Therapeutic drug monitoring

Last updated

Therapeutic drug monitoring (TDM) is a branch of clinical chemistry and clinical pharmacology that specializes in the measurement of medication levels in blood. Its main focus is on drugs with a narrow therapeutic range, i.e. drugs that can easily be under- or overdosed. [1] TDM aimed at improving patient care by individually adjusting the dose of drugs for which clinical experience or clinical trials have shown it improved outcome in the general or special populations. It can be based on a a priori pharmacogenetic, demographic and clinical information, and/or on the a posteriori measurement of blood concentrations of drugs (pharmacokinetic monitoring) or biological surrogate or end-point markers of effect (pharmacodynamic monitoring). [2]

Contents

There are numerous variables that influence the interpretation of drug concentration data: time, route and dose of drug given, time of blood sampling, handling and storage conditions, precision and accuracy of the analytical method, validity of pharmacokinetic models and assumptions, co-medications and, last but not least, clinical status of the patient (i.e. disease, renal/hepatic status, biologic tolerance to drug therapy, etc.). [3]

Many different professionals (physicians, clinical pharmacists, nurses, medical laboratory scientists, etc.) are involved with the various elements of drug concentration monitoring, which is a truly multidisciplinary process. Because failure to properly carry out any one of the components can severely affect the usefulness of using drug concentrations to optimize therapy, an organized approach to the overall process is critical. [3]

A priori therapeutic drug monitoring

A priori TDM consists of determining the initial dose regimen to be given to a patient, based on clinical endpoint and on established population pharmacokinetic-pharmacodynamic (PK/PD) relationships. These relationships help to identify sub-populations of patients with different dosage requirements, by utilizing demographic data, clinical findings, clinical chemistry results, and/or, when appropriate, pharmacogenetic characteristics. [2]

A posteriori therapeutic drug monitoring

The concept of a posteriori TDM corresponds to the usual meaning of TDM in medical practice, which refers to the readjustment of the dosage of a given treatment in response to the measurement of an appropriate marker of drug exposure or effect. TDM encompasses all aspects of this feedback control, namely: [2]

Characteristics of drugs candidate to therapeutic drug monitoring

In pharmacotherapy, many medications are used without monitoring of blood levels, as their dosage can generally be varied according to the clinical response that a patient gets to that substance. For certain drugs, this is impracticable, while insufficient levels will lead to undertreatment or resistance, and excessive levels can lead to toxicity and tissue damage.

Indications in favor of therapeutic drug monitoring include: [4] [5]

TDM determinations are also used to detect and diagnose poisoning with drugs, should the suspicion arise.

Examples of drugs widely analysed for therapeutic drug monitoring: [1]

TDM increasingly proposed for a number of therapeutic drugs, e.g. many antibiotics, small molecule tyrosine kinase inhibitors and other targeted anticancer agents, TNF inhibitors and other biological agents, antifungal agents, antiretroviral agents used in HIV infection, psychiatric drugs [7] etc.

Practice of therapeutic drug monitoring

Automated analytical methods such as enzyme multiplied immunoassay technique or fluorescence polarization immunoassay are widely available in medical laboratories for drugs frequently measured in practice. Nowadays, most other drugs can be readily measured in blood or plasma using versatile methods such as liquid chromatography–mass spectrometry or gas chromatography–mass spectrometry, which progressively replaced high-performance liquid chromatography. Yet, TDM is not limited to the provision of precise and accurate concentration measurement results, it also involves appropriate medical interpretation, based on robust scientific knowledge.

In order to guarantee the quality of this clinical interpretation, it is essential that the sample be taken under good conditions: i.e., preferably under a stable dosage, at a standardized sampling time (often at the end of a dosing interval), excluding any source of bias (sample contamination or dilution, analytical interferences) and having carefully recorded the sampling time, the last dose intake time, the current dosage and the influential patient's characteristics.

