40

Therapeutic drug monitoring

40

Therapeutic drug monitoring

40

Therapeutic drug monitoring

40

Therapeutic drug monitoring

  40 Therapeutic drug monitoring

Michael Oellerich

40.1 Introduction

Inappropriately high or low doses of therapeutic drugs are administered more often than is generally assumed. It is also recognized that the therapeutic effect of many drugs correlates better with their serum concentration than with the administered dose. The selection of drugs for which the serum concentration needs to be measured is based on certain criteria (Tab. 40-1 – Selection criteria for measuring serum concentrations of drugs).

Therapeutic drug monitoring (TDM) of the serum concentrations of the following drugs is recommended:

  • Antiarrhythmic agents (amiodarone, lidocaine, procainamide, quinidine, disopyramide)
  • Antibiotics (aminoglycosides, vancomycin)
  • Antidepressants (amitriptyline, nortriptyline, imipramine, desipramine, trimipramine, citalopram, clomipramine, norclomipramine, doxepin, nordoxepin, escitalopram, fluoxetine, norfluoxetine, mirtazapine, sertraline, venlafaxine, O-desmethylvenlafaxine)
  • Anticonvulsants (phenytoin, phenobarbital, primidone, carbamazepine, ethosuximide, valproic acid, lamotrigine)
  • Antifungal agents (itraconazole, posaconazole, voriconazole)
  • Antiretroviral drugs (efavirenz, nevirapine, indinavir, nelfinavir, ritonavir, saquinavir, lamivudine, zidovudine)
  • Cardiac glycosides (digoxin, digitoxin)
  • Immunosuppressive agents (mycophenolic acid; determination in whole blood: cyclosporine, tacrolimus, sirolimus, everolimus)
  • Lithium
  • Methotrexate
  • Neuroleptics (aripiprazole, clozapine, haloperidol, olanzapine, quetiapine, risperidone, 9-OH risperidone)
  • Theophylline.

To be able to determine and interpret serum drug concentrations, a knowledge of the pharmacokinetics is essential.

40.2 Indication

Suspected overdose, lack of therapeutic response, monitoring of patient compliance with prescribed medication.

Determination of dose (e.g., in conjunction with):

  • A lack of easily determinable, reliable efficacy parameters
  • Suspected drug interactions
  • Significant intraindividual and inter individual variability in pharmacokinetics
  • Highly variable first-pass effect
  • Diseases that influence drug absorption, protein binding, and elimination
  • Presence of a genetic polymorphism that affects drug absorption and metabolism
  • Accumulation of pharmacologically active metabolites
  • Suspected change in bioavailability
  • Persistent side effects, especially in patients who are critically dependent on the therapeutic effect of the medication.

40.3 Method of determination

Fluorescence polarization immunoassay, chemiluminescence immunoassay, enzyme immunoassay, turbidimetric immunoassay, nephelometric immunoassay, fluorescence immunoassay, radioimmunoassay /1/.

Gas chromatography (GC), liquid chromatography (LC), liquid chromatography-mass spectrometry (LC-MS/MS).

For information on individual methods, refer to

40.4 Specimen

Serum, plasma, whole blood: 1 mL

40.5 Clinical significance

Numerous factors may have an influence on the effect of a particular prescribed drug (Fig. 40-1 – Factors influencing the effect of a specific prescribed drug). Therefore, the serum concentration cannot be interpreted by relying exclusively on the therapeutic range.

Other points that need to be considered include the following /2/:

  • Timing of blood sampling
  • Route of administration
  • Occurrence of pharmacologically active metabolites with certain drugs
  • Presence of an adequate therapeutic response
  • The overall clinical presentation.

Therapeutic range: slight variations can be observed between the ranges specified in the literature. The ranges listed in this chapter are to be considered as recommendations only (Tab. 40-2 – Therapeutic ranges).

40.5.1 Timing of blood sampling

During the course of continuous therapy, blood should be collected when steady state has been reached (i.e., after the administration of a constant dose for at least 4 times the half life of the drug). Sampling should be performed according to the clinical situation, at the time of peak serum concentration and/or immediately prior to the administration of the next dose (trough serum concentration). Blood sampling to measure trough and peak serum concentrations is important for drugs with a narrow therapeutic range and a short half-life (e.g., theophylline, gentamicin, and certain antiarrhythmic agents). For some drugs (e.g., phenytoin and phenobarbital) the timing of blood sampling is not critical since the differences between trough and peak serum concentrations are relatively small once steady state has been reached. After intravenous drug administration, blood sampling should not be performed until the initial distribution phase has been completed. With most drugs, this is achieved after 1–2 h; with digoxin and digitoxin, it takes 6–8 h.

40.5.2 Pharmacologically active metabolites

With certain drugs, significant concentrations of active metabolites occur. For example, phenobarbital, the active metabolite of primidone, reaches higher serum concentrations than primidone itself.

40.5.3 Presence of an adequate therapeutic response

Occasionally, this occurs at a normally sub therapeutic serum level or else not until a potentially toxic level has been reached. The upper limit of the therapeutic range can shift to a higher level due to the development of tolerance associated with long term intake of a drug (e.g., phenobarbital). The limits of the therapeutic range can also change, as is the case with the additional intake of drugs with synergistic or antagonistic effects, with drugs that exhibit highly variable serum protein binding (e.g., phenytoin), and in the presence of factors that influence the effect of the drug at its site of action (e.g., digoxin). The overall clinical presentation must be taken into consideration when interpreting drug serum concentrations.

Causes for the occurrence of drug levels outside the therapeutic range:

  • Patient noncompliance
  • Incorrect dose
  • Malabsorption
  • Inadequate bioavailability of the drug preparation
  • Drug interactions
  • Renal disease
  • Hepatic disease
  • Altered serum protein concentrations
  • Febrile infections
  • Genetically determined differences in metabolism (e.g., rapid versus slow metabolism depending on genetic polymorphisms such as CYP2D6, CYP2C9, CYP2C19, UGT1A1, NAT2).

If the causes leading to sub therapeutic or toxic serum drug concentrations cannot be removed and the patient is compliant, a change in dose is required.

40.5.4 Dose finding

In the case of drugs with linear pharmacokinetics (with the exception of phenytoin, most drugs routinely monitored fall into this category), the new dose can be determined using a simple equation:

D N = D A × C N C A

DN , new dose; DO , old dose; CN , desired trough serum concentration at steady state; CO , current trough serum concentration at steady state

The desired steady state trough serum concentration can, however, only be reached if the dose intervals remain unchanged. Note also that, after dose adjustment, the trough and peak serum concentrations that occur during the dose intervals will change in different proportions. Using the described dose adjustment method can therefore lead to miscalculation of the required dose, especially in patients with extreme pharmacokinetic parameters.

With certain antidepressants (e.g., venlafaxine) or neuroleptics (e.g., clozapine), valuable conclusions can be drawn from the ratio of metabolite to parent drug /34/. A low ratio can indicate poor compliance, decreased metabolism due to enzyme inhibition (e.g., fluvoxamine), genetic polymorphism, or that the blood sample was collected at the wrong time. Conversely, a high ratio (e.g., venlafaxine) may indicate a very high rate of metabolism. For this drug, further information about whether a patient is an ultra rapid metabolizer can be obtained by performing CYP2D6 genotyping /5/.

In patients with difficult dose response control, pharmacokinetic methods can be used for dose finding /6/. With such methods, dose predictions can be made both prior to the start of therapy and during therapy.

The use of methods for dose prediction is particularly important for amino glycoside therapy in critically ill patients, for example. In such patients, the amino glycoside doses required by individual patients can differ by more than ten-fold. A method is available for predicting the amino glycoside maintenance dose using three precisely timed serum concentration measurements to estimate the individual clearance /7/.

In addition to such simple pharmacokinetic methods, more complex procedures have also been developed using population kinetics in combination with measured serum drug concentrations for dose prediction (Bayesian prediction method). With these methods, it is possible to make dose predictions during the course of therapy /8/.

Regardless of the method used, a dose prediction remains valid only if the drug clearance in a given patient does not change significantly during the further course of the disease. Rapid intraindividual changes in clearance require a dose review, preferably by monitoring the serum level of the drug in question. In such cases, it is possible to readjust the dose within a relatively short time interval to actual requirements with the help of serum concentration determinations and appropriate pharmacokinetic methods.

40.6 Serum drug concentrations and their significance

Refer to Tab. 40-3 – Serum drug concentrations and their significance.

40.7 Comments and problems

Method of determination

Although immunoassays have proved to be highly effective in routine diagnostics, cross reactions with metabolites and other substances as well as heterophilic antibodies (e.g., ACMIA tacrolimus assay) can interfere.

In the case of LC-MS/MS, mass interference as a result of in-source fragmentation of cyclosporine metabolites can lead to analytical problems. For this reason, metabolites should be separated before the sample is introduced to the ion source. A further problem associated with this method is ion suppression caused by the sample matrix, which can impair selectivity and specificity. Interference of this type can be prevented by separating the drug from endogenous substances using chromatography.

Cross reactions

Using immunoassays, cross reactions may occur with metabolites of the drug and with other structurally related substances. For example:

  • Immunoassays to measure digoxin show cross reactions with digoxin metabolites, other cardiac glycosides, an endogenous digoxin-like substance, and spironolactone metabolites. In the case of digitoxin determination, digitoxin-like immunoreactive substances simulate high values; these substances are steroids and lipids that can be extracted.
  • In patients with uremia, treatment with phenytoin or theophylline leads to the accumulation of metabolites that cause a significant cross reaction in certain immunoassays.
  • Cross reactions with related substances (e.g., 4-amino-4-deoxy-N10-methyl-pteroic acid and 7-hydroxy methotrexate have been observed in immunoassays used to determine methotrexate).
  • In immunoassays for the determination of quinidine, slightly higher values are seen when compared to the HPLC method due to a cross reaction with quinidine metabolites.
  • In immunoassays for cyclosporine and tacrolimus, cross reactions occurs due to cyclosporine and tacrolimus metabolites; the extent of cross reaction varies significantly between commercially available assays.

Stability

Serum and EDTA blood samples can be transported by mail at room temperature. The same applies to assays for cyclosporine, tacrolimus, sirolimus, everolimus, and mycophenolic acid if transport times are short.

40.8 Pharmacokinetics

The pharmacokinetics of a substance includes three processes: absorption, distribution, and elimination (metabolism, excretion). These processes generally overlap in time.

During the initial distribution of the drug in the organism (α phase), absorption and distribution into the various compartments play the dominant role. During this phase, rapid changes in serum and tissue concentrations of the drug occur and the serum drug level do not correlate with those at the site of action.

In contrast, during the equilibrium phase (β phase), the processes of elimination dominate. The serum drug concentration during this phase better reflects the concentration at the site of action.

