Journal List > Transl Clin Pharmacol > v.25(2) > 1082678

Atkinson: The stable isotope method for determining absolute bioavailability

Abstract

The bioavailability of a drug is usually assessed in healthy subjects. However, it is reasonable to expect that significant alterations in bioavailability may occur in actual patients with different diseases or in individuals belonging to special populations. Relatively few studies have been conducted to examine this possibility. The stable isotope method is well suited to compare absolute bioavailability in patients and healthy subjects. Studies in which this method was used indicate that significant changes in the bioavailability of some drugs are particularly likely in patients with advanced liver disease and in those whose splanchnic blood flow is reduced. The expectation is that bioavailability in neonates, children, and pregnant women may also differ from that in non-pregnant adults.

Introduction

Steady state drug concentrations are determined only by the absorbed dose and drug elimination clearance rate. Although considerable emphasis has been placed on characterizing the effects of disease on drug elimination, relatively little attention has been given to the impact of disease on drug absorption. In fact. the FDA guidance on conducting bioavailability and bioequivalence studies recommends that they be performed in healthy subjects where possible.[1]
The stable isotope method for measuring absolute bioavailability was introduced more than 40 years ago and remains a “gold standard” for measuring bioavailability because it eliminates the washout period inherent in the traditional crossover design used in bioavailability studies.[2] Because it provides detailed pharmacokinetic data at a single point in time, the stable isotope method is particularly suitable for studying drug bioavailability in various disease states and in special populations. Review of the results of 10 patient studies provides at least a preliminary indication of the patient groups in whom bioavailability is likely to vary from that measured in healthy subjects.

Background

The development of mass spectrometry and the identification of stable isotopes had a common origin in 1919 when Ashton built the first mass spectrometer to identify the two stable isotopes of neon.[3] However, it was not until 1935 that Schoenheimer and Rittenberg studied the conversion of stable isotope labeled linoleic acid to stearic acid and first used stable isotope labeled physiological compounds to elucidate pathways of intermediary metabolism.[4] In 1950, Maynert and Van Dyke were the first to use a stable isotope labeled drug to explore its possible metabolic pathways.[5] Oral doses of 15N-labeled pentobarbital were administered to dogs. About 60% of the 15N dose was excreted in the first 24 hours but less than 2% of this consisted of parent drug. Gas chromatography was the first separation technique to be directly coupled to mass spectrometers.[6] This greatly enhanced the utility of mass spectrometry and followed the development of molecule separators that enabled effluent from the gas chromatograph column to be transferred from the above atmospheric pressure of the gas chromatograph effluent to the high vacuum within the mass spectrometer.[7] This was accomplished by creating a small gap in the separator across which molecules of interest could pass, whereas a vacuum pump removed most of the much lighter helium carrier gas when it entered the gap.
In 1968, Hammar, Holmstedt, and Ryhage were the first to use gas chromatography-mass spectrometry to identify a drug and its metabolites in human plasma.[8] They also demonstrated that the analytical sensitivity of conventional mass spectrometry could be increased by focusing the mass spectrometer on only two or three prominent mass spectral ions from the analyte. They accomplished this by switching the accelerating voltage of a magnetic sector mass spectrometer to bring the selected ions into focus and termed this technique “mass fragmentography”. This technique is now commonly referred to as “selected ion monitoring” and continues to form the basis for most quantitative mass spectrometric applications. Unfortunately, the magnetic field that was varied to obtain a full mass spectrum had too much hysteresis for it to be switched rapidly enough for selected ion monitoring. Furthermore, the accelerating voltage could be varied over only a limited range and only two or three high voltages could be selected at a time. Subsequently, it was shown that a quadrupole mass spectrometer did not have these limitations and could be easily modified for selected ion monitoring.[9] For that reason, quadrupole-based mass spectrometers continue to be the primary ones used for quantitative work. These advances made it possible to introduce in 1975 the stable isotope method for assessing the absolute bioavailability of a drug.[2]

