Clinica Chimica Acta, 151 (1985) 193-216
193
Elsevies
CCA 03285
Critical Review
Free drug measurements: methodology and clinical significance Tai C. Kwong Depariment of Pathology and Laboratory Medicine, The University of Rochester Medical Center, 601 Efmwood Ave., Box 608, Rochester, NY I4642 (U.S.A.1 (Received
February
14th, 1985;
revision June 14th, 1985)
Key wor&: Free drug; Free drug level; Free drug concentration; Free drug measurement; Free drug rno~~tori~g
Introduction Drug molecules circulate in blood in two forms: those in association with blood components such as proteins, red cells, or platelets and those which are unbound and are dissolved in plasma water [l-3]. The extent of drug binding to plasma proteins varies widely among different types of drugs. The protein-bound fraction ranges from less than 0.1 for caffeine and ethosuximide to greater than 0.99 for highly bound drugs such as warfarin and dicumerol[4]. The effects of changes in the extent of protein binding on free drug fraction and on drug metabolism are significant only for highly bound drug (greater than 80% bound), since even a relatively small change in the degree of binding has a dramatic effect on the free fraction. Albumin is quantitatively the most important drug binding protein for many drugs, particuIarly the neutral and anionic (acidic) drugs such as warfarin, phenytoin, and valproic acid [.5]. In recent years, cationic (basic) drugs such as propranolol quinidine, and the tricyclic antidepressants have been shown to bind not only to albumin, but also to other blood proteins such as &,-acid glycoprotein (AAG) and lipoproteins f6-8). In some cases, binding to AAG and lipoproteins exceeds that to albumin. Fluctuations in plasma levels of AAG and lipoprotein fractions such as low density lipoprotein are common and point to the importance of understanding the interactions of cationic drugs with these proteins f&9,10]. Table I lists those drugs which are commonly encountered in clinical chemistry laboratories. Protein binding has an important effect on the pharmacokinetics of drugs. The impact of alterations in protein binding on drug clearance depends on the clearance characteristics of the drug. Restrictively cleared drugs are those that are removed from plasma by glomerular filtration through the kidneys or by passive uptake by the liver. Only the free fractions of such drugs are cleared by these organs. Thus, clearance of highly protein bound and restrictively cleared drugs are sensitive to
0009-8981/85/$03.30
Q 1985 Elsevier Science Publishers
B.V. (Biomedical
Division)
194 TABLE Protein
I binding
of drugs
Drug Acetaminophen N-Acetyl procainamide Amikacin Amitriptyline Carhamazepine Chloramphenicol Chlordiazepoxide Chlorpromazine Desipramine Diazepam Digitoxin Digoxin Disopyramide Ethosuximide Gentamicin Imipramine Lidocaine Lithium Methotrexate Nortriptyline Phenobarbital Phenytoin Primidone Procainamide Propranolol Quinidine Salicylic acid Theophylline Tolbutamide Valproic acid Warfarin ’ h ’ d
81% at 1.7 pg/ml; 66% 95% at 14 pg/ml; 80% 34-388 by equilibrium 95% at 30 pg/ml; 75%
s Bound
Proteina
0
II
10 4
96 82 53 97 89 92 99 90 25 66-81 L’ 0 leas than 10 89-94 70 0 45 92 51 89 20-25 16 88-95 71 80-95 34-45 93 93 “ 99
h L
at 5 pg/ml. at 300 pg/ml. dialysis; 41-45% at 160 pg/ml.
