which means
that there is no liberation phase. Others include a phase that combines distribution, metabolism and excretion
into a disposition phase. Other authors include the drug's toxicological aspect in what is known as ADME-Tox
or ADMET.
Each of the phases is subject to physico-chemical interactions between a drug and an organism, which can be
expressed mathematically. Pharmacokinetics is therefore based on mathematical equations that allow the
prediction of a drug's behavior and which place great emphasis on the relationships between drug plasma
concentrations and the time elapsed since the drug's administration.
Analysis
Bioanalytical methods
Bioanalytical methods are necessary to construct a concentration-time profile. Chemical techniques are
employed to measure the concentration of drugs in biological matrix, most often plasma. Proper bioanalytical
methods should be selective and sensitive. For example, microscale thermophoresis can be used to quantify
how the biological matrix/liquid affects the affinity of a drug to its target.[12][13]
Mass spectrometry
Pharmacokinetics is often studied using mass spectrometry because of the complex nature of the matrix (often
plasma or urine) and the need for high sensitivity to observe concentrations after a low dose and a long time
period. The most common instrumentation used in this application is LC-MS with a triple quadrupole mass
spectrometer. Tandem mass spectrometry is usually employed for added specificity. Standard curves and
internal standards are used for quantitation of usually a single pharmaceutical in the samples. The samples
represent different time points as a pharmaceutical is administered and then metabolized or cleared from the
body. Blank samples taken before administration are important in determining background and ensuring data
integrity with such complex sample matrices. Much attention is paid to the linearity of the standard curve;
however it is common to use curve fitting with more complex functions such as quadratics since the response
of most mass spectrometers is not linear across large concentration ranges.[14][15][16]
There is currently considerable interest in the use of very high sensitivity mass spectrometry for microdosing
studies, which are seen as a promising alternative to animal experimentation.[17] Recent studies show that
Secondary electrospray ionization (SESI-MS) can be used in drug monitoring, presenting the advantage of
avoiding animal sacrifice.[18]
Population pharmacokinetics
Population pharmacokinetics is the study of the sources and correlates of variability in drug concentrations
among individuals who are the target patient population receiving clinically relevant doses of a drug of
interest.[19][20][21] Certain patient demographic, pathophysiological, and therapeutical features, such as body
weight, excretory and metabolic functions, and the presence of other therapies, can regularly alter dose-
concentration relationships and can explain variability in exposures. For example, steady-state concentrations
of drugs eliminated mostly by the kidney are usually greater in patients suffering from kidney failure than they
are in patients with normal kidney function receiving the same drug dosage. Population pharmacokinetics
seeks to identify the measurable pathophysiologic factors and explain sources of variability that cause changes
in the dose-concentration relationship and the extent of these changes so that, if such changes are associated
with clinically relevant and significant shifts in exposures that impact the therapeutic index, dosage can be
appropriately modified. An advantage of population pharmacokinetic modelling is its ability to analyse sparse
data sets (sometimes only one concentration measurement per patient is available).
Clinical pharmacokinetics
Clinical pharmacokinetics (arising from the clinical use of population pharmacokinetics) is the direct
application to a therapeutic situation of knowledge regarding a drug's pharmacokinetics and the characteristics
of a population that a patient belongs to (or can be ascribed to).
An example is the relaunch of the use of ciclosporin as an immunosuppressor to facilitate organ transplant. The
drug's therapeutic properties were initially demonstrated, but it was almost never used after it was found to
cause nephrotoxicity in a number of patients.[22] However, it was then realized that it was possible to
individualize a patient's dose of ciclosporin by analysing the patients plasmatic concentrations
Drugs where pharmacokinetic monitoring is recommended
Antiepileptic Cardioactive Immunosuppressor Antibiotic
Phenytoin
Carbamazepine Ciclosporin
Gentamicin
Valproic acid Tacrolimus
Digoxin Tobramycin
Lamotrigine Sirolimus
Lidocaine Amikacin
Ethosuximide Everolimus
Vancomycin
Phenobarbital Mycophenolate
Primidone
(pharmacokinetic monitoring). This practice has allowed this drug to be used again and has facilitated a great
number of organ transplants.