TDM interpretation: an anticancer drug is given to a patient at a dosage of 400 mg every day at 8:00 am. A TDM sample is obtained at 6:00 am, showing a drug concentration of 0.46 mg/L. 1) Regarding "normality", the result is around the 25th percentile, suggesting a rather high drug clearance in this patient. 2) Regarding "appropriateness", the result suggests that the patient's most likely concentration curve (green dashed line) passes below the acceptance range of 0.75 to 1.5 mg/L at trough, raising concerns about treatment efficacy. 3) Regarding dosage adjustment to recommend, this TDM result suggests that a doubled dose of 800 mg daily might be suitable to drive the concentration curve close to target (blue dotted line). Note that this interpretation assumes that the patient's adherence to prescription is good, and that the sample measurement is accurate. TDM interpretation.tif
TDM interpretation: an anticancer drug is given to a patient at a dosage of 400 mg every day at 8:00 am. A TDM sample is obtained at 6:00 am, showing a drug concentration of 0.46 mg/L. 1) Regarding “normality”, the result is around the 25th percentile, suggesting a rather high drug clearance in this patient. 2) Regarding “appropriateness”, the result suggests that the patient's most likely concentration curve (green dashed line) passes below the acceptance range of 0.75 to 1.5 mg/L at trough, raising concerns about treatment efficacy. 3) Regarding dosage adjustment to recommend, this TDM result suggests that a doubled dose of 800 mg daily might be suitable to drive the concentration curve close to target (blue dotted line). Note that this interpretation assumes that the patient's adherence to prescription is good, and that the sample measurement is accurate.

The interpretation of a drug concentration result goes through the following stages : [8]

  1. Determine whether the observed concentration is in the “normal range” expected under the dosage administered, taking into account the patient's individual characteristics. This requires referring to population pharmacokinetic studies of the drug in consideration.
  2. Determine whether the patient's concentration profile is close to the “exposure target” associated with the best trade-off between probability of therapeutic success and risk of toxicity. This refers to clinical pharmacodynamic knowledge describing dose-concentration-response relationships among treated patients.
  3. If the observed concentration is plausible but far from the suitable level, determine how to adjust the dosage to drive the concentration curve close to target. Several approaches exist for this, from the easiest “rule of three” to sophisticated computer-assisted calculations implementing Bayesian inference algorithms based on population pharmacokinetics. [9]

Ideally, the usefulness of a TDM strategy should be confirmed through an evidence-based approach involving the performance of well-designed controlled clinical trials. In practice however, TDM has undergone formal clinical evaluation only for a limited number of drugs to date, and much of its development rests on empirical foundations.

Point-of-care tests for an easy performance of TDM at the medical practice are under elaboration. [10]

Related Research Articles

<i>In vitro</i> Latin term meaning outside a natural biological environment

In vitro studies are performed with microorganisms, cells, or biological molecules outside their normal biological context. Colloquially called "test-tube experiments", these studies in biology and its subdisciplines are traditionally done in labware such as test tubes, flasks, Petri dishes, and microtiter plates. Studies conducted using components of an organism that have been isolated from their usual biological surroundings permit a more detailed or more convenient analysis than can be done with whole organisms; however, results obtained from in vitro experiments may not fully or accurately predict the effects on a whole organism. In contrast to in vitro experiments, in vivo studies are those conducted in living organisms, including humans, and whole plants.

<span class="mw-page-title-main">Pharmacology</span> Branch of biology concerning drugs

Pharmacology is a branch of medicine, biology, and pharmaceutical sciences concerned with drug or medication action, where a drug may be defined as any artificial, natural, or endogenous molecule which exerts a biochemical or physiological effect on the cell, tissue, organ, or organism. More specifically, it is the study of the interactions that occur between a living organism and chemicals that affect normal or abnormal biochemical function. If substances have medicinal properties, they are considered pharmaceuticals.

The therapeutic index is a quantitative measurement of the relative safety of a drug. It is a comparison of the amount of a therapeutic agent that causes the therapeutic effect to the amount that causes toxicity. The related terms therapeutic window or safety window refer to a range of doses optimized between efficacy and toxicity, achieving the greatest therapeutic benefit without resulting in unacceptable side-effects or toxicity.

<span class="mw-page-title-main">Pharmacodynamics</span> Area of Academic Study

Pharmacodynamics (PD) is the study of the biochemical and physiologic effects of drugs. The effects can include those manifested within animals, microorganisms, or combinations of organisms.

In pharmacology, bioavailability is a subcategory of absorption and is the fraction (%) of an administered drug that reaches the systemic circulation.

<span class="mw-page-title-main">Bioequivalence</span>

Bioequivalence is a term in pharmacokinetics used to assess the expected in vivo biological equivalence of two proprietary preparations of a drug. If two products are said to be bioequivalent it means that they would be expected to be, for all intents and purposes, the same.