ALB.AAG,LP ALB. AA<;? ALB ALB ALB, AAG. LP ALB. AAG. LP ALB ALB ALB ALB. AAG
ALB, AAG. LP ALB. AAG ALB ALB, AAG ALB ALB ALB ALB, AAG. LP ALB. AAG, LP ALB ALB ALB ALB ALB
I2 13 14 IS.16 17 IX x.19 19.20 21 22 23 24.25 26 27 2X.29 30 31 32 14.33 34 35 36.37 12 3x 39.40 41 42.43 44 45 46
by ultrafiltration
alterations in protein binding. A higher free fraction will lead to higher clearance rates, shorter elimination half-lives and a larger fluctuation in peak and trough levels. Theophylline and warfarin are two of such ‘restrictively cleared’ drugs [42,46-481. The different clearance rates of theophylline and warfarin (0.7 and 0.05 ml/min/kg, respectively) are related to their free fractions (0.62 and 0.01, respectively). Non-restrictively cleared drugs are extracted by active mechanisms in both free and bound forms by the liver or the kidneys. Clearance rates are dependent on organ blood flow and relatively independent of protein binding [49]. Lidocaine and propranolol are examples of non-restrictively cleared drugs which are highly bound
195
and which are extensively cleared by the liver in one pass (first pass clearance) [50-521. A widely held view on the pharmacological effect of protein binding is that only the free form of the drug can cross capillary and cell membranes. It is therefore assumed that the free fraction in vitro is equivalent to the pharmacologically active fraction, in vivo, and free drug levels will correlate better with drug effects and clinical conditions of the patients. It should be appreciated that the free drug hypothesis is unproven. In fact, the basic assumption that only unbound drug can cross cell membrane has been challenged in recent studies on the lipop~lic drugs lidocaine and propranolol and some hormones [53,54]. It was elegantly demonstrated that globulin-bound lidocaine and propranolol were also transported into tissues such as brain [54]. Thus, equilibrium measurement of plasma free drug, in vitro, may underestimate the amount of exchangeable plasma drug, in vivo, that is available for transport into tissue. Such measurements, however, can still reflect the levels of exchangeable drugs, in vivo, but only when transport and tissue factors are constant. Under these conditions plasma free drug levels should correlate well with clinical conditions. Despite the limited understanding of the dynamics of exchangeable plasma drug, a great deal of interest has been focused on the .clinical significance of measuring free (unbound) drug levels in plasma or serum. Much effort also has been directed to understanding pathophysiological factors, in vivo, and experimental parameters, in vitro, that influence the binding of drugs to plasma proteins. The rationale for such studies has been that measurement of free drug levels will improve the therapeutic aspects of patient care. This review will consist of three major sections. In the first section the two most frequently used techniques in drug-protein binding studies will be assessed. The theoretical limitations, the nature of the errors inherent in these techniques, and the pitfalls in inte~retation of binding data will be critically examined. Part 2 consists of a review of the literature on alterations in drug binding and on the effects of such changes on drug metabolism and pharmacokinetics behavior. In Part 3 the clinical relevance of altered drug binding in the context of the material presented in the preceding sections will be discussed. Finally, the clinical utility for free drug measurement will be addressed. Analytical techniques
Many techniques have been used to study the binding of drugs to proteins and to measure the free drug fraction. Among these are equilibrium dialysis, ultrafiltration, ultracentrifugation, and gel-filtration [55-571. In all cases, a physical separation of the free drug from the binding macromolecules is required. Equilibrium dialysis is employed most frequently, although in recent years ultrafiltration has gained considerable popularity. This review will focus on ~uilib~um dialysis and ultrafiltration because these two techniques are the most likely candidates for adoption in clinical laboratory settings.
E~u~~~bri~~~ ~i~~~~~s~~s
Aithough equilibrium dialysis is considered by many to be the preferred method for determining drug-protein binding, certain theoretical limitations and their consequences have not always been fully appreciated. A number of reviews on equilibrium dialysis are available [55,58,59]. The technique is based on the principle that unbound drug will equilibrate across a semi-permeable membrane which is permeable to small drug molecules but not to drug binding proteins. The method is performed by placing serum or plasma in a chamber separated from buffer in another chamber by the membrane. At equilibrium, the drug concentration in the buffer compartment (dialysate) is considered to represent the unbound drug concentration which will be the same as the free drug concentration in the protein compartment (retentate). Under rigorously controlled experimental conditions such as temperature. time and buffer composition, equilibrium dialysis can give reproducible results. A lack of full understanding of equilibrium dialysis, (either of its theoretical concepts or practical problems) can result in improperly calculated data. A number of papers which critically addressed these problems have appeared ]58,60-663. The most common conceptual error is the failure to appreciate that during dialysis diffusion of free drug from protein compartment into the dialysate will result in a smaller total drug concentration in the plasma and a new equilibrium [60.66]. If binding of a drug to protein is dependent on drug concentration, then the free fraction that exits at post-dialysis equilibrium is related to the lower total drug concentration inside the protein compartment at equilibrium and not to that in the initial protein sample. In other words, the bound and unbound fractions at equilibrium are different from those of the original sample. The magnitude of the difference is dependent on the volume ratio of dialysis buffer to protein sample. the affinity constant and the concentrations of drug and protein samples [60,68]. The effect of varying the ratio of dialysis buffer volume to sample volume was tested with a testosterone albumin equilibrium dialysis system, with the initial concentrations of testosterone and albumin kept constant [66]. An increase in the buffer to sample ratio from 2: 1 to 20: 1 resulted in a change of the bound/free ratio from 3.22 to 4.43, and of the percent unbound from 23.7 to 18.4. When the initial concentrations of testosterone were varied to construct a Scatchard plot, the affinity constant K, was 1.02 x lo4 for a 2: 1 buffer to sample ratio and 0.30 X 10” for a 20: 1 ratio. The K, obtained from a plot constructed from data which have been corrected for post dialysis concentration was 2.37 X 104. Thus, a significant error can be incurred if dilutional shift is not taken into consideration. In experiments to measure the extent of drug binding of an endogenous drug in plasma, an unbound labeled drug is added to the buffer compartment. A new equilibrium will be established based on the new total drug concentration which is the sum of the endogenous drug in plasma and the added labeled drug. Determination of free and bound drug will give values which are relevant to the new post-dialysis equilibrium and are not the same as those in the original sample unless the labeled drug is added at a concentration identical to the concentration of the endogenous free drug in the plasma under examination [62].
