Journal of Clinical Pathology and Laboratory Medicine

All submissions of the EM system will be redirected to Online Manuscript Submission System. Authors are requested to submit articles directly to Online Manuscript Submission System of respective journal.
Reach Us +1 (202) 780-3397

Review Article - Journal of Clinical Pathology and Laboratory Medicine (2024) Volume 6, Issue 1

Therapeutic Drug Monitoring (TDM) from a clinical perspective: Challenges and approaches

Divya.Dara*

Geethanjali College of Pharmacy, Keesara, Hyderabad, Telangana

*Corresponding Author:
Divya.Dara
Geethanjali College of Pharmacy, Keesara, Hyderabad, Telangana, India
Geethanjali College of Pharmacy
India
E-mail:divyadara528@gmail.com

Received:24-Jan-2024, Manuscript No. AACPLM-24-126518; Editor assigned: 27-Jan-2024, PreQC No. AACPLM-24-126518(PQ); Reviewed:10-Feb-2024, QC No. AACPLM-24-126518; Revised:15-Feb-2024, Manuscript No. AACPLM-24-126518(R); Published:22-Feb-2024, DOI:10.35841/ aacplm-6.1.187

Citation: Dara D. Therapeutic Drug Monitoring (TDM) from a clinical perspective: Challenges and approaches. J Clin Path Lab Med. 2024;6(1):187

Abstract

The clinical technique of measuring particular pharmaceuticals at predetermined intervals to keep their level in a patient's bloodstream constant and optimize their individual dosing regimens is known as Therapeutic Drug Monitoring. For most drugs, TDM is not necessary; it is primarily used to monitor drugs with limited therapeutic ranges, drugs with significant pharmacokinetic variability, drugs whose target concentrations are challenging to monitor, and drugs that are known to have both beneficial and negative effects. The foundation of TDM is the belief that there is a measurable correlation between dosage and drug concentration in the blood or plasma, as well as between concentration and therapeutic outcomes.

Abstract

The clinical technique of measuring particular pharmaceuticals at predetermined intervals to keep their level in a patient's bloodstream constant and optimize their individual dosing regimens is known as Therapeutic Drug Monitoring. For most drugs, TDM is not necessary; it is primarily used to monitor drugs with limited therapeutic ranges, drugs with significant pharmacokinetic variability, drugs whose target concentrations are challenging to monitor, and drugs that are known to have both beneficial and negative effects. The foundation of TDM is the belief that there is a measurable correlation between dosage and drug concentration in the blood or plasma, as well as between concentration and therapeutic outcomes.

Keywords

Therapeutic Drug Monitoring, Plasma drug concentrations, Blood or plasma.

Introduction

A common definition of Therapeutic Drug Monitoring (TDM) is the clinical laboratories assessment of a chemical parameter that would directly affect drug prescribing practices when properly interpreted by a physician. If not, TDM describes the individualized administration of medication by preserving drug concentrations in the blood or plasma within specific therapeutic intervals [1, 2].

The basic concept of TDM is that there is a defined correlation between drug concentration in the blood or plasma and pharmacodynamic effects, as well as between the two. Measuring the medication's concentration at steady-state and adjusting the dosage to reach a target drug concentration that is known to be connected with efficacy rather than toxicity might help determine how much pharmacokinetic variability contributes to variations in dosage requirements. Developments in analytical technology, high-throughput computerization, mapping of drug pharmacokinetic properties, and growing understanding of drug concentration-response connections all contributed to the rise of clinical pharmacokinetic monitoring. An interdisciplinary strategy is necessary for TDM performance. Only with full cooperation from a TDM team-which usually consists of scientists, doctors, nurses, and pharmacists-can precise and clinically significant drug concentrations be achieved[3].

Methodology Adequacy, adherence, medication-drug interactions, toxicity prevention, and therapy discontinuation monitoring are now among the expanded list of indications for drug monitoring. Measuring plasma drug concentrations alone may be useful in a number of situations, albeit not all drugs will benefit from every indication. However, measuring plasma concentrations might be useful because low values indicate either under treatment or poor recent compliance. When a patient is prescribed a dose that is unlikely to result in a low concentration that is observed, or when a prior measurement shows that the level of plasma should be higher for the recommended dose, poor compliance is implicated[4, 5].

Measurements of the drug's plasma concentration and individual dosage adjustments may be helpful to the doctor when starting medication therapy. A portion of TDM that offers professional clinical assessments of drug concentration and assessment grounded in pharmacokinetic principles is therapeutic drug measurement. To provide maximum clinical benefit, a medication concentration measurement must be interpreted expertly[6].