The bioavailability of a drug is determined by the proportion of the administered dose reaching the systemic circulation in its pharmacologically active form and by the speed of this process.

The first pass effect describes the phenomenon by which enzymatic metabolism of the drug occurs during absorption and the first passage through the liver.

Accumulation can occur with continuous infusion or repeated intermittent administration of a drug. The serum concentration of the substance increases within a particular period of time until it reaches a plateau (steady state concentration). This steady state is usually almost reached after the administration of a constant dose over five half lives. The levels of the steady state peak and trough concentrations depend on the amount of drug administered, the dosing interval, and the elimination half life. The time taken to reach the corresponding plateau depends only on the elimination half life. If the dosing interval is short in comparison to the elimination half life, only relatively minor variations between the minimum and maximum steady state serum concentrations will occur.

Many drugs are characterized by linear kinetics. This implies that a particular percentage of the total amount of drug in the body is eliminated per unit of time (first order reaction). The serum concentration in the β phase therefore shows a linear decline on the concentration-time curve (as depicted in a semi-logarithmic coordinate system). The elimination half life (t1/2 = time period until the serum concentration declines by 50% after completion of all distribution processes) and the elimination constant (K) are independent of the administered dose or the drug serum concentration: K = 0.693/t1/2. A doubling of the dose therefore also leads to a doubling of the serum concentration at steady state. In contrast, some drugs (e.g. phenytoin) are characterized by non linear kinetics. This implies that only a certain amount of the drug is eliminated per unit time, independent of the dose or the serum concentration (zero order reaction). The concentration-time curve during the β phase does not show a linear decline if depicted in a semi-logarithmic coordinate system. This is because the systems of the body that are responsible for the elimination are already saturated at therapeutic serum levels of the drug. The elimination half life of the drug therefore increases with rising dose and increasing serum concentration. Thus, a doubling of the dose usually increases the steady state concentration more than two-fold.

For most drugs, there is a rough correlation between the administered dose and the intensity of the pharmacological effect. The high degree of variability of the dose-response relationship in patients is based on a multitude of possible biological influence factors (Fig. 40-1 – Factors influencing the effect of a specific prescribed drug).

The determination of the serum drug concentration is relevant for adjusting therapy if:

  • The serum concentration of a drug with reversible effect is in equilibrium with the concentration at the site of action
  • The drug does not result in the development of tolerance at the receptor
  • A therapeutic range can be defined based on the effect of the drug.

With certain drugs, a better prediction of the effect can be made based on the serum concentration rather than on the dose because the relationship between the serum concentration and effect is influenced by far fewer factors than the dose-response relationship. If, however, the site of action of the drug is in an organ or tissue represented by a deep compartment, measurement of the serum concentration does not help in adjusting therapy. In this case, the effect of the drug can persist despite non detectable levels in the serum. Furthermore, the measurement of serum concentration is usually less appropriate for substances that exert irreversible effects, accumulate at the site of action, or require active transport to the site of action.

In the area of transplant medicine, for example, pharmacodynamic principles are becoming increasingly important /40/. It has been shown that the effects of immunosuppressive drugs can vary significantly despite the presence of comparable blood levels. For this reason, suitable biomarkers are being sought that may allow individualized pharmacotherapy. Although the primary value of drug monitoring lies in the prevention of toxicity, significant inter individual variability exists with respect to the effects of immunosuppressive drugs, which means that individual patients may require more or less robust immunosuppressive therapy. To date, the inability to measure the effects of immunosuppressive drugs on immune cells in vivo has significantly hindered the optimum use of these drugs in transplant medicine. As a result, the risk of chronic over suppression or under suppression of the immune system is still high. The long term survival of transplants is negatively affected by chronic rejection and the side effects of standard immunosuppressive regimens. In the future, suitable combinations of such biomarkers will be a useful adjunct to therapeutic drug monitoring, which is currently based on pharmacokinetic measure­ments /4044/.

Multiple physiological factors need to be taken into account with drug therapy. The amount of drug prescribed depends, among other factors, on the weight and body surface. Drugs that are only minimally taken up by fatty tissue should be dosed according to the lean rather than the actual body weight.

The function of the gastrointestinal tract affects the absorption of the drug, the composition of the serum proteins its distribution, the liver function its metabolism, and the function of the liver and kidneys its excretion. In addition, cardiac function plays an important role in the absorption, distribution, biotransformation, and excretion of drugs due to its influence on the regional blood flow of the relevant organs.

Most drugs in the blood are bound to serum proteins to a varying extent. Acidic drugs are usually bound to albumin, while basic drugs are frequently bound to α1-acid glycoprotein and lipoproteins. Drugs with higher affinity can displace those that bind less strongly. In such instances, the free fraction of the drug can increase despite a constant total concentration, resulting in toxic side effects. An equilibrium is established between the protein bound and free proportion of the drug. In certain cases, the free proportion of the drug, which is able to pass through membranes and diffuse into tissue, correlates better with the concentration at the site of action than with its total serum concentration.

The binding capacity is defined by the maximum amount of drug that can be bound by the serum proteins at equilibrium. At therapeutic serum concentrations, usually only a small portion of the available binding sites are occupied by the drug. Therefore, the free fraction is relatively constant and independent of the drug concentration. If, however, the binding capacity is exceeded at high doses, the free fraction of the drug increases disproportionately and an unexpected enhancement of the pharmacological effect can occur as a result. Disopyramide and lidocaine already display such concentration dependent binding within the therapeutic range.

The most important site for the biotransformation of drugs is the liver. The activity of microsomal enzyme systems, by which many drugs are metabolized, can be induced by certain substances (e.g., phenobarbital). The serum concentration of a drug as well as the extent and duration of its effect can be reduced due to the resulting enhanced metabolism.

For some substances (e.g., procainamide) isoniazid, and some sulfonamides, acetylation is an important step in the biotransformation process. It is genetically determined, belongs to the non microsomal systems of biotransformation, and is not associated with induction. Patients with slow acetylation have a higher risk of toxic side effects.

Genetic polymorphisms are also known to play an important role in the absorption, metabolism, and receptor interactions of many other drugs /41/. Examples include the production of the active tamoxifen metabolite endoxifen in association with CYP2D6 polymorphism or the dependence of the effects of the anticoagulant warfarin on VKORC1 and CYP2C9 polymorphism. Substances with non linear pharmacokinetics (e.g., phenytoin) show saturation of hepatic enzymes at therapeutic serum concentrations. The resulting limited metabolism can lead to unexpectedly high serum levels of these drugs.

Furthermore, liver diseases and alterations in hepatic perfusion can critically influence the metabolism of drugs.

When using a new drug formulation, it is important to ensure that the preparations are equivalent for the purposes of therapeutic drug monitoring. It must be clarified in each case whether the therapeutic range of a drug is still valid if the galenic characteristics of the drug have changed. For example, the tacrolimus preparation A shows, in comparison to the same dose of the preparation B a higher C(max) and AUC despite similar trough levels of tacrolimus for both preparations /42/. In other words, although the trough levels for both preparations are the same, the levels of drug exposure differ. It is obvious, therefore, that due to a lack of bioequivalence, the TDM approach that was developed for B is not appropriate for A. A further example of this is provided by the mycophenolic acid preparations C and M. Exposure to mycophenolic acid is similar for both preparations at equimolar doses; however, mycophenolic acid C0 levels are significantly higher with C than with M /43/. In this example, the same TDM approach cannot be used for C and M. With the increasing availability of generic preparations, similar effects can also be seen for other drugs.

References

1. Oellerich, M. Enzyme-immunoassay: a review. J Clin Chem Clin Biochem 1984; 22: 895–904.

2. Koch-Weser, J. Serum drug concentrations as therapeutic guides. N Engl J Med 1972; 287: 227–31.

3. Couchman L, Morgan PE, Spencer EP, Flanagan RJ. Plasma clozapine, norclozapine, and the clozapine: norclozapine ratio in relation to prescribed dose and other factors: data from a therapeutic drug monitoring service, 1993–2007. Ther Drug Monit 2010; 32: 438–47.

4. Reis M, Aamo T, Spigset O, Ahlner J. Serum concentrations of antidepressant drugs in a naturalistic setting: compilation based on a large therapeutic drug monitoring database. Ther Drug Monit 2009; 31: 42–56.

5. Baumann P, Hiemke C, Ulrich S, Eckermann G, Gaertner I, Gerlach M, Kuss HJ, Laux G, Müller-Oerlinghausen B, Rao ML, Riederer P, Zernig G. The AGNP-TDM expert group consensus guidelines: Therapeutic drug monitoring in psychiatry. Pharmacopsychiatry 2004; 37: 243–65.

6. Sheiner LB, Beal S, Rosenberg B, Marathe VV. Forecasting individual pharmacokinetics. Clin Pharmacol Therap 1979; 26: 294–305.

7. Böttger HCh, Oellerich M, Sybrecht GW. Use of aminoglycosides in critically ill patients: individualisation of dosage using Bayesian statistics and pharmacokinetic principles. Ther Drug Monit 1988; 10: 280–6.

8. Oellerich M, Schneider B. Clinical biochemistry; 2. Data presentation, interpretation. In: Keller H, Trendelenburg CH, eds. Single-point and Bayesian approach for individualizing theophylline dosage. Berlin; Walter de Gruyter Verlag, 1989.

9. Oellerich M, Müller-Vahl H. The EMIT free level ultrafiltration technique compared with equilibrium dialysis and ultra-centrifugation to determine protein binding of phenytoin. Clin Pharmacokinet 1984; 9 (Suppl. 1): 61–70.

10. Johannessen SI, Battino D, Berry DJ, Bialer M, Krämer G, Tomsen T, Patsalos PN. Therapeutic drug monitoring of the newer antiepileptic drugs. Ther Drug Monit 2003; 25: 340–63.

11. Schobben F, van der Kleijn E, Vree TB. Therapeutic monitoring of valproic acid. Ther Drug Monit 1980; 2: 61–71.

12. Rathore SS, Curtis JP, Wang J, Bristow MR, Krumholz HM. Association of serum digoxin concentration and outcomes in patients with heart failure. JAMA 2003; 289: 871–8.

13. Jortani SA, Valdes Jr. R. Digoxin and its related endogenous factors. Crit Rev Clin Lab Sci 1997; 34: 225–74.

14. Oellerich M, Armstrong VW. The MEGX test: a tool for the real-time assessment of hepatic function. Ther Drug Monit 2001; 23: 81–92.

15. Streit F, Niedmann PD, Shipkova M, Oellerich M. Rapid and sensitive liquid chromatography-tandem mass spectrometry method for determination of monoethylglycinexylidide. Clin Chem 2001; 47: 1853–6.