Clinical monitoring is usually carried out by determination of plasma concentrations as this data is usually the
easiest to obtain and the most reliable. The main reasons for determining a drug's plasma concentration
include:[23]
Ecotoxicology
Ecotoxicology is the branch of science that deals with the nature, effects, and interactions of substances that
are harmful to the environment.[24][25]
See also
Blood alcohol Idiosyncratic drug Bioequivalence
concentration reaction Generic drugs
Biological half-life Drug interaction Physiologically based
Bioavailability Patlak plot pharmacokinetic modelling
Enzyme kinetics Pharmacometrics Plateau principle
Pharmacodynamics Pharmacy Toxicokinetics
References
1. Nordberg M, Duffus J, Templeton DM (1 January 2004). "Glossary of terms used in
toxicokinetics (IUPAC Recommendations 2003)". Pure and Applied Chemistry. 76 (5): 1033–
1082. doi:10.1351/pac200476051033 (https://doi.org/10.1351%2Fpac200476051033).
2. Pharmacokinetics. (2006). In Mosby's Dictionary of Medicine, Nursing & Health Professions.
Philadelphia, PA: Elsevier Health Sciences. Retrieved December 11, 2008, from
http://www.credoreference.com/entry/6686418
3. Knights K, Bryant B (2002). Pharmacology for Health Professionals. Amsterdam: Elsevier.
ISBN 0-7295-3664-5.
4. Koch HP, Ritschel WA (1986). "Liberation". Synopsis der Biopharmazie und Pharmakokinetik
(in German). Landsberg, München: Ecomed. pp. 99–131. ISBN 3-609-64970-4.
5. Ruiz-Garcia A, Bermejo M, Moss A, Casabo VG (February 2008). "Pharmacokinetics in drug
discovery". Journal of Pharmaceutical Sciences. 97 (2): 654–90. doi:10.1002/jps.21009 (https://
doi.org/10.1002%2Fjps.21009). PMID 17630642 (https://pubmed.ncbi.nlm.nih.gov/17630642).
6. AGAH working group PHARMACOKINETICS (2004-02-16). "Collection of terms, symbols,
equations, and explanations of common pharmacokinetic and pharmacodynamic parameters
and some statistical functions" (https://web.archive.org/web/20160508185420/http://www.agah.
eu/fileadmin/_migrated/content_uploads/PK-glossary_PK_working_group_2004.pdf) (PDF).
Arbeitsgemeinschaft für Angewandte Humanpharmakologie (AGAH) (Association for Applied
Human Pharmacology). Archived from the original (http://www.agah.eu/fileadmin/_migrated/con
tent_uploads/PK-glossary_PK_working_group_2004.pdf) (PDF) on 2016-05-08. Retrieved
2011-04-04.
7. Pharmacokinetics Online Resource (https://sepia.unil.ch/pharmacology/index.php?
id=100)University of Lausanne Faculty of Biology and Medicine (FBM)
8. Milo Gibaldi, Donald Perrier. FarmacocinéticaReverté 1982 pages 1–10. ISBN 84-291-5535-X,
9788429155358
9. Gill SC, Moon-Mcdermott L, Hunt TL, Deresinski S, Blaschke T, Sandhaus RA (Sep 1999).
"Phase I Pharmacokinetics of Liposomal Amikacin (MiKasome) in Human Subjects: Dose
Dependence and Urinary Clearance" (http://gateway.nlm.nih.gov/MeetingAbstracts/ma?f=1022
44695.html). Abstr Intersci Conf Antimicrob Agents Chemother. 39: 33 (abstract no. 1195).