Clinical Studies

Between 1975 and 2015, thirty-one published studies were identified that were conducted using stable isotope labeled drugs to assess absolute bioavailability. Most of these were carried out in healthy subjects and appeared to be incorporated in the development program of the studied drug. In addition, specially focused studies were conducted to evaluate nifedipine absorption from four intestinal segments,[10] the effect of administration rate on verapamil absorption,[11] and the comparable absolute bioavailability of three transdermal nitroglycerin formulations.[12] Additional special studies reported the absolute bioavailability of phenytoin in patients under steady-state conditions, in order to avoid dealing with its non-linear kinetics,[13] the absorption of different isomers of verapamil,[14] and the absence of a chronopharmacologic effect on the absolute bioavailability of a modified formulation of verapamil.[15]
Of particular interest are ten studies that were conducted either in patients with a particular disorder or in the special populations of elderly, neonatal, or pregnant patients (Table 1). Six of the comparisons were made to healthy subject studies in which the stable isotope method was used but only three of these were done concurrently with the patient study.[161721] The most significant change was the increase in bioavailability that was reported in patients with hepatic cirrhosis.[16] This was attributed to the intrahepatic and extrahepatic shunting that accompanies this condition. When compared to healthy subjects, bioavailability more than doubled in liver disease patients and, combined with a 50% reduction in elimination clearance, was estimated to increase steady state plasma concentrations by five-fold if the same verapamil dose had been given to both groups of study subjects. In one study, slow gastric emptying with increased gastric absorption was suggested as the cause for the apparent increase in the bioavailability of cibenzoline in patients with impaired renal function.[17] However, this difference was not statistically significant, perhaps because only eight patients were included in each study group. In the study of patients with heart disease,[20] absorption of N-acetylprocainamide (NAPA) was reduced to a mean of 78% from the mean of 92% reported separately for four healthy subjects.[2] In four of the studies, there was no difference in bioavailability between the patient and control groups.[18212426] The conclusion reached in the comparison study for methadone[22] is particularly suspect because the bioavailability for the control group was estimated from the pharmacologic response of cancer patients who were receiving methadone to relieve chronic pain.[23]
Special populations were the subject of four studies. Two of these focused on elderly patients.[2426] Of particular note is the study of phenytoin absorption in three neonates.[28] Absolute bioavailability was 100% in two of them but only 50% in the patient with the youngest gestational age. A comparator study in healthy neonates was not conducted and probably was not feasible. A study of calcium carbonate absorption in pregnant patients was conducted to compare the absolute bioavailability of a conventional formulation with an extended release formulation in which microencapsulation was designed to release calcium at pH ≥ 5.5.[29] Three different doses were examined but results are shown only for the 500 mg dose that is recommended by the WHO. Each patient served as her own control for only one of the three doses. It was found that the extended release formulation had reduced bioavailability. Unfortunately, the study was not designed to include a comparison of calcium carbonate absorption in pregnant and non-pregnant women.