16. Karlsson E. Clinical pharmacokinetics of procainamide. Clinical Pharmacokinet 1978; 3: 97–107.

17. Myerburg RJ, Conde C, Sheps DS, Appel RA, Kiem J, Sung RJ, Castellanos A. Antiarrhythmic drug therapy in survivors of prehospital cardiac arrest: comparison of effects on chronic ventricular arrhythmias and recurrent cardiac arrest. Circulation 1979; 59: 855–63.

18. Raude E, Oellerich M, Weinel P, Freund M, Schrappe M, Riehm H, Poliwoda H. High-dose methotrexate: pharmacokinetics in children and young adults. Int J Clin Pharmacol Ther Toxicol 1988; 26: 364–70.

19. Bettinger TL, Crismon ML. Lithium. In: Burton ME, Shaw LM, Schentag JJ, Evans WE (eds). Applied pharmacokinetics: Principles of therapeutic drug monitoring. Philadelphia; Lippincott Williams, 2006.

20. Blaser J, König C, Simmen HP, Thurnheer U. Monitoring serum concentrations for once-daily netilmicin dosing regimens. J Antimicrob Chemother 1994; 33: 341–8.

21. Tulkens, PM. Efficacy and safety of aminoglycosides once-a-day: experimental and clinical data. Scand J Infect Dis 1990; Suppl 74: 249–57.

22. Rybak MJ, Lomaestro BM, Rotschafer JC, Moellering RC Jr., Craig WA, Billeter M, et al. Vancomycin therapeutic guidelines: a summary of consensus recommendations from the infectious diseases Society of America, the American Society of Health-System Pharmacists, and the Society of Infectious Diseases Pharmacists. CID 2009; 49: 325–7.

23. Oellerich M, Armstrong VW, Kahan B, Shaw L, Holt DW, Yatscoff R, et al. Lake Louise consensus conference on cyclosporin monitoring in organ transplantation: report of the consensus panel. Ther Drug Monit 1995; 17: 642–54.

24. Knight SR, Morris PJ. The clinical benefits of cyclosporine C2-level monitoring: a systematic review. Transplantation 2007; 83: 1525–35.

25. Fahr A. Cyclosporin clinical pharmacokinetics. Clin Pharmacokinet 1993; 24: 402–95.

26. Streit F, Armstrong VW, Oellerich M. Rapid liquid chromatography-tandem mass spectrometry routine method for simultaneous determination of sirolimus, everolimus, tacrolimus, and Cyclosporin A in whole blood. Clin Chem 2002; 48: 955–8.

27. De Jonge H et al. Reduced C0 concentrations and increased dose requirements in renal allograft recipients converted to the novel once-daily tacrolimus formulation. Transplantation 2010, 90: 523–9.

28. Undre A, Tacrolimus modified release kidney study group. Use of a once daily modified release regimen in de novo kidney transplant recipients. Am J Transplant 2005, 5: 190.

29. Armstrong VW, Oellerich M. New developments in the immunosuppressive drug monitoring of cyclosporine, tacrolimus and azathioprine. Clin Biochem 2001; 34: 9–16.

30. Lennard L, Van Loon JA, Lilleyman JS, Weinshilboum RM. Thiopurine pharmacogenetics in leukemia: correlation of erythrocyte thiopurine methyltransferase activity and 6-thioguanine nucleotide concentrations. Clin Pharmacol Ther 1987; 41: 18–25.

31. Schaeffeler E, Fischer C, Brockmeier D, Wernet D, Moericke K, Eichelbaum M, et al. Comprehensive analysis of thiopurine S-methyltransferase phenotype-genotype correlation in a large population of German caucasians and identification of novel TPMT variants. Pharmacogenetics 2004; 14: 407–14.

32. Kahan BD, Camardo JS. Rapamycin: clinical results and future opportunities. Transplantation 2001; 72: 1181–93.

33. Oellerich M, Armstrong VW. The role of therapeutic drug monitoring in individualizing immunosuppressive drug therapy: recent developments. Ther Drug Monit 2006; 28: 720–5.

34. van Gelder T, Le Meur Y, Shaw LM, Oellerich M, DeNofrio D, Holt C, et al. Therapeutic drug monitoring of mycophenolate mofetil in transplantation. Ther Drug Monit 2006; 28: 145–54.

35. Kuypers DRJ, Le Meur Y, Cantarovich M, Tredger MJ, Tett SE, Cattaneo D, et al. for the Transplantation Society (TTS) Consensus Group of TDM of MPA. Consensus report of therapeutic drug monitoring of mycophenolic acid in solid organ transplantation. Clin J Am Soc Nephrol 2010, 5: 341–58.

36. van Gelder T, Silva HT, de Fijter JW, Budde K, Kuypers D, Tyden G, et al. Comparing mycophenolate mofetil regimens for de novo renal transplant recipients: the fixed-dose concentration-controlled trial. Transplantation 2008; 86: 1043–51.

37. Le Meur Y, Büchler M, Thierry A, Caillard S, Villemain F, Lavaud S, et al. Individualized mycophenolate mofetil dosing based on drug exposure significantly improves patient outcomes after renal transplantation. Am J Transplant 2007; 7: 2496–503.

38. Gaston RS, Kaplan B, Shah T, Cibrik D, Shaw LM, Angelis M, et al. Fixed- or controlled-dose mycophenolate mofetil with standard- or reduced-dose calcineurin inhibitors: the opticept trial. Am J Transplant 2009, 9: 1–13.

39. Brandhorst G, Marquet P, Shaw LM, Liebisch G. Schmitz G, Coffing MJ, et al. Multicenter evaluation of a new inosine monophosphate dehydrogenase inhibition assay for quantification of total mycophenolic acid in plasma. Ther Drug Monit 2008, 30: 428–33.

40. Wieland E, Olbricht CJ, Süsal C, Gurragchaa P, Böhler T, Israeli M, et al. Biomarkers as a tool for management of immunosuppression in transplant patients. Ther Drug Monit 2010; 32: 560–72.

41. Ingelman-Sundberg M. Pharmacogenetics: an opportunity for a safer and more efficient pharmacotherapy. J Intern Med 2001; 250: 186–200.

42. Park K, Kim YS, Kwon Kl, Park MS, Lee YJ, Kim KH. A randomized, open-label, two-period, crossover bioavailability study of two oral formulations of tacrolimus in healthy Korean adults. Clin Ther 2007; 29: 154–62.

43. Wallemacq P, Armstrong VW, Brunet M, Haufroid V, Holt DW, Johnston A, et al. Opportunities to optimize tacrolimus therapy in solid organ transplantation: Report of the European Consensus Conference. Ther Drug Monit 2009; 31: 139–52.

44. Oellerich M, Schütz E, Kanzow P, Schmitz J, Beck J, Kollmar O, et al. Use of graft-derived cell-free DNA as an organ integrity biomarker to reexamine effective tacrolimus trough concentrations after liver transplantation. Ther Drug Monit 2014; 36: 136–40.

Table 40-1 Selection criteria for measuring serum concentrations of drugs

  • Serious toxicity in conjunction with poorly defined clinical endpoint for therapeutic efficacy
  • Steep dose response curve
  • Narrow therapeutic range
  • Long term therapy
  • Life threatening illnesses
  • Significant inter individual differences in pharmacokinetics
  • Non linear pharmacokinetics
  • Frequent administration of the drug
  • Reliable methods of determination

Table 40-2 Therapeutic ranges

Antibiotics (*multiple daily doses)

  • P amikacin* (trough)

5–10

mg/L

  • P amikacin* (peak)

20–30

mg/L

  • P gentamicin* (trough)

≤ 2

mg/L

  • P gentamicin* (peak)

5–10

mg/L

  • P tobramycin* (trough)

≤ 2

mg/L

  • P tobramycin* (peak)

5–10

mg/L

  • P vancomycin (trough)

5–20

mg/L

Antidepressants

  • P amitriptyline + nortriptyline

80–200*

μg/L

  • P citalopram

30–130

μg/L

  • P clomipramine + norclomipramine

175–450*

μg/L

  • P desipramine

100–300

μg/L

  • P doxepin + nordoxepin

50–150*

μg/L

  • P duloxetine

20–80

μg/L

  • P escitalopram

15–80

μg/L

  • P fluoxetine + norfluoxetine

120–300*

μg/L

  • P fluvoxamine

150–300

μg/L

  • P imipramine + desipramine

175–300*

μg/L

  • P maprotiline

125–200

μg/L

  • P mirtazapine

40–80

μg/L

  • P nortriptyline

70–170

μg/L

  • P paroxetine

70–120

μg/L

  • P sertraline

10–50

μg/L

  • P trimipramine

150–350

μg/L

  • P venlafaxine + O-desmethylvenlafaxine

195–400*

μg/L

* Total of parent substance plus pharmacologically active metabolite

Anticonvulsants

  • P carbamazepine

4–12

mg/L

  • P 10-OH-carbazepine

13–35***

mg/L

  • P ethosuximide

40–100

mg/L

  • P gabapentin

4–16

mg/L

  • P lamotrigine

3–14**

mg/L

  • P levetiracetam

10–40**

mg/L

  • P phenobarbital

10–40

mg/L

  • P phenytoin, total

10–20

mg/L

  • P phenytoin, free

1–2

mg/L

  • P primidone

5–15

mg/L

  • P topiramate

5–20**

mg/L

  • P valproic acid

50–100

mg/L

  • P zonisamide

10–40

mg/L

** Indicative value. *** If eslicarbazepine acetate is used, proportion of active S-enantiomer of 10-OH-carbazepine is 8% higher.