<span class="mw-page-title-main">Pharmacogenomics</span> Study of the role of the genome in drug response

Pharmacogenomics is the study of the role of the genome in drug response. Its name reflects its combining of pharmacology and genomics. Pharmacogenomics analyzes how the genetic makeup of an individual affects their response to drugs. It deals with the influence of acquired and inherited genetic variation on drug response in patients by correlating DNA mutations with pharmacokinetic, pharmacodynamic, and/or immunogenic endpoints.

<span class="mw-page-title-main">Zopiclone</span> Hypnotic medication

Zopiclone, sold under the brand name Imovane among others, is a nonbenzodiazepine used to treat difficulty sleeping. Zopiclone is molecularly distinct from benzodiazepine drugs and is classed as a cyclopyrrolone. However, zopiclone increases the normal transmission of the neurotransmitter gamma-aminobutyric acid (GABA) in the central nervous system, via modulating GABAA receptors similarly to the way benzodiazepine drugs do.

<span class="mw-page-title-main">Perhexiline</span>

Perhexiline (Pexsig) is a prophylactic antianginal agent used primarily in Australia and New Zealand. Perhexiline is thought to act by inhibiting mitochondrial carnitine palmitoyltransferase-1. This shifts myocardial metabolism from fatty acid to glucose utilisation which results in increased ATP production for the same O2 consumption and consequently increases myocardial efficiency. Its clinical use has been limited by its narrow therapeutic index and high inter- and intra-individual pharmacokinetic variability. It was outlawed in many countries due to its adverse effects on poor metabolisers (PM). The product has been reintroduced for patients who have contraindications, or have not responded to other treatments for angina.

Biological half-life is the time taken for concentration of a biological substance to decrease from its maximum concentration (Cmax) to half of Cmax in the blood plasma, and is denoted by the abbreviation .

<span class="mw-page-title-main">Tolcapone</span> Chemical compound

Tolcapone, sold under the brand name Tasmar, is a medication used to treat Parkinson's disease (PD). It is a selective, potent and reversible nitrocatechol-type inhibitor of the enzyme catechol-O-methyltransferase (COMT). It has demonstrated significant liver toxicity, which has led to suspension of marketing authorisations in a number of countries.

<span class="mw-page-title-main">Phenprocoumon</span> Drug

Phenprocoumon is a long-acting blood thinner drug to be taken by mouth, and a derivative of coumarin. It acts as a vitamin K antagonist and inhibits blood clotting (coagulation) by blocking synthesis of coagulation factors II, VII, IX and X. It is used for the prophylaxis and treatment of thromboembolic disorders such as heart attacks and pulmonary (lung) embolism. The most common adverse effect is bleeding. The drug interacts with a large number of other medications, including aspirin and St John's Wort. It is the standard coumarin used in Germany, Austria, and other European countries.

<span class="mw-page-title-main">Acecainide</span>

Acecainide is an antiarrhythmic drug. Chemically, it is the N-acetylated metabolite of procainamide. It is a Class III antiarrhythmic agent, whereas procainamide is a Class Ia antiarrhythmic drug. It is only partially as active as procainamide; when checking levels, both must be included in the final calculation.

Pharmacokinetics, sometimes abbreviated as PK, is a branch of pharmacology dedicated to determining 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.

In the field of pharmacokinetics, the area under the curve (AUC) is the definite integral of the concentration of a drug in blood plasma as a function of time. In practice, the drug concentration is measured at certain discrete points in time and the trapezoidal rule is used to estimate AUC.

The Infectious Disease Pharmacokinetics Laboratory (IDPL) is a research facility that is affiliated with the College of Pharmacy at the University of Florida.

PK/PD modeling is a technique that combines the two classical pharmacologic disciplines of pharmacokinetics and pharmacodynamics. It integrates a pharmacokinetic and a pharmacodynamic model component into one set of mathematical expressions that allows the description of the time course of effect intensity in response to administration of a drug dose. PK/PD modeling is related to the field of pharmacometrics.

The Pharmacogenomics Knowledgebase (PharmGKB) is a publicly available, online knowledge base responsible for the aggregation, curation, integration and dissemination of knowledge regarding the impact of human genetic variation on drug response. It is funded by the National Institutes of Health (NIH) National Institute of General Medical Sciences (NIGMS), and is a partner of the NIH Pharmacogenomics Research Network (PGRN). It has been managed at Stanford University since its inception in 2000.