Feasibility of Implementation

The feasibility of implementing the stable isotope method for determining bioavailability depends on both the advantages and disadvantages of the technique and on certain technical considerations. The method has obvious advantages over the conventional two-stage crossover method. Only a single study and set of blood samples is required, thus shortening study time, minimizing between study variation, and increasing patient comfort and safety. In addition, both labeled and unlabeled drug concentrations can be measured from only a single set of blood samples. Finally, the reduction in between-test variation enables statistically valid results to be obtained with fewer subjects. Eichelbaum estimated that use of the stable isotope method in bioavailability studies can reduce the number of subjects needed by at least 50%.[30] In an FDA-sponsored study, Hect et al. estimated that 28 to 36 subjects would be needed to establish a clinically significant difference in the relative bioavailability of two commercially available formulations of imipramine if the crossover method were used. However, only 4 to 6 subjects would be needed if a solution of a stable isotope standard were administered orally together with each formulation.[31]
Two major disadvantages to stable isotope methods that have been cited in the past are the need for mass spectrometric analysis and the high cost of purchasing stable isotope labeled drugs or the chemical intermediates needed for their synthesis.[32] However, suitable analytical instrumentation is now more generally available and the cost of stable isotope labeled compounds has decreased as the number of competing suppliers has increased. In addition, the cost of these studies to the pharmaceutical industry could be amortized if optimal use were made of stable isotope methodology for other applications throughout early drug development. Browne estimated that this could result in significant reductions in time as well as in the overall costs of these studies.[33]
A technical consideration is that the choice of a stable isotope labeled compound is limited by requirements regarding the selection and positioning of the label. It is critically important to examine the drug's mass spectral fragmentation pattern to make sure that the fragment ion selected for monitoring actually contains the isotope label. Deuterium, 13C and 15N labels are usually chosen for the isotope labeled drug that is administered to study subjects but the choice of isotope and the number isotope atoms that can be incorporated during synthesis may be limited. Deuterium labeling is generally the least expensive and easiest to perform. However, deuterium can alter enzymatic reactions if placed at or near a reaction site, and exchanges with water if linked to oxygen or nitrogen atoms.[34] Therefore, drugs labeled with 13C and 15N are preferred as they do not have these problems and their behavior replicates that of the test drug. It is generally desirable to include two or more labels in synthesizing these compounds to avoid interference from the natural abundance of stable isotopes in the test drug.[35] However, the contributions of the unlabeled and isotope labeled drugs to a monitored mass spectral peak can be separated by deconvolution if this interference cannot be avoided.[36]
With respect to the safety of stable isotope administration, Klein and Klein point out that with respect to effects on enzymatic, cellular, and physiologic processes, stable isotopes can be subdivided into two categories: deuterium and those that involve all the other elements found in an organism.[37] The relative mass difference between hydrogen and deuterium results in a marked difference in the reactivity of their chemical bonds, and this accounts for deuterium's biological effects. Nonetheless, there is a wide margin of safety and the enrichment of total body water with deuterium may be as high as 1% to 2% without adverse consequences. This level of deuterium enrichment is never attained in pharmacokinetic studies and the stable isotopes of carbon, oxygen, and nitrogen can be administered without any safety concerns.
A final concern is the regulatory status of these studies. The FDA has now included guidance to help determine whether certain kinds of human research can be conducted without an IND.[38] A section on “cold isotopes” is included that exempts stable isotope studies provided that certain conditions are met. Unfortunately, there is a stipulation that the administered doses of the labeled compounds should not have a “clinically detectable pharmacological effect in humans”. Since all organic compounds contain stable isotopes, this stipulation seems unwarranted. It might clarify thinking in this regard to refer to stable isotope labeled compounds simply as “stable isotope enriched" rather than as “cold”.

Clinical Perspective

Based on the limited number of results shown in Table 1, it appears that disease-related decreases in drug absorption are likely to be greatest in patients with liver disease. In the verapamil study conducted by Somogyi et al., the combined effects of increased absorption and reduced elimination clearance were shown to have a multiplicative rather than an additive effect that would necessitate a five-fold reduction in oral dose to provide these patients with the same exposure as healthy subjects.[16] Although bioavailability studies are not recommended in the FDA guidance on pharmacokinetic studies in patients with impaired liver function, they would seem to be desirable, particularly for drugs like verapamil that normally have extensive first pass hepatic metabolism.[39]
The cause of reduced NAPA absorption in patients with heart disease suggests that reduced splanchnic blood flow could impair the bioavailability of at least some drugs. As shown in Figure 1, the extent of NAPA absorption was markedly reduced in both patients and one healthy subject when the rapid intercompartmental clearance (CLF) between the intravascular space and splanchnic organs fell below an apparent threshold of 1.1 L/min. According to the following equation, CLF in this case is a function of both the compartmental permeability coefficient-surface area product (P • S) and blood flow (Q)
CLF = Q (1−e−P·S/Q)
so CLF is to some extent an indicator of splanchnic blood flow when it represents transfer from the intravascular space to splanchnic organs. Reduced splanchnic blood flow has previously been thought to account for slow and incomplete absorption of procainamide[40] and has been shown in animals to reduce bioavailability.[41] So this explanation is plausible but requires further study.
In patients with renal disease, the increase in cibenzoline absolute bioavailability was relatively modest and not readily explained.[17] In addition, there was no difference in nitrendipine bioavailability between renal disease patients and healthy subjects.[1819] Epilepsy, and age did not affect drug absorption and the non-pharmacokinetic evaluation of methadone absorption in the cancer patient controls was methodologically flawed.
Neonates and children are special populations in which few formal pharmacokinetic studies have been reported. However, Malik has demonstrated that pharmacokinetic studies are feasible even in neonates.[28] In future studies, the stable isotope method can utilize dried blood spot sampling to provide a complete pharmacokinetic analysis with only one set of microsamples.[42] A thorough characterization of pharmacokinetics in pregnant women requires separate studies during each trimester with a comparator study usually conducted in the postpartum period. Although Roth studied pregnant women only in the third trimester, he demonstrated the clinical feasibility of using the stable isotope method in these women even though calcium carbonate obviously differs from most drugs.[29]
Based on only limited data, it can be concluded that the stable isotope method of determining absolute bioavailability is feasible from the technical standpoint and is likely to efficiently characterize differences from normal absorption, particularly in patients with liver disease or reduced splanchnic blood flow. It is also the optimal way to characterize drug absorption in neonates, infants, and pregnant women.