197
Other problems which should be considered in obtaining accurate results are: (a) significant Donnan effect; (b) osmotic dilution; (c) nonspecific adsorption of free drug, protein molecules or drug-protein complexes to the surface of dialysis apparatus; (d) lack of attainment of equilibrium; (e) radiochemical impurity and (f) protein leakage into dialysate. The Donnan equilibrium effect stems from unequal distribution of diffusable charged ions (such as ionized drug molecules) due to the confinement of nondiffusable charged protein molecules to one compartment by the semi-permeable membrane f67]. It is well-known that the Donnan effect will affect the determination of drug binding by equilibrium dialysis [68j, particularly for those drugs which are highly ionized and weakly bound. In principle the Donnan effect can be minimized by increasing the concentration of diffusible electrolytes or by choosing a pH close to the isoelectric point of the protein. Addition of small electrolytes (NaCl or KCl) to a final concentration of 0.15 mol/l in the system is expected to abolish the unequal distribution of diffusible ions [67]. These remedies, however, may not be applicable if drug-protein interaction is primarily electrostatic in nature, or if the binding protein precipitates at its isoelectric point. Appropriate correction of equilibrium dialysis results is possible using the Donnan ratio which can be calculated from drug ionization and distribution of non-binding ion, such as cesium ion [69]. Dilution of protein sample (retentate) due to osmotic pressure of plasma protein can lead to considerable variation in retentate volumes at final equilibrium. Accurate and precise determination of retentate volume is more difficult than for protein concentration. Thus, measurement of starting and retentate plasma protein concentrations will allow correction for dihrtion. Drug, drug-protein complex or protein may adsorb to dialysis membrane or to surfaces of the dialysis apparatus. Adsorption appears to be highest among highly ionized drugs and may be greater than 50% for some drugs as streptomycin [55]. Adsorption is dependent on the concentration of the drug. For example, as the amount of suxamethonium adsorbed to dialysis membrane increases with increasing drug concentration, the binding data could be erroneously interpreted to suggest that suxamethonium binding to protein decreases with increasing drug concentration. Blanks have to be made to eliminate the error due to adsorption. Non-specific loss of drug does not affect the calculation of free fraction if both the dialysate and retentate are assayed for drug levels [SS]. For drugs whose binding to protein varies with drug concentration, the free fraction determined after equilibrium dialysis relates to the equilibrium total drug concentration after changes due to osmotic dilution, nonspecific adsorption to membrane, and loss of free drug to the dialysate. Thus, the true free fraction of variably bound drug in the starting plasma must be calculated [60,70]. Other factors such as temperature, pH, and buffer may significantly alter the determination of drug binding to proteins, in vitro, and may account for the variation in reported values of free fractions of some drugs. In general, protein binding decreases at higher temperature; binding of theophylline was 8.9% and quinidine was 6.0% lower at 37 than at 25°C [42,71]. Dependency of protein binding on pH has been reported for a number of drugs including warfarin [72], propranolol
[73]. imipramine [74], theophylline [42.75]. quinidine and prazosin [75]. The average bound fraction of theophylline to human serum adjusted to pH 7.4 was 10.9% lower than that for the same sera without pH adjustment (pH ranged from 7.6 to 8.7) [42]. The implication of variation in binding with pH is that without careful control of pH during equilibrium dialysis, the binding data may not reflect conditions. in vivo. Buffer composition significantly influences binding values determined by equilibrium dialysis [75-771. Krebs-Ringer bicarbonate and phosphate buffers gave higher unbound fractions than isotonic phosphate buffer for quinidine 1771 and theophylline [42]. Lowering the chloride concentration in the Krehs-Ringer buffers by half decreased the free fraction of quinidine in normal serum from 22.2 to 13.5%. This is consistent with previous Findings that chloride displaces quinidine from plasma proteins (781. Ultrufiltration