Antifungal agents

  • P itraconazole

0.5–2.0

mg/L

  • P voriconazole

1–6

mg/L

Antineoplastic agents

  • P imatinib (trough)

> 1,002*

μg/L

  • P methotrexate 24 h

≤ 10**

μmol/L

  • P methotrexate 48 h

≤ 1.0**

μmol/L

  • P methotrexate 72 h

≤ 0.1**

μmol/L

* Indicative value for ”major molecular response” in CML (chronic phase). ** High-dose therapy; infusion over 4–6 h

Immunosuppressive agents

  • B cyclosporine (trough)

100–300

μg/L

 

Initial therapy (approx.
≤ 3 months after
transplantation)

Maintenance therapy

Kidney

150–225 μg/L

100–150 μg/L

Liver

225–300 μg/L

100–150 μg/L

Heart

250–350 μg/L

150–250 μg/L

GVHD prophylaxis < 100 days

GVHD treatment (with additional steroid administration)

Stem cells

200–250 (300) μg/L

150–200 μg/L

  • B tacrolimus (trough)

4–15 μg/L

Initial therapy (approx.
≤ 3 months after
transplantation)

Maintenance therapy

Kidney

9.0–13.0 μg/L

4.0–9.0 μg/L

Liver

9.0–13.0 μg/L

4.0–9.0 μg/L

Heart

9.0–15.0 μg/L

7.0–13.0 μg/L

GVHD prophylaxis

GVHD treatment (with additional steroid administration)

Stem cells

4.0–10.5 μg/L

4.0–10.5 μg/L

P mycophenolic acid (MPA, trough)

  • Concomitant cyclosporine

1.3–3.5*

mg/L

  • Concomitant tacrolimus

1.9–4.0*

mg/L

  • P MPA AUC (0–12 h)

30–60*

mg × h/L

* Tentative therapeutic range for MPA in the early phase (< 3 months) after kidney transplantation

B sirolimus (trough)

  • Kidney transplantation – Triple therapy with cyclosporine, corticosteroids, and sirolimus

4.0–12.0

μg/L

  • Dual therapy with sirolimus and corticosteroids

12.0–20.0

μg/L

Liver transplantation

  • Therapy with cyclosporine or tacrolimus, sirolimus, +/- corticosteroids

3.0–6.0

μg/L

  • Therapy with sirolimus +/- corticosteroids

5.0–8.0

μg/L

B everolimus (trough)

  • Triple therapy with cyclosporine, corticosteroids, and everolimus

3.0–8.0

μg/L

E 6-thioguanine nucleotide (6-TGN)

  • Organ transplantation (triple therapy with azathioprine)

100–450

pmol/0.8 × 109 Erys

  • Chronic inflammatory bowel disease (azathioprine)

250–450

 

  • Chemotherapy (6-mercaptopurine)

500–1,000

 

  • E 6-methylmercaptopurine (6-MMP)

< 5,700*

pmol/0.8 × 109 Erys

* Indicative value

Cardiac drugs

  • P amiodarone

1.0–2.5

mg/L

  • P digitoxin

10–30

μg/L

  • P digoxin, in heart failure (sinus rhythm): 0.5–0.8 μg/L

0.6–1.2

μg/L

  • P lidocaine

1.5–5.0

mg/L

Neuroleptics

  • P aripiprazole

150–250

μg/L

  • P clozapine

350–600

μg/L

  • P haloperidol

5–17

μg/L

  • P olanzapine

20–80

μg/L

  • P quetiapine

70–170

μg/L

  • P risperidone + 9-OH risperidone

20–60*

μg/L

*Total of parent substance plus pharmacologically active metabolite

Other

  • S lithium

0.60–1.20

mmol/L

  • P theophylline

8–20

mg/L

  • P theophylline, premature infants

6–11

mg/L

B, blood; E, erythrocytes; P, plasma; S, serum; peak: maximum plasma concentration

Table 40-3 Serum drug concentrations and their significance

Clinically and laboratory findings

Anticonvulsants – Generally

Indication for measuring concentration: therapy failure, e.g. due to unreliable patient compliance or pharmacokinetic reasons; suspected toxicity, e.g. cases with unclear neurological or psychiatric symptoms; drug therapy in patients with altered pharmacokinetics (e.g., in pregnant patients or patients on dialysis).

– Phenytoin

Therapeutic range: 10–20 mg/L adults, children; 6–14 mg/L premature and full-term newborns, infants (age 2–12 weeks). The therapeutic range for the free phenytoin concentration in the serum is 1–2 mg/L.

Elimination half-life: it does not make sense to specify a half-life since phenytoin has dose-dependent pharmacokinetics. The elimination of this drug is better described by the maximum rate of metabolism (Vmax) and the concentration at which the half-maximum rate of metabolism (Km) is present: Vmax 100–1,000 mg/d; Km 1–15 mg/L.

Recommendation for blood sampling: during the dose interval.

Elimination: metabolism in the liver; the metabolites have no significant anticonvulsive effect.

Clinical aspects: because phenytoin exhibits non-linear pharmacokinetics, no definite prediction can be made about the expected serum concentration after a specific dose. Doses should be increased in small increments only, otherwise toxicity can occur due to a disproportionate rise in the serum concentration. Toxic doses can have a pro convulsive effect. In the presence of chronic hepatic disease, the rate of phenytoin metabolism is reduced. During pregnancy, there is generally a decline in the serum phenytoin concentration. An increase in phenytoin dose seems to be necessary in patients with brain tumors or brain metastases undergoing chemotherapy (e.g., cisplatin) to maintain the phenytoin concentration within the therapeutic range. The concentration of free phenytoin not bound to serum proteins correlates generally with its concentration in the cerebrospinal fluid and saliva. In patients with altered serum protein binding or when an unexpected effect occurs, the serum free phenytoin concentration allows a better estimate of toxicity and efficacy than does the total phenytoin concentration /9/. Altered protein binding of phenytoin, for example, is observed with hypoalbuminemia, displacement from albumin binding (e.g., by valproic acid), hyperbilirubinemia, and uremia. Phenytoin concentrations > 20 mg/L are associated with side effects (nystagmus, ataxia). Concentrations > 35 mg/L are associated with increased tendency to seizures.

– Phenobarbital

Therapeutic range: 10–40 mg/L.

Elimination half-life: 50–120 h (adults), 40–70 h (children).

Recommendation for blood sampling: during the dose interval.

Elimination: metabolism in the liver; no known metabolites with anticonvulsive effect.

Clinical aspects: the interpretation of the serum phenobarbital concentration is complicated by the fact that tolerance to the effect on the central nervous system can develop. The upper limit of the therapeutic range can vary between patients. Therefore, the measurement of phenobarbital is of limited value. Because of its ability to induce hepatic microsomal enzymes, phenobarbital can influence the metabolism of other drugs.

– Primidone

Therapeutic range: 5–15 mg/L.

Elimination half-life: 4–22 h.

Recommendation for blood sampling: measure peak level 2–4 h after the last dose; measure trough level immediately prior to the next dose.

Elimination: primidone is metabolized in the liver into two metabolites with anticonvulsive effect (phenobarbital, phenyl ethyl malonamide) and one without anticonvulsive activity. Approximately 25% of the administered dose of primidone is metabolized to phenobarbital, which, due to its long half-life, accumulates and reaches serum concentrations more than double those of primidone.

Clinical aspects: phenobarbital resulting from the conversion of primidone is responsible for most of the anticonvulsive effect of primidone. To monitor therapy with primidone, multiple measurements of both primidone and phenobarbital are undertaken, with phenobarbital being of greater significance. Primidone concentrations > 15 mg/L are usually considered toxic.

– Carbamazepine

Therapeutic range: 4–10 mg/L.

Elimination half-life: 10–25 h (continuous therapy).

Recommendation for blood sampling: Measure peak level 6–18 h after the last dose; measure trough level immediately prior to the next dose.

Elimination: carbamazepine is metabolized in the liver. Only small amounts (approx. 1%) of the parent drug are excreted in the urine. Multiple metabolites are created, of which carbamazepine-10, 11-epoxide has shown anticonvulsive efficacy in animal studies. How much this metabolite contributes to the therapeutic effect of carbamazepine is unclear. The serum concentration of carbamazepine-10, 11-epoxide is 5–81% (average: 30%) of that of carbamazepine. The metabolism of carbamazepine can be induced by the substance itself or by other anticonvulsants (e.g., phenytoin or phenobarbital). The clearance of carbamazepine is reduced in the presence of underlying liver diseases. During pregnancy, a decrease in the carbamazepine serum concentration is observed.

Clinical aspects: the serum concentration of carbamazepine is difficult to estimate due to slow and incomplete intestinal absorption, low renal excretion, and large interindividual differences in the half-life. Therefore, it is important to monitor it. In general, the measurement of carbamazepine-10, 11-epoxide appears to be unnecessary for routine clinical practice. Toxic doses of carbamazepine can have a pro convulsive effect. Carbamazepine concentrations > 15 mg/L are associated with side effects (visual disturbances, ataxia, nystagmus).

– Oxcarbazepine

Therapeutic range: oxcarbazepine is a prodrug that is metabolized rapidly and almost completely to the active metabolite 10-hydroxycarbazepine. A therapeutic range for 10-hydroxycarbazepine has not yet been established. In seizure-free adults, plasma concentrations of 13–35 mg/L have been reported /10/. Concentrations of 15–55 mg/L have been reported in children. Side effects are more frequent at plasma concentrations of 35–40 mg/L.

Elimination half-life: 8–10 h for 10-hydroxycarbazepine.

Recommendation for blood sampling: measure trough concentration immediately prior to the next dose.

Elimination: 10-hydroxycarbazepine is eliminated mainly by conjugation to glucuronic acid, which is excreted by the kidneys. In contrast to carbamazepine, repeated doses of oxcarbazepine do not affect its own metabolism. Induction and/or inhibition of the cytochrome P450 system has little effect on the pharmacokinetics of oxcarbazepine and its active metabolites.

Clinical aspects: since there appears to be a linear relationship between the oxcarbazepine dose and the 10-hydroxycarbazepine plasma concentration, monitoring the plasma concentration of the active metabolite is of limited usefulness. An advantage of oxcarbazepine is that 10-hydroxycarbazepine does not undergo epoxidation, thereby preventing the production of carbamazepine epoxide, which is primarily responsible for the toxic side effects of carbamazepine. HPLC methods are available for determining 10-hydroxycarbazepine.

– Lamotrigine

Therapeutic range: 3–14 mg/L. These values are tentative, however, since a therapeutic range has not yet been firmly established /10/.

Elimination half-life: approximately 25 h (15–35 h).

Recommendation for blood sampling: measure trough concentration immediately prior to the next dose.

Elimination: mainly renal, as glucuronide.

HPLC and LC-MS/MS methods are available for determining plasma concentrations.

Clinical aspects: therapeutic drug monitoring is recommended for lamotrigine because of significant inter individual differences in its pharmacokinetics. The clearance of lamotrigine is age-dependent. It is higher in children than in adults and declines even further in elderly patients. It also appears to be significantly increased in the later stages of pregnancy. Lamotrigine is associated with significant drug interactions. Phenytoin, carbamazepine, and barbiturates induce the metabolism of lamotrigine, reducing its half-life to 15 h (8–20 h). Conversely, valproic acid inhibits the metabolism of lamotrigine, increasing its half-life to 60 h (30–90 h). HPLC methods are available for determining plasma concentrations of lamotrigine /10/.

– Topiramate

Therapeutic range: 5–20 mg/L /10/. These values are tentative.

Elimination half-life: 20–30 h.

Recommendation for blood sampling: Measure trough concentration immediately prior to the next dose.

Elimination: topiramate is mainly eliminated unchanged in the urine; only a certain proportion is metabolized by oxidation.