By closely observing the physiological markers of therapeutic reactions, such as blood pressure, coagulation, glucose, and lipid concentrations, clinicians can regularly evaluate the pharmacodynamics of drugs. For many medications, the approach is not sensitive enough, or there isn't a readily available measure of effect[7].

As a result, the prediction of TDM is predicated on the idea that there is a measurable correlation between dosage and blood drug concentration, also known as Plasma Drug Concentration, and pharmacodynamic consequences. Pharmaceutical pharmacokinetics, pharmaceutical dynamics, and laboratory analysis are among the disciplines that must be coordinated for the purpose of therapeutic drug monitoring[8].

It is not widely recognized how analytical methods affect the determination of pharmacokinetic parameters. In therapeutic drug monitoring, analytical goals should be defined by identifying the type of problem to be solved, choosing the best matrix and methodology, and creating reliable analytical schemes that are carried out skilfully, appropriately, and understood in relation to the problem[9].

Evaluating TDM effectiveness involves determining the frequency of adverse medication reactions, cure, mortality, and cost-saving measures related to a TDM service. Adult individuals with general tonic-clonic epilepsy who received TDM had significantly better seizure control, fewer adverse reactions, higher earning potential, lower patient costs, and savings from fewer hospitalizations per seizure, and higher chances of remission, according to a pharmcoeconomic analysis[10].

Conclusion

Pharmacokinetic, pharmacodynamic and pharmacological methodologies and analyses must all be coupled when using TDM. More than just measuring a patient's blood medication concentration and comparing it to a target range is necessary for the proper use of TDM. Instead, TDM is crucial to the creation of therapeutic drugs that are both safe and efficacious as well as to the personalization of these drugs. TDM can also be used to detect medication compliance issues in noncompliant patient instances. A few things to keep in mind when interpreting drug concentration values are the patient's response, the planned clinical targets, the dosage history, and the period of sampling in relation to the dose. With this data, the best dosage schedule for achieving the desired effect with the least amount of toxicity can be determined.

References

  1. Levy G, Ebling WF, Forrest A. Concentration?or effect?controlled clinical trials with sparse data. Clin Pharmacol Ther. 1994;56(1):1-8.
  2. Indexed at, Google Scholar, Cross Ref

  3. Johnston A, Holt DW. Therapeutic drug monitoring of immunosuppressant drugs. Br J Clin Pharmacol. 1999;47(4):339.
  4. Indexed at, Google Scholar, Cross Ref

  5. Shenfield GM. Therapeutic drug monitoring beyond 2000. Br J Clin Pharmacol. 2001;52(1).
  6. Indexed at, Google Scholar, Cross Ref

  7. Aronson JK, Hardman M. ABC of monitoring drug therapy. Measuring plasma drug concentrations. Br Med J. 1992;305(6861):1078.
  8. Indexed at, Google Scholar, Cross Ref

  9. Steijns LS, Bouw J, van der Weide J. Evaluation of fluorescence polarization assays for measuring valproic acid, phenytoin, carbamazepine and phenobarbital in serum. Ther Drug Monit. 2002;24(3):432-5.
  10. Indexed at, Google Scholar, Cross Ref

  11. Patel JA, Clayton LT, LeBel CP, et al. Abnormal theophylline levels in plasma by fluorescence polarization immunoassay in patients with renal disease. Ther Drug Monit. 1984;6(4):458-60.
  12. Indexed at, Google Scholar, Cross Ref

  13. Hicks JM, Brett EM. Falsely increased digoxin concentrations in samples from neonates and infants. Ther Drug Monit. 1984;6(4):461-4.
  14. Indexed at, Google Scholar, Cross Ref

  15. Frank EL, Schwarz EL, Juenke J, et al. Performance characteristics of four immunoassays for antiepileptic drugs on the IMMULITE 2000 automated analyzer. Am J Clin Pathol. 2002;118(1):124-31.
  16. Indexed at, Google Scholar, Cross Ref

  17. Wu SL, Li W, Wells A, Dasgupta A. Digoxin-like and digitoxin-like immunoreactive substances in elderly people: Impact on therapeutic drug monitoring of digoxin and digitoxin concentrations. Am J Clin Pathol. 2001;115(4):600-4.
  18. Indexed at, Google Scholar, Cross Ref

  19. Somerville AL, Wright DH, Rotschafer JC. Implications of vancomycin degradation products on therapeutic drug monitoring in patients with end?stage renal disease. Pharmacotherapy. 1999;19(6):702-7.
  20. Indexed at, Google Scholar, Cross Ref

Get the App