Ultrafiltration is the other commonly used technique for studying drug binding to protein. A pressure gradient, which can be conveniently generated by centrifugation, forces plasma water and small molecules through a semi-permeable membrane. Ultrafiltration is an attractive alternative to equilibrium dialysis because of the ease and speed with which it can be accomplished. Speed is an important consideration in studying the binding of drugs to albumin since binding of some drugs may be decreased by fatty acids [79,80]. Fatty acid released through lipoiysis of plasma triglycerides can be significant in tong storage and in dialysis of long duration [81,82]. A major concern in using ultrafiltration has been the effect of the removal of free drug on physiologic binding equilibrium. For this reason it has been suggested that only a small fraction of the totat sample (often less than 10%) should be ultrafiltered to avoid disturbance of protein binding equilibrium [57,83,84]. This, however, makes measurements of low levels of drug in a small volume of ultrafiltrate analytically difficult. Recent reexamination of the thermodynamic aspects of ultrafiltration has indicated that ultrafiltration does not inherently alter binding equilibria [57.83,85]. Even as protein and total drug concentration increase markedly during ultrafiltration, the drug-protein molar binding ratio remains constant. Therefore, the drug concentration in the ultrafiltrate also remains constant and will be the same as the equilibrium free drug concentration in the original sample. Drug concentrations in the ultrafiltrate will also be independent or the volume of ultrafiltrate. The above theoretical prediction holds true if the association constant and the number of binding sites per protein molecule remain unchanged. The association constant may vary at very high protein concentration which may lead to protein ~onformati#nal changes or formation of aggregates. The membrane used for ultrafiltration is assumed to act as a perfect molecular sieve with no discrimination between water molecules and the larger drug molecules. A non-ideal membrane will allow higher fiitration rate for water over drug molecules. This will result in dilution of drug concentration in the uItrafiltrate which wit1 be exaggerated at higher gradient pressure. Thus, it has been observed that unbound digoxin varied from 40-75% and unbound ouabain varied from BO-97% when ultrafiltration was performed at
199
pressure ranging from 45 to 2 pound per square inch [86]. It can be expected that this molecular sieving effect will be greater for drug molecules of higher molecular weight. The error in a protein free solution is about 2% for a drug of molecular weight of 300, increasing rapidly to 13% for streptomycin (mol. wt. 581) [Ml. Ultrafiltration assays need to be vigorously validated and controlled. For each drug binding system, the effects of pH and temperature variation must be established, In general, drug binding decreases with increasing temperature, and ultrafiltration is best performed in a temperature controlled centrifuge to minimize the effect of heat generated by the centrifuge motor. Pressure gradient should be low but sufficient to generate acceptable filtration rate and is best maintained by low speed centrifugation (1000-2000 x g). Sieve effects due to polarization of protein on membrane may be minimized by centrifugation in a fixed angle rotor. Adsorptive loss to membrane has to be measured. The presence of Donnan effect in ultrafiltration is well-documented and can be corrected by the method similar to that for equilibrium dialysis [57]. Since the limitation of ultrafiltrate volume to lo-20% is arbitrary and has no theoretical basis, larger volume of ultrafiltrate can be generated to facilitate drug measurement. The volume of ultrafiltrate that can be obtained is limited by practical considerations such as the viscosity of the remaining serum and the length of time for centrifugation. Ultrafiltrate drug concentration remains the same when up to 40% of the total volume has filtered. Factors influencing drug binding, in vivo Physiologic and pathologic factors that change the extent of drug binding will result in an altered free fraction. Depending on the mechanism of drug metabolism, free drug concentration and drug clearance can also be affected. The following sections will examine these factors which affect drug binding in vivo. Ph~s~o~ogic~lfactors
There have been a limited number of studies focusing on sex-related differences in protein binding of drugs [87]. Lower protein binding in women than in men of comparable age have been observed for imipramine [88], chlordiazepoxide [89], and nitrazepam [90]. The differences are generally small but statistically significant. Reports on sex-related differences in protein binding of diazepam are conflicting [91-961. Studies on protein binding of lidocaine [93], lorazepam [97], oxazepam [98] and propranolol[96] did not reveal any differences between men and non-pregnant women. Women on oral contraceptives had significantly lower binding for chlordiazepoxide, lidocaine and diazepam than contraceptive-free females [89,93]. AAG, a major protein binder for lidocaine is as much as 35% lower in plasma concentration in women taking estrogen-containing oral contraceptives 193,991.The reduced binding of lidocaine in females on oral contraceptives has been attributed to the lower plasma AAG concentrations. Pregnancy is associated with physiological changes which have important influence on drug metabolism and disposition [lOO,~Ol]. Changes in serum levels of protein, free fatty acids, or other endogenous substances affect the extent of drug binding [102,103]. Increase in free drug fraction in patients during the last trimester