Clinical aspects: because the correlation between concentration and effect has not been studied systematically to date, the available therapeutic range is only tentative. In clinical studies, serum topiramate concentrations between 3.4 and 5.2 mg/L have been associated with an adequate anticonvulsive effect. In refractory patients, concentrations > 4 mg/L were necessary for an adequate response, and there are indications that concentrations > 10 mg/L may be required. Further studies are required to define the therapeutic window more clearly. Phenytoin and carbamazepine induce the metabolism of topiramate, leading to a significantly reduced serum concentration. Valproic acid can also lower the concentration of topiramate, but to a lesser extent. Due to these pharmacokinetic interactions, therapeutic drug monitoring for topiramate would appear to be useful in selected patients /10/.

– Ethosuximide

Therapeutic range: 40–100 mg/L.

Elimination half-life: 30–60 h (adults), 20–55 h (children).

Recommendation for blood sampling: during the dose interval.

Elimination: pharmacologically active metabolites do not occur; approximately 20% of the daily dose is excreted unchanged in the urine.

Clinical aspects: ethosuximide is used to treat petit mal epilepsy. Serum concentrations of greater than 150 mg/L are usually considered toxic. The frequency of absences can increase with toxic doses.

– Valproic acid

Therapeutic range: 50–100 mg/L.

Elimination half-life: 10–16 h.

Recommendation for blood sampling: measure peak level 1–4 (–8) h after the last dose; measure trough level immediately prior to the next dose.

Elimination: metabolism in the liver; 1–3% of the dose is excreted unchanged in the urine.

Clinical aspects: there does not seem to be a direct correlation between the serum concentration of valproic acid and its efficacy /11/. Routine serum measurements of valproic acid are therefore of limited value for optimizing the dose. Measurement of the valproic acid concentration is, however, clinically useful for checking patient compliance.

Cardiac glycosides – Generally

Indication for measuring concentration: suspected toxicity, e.g. arrhythmias; therapy failure, e.g. due to unreliable patient compliance or pharmacokinetic reasons. Therapeutic use in patients with altered pharmacokinetics, e.g. in renal failure, severe decompensated heart failure, thyroid dysfunction, or drug interactions. Lack of information about prior therapeutic use of cardiac glycosides (e.g., in the case of unconscious patients). In the case of a non-visible digitalis effect due to preexisting ECG abnormalities (e.g., in patients with a cardiac pacemaker or in ventricular bundle branch block).

– Digoxin

Therapeutic range: 0.8–2.0 μg/L; 0.5–0.8 μg/L (heart failure in sinus rhythm) /12/.

Elimination half-life: 1–2 d.

Recommendation for blood sampling: 8–24 h after the last dose.

Elimination: only a small percentage is converted in the liver to metabolites with cardiac effects and the significantly less active metabolite dihydrodigoxin. Elimination is predominantly renal.

Clinical aspects: because the effects of digitalis are dependent on numerous factors, there is an overlap between the therapeutic and toxic ranges for digoxin /13/. The interpretation of serum values is therefore only meaningful in conjunction with the assessment of the overall clinical situation. Factors that influence the serum concentration or efficacy of cardiac glycosides at therapeutic doses are:

Disturbances of electrolyte status and acid-base balance. A serum digoxin concentration usually considered therapeutic can exert toxic effects due to the presence of hypokalemia, e.g. as found with the combination of digoxin plus diuretics, hypercalcemia, and hypomagnesemia. Tolerance to digoxin is also reduced in acid-base disorders, tissue hypoxia, acute myocardial infarction, cardiomyopathies, and valvular heart diseases.

Concomitant diseases or conditions that result in a change in the pharmacokinetics of digoxin. These include decreased glomerular filtration in patients with advanced renal failure and in elderly patients and impaired intestinal absorption in patients with malabsorption syndrome. Patients with hypothyroidism generally seem to have higher, and patients with hyperthyroidism on average lower, digoxin concentrations than would be expected from the dose.

Drug interactions. A significant increase in the serum digoxin concentration can result from concomitant use of quinidine at a customary dose (750–1500 mg/d). Digoxin concentrations that do not correlate with the administered dose can occur with concomitant use of drugs that impair the intestinal absorption of digoxin (e.g., cholestyramine, neomycin, antacids, or kaolin/pectin).

High digoxin concentrations are frequently measured in patients with cardiac pacemakers because glycoside-induced ECG changes cannot be discerned.

– Digitoxin

Therapeutic range: 10–25 μg/L.

Elimination half-life: 6–8 d.

Recommendation for blood sampling: 8–24 h after the last dose. Digitoxin is much more strongly bound to protein (90–97%) than digoxin.

Elimination: digitoxin is metabolized in the liver. During this process, approximately 10% of the administered dose is converted into digoxin and 30% is eliminated renally.

Clinical aspects: see under digoxin.

Antiarrhythmic agents – Lidocaine

Therapeutic range: 1.5–5.0 mg/L.

Elimination half-life: 70–140 min.

Recommendation for blood sampling: during the infusion.

Elimination: metabolized in the liver. The clearance of lidocaine is reduced in the presence of liver diseases (e.g., cirrhosis) and reduced hepatic perfusion due to heart failure.

Clinical aspects: parenteral therapy of ventricular arrhythmias. See under disopyramide. Lidocaine can also be used as a test substance for the purpose of assessing liver function /14/. In this context, the cytochrome-P450-mediated formation of the lidocaine metabolite monoethylglycinexylidide (MEGX) serves as an indicator of liver function. Decreased cytochrome P450 3 A4 activity in the hepatocyte as well as disturbances of hepatic perfusion lower the rate of formation of this metabolite. The MEGX test was developed based on this concept. This test is particularly useful in the field of liver transplantation, where it is used to assess the quality of donor organs, to estimate the prognosis of transplant candidates, and to evaluate the function of the graft during the early postoperative course. In addition, the formation of MEGX is an early predictor of posttraumatic multi organ failure.

MEGX test protocol: blood sampling before, and 15 or 30 min. after, an i.v. bolus injection (administered over 2 min.) of 1 mg/kg lidocaine hydrochloride. Measurement of MEGX in the serum by HPLC, LC-MS/ MS /15/. If the MEGX concentration measured before the test dose is administered is above the detection limit (3 mg/L), this concentration is to be subtracted from the MEGX values at 15 and 30 min respectively. This test must not be performed in patients with known lidocaine allergy or cardiac injury.

Interpretation of test results /14/:

  • MEGX 15 or 30 min. levels > 50 mg/L in liver donors are consistent with a functionally intact donor organ.
  • MEGX 30 min. levels < 10 mg/L in transplant candidates with chronic liver disease indicate a high mortality risk.
  • MEGX 15 min. levels < 20 mg/L within the first 3 days after liver transplantation are consistent with hepatic dysfunction and reduced survival of the transplanted organ.
  • MEGX 15 min. levels < 30 mg/L on the 3rd day post injury in a patient with multiple trauma are predictive of multiple organ failure.

Based on current data, MEGX test results < 20 mg/L appear to be associated with hepatic dysfunction in adults. When interpreting the MEGX test results, it is important to consider the overall clinical picture, because extrahepatic factors, e.g. heart failure or therapeutic drugs, can affect hepatic perfusion or the activity of the cytochrome P450 system.

– Phenytoin

Therapeutic range: 10–18 mg/L.

Blood sampling, elimination: see under anticonvulsants.

Clinical aspects: oral and intravenous therapy for ventricular arrhythmias.

– Procainamide, N-acetylprocainamide

Therapeutic range: procainamide 4–10 mg/L, N-acetylprocainamide 6–20 mg/L. For the total serum concentration of procainamide and its active metabolite N-acetylprocainamide, a therapeutic range of 10–30 mg/L has been proposed. The clinical significance of this range, however, has not been adequately clarified.

Elimination half-life: procainamide 3–5 h, N-acetylprocainamide 6–10 h.

Recommendation for blood sampling: measure trough level immediately prior to the next dose; measure peak level 1–5 h after the last oral dose.

Elimination: metabolized in the liver to the active metabolite N-acetylprocainamide (20–50%). Due to N-acetyltransferase polymorphism, significant interindividual differences in the metabolism of this substance occur. Approximately 50% of the dose is excreted unchanged in the urine /16/. An increase in the serum concentration is seen in heart failure and renal failure.

Clinical aspects: prophylaxis and therapy of atrial and ventricular arrhythmias.

– Quinidine

Therapeutic range: 2–5 mg/L.

Elimination half-life: approximately 6 h.

 Recommendation for blood sampling: measure peak level 1–3 h after dose (for sustained release preparations, approximately 8 h); measure trough level immediately prior to the next dose.

Elimination: metabolized in the liver to metabolites that are likely to be active; 10–20% is excreted unchanged in the urine. Reduced elimination can result from chronic liver disease, heart failure, or urinary alkalinization.

Clinical aspects: continuous oral therapy and prophylaxis of atrial and ventricular cardiac arrhythmias.

– Disopyramide

Therapeutic range: 2–5 mg/L.

Elimination half-life: 7 h.

Recommendation for blood sampling: measure peak level 2–3 h after dose; measure trough level immediately prior to the next dose.

Elimination: N-desisopropyldisopyramide, a metabolite formed in the liver, exerts a lesser antiarrhythmic effect in animals. Approximatey 40–60% of the administered dose is excreted unchanged in the urine.

Clinical aspects: continuous oral therapy and prophylaxis of atrial and ventricular cardiac arrhythmias.

Clinical aspects of antiarrhythmic agents: monitoring the serum concentration of antiarrhythmic agents is clinically relevant for detecting the therapeutic endpoint. A clear relationship exists between the serum concentrations of some antiarrhythmic agents and their therapeutic efficacy. For instance, in patients with stable therapeutic serum concentrations of quinidine or procainamide, no new episodes of ventricular fibrillation were observed /17/. High serum concentrations of quinidine, procainamide, and N-acetylprocainamide can induce ventricular arrhythmias.

Theophylline

Therapeutic range: 8–20 mg/L (adults, children), 6–11 mg/L (premature infants).

Elimination half-life: 3–12 h adults; 2–6 h children (1–17 years) and smokers; approximately 30 h premature infants and adults with liver cirrhosis.

Recommendation for blood sampling: during continuous infusion until steady state is reached 4–8, 12, 24, 48 h after the start of the infusion and during further therapy for dose adjustments. Measure trough level immediately prior to the next maintenance dose; measure peak level approximately 1 h (for sustained release preparations, approximately 4 h) after the last dose.

Elimination: theophylline is largely converted to relatively inactive metabolites in the liver. In newborns, theophylline is also metabolized to caffeine. Only approximately 10% of the dose is excreted unchanged in the urine.

Clinical aspects: theophylline is used as a bronchodilator and for the treatment of apnea in premature infants. The clearance of theophylline is reduced in the presence of heart failure, liver cirrhosis, acute viral respiratory infections, and during the concomitant use of certain drugs (e.g., cimetidine, erythromycin, or allopurinol). Due to significant interindividual differences in pharmacokinetics, theophylline dose regimens based on body weight alone are unreliable and sometimes even dangerous for the patient.