10. Weiner D, Gabrielsson J (2000). "PK24 – Non-linear kinetics – flow II" (https://books.google.co
m/?id=K-gT2_Ukk0QC&lpg=PA529&dq=pharmacokinetic%20alpha%20beta%20gamma%20p
hase&pg=PA528#v=onepage&q&f=false). Pharmacokinetic/pharmacodynamic data analysis:
concepts and applications. Apotekarsocieteten. pp. 527–36. ISBN 91-86274-92-9.
11. Michael E. Winter, Mary Anne Koda-Kimple, Lloyd Y. Young, Emilio Pol Yanguas
Farmacocinética clínica básica Ediciones Díaz de Santos, 1994 pgs. 8–14 ISBN 84-7978-147-
5, 9788479781477 (in Spanish)
12. Baaske P, Wienken CJ, Reineck P, Duhr S, Braun D (March 2010). "Optical thermophoresis for
quantifying the buffer dependence of aptamer binding". Angewandte Chemie. 49 (12): 2238–
41. doi:10.1002/anie.200903998 (https://doi.org/10.1002%2Fanie.200903998).
PMID 20186894 (https://pubmed.ncbi.nlm.nih.gov/20186894). Lay summary (http://www.physor
g.com/news186225693.html) – Phsyorg.com.
13. Wienken CJ, Baaske P, Rothbauer U, Braun D, Duhr S (October 2010). "Protein-binding
assays in biological liquids using microscale thermophoresis" (https://doi.org/10.1038/ncomms
1093). Nature Communications. 1 (7): 100. Bibcode:2010NatCo...1..100W (https://ui.adsabs.har
vard.edu/abs/2010NatCo...1..100W). doi:10.1038/ncomms1093 (https://doi.org/10.1038%2Fnco
mms1093). PMID 20981028 (https://pubmed.ncbi.nlm.nih.gov/20981028).
14. Hsieh Y, Korfmacher WA (June 2006). "Increasing speed and throughput when using HPLC-
MS/MS systems for drug metabolism and pharmacokinetic screening". Current Drug
Metabolism. 7 (5): 479–89. doi:10.2174/138920006777697963 (https://doi.org/10.2174%2F138
920006777697963). PMID 16787157 (https://pubmed.ncbi.nlm.nih.gov/16787157).
15. Covey TR, Lee ED, Henion JD (October 1986). "High-speed liquid chromatography/tandem
mass spectrometry for the determination of drugs in biological samples". Analytical Chemistry.
58 (12): 2453–60. doi:10.1021/ac00125a022 (https://doi.org/10.1021%2Fac00125a022).
PMID 3789400 (https://pubmed.ncbi.nlm.nih.gov/3789400).
16. Covey TR, Crowther JB, Dewey EA, Henion JD (February 1985). "Thermospray liquid
chromatography/mass spectrometry determination of drugs and their metabolites in biological
fluids". Analytical Chemistry. 57 (2): 474–81. doi:10.1021/ac50001a036 (https://doi.org/10.102
1%2Fac50001a036). PMID 3977076 (https://pubmed.ncbi.nlm.nih.gov/3977076).
17. Committee for Medicinal Products for Human Use (CHMP) (December 2009). "ICH guideline
M3(R2) on non-clinical safety studies for the conduct of human clinical trials and marketing
authorisation for pharmaceuticals" (http://www.ema.europa.eu/docs/en_GB/document_library/S
cientific_guideline/2009/09/WC500002720.pdf) (PDF). European Medicines Agency,
Evaluation of Medicines for Human Use. EMA/CPMP/ICH/286/1995. Retrieved 4 May 2013.
18. Li, Xue; Martinez-Lozano Sinues, Pablo; Dallmann, Robert; Bregy, Lukas; Hollmén, Maija;
Proulx, Steven; Brown, Steven A.; Detmar, Michael; Kohler, Malcolm; Zenobi, Renato (2015-06-
26). "Drug Pharmacokinetics Determined by Real-Time Analysis of Mouse Breath".