200
when compared to controls have been observed for phenytoin (l&20%) [104 1061, phenobarbital (12%) [104]. valproic acid (55’%} 11041, diazepam (44462%) [46,105-1071, lidocaine (50%) [961, salicyhc acid (80%) [105,108], sulfisoxazole [105], and propranolol (25%) [96]. Furthermore, the free fraction in pregnant women correlated negatively with serum albumin concentrations which progressively decline during pregnancy. Other pregnancy-associated factors which may be important in influencing binding capacity are elevated free fatty acid levels [109], as well as reduced binding affinity of albumin of which there is some evidence [108,110]. The effect of age on protein binding and drug disposition have been extensively reviewed [ill-1131. The free fraction of several drugs such as hdocaine [96.111], bupivacaine 11151, meperidine [116], and propranotol [96] is much higher in fetal (cord) plasma compared to maternal plasma. As binding of these drugs to AAG is substantial, the difference in maternal and fetal protein binding was probably accounted for by the two to three times greater plasma concentration of AAG in the mother than in the fetus 190.1141. For drugs which bind primarily to albumin such as diazepam [117,1 IS], valproic acid [119,120], phenobarbital and phenytoin [34], clonazepam [121] and salicylate [122], the fetal free fraction is lower. This may be because of the higher concentrations of albumin in fetal plasma and therefore higher binding [122]. Furthermore, the higher maternal concentrations of free fatty acids can competitively displace drug from albumin binding sites, making a greater fraction of drug available for placenta1 transfer. This transplacental binding gradient explains the observation that total drug levels at delivery are higher in the newborn than in the mother [117,120]. In the neonatal period, the doubling of diazepam free fraction during the first day of life has been correlated with the greatly increased free fatty acid concentrations observed shortly after birth. This. in conjunction with slow drug metabolism due to hepatic immaturity can result in unexpectedly high free drug concentration which may explain the adverse effects observed in some neonates of diazepam-treated mothers [ 118 J. Reduced protein binding in neonates also may be related to the quaIitatively different fetal albumin. Albumin in newborns differs from that of the adult [123] in having lower binding affinities for drugs [117,124,125] but higher affinity for bilirubin [124]. High levels of unconjugated bilirubin have been reported to affect the binding of diazepam 11241 and phenytoin [126,127]. In the geriatric population, decrease in drug binding to albumin has been correlated to a decline in plasma albumin concentration [128]. The decrease in albumin levels may be significantly large among poorly nourished and severely ill elderly patients [128-1301. Pathological factors
Various disease states influence the binding of drugs to plasma proteins by pathology-induced changes in the plasma concentrations of the major drug-binding proteins. For drugs which bind primarily to albumin, the literature abounds with reports of reduced drug binding in disease states associated with hypoalbumin~mia. These have included liver disease, renal failure, nephrotic syndrome, protein losing enteropathy and burns [5,131].
201
In patients with liver disease, hypoalbuminemia is primarily due to albumin loss into interstitial spaces as well as to compromised synthetic capacity for albumin. For a variety of drugs, binding to plasma proteins is reduced in patients with liver disease [132,133]. These have included phenytoin [35,134], quinidine [135], propranolol [136], the benzodiazepines [137-1391, and verapamil [140]. The lower binding of phenytoin has been associated with acute viral hepatitis [141], alcoholic liver disease [135], and liver failure [134], and have been correlated with albumin [35,135,141] and total bilirubin concentrations [142]. The association of renal failure with abnormal drug binding to albumin has long been appreciated [143,144]. Since the effect of uremia on drug binding relates to albumin, the drugs most affected are the anionic and neutral drugs such as phenytoin [22,35,134,143,144-1461, valproic acid [147-1491, phenobarbital 11501, phenylbutazone [151], salicylate [150,152], thyroxine (1531, and warfarin [151,154], and those cationic drugs such as diazepam [155] and oxazepam [156] which have significant binding to albumin. As expected, protein binding of those cationic drugs such as propranolol 11571, quinidine [157,158] and verapamil [159] which bind to AAG and lipoproteins are minimally affected by renal disease. Decrease in drug binding due to hypoalbuminemia in the nephrotic syndrome has been studied. It was observed that for each 1 g/l fall in serum albumin below 37 g/l, there was a 1% decrease in the percentage of bound phenytoin [160]. In uremic patients reduced binding has been attributed to qualitatively different albumin molecules, or to displacement of drug molecules