Indication for measuring theophylline concentration: suspected toxicity, therapy failure (e.g., due to unreliable patient compliance or pharmacokinetic reasons), or administration of theophylline as a continuous i.v. infusion (e.g., in severe respiratory obstruction); lack of information about any prior therapeutic use of theophylline; or therapeutic use in patients with altered pharmacokinetics (e.g. in cases of concomitant disease, changes in patterns of tobacco use, or drug interactions). Pharmacokinetic methods can be used for individual dose adjustments /8/. Based on the theophylline concentration in the saliva, it is possible to estimate whether the correct dose was chosen and adequate patient compliance is present. The ratio of the saliva/serum concentration of theophylline is on average 0.68 (ranging from 0.5 to 0.85). In cases of suspected toxicity and for optimizing the dose, however, it is necessary to measure the theophylline concentration in the serum.

Methotrexate

Therapeutic range: a therapeutic range does not exist. To avoid serious toxic side effects, the serum methotrexate concentrations during high-dose methotrexate therapy (infusion over a period of 4–6 h) should be kept below the following values:

  • ≤ 10 μmol/L 24 h after the start of the infusion
  • ≤ 1.0 μmol/L 48 h after the start of the infusion
  • ≤ 0.1 μmol/L 72 h after the start of the infusion.

Recommendation for blood sampling: 24, 48, and frequently also 72 h after the start of the methotrexate infusion. In cases of delayed elimination, further blood sampling is required until the methotrexate concentration is < 0.05–0.10 μmol/L.

Elimination half-life: biexponential decline in serum concentration with half-life of 2–4 h and 10–20 h respectively /18/.

Elimination: methotrexate is largely (80%) excreted unchanged in the urine. A small percentage of the dose reaches the intestine in the bile and undergoes enterohepatic circulation. It would appear that methotrexate can be metabolized by the intestinal bacterial flora to 4-amino-4-deoxy-N10-methylpteroic acid. 7-hydroxymethotrexate is another important metabolite, which is considered to be potentially nephrotoxic. Both metabolites exhibit significantly less inhibition of dihydrofolate reductase than methotrexate.

Clinical aspects: methotrexate is an inhibitor of dihydrofolate reductase. It is used in chemotherapy for malignant diseases (e.g., osteogenic sarcoma). Methotrexate clearance is reduced in the presence of renal and hepatic diseases as well as in aciduria. In the case of ascites or pleural effusions, the half-life of methotrexate is prolonged. Toxic side effects occur when the concentration exceeds the specified threshold values. The severity of toxicity is determined more by the length of time by which the time limit is exceeded than by the extent of the rise in the methotrexate concentration above the concentration limit. Monitoring the methotrexate concentration allows any possible risk to the patient to be detected at an early stage. If necessary, life-threatening side effects can then be prevented by administering the antidote leucovorin (citrovorum factor) and implementing measures to increase the renal excretion of methotrexate. Administration of leucovorin should be continued until the serum methotrexate concentration has decreased below 0.10 μmol/L), or better still, 0.01 μmol/L.

Lithium

Therapeutic range: 0.6–1.2 mmol/L /19/.

Elimination half-life: 14–33 h.

Recommendation for blood sampling: 12 h after the last dose.

Elimination: renal excretion, which is increased by a high intake of sodium and water.

Clinical aspects: even if renal function is normal and a standard maintenance dose is administered, significant variations in the serum lithium concentration can occur. Toxic side effects such as muscle twitching, ataxia, and drowsiness occur at concentrations > 1.5 mmol/L; seizures, dehydration, and coma occur at concentrations > 3.0 mmol/L; concentrations > 4.0 mmol/L are potentially lethal. It is therefore necessary to monitor the serum lithium concentration during continuous lithium therapy for depressive and manic-depressive disorders.

Amino glycosides

Therapeutic ranges:

  • Amikacin: 20–30 mg/L (peak), < 5 (8) mg/L (trough)
  • Gentamicin: 5–10 mg/L (peak), < 2 mg/L (trough)
  • Netilmicin: 5–12 mg/L (peak), < 3 mg/L (trough)
  • Tobramycin: 5–10 mg/L (peak), < 2 mg/L (trough)

Elimination half-life: 0.5–3.0 h in the presence of normal renal function.

Recommendation for blood sampling: measure peak level 30 min. after completion of an i.v. infusion over 30 min. or 1 h after i.m. administration; measure trough level immediately prior to the next dose.

Elimination: excreted unchanged in the urine. No known metabolites.

Clinical aspects: amino glycosides are used to treat serious infections involving gram-negative bacteria. Impaired renal function results in reduced clearance of amino glycosides. In dehydration, the distribution volume decreases. High concentrations of certain penicillins, e.g. carbenicillin, can inactivate amino glycosides in vitro and in vivo. Accumulation of amino glycosides in the tissues can result in ototoxicity and nephrotoxicity. The trough serum concentration (prior to the administration of the next dose) is an important indicator for the accumulation of these substances. A suitable dosing regimen has to be set up such that, on the one hand, an adequate peak serum concentration of the amino glycoside above the minimum inhibitory concentration for the pathogen in question can be achieved, while on the other hand, the trough serum concentration remains below the threshold above which toxicity can be expected. The use of pharmacokinetic methods for individual dosage adjustment is highly recommended /7/. For once-daily dosing of amino glycosides, no particular recommendations exist with regard to drug monitoring /20/. Once-daily dosing with 6.6 mg netilmicin/kg, for instance, resulted in an average peak serum concentration of 21.3 mg/L /21/.

Vancomycin

Therapeutic range: 5–20 mg/l (trough) /22/.

Elimination half-life: 4–10 h (adults), 2–3 h (children), 6–10 h (newborns).

Recommendation for blood sampling: measure peak level 1 h after completion of an i.v. infusion; measure trough level immediately prior to the next dose.

Elimination: more than 90% of the administered dose is excreted unchanged in the urine.

Clinical aspects: vancomycin is effective against gram-positive bacteria. A decline in renal function can cause the serum vancomycin concentration to increase to a toxic level. In patients with reduced liver function, elimination of vancomycin is delayed. At serum concentrations above the therapeutic range, vancomycin exerts ototoxic and nephrotoxic effects. The degree of ototoxicity is increased by concomitant use of amino glycosides or furosemide.

Cyclosporine

Therapeutic range: this is difficult to define since there are no easy-to-measure parameters for assessing the immunosuppressive effect. The trough serum concentration of cyclosporine prior to the next dose is generally used to guide cyclosporine therapy after organ transplantation /23/. The following tentative range for the cyclosporine trough concentration in whole blood is recommended for specific analytic methods such as LC-MS/MS: 100–300 μg/L. Therapeutic ranges for trough concentrations of cyclosporine are shown in Tab. 40-2 – Therapeutic ranges. During the early postoperative phase, cyclosporine concentrations should be maintained within the upper half of the therapeutic range. Usually, the dose of cyclosporine is reduced in a stepwise manner within 3–6 months after transplantation. The cyclosporine concentrations during maintenance therapy are adjusted to lie within the lower part of the therapeutic range. Cyclosporine concentrations > 400 μg/L are associated with a high risk of toxicity /23/. It is still unclear whether lower therapeutic ranges are adequate when cyclosporine is used in combination with other immunosuppressive drugs such as mycophenolate mofetil or sirolimus. An alternative approach is C2 monitoring. The cyclosporine concentration (C2) determined 2 h after the administration of the micro emulsion formulation Sandimmun® Optoral allows a better estimate of cyclosporine exposure and the risk of acute rejection or toxicity than an estimate based on the trough level. The most significant inhibition of calcineurin activity and maximum reduction in IL-2 production by cyclosporine occur in the first two hours after cyclosporine administration. The particular importance of adequate cyclosporine exposure within the first 3–5 days after transplantation to prevent acute rejection has been documented in prospective studies of patients who have received kidney or liver transplants. However, prospective studies have been unable to provide evidence to support the theoretical benefit of C2 monitoring /24/.

Elimination half-life: terminal half-life in healthy individuals 6.3 (4.7–9.5) h, in kidney transplant patients 10.7 (4.3–53.4) h, and in patients with liver cirrhosis 20.4 (10.8–48.0) h /25/.

Recommendation for blood sampling: measure trough level immediately prior to the next dose, e.g. 12 h after each dose with twice-daily dosing. C2 concentration: 2 h ± 15 min. after the last dose. Additional subsequent blood sampling, e.g. 6 h post-dose, is necessary in patients with reduced or delayed absorption. EDTA is the anticoagulant of choice.

Elimination: 99% of cyclosporine is metabolized in the liver and exhibits a first-pass effect. Multiple metabolites are produced /25/. Less than 1% of the dose is excreted unchanged in the urine or bile.

Clinical aspects: Cyclosporine metabolites are excreted mainly in the bile. Other immunosuppressive drugs such as mycophenolate mofetil or sirolimus. An alternative approach is C2 monitoring. The cyclosporine concentration (C2) determined 2 h after the administration of the microemulsion formulation Sandimmun® Optoral allows a better estimate of cyclosporine exposure and the risk of acute rejection or toxicity than an estimate based on the trough level. The most significant inhibition of calcineurin activity and maximum reduction in IL-2 production by cyclosporine occur in the first two hours after cyclosporine administration. The particular importance of adequate cyclosporine exposure within the first 3–5 days after transplantation to prevent acute rejection has been documented in prospective studies of patients who have received kidney or liver transplants. However, prospective studies have been unable to provide evidence to support the theoretical benefit of C2 monitoring /24/.

Elimination half life: terminal half-life in healthy individuals 6.3 (4.7–9.5) h, in kidney transplant patients 10.7 (4.3–53.4) h, and in patients with liver cirrhosis 20.4 (10.8–48.0) h /25/.

Recommendation for blood sampling: measure trough level immediately prior to the next dose (e.g., 12 h after each dose with twice-daily dosing). C2 concentration: 2 h ± 15 min. after the last dose. Additional subsequent blood sampling (e.g., 6 h post dose) is necessary in patients with reduced or delayed absorption. EDTA is the anticoagulant of choice.

Elimination: 99% of cyclosporine is metabolized in the liver and exhibits a first-pass effect. Multiple metabolites are produced /25/. Less than 1% of the dose is excreted unchanged in the urine or bile. Cyclosporine metabolites are excreted mainly in the bile.