Angewandte Chemie International Edition. 54 (27): 7815–7818. doi:10.1002/anie.201503312
(https://doi.org/10.1002%2Fanie.201503312). hdl:20.500.11850/102558 (https://hdl.handle.net/
20.500.11850%2F102558). PMID 26015026 (https://pubmed.ncbi.nlm.nih.gov/26015026).
19. Sheiner LB, Rosenberg B, Marathe VV (October 1977). "Estimation of population
characteristics of pharmacokinetic parameters from routine clinical data". Journal of
Pharmacokinetics and Biopharmaceutics. 5 (5): 445–79. doi:10.1007/BF01061728 (https://doi.o
rg/10.1007%2FBF01061728). PMID 925881 (https://pubmed.ncbi.nlm.nih.gov/925881).
20. Sheiner LB, Beal S, Rosenberg B, Marathe VV (September 1979). "Forecasting individual
pharmacokinetics". Clinical Pharmacology and Therapeutics. 26 (3): 294–305.
doi:10.1002/cpt1979263294 (https://doi.org/10.1002%2Fcpt1979263294). PMID 466923 (http
s://pubmed.ncbi.nlm.nih.gov/466923).
21. Bonate PL (October 2005). "Recommended reading in population pharmacokinetic
pharmacodynamics" (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2750974). The AAPS
Journal. 7 (2): E363–73. doi:10.1208/aapsj070237 (https://doi.org/10.1208%2Faapsj070237).
PMC 2750974 (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2750974). PMID 16353916 (http
s://pubmed.ncbi.nlm.nih.gov/16353916).
22. O'Valle, F.; García del Moral, R.; Andujar, M. (1995). "Mecanismos de nefrotoxicidad por
ciclosporina A a nivel celular" (http://www.revistanefrologia.com/es-publicacion-nefrologia-artic
ulo-mecanismos-nefrotoxicidad-por-ciclosporina-a-nivel-celular-X0211699595022690).
Nefrologia (in Spanish). 15 Supplement 1.
23. Joaquín Herrera Carranza Manual de farmacia clínica y Atención Farmacéutica (https://books.g
oogle.com/books?id=7Vw7a4wBzRgC) (in Spanish). Published by Elsevier España, 2003;
page 159. ISBN 84-8174-658-4
24. Jager T, Albert C, Preuss TG, Ashauer R (April 2011). "General unified threshold model of
survival--a toxicokinetic-toxicodynamic framework for ecotoxicology". Environmental Science &
Technology. 45 (7): 2529–40. Bibcode:2011EnST...45.2529J (https://ui.adsabs.harvard.edu/ab
s/2011EnST...45.2529J). doi:10.1021/es103092a (https://doi.org/10.1021%2Fes103092a).
PMID 21366215 (https://pubmed.ncbi.nlm.nih.gov/21366215).
25. Ashauer R. "Toxicokinetic-Toxicodynamic Models – Ecotoxicology and Models" (https://web.arc
hive.org/web/20120405200941/http://www.ecotoxmodels.org/research-publications-projects/tox
icokinetic-toxicodynamic-models/). Swiss Federal Institute of Aquatic Science and Technology.
Archived from the original (http://www.ecotoxmodels.org/research-publications-projects/toxicoki
netic-toxicodynamic-models/) on 2012-04-05. Retrieved 2011-12-03.
External links
Software
Noncompartmental
Compartment based
Physiologically based
Population PK
Educational centres
Global centres with the highest profiles for providing in-depth training include the Universities of Buffalo,
Florida, Gothenburg, Leiden, Otago, San Francisco, Beijing, Tokyo, Uppsala, Washington, Manchester,
Monash University, and University of Sheffield.[1]
Text is available under the Creative Commons Attribution-ShareAlike License; additional terms may apply. By using this
site, you agree to the Terms of Use and Privacy Policy. Wikipedia® is a registered trademark of the Wikimedia
Foundation, Inc., a non-profit organization.