from albumin binding sites by the organic molecules which accumulate in uremia. Evidence supporting either hypothesis has been summarized in a recent review [161]. The preponderance of the evidence, however, suggests that in uremic patients the accumulation of endogenous material which competes for albumin binding sites is probably the more important mechanism of reduced binding to albumin. As a response to tissue injury, a number of plasma proteins increase in concentrations. Among these acute phase reactants are alpha-l acid glycoproteins (AAG) and lipoproteins [10,162]. The cationic drugs such as lidocaine, propranolol, quinidine, imipramine and diltiazem bind not only to albumin, but also significantly to AAG and lipoproteins [6,9]. As AAG and lipoprotein serum levels increase following tissue injury, protein binding of cationic drug will also increase [6]. This has been reported for propranolol, lidocaine, disopyramide and i~pra~ne after acute myocardial infarction [163-l@], quinidine and propranolol after surgery [169,170], lidocaine, propranolol and methadone in cancei patients [171-1731, lidocaine in trauma [174], imipramine after severe burn injury [175], and propranolol and chlorpromazine in patients with Crohn’s disease and inflammatory arthritis 11761. Concentration-dependent
binding
At therapeutic concentrations, most drugs do not saturate the binding capacity of the binding proteins. Notable exceptions are valproic acid [17’7],salicylate [178,179] and disopyra~de [180]. Higher ~ncentrations of these drugs are associated with higher free fractions. Valproic acid free fraction jumped from 0.05 to 0.25 at plasma levels between 30 and 160 pg/ml. It was also observed that valproic acid free
fraction varied more than two-fold during a single dosing interval (177). Since valproic acid has a low extraction ratio and it restrictively cleared bv the liver, increase in free fraction after increasingly higher doses will lead to higher drug clearance and, as a result. lower steady state total drug levels. Thus. the hyperbolic level vs dose relationship for valproic acid is attributable to its concentration-dependent binding to albumin [177]. Within therapeutic range. the free fraction of salicylate increases by approximately 50% as plasma salicylate concentration increased from 200 to 300 mg/I [17X]. The increase is much more hazardous in toxic levels (181). The concentration dependent binding of disopyramide was first documented in 1974 [182] and has been confirmed by many laboratories [180,183-1851. The free disopyramide fraction at a total concentration of 4.6 pmol,/l is 0.19. but it increases to 0.46 when the total concentration is raised to 18.3 pmol/l [183]. This represents a 2.4-fold increase in the free fraction within the therapeutic range for total disopyramide which is 6-15 pmol/l. Thus, one would expect the disposition of disopyramide to be non-linear when based on total concentration in plasma. and that it would be necessary to consider free disopyramide concentration [186,187]. Protein binding of disopyramide is also altered by competitive binding from its monodealkylated metabolite. The concentrations of the metabolite are highly variable. Among patients receiving enzyme inducing drugs and in patients with renal dysfunction, the metabolite can accumulate to levels higher than its parent compound (25,188) Drug interactions and displacements Drug interaction can be either direct competition by two drugs for the same binding sites, thus resulting in the displacement of the one with weaker binding, or it can be one drug affecting the metabolism and clearance of another drug. Literature on drug interactions is volu~nous but there are only a limited number of clinically significant drug interactions relating to alterations in protein binding. Two of the most clinically significant displacement interactions are the displacement of phenytoin with valproic acid 1189%1941 and warfarin by phenylbutazone or other nonsteroidal anti-inflammatory drugs [195,196]. Valproic acid displaces phenytoin from their common binding site on albumin [194]. It has been shown that this displacement of phenytoin resulted in a 100% increase in the phenytoin free fraction of plasma [194]. As hepatic clearance of phenytoin is dependent on free fraction, the increased free fraction due to displacement will increase phedytoin clearance. The net result should be reduced total phenytoin concentration while the free concentration remains unchanged. Different studies of phenytoin plasma levels following the addition of valproic acid to patients stabilized on phenytoin, however, have shown that total phenytoin levels have either increased or decreased, and that free phenytoin levels have increased [197]. Increased total and free phenytoin levels could be explained if, in addition to displacement. there were inhibition of phenytoin metabolism by valproic acid. This proposed dual mechanism of phenytoin-valproic acid interaction has been complicated by a recent observation that free phenytoin concentration was not altered by valproic acid [19S].