Clinical aspects: cyclosporine is a highly effective immunosuppressive substance used to prevent graft rejection (kidney, liver, heart, lung, pancreas, bone marrow). It inhibits calcineurin and suppresses the synthesis of cytokines (e.g., IL-2). As a result of immunosuppression, there is a risk of infection, which is significantly higher in the presence of toxic cyclosporine concentrations. Cyclosporine can also cause serious nephrotoxic, hepatotoxic, and neurotoxic effects. These side effects are usually dose-dependent. In the event of deteriorating renal function, e.g. serum creatinine concentration > 2 mg/dL (177 μmol/L) 3–6 months after transplantation, cyclosporine can be reduced or discontinued and the use of other immunosuppressive drugs such as mycophenolate mofetil and sirolimus can be increased. The pharmacokinetics of cyclosporine show significant intraindividual and inter individual differences. After oral administration of the micro emulsion preparation Sandimmun® Optoral in comparison to Sandimmun®, less variability in the cyclosporine kinetics is observed as well as less dependence on bile acids and no interaction with food intake. The time required to reach the peak blood concentration after oral intake of Sandimmun® Optoral is 2–3 h. Multiple drug interactions have been observed. Certain drugs (e.g., ketoconazole, phenytoin, phenobarbital, and steroids) affect the metabolism of cyclosporine whereas other substances (e.g., amino glycosides and cephalosporins) increase its nephrotoxicity.

Cyclosporine significantly decreases exposure to mycophenolic acid, an immunosuppressive agent frequently used in combination with cyclosporine. In patients with abnormal liver function, reduced absorption and delayed elimination of cyclosporine mean that lower doses are generally required. Children have a higher cyclosporine clearance than adults.

Monitoring of the blood concentration of cyclosporine is necessary to optimize the dose regimen. Recent studies have shown that acute rejection in the first year following kidney transplantation negatively impacts long-term graft survival.

Because numerous factors (e.g., temperature, lipoprotein concentration, and hematocrit) influence the blood/plasma concentration ratio, cyclosporine should be measured in hemolyzed blood. During the early postoperative phase in patients with kidney, liver, and heart transplants, cyclosporine blood monitoring is carried out 4–7 times per week. Following this period of close monitoring, the frequency of measurement is reduced in a stepwise manner. For instance, in kidney transplant patients with an uncomplicated course, the cyclosporine concentration should be monitored once a month during the first year and every 1–3 months thereafter. Additional measurements are necessary if the clinical situation requires a change in dose. This is also the case during concomitant treatment with drugs known to interact with the metabolism of cyclosporine. Changes in dose should be monitored the following week by measuring the cyclosporine level.

Specific HPLC and LC-MS/MS methods or immunoassays are available for measuring cyclosporine in the blood. For individual dose adjustment, especially in patients post liver transplantation or with abnormal liver function, specific methods such as LC-MS/MS /26/ or a specific immunoassay should ideally be used to determine the trough level. With regard to C2 monitoring, differences in the specificities of the immunoassays are less relevant (more favorable ratio of parent substance to cross-reactive metabolites), which means that assay-specific reference values are unnecessary. Impaired liver function leads to the accumulation of cyclosporine metabolites in the blood. Routine measurements of metabolite concentrations, however, do not appear to be justified.

Tacrolimus

Therapeutic range: as in the case of cyclosporine, it is difficult to define a specific range. The trough serum concentration of tacrolimus prior to the next dose is used to guide tacrolimus therapy following organ transplantation. If commercial tacrolimus assays are used, a tentative trough concentration range of 5–18 μg/L should be used while, based on our experiences, the corresponding range for LC-MS/MS is 4–15 μg/L.

During the early postoperative phase, tacrolimus concentrations should be maintained within the upper half of the therapeutic range. The dose of tacrolimus is usually reduced in a stepwise manner within 3–6 months after transplantation (Tab. 40-2 – Therapeutic ranges). Concentrations > 20 μg/L are associated with an increased incidence of toxic side effects.

Elimination half-life: terminal half life approximately 16 h in patients with kidney transplant and approximately 12 h in patients with liver transplant; if hepatic function is severely impaired, the elimination half-life may be > 50 h.

Recommendation for blood sampling: immediately prior to the next dose, e.g. 12 h after each dose with twice-daily dosing. EDTA is the anticoagulant of choice.

Elimination: tacrolimus is mainly metabolized by cytochrome P450 3A in the liver. Multiple metabolites are produced. Less than 5% of the dose is excreted unchanged in the bile. Tacrolimus metabolites are excreted mainly in the bile.

Clinical aspects: tacrolimus is a highly effective immunosuppressive substance used to prevent graft rejection (e.g., liver, kidney, heart). Like cyclosporine, it inhibits calcineurin. As a result of immunosuppression, there is a risk of infection, which is significantly higher in the presence of toxic tacrolimus concentrations. Like cyclosporine, tacrolimus can cause dose-dependent nephrotoxic and neurotoxic side effects (such as tremor). Another side effect is hypertension. Tacrolimus also has a diabetogenic effect. However, it does not appear to have a significant effect on the plasma cholesterol concentration. The peak concentration of tacrolimus is reached after approx. 2 h (0.7–6 h). Taking tacrolimus with food delays the increase in the blood concentration. Multiple drug interactions have been observed. Certain therapeutic drugs (e.g., ketoconazole, erythromycin, fluconazole, diltiazem, cimetidine) and high-dose steroids increase the tacrolimus concentration while other substances (e.g., rifampicin, barbiturates, and carbamazepine) taken at the same time reduce the concentration. The pharmacokinetics of tacrolimus show significant intraindividual and inter individual differences. Monitoring of the blood concentration of tacrolimus is necessary to optimize the dose regimen. During the first two weeks after organ transplantation, tacrolimus levels should be measured four to seven times per week. The frequency of measurement can then be reduced in a step wise manner (as with cyclosporine). A sustained release formulation is available for once daily dosing. In concentration a formulation licensed for twice daily dosing, the trough concentrations achieved with once daily formulation are on average 12.7% lower at the same dose /27/. The two formulations are considered to be bioequivalent. The same TDM approach to the therapeutic range of tacrolimus applies for both formulations but the differences in the trough levels must be taken into account when interpreting the results /28/.

Immunoassays and highly specific LC-MS/MS /26/ methods are available for measuring tacrolimus in the blood. Cross reactivity with tacrolimus metabolites (M-II, M-III, M-V) is observed when immunoassays are used. Accumulation of tacrolimus metabolites (e.g. due to cholestasis) can therefore lead to falsely high tacrolimus concentrations if certain immunoassays are used.

Azathioprine

Therapeutic range: azathioprine itself is a prodrug and does not have immunosuppressive effects. It is converted into the active metabolite 6-thioguanine nucleotide (6-TGN). The tentative therapeutic range for the erythrocyte 6-TGN concentration in transplant recipients undergoing triple therapy with cyclosporine and prednisone is 100–450 pmol/0.8 × 109 erythrocytes and in patients receiving azathioprine therapy for chronic inflammatory bowel disease is 250–450 pmol/0.8 × 109 erythrocytes /29/. For chemotherapy with 6-mercaptopurine, a 6-TGN concentration of 500–1,000 pmol/0.8 × 109 erythrocytes has been proposed. An increased risk of azathioprine intolerance is observed in patients with thiopurine methyl transferase (TPMT) deficiency /29/. Erythrocyte TPMT activity < 2.8 nmol/(mL Ery × h) indicates homozygous TPMT deficiency while TPMT activity of 2.8–10.0 nmol/(mL Ery × h) indicates heterozygous TPMT deficiency. During therapy with thiopurines (e.g., azathioprine, 6-mercaptopurine), induction of TPMT activity occurs, so that even patients with constitutively intermediate activity levels can have values > 10 nmol/(mL Ery × h).

Recommendation for blood sampling: during the dose interval. The measurement is performed in erythrocytes from heparinized or EDTA whole blood.

Elimination: in vivo, azathioprine is metabolized via 6-mercaptopurine to 6-TGN, by xanthine oxidase to 6-thiouric acid, and by TPMT to 6-methyl mercaptopurine. In TPMT deficiency, relatively more 6-TGN is produced.

Clinical aspects: azathioprine is used to treat chronic inflammatory bowel disease (e.g. Crohn’s disease) and following organ transplantation. Patients with a deficiency of the enzyme TPMT are at risk of azathioprine intolerance. In patients with homozygous TPMT deficiency, which is observed in 0.3% of Caucasians, normal doses of azathioprine lead to severe bone marrow suppression. Determination of TPMT activity can therefore be used to screen patients for homozygous TPMT deficiency before starting azathioprine therapy. In these patients, the possibility of using an alternative form of immunosuppressive therapy must be examined. If azathioprine is still to be used, it is essential to monitor the 6-TGN concentration. In these cases, approximately one-tenth of the normal dose must be used. The risks associated with TPMT deficiency are also relevant for the use of 6-mercaptopurine or thioguanine in chemotherapy, for acute juvenile leukemia in particular /30/. If the TPMT activity is consistent with a ”wild type” genotype [(10–20) nmol/mL Ery × h], therapy with standard doses of thiopurine drugs (e.g., azathioprine, 6-mercaptopurine) can be started immediately in accordance with the usual level of monitoring recommended by specialists (blood count, including platelet count). If intermediate TPMT activity [(2.8–9.9) nmol/(mL Ery × h] is demonstrated, typically due to a heterozygous TPMT gene mutation, therapy should be commenced at 30–50% of the standard dose and, if tolerated (WBC count, 6-TGN concentration), increased weekly until the target dose is reached. Note that in vitro assays cannot clearly identify inhibition of TPMT activity by concomitantly administered drugs (e.g., amino salicylate, sulfasalazine). In these cases, measurement of the 6-TGN concentration should be used to guide therapy. In patients with increased TPMT activity (> 20 nmol/mL Ery × h), there is evidence to suggest that using standard doses results in lower concentrations of active 6-TGN, which can lead to therapy failure. In these patients, increasing the dose can result in increased production of hepatotoxic metabolites (6-MMP, 6-methyl mercaptopurine; 6-MMPR, 6-methyl mercaptopurine ribonucleotide). The measured 6-TGN concentration in conjunction with monitoring of aminotransferases should be used to guide therapy.

An HPLC method is used to determine the 6-TGN concentration in erythrocytes. The activity of TPMT in the erythrocytes is also measured using suitable HPLC methods. However, it is not possible to determine the TPMT phenotype reliably based on the TPMT activity for 30–60 days after a blood transfusion. In such cases, and in patients with diseases associated with over-aged erythrocytes, genotyping must be performed. Twenty allelic variants for the TPMT gene are currently known /31/. Due to a lack of standardization, the ranges specified for the 6-TGN concentration and TPMT activity depend on the method used.

Sirolimus

Therapeutic range:

  • 4–12 μg/L (trough) for triple therapy with cyclosporine, corticosteroids, and sirolimus in kidney transplant patients
  • 12–20 μg/L (trough) for dual therapy with corticosteroids and sirolimus in kidney transplant patients
  • 3–6 μg/L (trough) for liver transplant patients with secondary immunosuppression (e.g., due to calcineurin inhibitor nephrotoxicity) with a 50% reduction in the calcineurin trough concentration
  • 5–8 μg/L (trough) for liver transplant patients without calcineurin.