Figures and Tables

Figure 1

Relationship between bioavailability and CLF as an indirect indicator of splanchnic blood flow. Absorption of NAPA was reduced in both healthy subjects (■) and patients (■) with CLF less than 1.1 L/min (p = .0286, Fisher's exact test). (Data from Strong et al. (2) and Atkinson et al. (20)).

tcp-25-53-g001
Table 1

SUMMARY OF COMPARATIVE STUDIES IN PATIENTS

tcp-25-53-i001
CLINICAL CONDITION DRUG BA RESULT METHOD REF.
LIVER DISEASE
Verapamil P 52%, H 22% SI 16
KIDNEY DISEASE
Cibenzoline P 90%, H 83% SI 17
Nitrendipine 21% SI 18
23% SI 19
HEART DISEASE
NAPA P 78% SI 20
H 92% SI 2
EPILEPSY
Phenytoin P 100%, H 100% SI 21
ADDICTS
Methadone Addict 79% SI 22
Cancer 50%? Response 23
ELDERLY
Verapamil S 14%
R 38%
SI 24
S 16%
R 38%
Conventional 25
Propafenone Extensive 30%
Poor 83%
SI 26
Extensive 30%
Poor 81%
SI 27
NEONATES
Phenytoin 50 - 100% SI 28
No comparator
PREGNANCY
Ca-carbonate Non-EC 22%
EC 3%
SI 29

* Abbreviations: H = healthy subjects, P = patients, BA-bioavailability, Ref.-reference number, S&R-enantiomers, Extensive and Poor metabolizers, EC-enteric coated, SI-stable isotope method. Healthy subject results are listed on the same line for concurrent studies and on the line below for studies done prior to or after the patient studies.