203
The other major ~ticon~lsant drugs, including phenytoin, carbamazepine and phenobarbital, have no effect on valproic acid binding to albumin. The reasons are that valproic acid is more tightly bound to albumin than the other anticonvulsant drugs; that the binding constants for valproic acid, phenytoin and carbamazepine are 6.9 x 104, 1.3 x lo4 and 1.4 x 103, respectively [199]; and also that the molar concentration of valproic acid is higher than those of the other drugs. The displacement of valproic acid by salicylate, in vitro and in vivo, has been demonstrated [200-2023. The mechanism of interaction between these drugs appears to be more than just displacement. The increase in both free valproic acid concentration and half-life of elimination suggest that salicylate inhibits valproic acid metabolism [201]. The highly protein bound anti-coagulant warfarin is dispiaced from albumin binding sites by phenylbutazone f19.51 and other nonsteroidal anti-infl~matory/ analgesic agents including azapropazone, mefenamic acid, naproxen, indomethacin, ketoprofen and iburofen [196]. At therapeutic concentrations, however, only azapropazone and phenylbutazone are known to cause clinically significant potentiation of the anticoagulant effect of warfarin [195,196,203,204]. Relatively few studies have been reported on drug displacement relating to basic drugs which bind not only to albumin but also to &,-acid glycoprotein. The reasons for this may have to do with the fact that albumin binding of cationic drugs is of high capacity and low affinity, and also that the cationic drugs have large volumes of distribution and drug concentrations in plasma water are usually low. Furthermore, binding of the cationic drugs is distributed to more than” one protein (albumin, AAG, lipoproteins), and displacement from one protein may not result in a measurable change in the free fraction of the displaced drug. Thus, even though disopyramide displaces lidocaine and propranolol from AAG, no significant displacement is observed in serum (2051. Other reported drug displacement interactions involving cationic drugs are meperidine and bupivacaine 12061, meperidine and lidocaine [207], and bupivacaine and mepvacaine [208]. Endogenous substances which alter drug binding are free fatty acids and bilirubin, both of which at elevated serum levels are presumed to displace bound drugs from their albumin binding sites. In vitro demonstration of reduced valproic acid binding to serum albumin and patient sera due to increased free fatty acid levels [209-2111 have been confirmed by in vivo studies on patients who are on intravenous fat emulsion therapy and on fasting volunteers [211,212]. A six-fold increase in unbound warfarin and a two-fold increase in unbound phenytoin were produced by increasing plasma free fatty acid concentrations to 2 mEq/l [213]. Other drugs which exhibit reduced binding to albumin in the presence of elevated free fatty acids are the benzodi~epines [120,123,214]. Heparin activates lipoprotein lipase which hydrolyses triglycerides to free fatty acids. It has been reported that the administration of as little as 100 units of heparin (an amount that is likely to enter the bloodstream from an indwelling catheter) produced a 260% increase in unbound diazepam [215]. The high con~ntration of free fatty acid may be artifactual since the degradation of triglycerides continued in vitro after blood collection [216]. This has been confirmed in recent studies which
showed that a large single dose of heparin did not affect the protein binding of prazosin and phenytoin if a lipase inhibitor was present at the time of blood collection [217--2191. In the absence of a lipase inhibitor, the large increases in fatty acids, in vitro, will distort the true extent of drug binding. Bilirubin can also displace drugs from albumin binding sites. High plasma levels of bil~rubin have been reported to have a positive correlation with the free drug fraction of phenytoin and diazepam [124,128,129]. Clinical significance
of altered drug binding
In principle, therapeutic drug monitoring based on the measurement of free drug should be more meaningful than that of total drug because free drug reflects drug concentration at drug receptor sites. Unfortunately, the technology to date for free drug measurement is complex compared to the routine assays for total drug. Hence clinical correlation of drug effects with plasma levels has been based on total drug, and therapeutic ranges are defined for total drug. As long as protein binding of drug is normal and constant (within and between patients~, free drug is a fairly constant fraction of the total drug. As such, measurement of total drug is an adequate estimation of free drug, making routine measurement of free drug unnecessary. Indeed, this has been shown to be the case in a recent report on epileptic patients who were on phenytoin 12201. This relationship between free and total drug does not hold, however, when protein binding is variable or abnormal. In these situations, pharmacological effect may correlate better with free drug and the use of therapeutic ranges based on total drug may be invalid. Altered drug binding due to abnormal protein concentrations, drug interaction and displacement, and concentration-dependent binding is well-documented and summarized in the preceding section, but the clinical significance of altered protein binding has not been studied as thorou~ly. It is clear, however, that nor uil abnormaf drug binding results in free drug concentration which will be of clinital concern. At this juncture it is important to emphasize the distinction between free drug fraction and free drug concentration. Free drugfraczion is the fraction of total drug which is not bound to protein. Free drug ~oncentraf~on is the actual concentration of drug which is not bound to protein. Therefore, depending on the total drug concentration, a normal free drug concentration can actually constitute an abnormal free drug fraction. It should be appreciated that pharmacokinetic principles have predicted that variation in protein binding of drug does not change free drug concentration when steady state is regained at the same rate of drug administration and free drug clearance. Alteration in drug binding, however, does affect total drug clearance. Therefore, total drug concentration at steady state will be different and with it an altered free drug fraction, but the same free drug concentration. Based on this pharmacokinetic consideration, it is evident that there is little clinical significance to many of the reports of altered protein binding resulting from changes in protein concentrations, since free drug clearance in these reports was not affected. Even when free drug concentration was altered as a consequence of altered protein binding, the clinical significance has to be assessed in the context of the width