Elimination half-life: 59 ± 19 h (adults), 10–23 h (children aged 5–11 yrs.), approx. 110 h (patients with abnormal liver function) /32/.

Recommendation for blood sampling: immediately prior to the next dose. EDTA is the anticoagulant of choice.

Elimination: sirolimus is mainly metabolized by CYP3A4 in the liver into multiple relatively inactive metabolites. Approximately 2.2% of the dose is excreted by the kidneys.

Clinical aspects: sirolimus (rapamycin) is a highly effective immunosuppressive substance used to prevent graft rejection (e.g. kidney, heart, liver). It inhibits T-cell activation and proliferation by inhibiting mTOR (mammalian target of rapamycin). The pharmacokinetics of sirolimus show significant intraindividual and inter individual differences. It has a bioavailability of approximately 14%. The low bioavailability of sirolimus following oral administration is due to its extensive metabolism by CYP3A4 in the intestine and liver and P-glycoprotein counter transport in the intestine. Peak blood concentrations are achieved after 1–2 h and the plasma free fraction is only about 8%. Multiple drug interactions have been observed. Ketoconazole, itraconazole, cyclosporine, erythromycin, nelfinavir, and diltiazem increase sirolimus exposure while rifampicin decreases it. Conversely, sirolimus increases cyclosporine exposure approximately 2-fold /32/. Sirolimus does not affect mycophenolic acid exposure. Although sirolimus is nephrotoxic only in the presence of cyclosporine, it has dose-dependent side effects such as hyperlipidemia and thrombocytopenia. According to current tentative drug monitoring recommendations, the sirolimus blood concentration should be determined as follows: following administration of the loading dose, following the establishment of a new steady state after a change in dose (approx. 7–10 days after the dose change), and after starting or discontinuing inhibitors or inducers of CYP3A4 and the P-glycoprotein transporter. The sirolimus trough level should be measured weekly for the first month after transplantation and every 2 months thereafter. Monitoring is also advisable in the event of a change in the dose or steady state concentration of cyclosporine; a change in the relative dosing intervals of sirolimus and cyclosporine; hyperlipidemia, leukopenia, thrombocytopenia, or liver disease; and to check compliance. Close monitoring is recommended for pediatric patients and patients with abnormal liver function.

Highly specific LC-MS/MS methods /26/ or LC-UV methods and a less specific immunoassay are available for measuring blood sirolimus levels. The specified therapeutic ranges only apply to specific methods (e.g., LC-MS/MS).

Everolimus

Therapeutic range: 3–8 μg/L (trough) for triple therapy with cyclosporine, corticosteroids, and everolimus in kidney transplant patients /33/.

Elimination half-life: 28 (24–35) h.

Recommendation for blood sampling: immediately prior to the next dose. EDTA is the anticoagulant of choice.

Elimination: everolimus is mainly metabolized by CYP3A4 in the liver.

Clinical aspects: everolimus is a highly effective immunosuppressive substance used to prevent graft rejection (e.g., kidney, heart, liver). It is also used to treat renal cell carcinoma and sub ependymal giant cell astrocytoma [SEGA]. In this setting, a tentative therapeutic range of 5–15 μg/L is used. Everolimus works by inhibiting mTOR. The pharmacokinetics of everolimus show significant intraindividual and inter individual differences. It has a bioavailability of 5–26%. The low bioavailability of everolimus following oral administration is due to its extensive metabolism by CYP3A4 in the intestine and liver and P-glycoprotein counter transport in the intestine. Peak blood concentrations are achieved after 1–2.2 h and the plasma free fraction is about 26%. As with sirolimus, multiple drug interactions have been observed. The everolimus trough level should be measured weekly for the first month after transplantation and every 2 months thereafter. The indications for drug monitoring are analogous to those for sirolimus.

LC-MS/MS methods /26/ and an immunoassay are available for measuring everolimus levels. The specified therapeutic range applies if LC-MS/MS is used.

Mycophenolic acid

Therapeutic range:

  • 1.3–3.5 mg/L (plasma concentration prior to the administration of the next dose, C0) for triple therapy with cyclosporine, corticosteroids, and mycophenolate mofetil (MMF) in kidney transplant patients in the early post-transplantation phase (≤ 3 months)
  • 1.9–4.0 mg/L with concomitant use of tacrolimus
  • (30–60) mg × h/L MPA AUC012for triple therapy with cyclosporine or tacrolimus, corticosteroids, and MMF /3334/.

Algorithms have been developed to determine the MPA AUC using three measurement points /34/. The MPA AUC can also be estimated using Bayesian forecasting (https://pharmaco.chu-limoges.fr). The ranges specified apply if HPLC or LC-MS/MS is used. According to provisional results, the therapeutic ranges for liver and heart transplantation are similar. Plasma MPA concentrations below the specified therapeutic ranges are associated with an increased risk of rejection. Studies performed to date have shown that MPA concentrations above the therapeutic ranges do not appear to be associated with increased efficacy. Significant correlations between toxicity and the height of the MPA AUC or the C0 concentration have not been established to date. It appears that the free MPA concentration is a better indicator of leukopenia as a side effect of MPA. For EC-MPS, the MPA concentrations of C0 are higher than those for MMF at an equimolar dose, which means that the therapeutic range cannot be used for EC-MPS.

Elimination half-life: 17 h (healthy adults).

Recommendation for blood sampling: immediately prior to the next dose (C0) or AUC estimation using three measurement points. MPA is determined in the plasma and EDTA is the anticoagulant of choice. Storing samples at room temperature can lead to an increase in the plasma MPA concentration.

Elimination: mycophenolic acid is largely metabolized, mainly into inactive mycophenolic acid glucuronide (MPAG), which is excreted primarily by the kidneys but also in the bile. In the intestine, MPAG undergoes cleavage by bacterial glucuronidases to MPA, which is subsequently reabsorbed. MPA is also metabolized to the active metabolite mycophenolic acid glucuronide (AcMPAG), an inactive 7-O-glucoside, and a cytochrome P450 oxidation product.

Clinical aspects: mycophenolic acid is a highly effective immunosuppressive substance that is available as mycophenolate mofetil and EC-MPS and is used to prevent graft rejection (e.g., kidney, heart, liver). MMF is hydrolyzed in vivo to mycophenolic acid (MPA), which is a selective, reversible inhibitor of inosine mono phosphate dehydrogenase (IMPDH) in de novo purine synthesis. By inhibiting this enzyme, it blocks the conversion of inosine mono phosphate to xanthine mono phosphate and subsequently to guanosine mono phosphate. In contrast to other cells, T and B lymphocytes do not have a salvage pathway. Therefore, inhibition of IMPDH leads to depletion of intracellular guanosine triphosphate and reduced proliferation. The pharmacokinetics of mycophenolic acid show significant inter individual differences. There are various reasons for this. Hypoalbuminemia leads to increased MPA clearance as a result of reduced protein binding. Dis continuation of corticosteroids leads to an increase in MPA exposure due to a reduction in UDP glucuronyltransferase (UGT) activity. Cyclosporine reduces the enterohepatic recirculation of MPA by inhibiting the MRP2 transporter. Decreased creatinine clearance leads to increased MPA exposure. Low UGT1A9 activity increases MPA exposure, while UGT1A9 promoter polymorphism increases MPA clearance, which results in decreased MPA exposure. In addition, numerous drug interactions have been observed.

Antacids, cholestyramine, metronidazole, and norfloxacin all decrease MPA exposure. Rifampicin also reduces MPA exposure by inducing UGT. Mycophenolic acid has no nephrotoxic or neurotoxic side effects. Furthermore, it does not influence lipoprotein metabolism. It is not diabetogenic and does not cause hypertension. Elevated free MPA levels can lead to infections and leukopenia.

Another side effect is anemia. Gastrointestinal side effects (e.g., diarrhea) can also occur; these are primarily dose-dependent. The active MPA metabolite AcMPAG is involved in the development of diarrhea.

A drug monitoring approach for MPA has not yet been established /35/. Three prospective studies have investigated the therapeutic benefits of TDM for mycophenolic acid. In the FDCC study /36/, a significant correlation was observed between the MPA AUC in the early post transplant phase and the incidence of acute rejection during the first year after kidney transplantation. However, a clear overall benefit for drug monitoring was not established. In contrast to the FDCC study, systematic dose adjustment was carried out in the APOMYGRE study, based on the MPA AUC estimated using the Bayesian method. A significantly lower rejection rate was observed in patients who underwent concentration-controlled dose adjustment /37/. The Opticept study /38/ showed a potential benefit for MPA C0 monitoring when reducing the dose of calcineurin inhibitors.

The following tentative recommendations for measuring MPA plasma levels have been proposed: it would seem reasonable to aim for therapeutic MPA concentrations in the early post transplantation phase (day 3–5). Drug monitoring for MPA is recommended for heart transplant patients due to their minimal tolerance for rejection episodes and in liver transplant patients due to significant inter individual differences in pharmacokinetics. In kidney transplant patients, MPA monitoring is useful in certain circumstances, e.g. reduced CNI exposure, delayed graft function, poor HLA matching, compliance problems in young patients, and suspected drug interactions /34/.

With respect to the frequency of MPA monitoring in kidney transplant patients, an initial evaluation (preferably an abbreviated AUC estimation) is recommended once during the first week after transplantation (day 3–5). In addition, either measurement of the pre-dose concentration or, preferably, an abbreviated MPA AUC estimation should be carried out once between days 10 and 14 and again after 3–4 weeks. If the MPA AUC is in the range (30–60) mg × h/L, a dose adjustment is generally not necessary. If the MPA AUC is < 30 mg × h/L, a dose adjustment is recommended. The same applies if the MPA AUC is > 60 mg × h/L and side effects are present. A confirmatory analysis is then required after 3–5 days, by which time a new steady state plasma concentration has been reached. A therapeutic range for long-term therapy has not yetbeen established.

HPLC or LC-MS/MS can be used to measure MPA. It can also be measured using EMIT, PETINIA, or an IMPDH-based enzyme inhibition assay. The IMPDH-based enzyme inhibition assay displays a lack of significant AcMPAG cross reactivity /39/.

Figure 40-1 Factors influencing the effect of a specific prescribed drug, modified from Ref. /2/

Prescribed dose Compliance with intake Erroneous prescription Administered dose Absorption Distribution Biotransformation Excretion Drug serum concentration Regional blood flow Serum protein binding Transport mechanisms Drug concentration at site of action (receptor) Tissue responsiveness Other drugs Diseases Age Placebo effects Intensity of pharmacological effect
Goto top <