Leon Aarons is an Australian pharmacist who researches and teaches in the areas of pharmacodynamics and pharmacokinetics. He lives in the United Kingdom and from 1976 has been a professor of pharmacometrics at the University of Manchester. In the interest of promoting the effective development of drugs, the main focus of his work is optimizing pharmacological models, the design of clinical studies, and data analysis and interpretation in the field of population pharmacokinetics. From 1985 to 2010 Aarons was an editor emeritus of the Journal of Pharmacokinetics and Pharmacodynamics and is a former executive editor of the British Journal of Clinical Pharmacology.

<span class="mw-page-title-main">Cancer pharmacogenomics</span>

Cancer pharmacogenomics is the study of how variances in the genome influences an individual’s response to different cancer drug treatments. It is a subset of the broader field of pharmacogenomics, which is the area of study aimed at understanding how genetic variants influence drug efficacy and toxicity.

References

  1. 1 2 Marshall WJ, Bangert SK. Clinical Chemistry, 6th Edition. Edinburgh, London: Mosby Elsevier. 2008. ISBN   978-0723434559
  2. 1 2 3 IATDMCT Executive Committee. "Definition of TDM", 2004, accessed May 5, 2020.
  3. 1 2 Burton ME, Shaw LM, Schentag JJ, Evans, WE. Applied Pharmacokinetics & Pharmacodynamics, 4th Edition. Baltimore, Philadelphia: Lippincott Williams & Wilkins. 2006. ISBN   978-0781744317
  4. Ensom MH, Davis GA, Cropp CD, Ensom RJ (April 1998). "Clinical pharmacokinetics in the 21st century. Does the evidence support definitive outcomes?". Clinical Pharmacokinetics. 34 (4): 265–79. doi:10.2165/00003088-199834040-00001. PMID   9571300. S2CID   37921089.
  5. Gross AS (April 1998). "Best practice in therapeutic drug monitoring". British Journal of Clinical Pharmacology. 46 (2): 95–9. doi:10.1046/j.1365-2125.1998.00770.x. PMC   2014621 . PMID   11564048.
  6. Holford NH, Buclin T (May 2012). "Safe and effective variability -- a criterion for dose individualization". Therapeutic Drug Monitoring. 34 (5): 565–8. doi:10.1097/FTD.0b013e31826aabc3. PMID   22960736. S2CID   5229360.
  7. Schoretsanitis G, Paulzen M, Unterecker S, Schwarz M, Conca A, Zernig G, Gründer G, Haen E, Baumann P, Bergemann N, Clement HW, Domschke K, Eckermann G, Egberts K, Gerlach M, Greiner C, Havemann-Reinecke U, Hefner G, Helmer R, Janssen G, Jaquenoud-Sirot E, Laux G, Messer T, Mössner R, Müller MJ, Pfuhlmann B, Riederer P, Saria A, Schoppek B, Silva Gracia M, Stegmann B, Steimer W, Stingl JC, Uhr M, Ulrich S, Waschgler R, Zurek G, Hiemke C (April 2018). "TDM in psychiatry and neurology: A comprehensive summary of the consensus guidelines for therapeutic drug monitoring in neuropsychopharmacology, update 2017; a tool for clinicians". World Journal of Biological Psychiatry. 19 (3): 162–174. doi:10.1080/15622975.2018.1439595. PMID   29493375. S2CID   3743390.
  8. Buclin T, Thoma Y, Widmer N, André P, Guidi M, Csajka C, Decosterd LA (March 2020). "The Steps to Therapeutic Drug Monitoring: A Structured Approach Illustrated With Imatinib". Frontiers in Pharmacology. 11 (3): 177. doi: 10.3389/fphar.2020.00177 . PMC   7062864 . PMID   32194413.
  9. Fuchs A, Csajka C, Thoma Y, Buclin T, Widmer N (January 2013). "Benchmarking therapeutic drug monitoring software: a review of available computer tools". Clinical Pharmacokinetics. 52 (1): 9–22. doi:10.1007/s40262-012-0020-y. PMID   23196713. S2CID   21386054.
  10. Meneghello A, Tartaggia S, Alvau MD, Polo F, Toffoli G (2018). "Biosensing technologies for therapeutic drug monitoring". Current Medicinal Chemistry. 25 (34): 4354–77. doi:10.2174/0929867324666170720101736. PMID   28724346.