Acknowledgements

Nothing to declare

Notes

Conflict of interest Nothing to declare

References

1. CDER. Draft Guidance for Industry. Bioavailability and bioequivalence studies submitted in NDAs or INDs - general; considerations. Silver Spring, MD: FDA;03. 2014. Accessed 1 December 2016. http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM389370.pdf.
2. Strong JM, Dutcher JS, Lee WK, Atkinson AJ Jr. Absolute bioavailability of N-acetylprocainamide determined by a novel stable isotope method. Clin Pharmacol Ther. 1975; 18:613–622.
crossref
3. Aston FW. Untitled announcement. Science. 1919; 104:134.
4. Schoenheimer R, Rittenberg D. Deuterium as an indicator in the study of intermediary metabolism. J Biol Chem. 1935; 111:163–168.
crossref
5. Maynert EW, Van Dyke HB. The metabolic fate of pentobarbital. Isotope dilution experiments with urine after the administration of labeled pentobarbital. J Pharmacol Exp Ther. 1950; 98:174–179.
6. Gohlke RS, McLafferty FW. Early gas chromatography/mass spectrometry. J Am Soc Mass Spectrom. 1993; 4:367–371.
crossref
7. Ryhage R. Use of a mass spectrometer as a detector and analyzer for effluents emerging from high temperature gas liquid chromatography columns. Anal Chem. 1964; 36:759–764.
crossref
8. Hammar CG, Holmstedt B, Ryhage R. Mass fragmentography. Identification of chrolopromazine and its metabolites in human blood by a new method. Anal Biochem. 1968; 25:532–548.
9. Strong JM, Atkinson AJ Jr. Simultaneous measurement of plasma concentrations of lidocaine and its desethylated metabolite by mass fragmentography. Anal Chem. 1972; 44:2287–2290.
crossref
10. Bode H, Brendel E, Ahr G, Fahr U, Harder S, Staib AH. Investigation of nifedipine absorption in different regions of the human gastrointestinal (GI) tract after simultaneous administration of 13C- and 12C-nifedipine. Eur J Clin Pharmacol. 1996; 50:195–201.
crossref
11. Glaeser H, Drescher S, Hofmann U, Heinkele G, Somogyi AA, Eichelbaum M, et al. Impact of concentration and rate of intraluminal drug delivery on absorption and gut wall metabolism of verapamil in humans. Clin Pharmacol Ther. 2004; 76:230–238.
crossref
12. Sun JX, Piraino AJ, Morgan JM, Joshi JC, Cipriano A, Chan K, et al. Comparative pharmacokinetics and bioavailability of nitroglycerin and its metabolites from Transderm-Nitro, Nitrodisc, and Nitro-Dur II systems using a stable isotope technique. J Clin Pharmacol. 1995; 35:390–397.
crossref
13. Aliwarga T, Cloyd JC, Goel V, Brundage RC, Marino SE, Leppik IE, et al. Excretion of the principal urinary metabolites of phenytoin and absolute oral bioavailability determined by use of a stable isotope in patients with epilepsy. Ther Drug Monit. 2011; 33:56–63.
crossref
14. Vogelgesang B, Echizen H, Schmidt E, Eichelbaum M. Stereoselective first-pass metabolism of highly cleared drugs: studies of the bioavailability of L- and D-verapamil examined with a stable isotope technique. Br J Clin Pharmacol. 1984; 18:733–740.
crossref
15. Dilger K, Eckhardt K, Hofmann U, Kucher K, Mikus G, Eichelbaum M. Chronopharmacology of intravenous and oral modified release verapamil. Br J Clin Pharmacol. 1999; 47:413–419.
crossref
16. Somogyi A, Albrecht M, Kliems G, Schäfer K, Eichelbaum M. Pharmacokinetics, bioavailability and ECG response of verapamil in patients with liver cirrhosis. Br J Clin Pharmacol. 1981; 12:51–60.
crossref
17. Aronoff G, Brier M, Mayer ML, Barbalas M, Aogaichi K, Sloan R, et al. Bioavailability and kinetics of cibenzoline in patients with normal and impaired renal function. J Clin Pharmacol. 1991; 31:38–44.
crossref
18. Mikus G, Mast V, Fischer C, Machleidt C, Kuhlmann U, Eichelbaum M. Pharmacokinetics, bioavailability, metabolism and acute and chronic antihypertensive effects of nitrendipine in patients with chronic renal failure and moderate to severe hypertension. Br J Clin Pharmacol. 1991; 31:313–322.
crossref
19. Mikus G, Fischer C, Heuer B, Langen C, Eichelbaum M. Application of stable isotope methodology to study the pharmacokinetics, bioavailability, and metabolism of nitrendipine after i.v. and p.o. administration. Br J Clin Pharmacol. 1987; 24:561–569.
crossref
20. Atkinson AJ Jr, Ruo TI, Pierges AA, Breiter HC, Connelly TJ, Sedek GS, et al. Pharmacokinetics of N-acetylprocainamide in patients profiled with a stable isotope method. Clin Pharmacol Ther. 1989; 46:182–189.
crossref
21. Kasuya Y, Mamada K, Baba S, Matsukura M. Stable-isotope methodology for the bioavailability study of phenytoin during multiple-dosing regimens. J Pharm Sci. 2985; 74:503–507.
crossref
22. Meresaar U, Nilsson MI, Holmstrand J, Änggård E. Single dose pharmacokinetics and bioavailability of methadone in man studied with a stable isotope method. Eur J Clin Pharmacol. 1981; 20:473–478.
crossref
23. Beaver WT, Wallenstein SL, Houde RW, Rogers A. A clinical comparison of the analgesic effects of methadone and morphine administered intramuscularly, and of orally and parenterally administered methadone. Clin Pharmacol Ther. 1967; 8:415–426.
crossref
24. Fromm MF, Dilger K, Busse D, Kroemer HK, Eichelbaum M, Klotz U. Gut wall metabolism of verapamil in older people: effects of rifampin-mediated enzyme induction. Br J Clin Pharmacol. 1998; 45:247–255.
crossref
25. Fromm MF, Busse D, Kroemer HK, Eichelbaum M. Differential induction of prehepatic and hepatic metabolism of verapamil by rifampin. Hepatology. 1996; 24:796–801.
crossref
26. Dilger K, Hofmann U, Klotz U. Enzyme induction in the elderly: Effect of rifampin on the pharmacokinetics and pharmacodynamics of propafenone. Clin Pharmacol Ther. 2000; 67:512–520.
crossref
27. Dilger K, Greiner B, Fromm MF, Hofmann U, Kroemer HK, Eichelbaum M. Consequences of rifampin treatment on propafenone disposition in extensive and poor metabolizers of CYP2D6. Pharmacogenetics. 1999; 9:551–559.
crossref
28. Malik SI, Painter MJ, Venkataramanan R, Alvin JD. Phenytoin and phenobarbital stable isotope studies in neonates. Pediatr Neurol. 2003; 29:376–380.
crossref
29. Roth DE, Pezzack B, Al Mahmud A, Abrams SA, Islam M, Aimone Phillips A, et al. Bioavailability of enteric-coated microencapsulated calcium during pregnancy: a randomized crossover trial in Bangladesh. Am J Clin Nutr. 2014; 100:1587–1595.
crossref
30. Eichelbaum M, von Unruh GE, Somogyi A. Application of stable isotope labeled drugs in clinical pharmacokinetic investigations. Clin Pharmacokinet. 1982; 7:490–507.
crossref
31. Heck HA, Buttrill SE Jr, Flynn NW, Dyer RL, Anbar M, Cairns T, et al. Bioavailability of imipramine tablets relative to a stable isotope-labeled internal standard: Increasing the power of bioavailability tests. J Pharmacokinet Biopharm. 1979; 7:233–248.
crossref
32. Browne TR. Stable isotopes in clinical pharmacokinetic investigations: advantages and disadvantages. Clin Pharmacokinet. 1990; 18:423–433.
33. Browne TR. Stable isotope techniques in early drug development: an economic evaluation. J Clin Pharmacol. 1998; 38:213–220.
crossref
34. Blake MI, Crespi HL, Katz JJ. Studies with deuterated drugs. J Pharm Sci. 1975; 64:367–391.
crossref
35. Wolen RL. The application of stable isotopes to studies of drug bioavailability and bioequivalence. J Clin Pharmacol. 1986; 26:419–424.
crossref
36. Dutcher JS, Strong JM, Lee W-K, Atkinson AJ Jr. Stable isotope methods for pharmacokinetic studies in man. In : Klein R, editor. Proceedings of the Second International Conference on Stable Isotopes. Washington, DC: US Govt Printing Office;1976. (CONF-751027).
37. Klein PD, Klein ER. Stable isotopes: origins and safety. J Clin Pharmacol. 1986; 26:378–382.
crossref
38. CDER. Guidance for Clinical Investigators, Sponsors, and IRB's. Investigational new drug applications -(INDS) - determining whether human research studies can be conducted without an IND. Rockville, MD: FDA;09. 2013. Accessed 1 December 2016. http://www.fda.gov/downloads/drugs/guidances/ucm229175.pdf.
39. CDER, CBER. Guidance for industry. Pharmacokinetics in patients with impaired hepatic function: study design, data analysis, and impact on dosing and labeling. Rockville, MD: FDA;05. 2003. Accessed 1 December 2016. http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM072123.pdf.
40. Koch-Weser J. Pharmacokinetics of procainamide. Ann N Y Acad Sci. 1971; 179:370–382.
41. Crouthamel WG, Diamond L, Dittert LW, Dolusio JT. Drug absorption VII: influence of mesenteric blood flow on intestinal drug absorption in dogs. J Pharm Sci. 1975; 64:664–671.
crossref
42. Samardzic J, Turner MA, Bax R, Allegaert K. Neonatal medicines research: challenges and opportunities. Expert Opin Drug Metab Toxicol. 2015; 11:1041–1052.
crossref
TOOLS
Similar articles