205
of the therap~tic index of that drug. An altered free phenytoin level will be likely to be a significant change relative to the narrow therapeutic range fur phenytoin. An altered free propranolol level may very well be of little consequence since the therapeutic range for propranolol is wide and not well defined; the minimum concentration for effective beta-bl~kade is ill defined and the patient tolerance for high propranolol levels are well known. Nevertheless, the importance of altered protein binding in the interpretation of serum or plasma drug levels should be emphasized [221]. It is possible to be misled by an abnormal total drug concentration inte~reted in reference to a therapeutic range based on total drug concentration when the free drug concentration has remained therapeutically correct. Such a misinterpretation of total drug levels can lead to unnecessary dosage adjustments to target total drug concentration within the therapeutic range defined for total drug. As a result, free drug concentration may now be inappropriate and the patient may exhibit signs of therapeutic failure or toxicity despite an acceptable total drug concentration. Clinical utility of free drug measurement
Altered protein binding can result in clinically significant alterations in free drug concentration for many patients. In these instances, total drug levels are misleading and free drug levels may be required for optimal patient management. In the case of hypoalbuminemia due to liver disease, reduction in binding to albumin is generally too slow and gradual in onset to be of clinical significance. If free drug clearance is not altered, compensatory mechanisms will increase total drug clearance to maint~n steady state free drug concentration at a lower total drug level. As long as it is recognized that therapeutic range for total drug has been shifted down, neither free drug measurement nor dosage adjustment are necessary. AS liver dysfunc-
tion progresses, the capacity for drug biotransformation is impaired, and free drug clearance eventually will be reduced. Consequently, free drug concentration can no longer be maintained at steady-state level. Thus, for highly protein-bound, restrictively cleared drugs such as phenytoin and valproic acid, advanced liver disease can be accompanied by reduced total drug concentration, increased free drug fraction, and an unpredictable free drug level. In such instances of unpredictability during advanced state of liver disease, measurement of free phenytoin and valproic acid concentrations should be performed. Furthermore, because it is not possible to correlate the extent of liver damage and the onset of compromised free drug clearance, patients with rapidIy progressing liver disease should be monitored by free drug measurement.
Reduction in drug binding in renal disease also lowers steady-state total drug con~ntration and increases free fraction, whereas the free drug ~ncentration will remain unchanged. As a result, therapeutic total drug levels will be lower than the therapeutic range defined for total drug. A number of studies of protein binding of phenytoin and valproic acid in renal patients has shown that free drug fraction is proportional to the degree of renal failure. Therefore, it is possible to calculate at different serum creatinine levels the total drug concentration that will be required to produce therapeutic free drug level. A nomogram can then be constructed to relate
206
total drug therapeutic range to different serum creatinine levels [ 146.149.15 1.2221. While such nomograms are useful as general guidelines. free phenytoin and vatproic acid levels, if available. are the more reliable ways to monitor the complicated uremic patients. An abrupt reduction in protein binding due to drug displacement from binding sites by another drug can lead to a sudden release of bound drug. This is particularly significant if the displacing agent is administered as a bolus by intravenous injection. A transient increase in free drug can be of sufficient magnitude to precipitate an adverse drug reaction, if the drug has a small volume of distribution, a narrow therapeutic index, and is restrictively cleared. The number of~iinjcaf~~ ~~~~n~~cantdrug interactions is smull. The displacement of the anticoagulant warfarin by the nonsteroidal anti-inflammatory agent phenylbutazone and of phenytoin by valproic acid are examples of drug interactions which result in clinically significant toxicity. Addition of valproic acid to patients maintained on phenytoin has precipitated toxicity in at least 25% of the study patients, as well as decreased seizure control in up to 13% [223-2261. In these cases of drug interaction, free drug levels will have better correlation with clinical status than total drug levels. This is particularly evident in the case of phenytoin because of the conflicting observations on free and total phenytoin levels following the addition of valproic acid to the drug regimen, Thus, for patients treated ~vjth Fhen_~toi~l and 15a~pro~~.acid, monitoring of free phenytoin /eve& is recommended, particular@ when an unexpected therupeutic outcome occurs. Concentration-dependent binding of valproic acid and disopyramide should be taken into consideration during optimization of drug therapy for these drugs. As the ratio of free to bound drug varies with total drug concentration, quantitation of total drug concentration is no longer an indirect measure of free drug concentration. This phenomenon is also responsible, at least in part, for the nonlinear plasma total level-dose relationship, although free drug level at steady state is linearly related to dose. Thus, a dosage increase can yield a lower than expected increase in total drug level while free drug level has, in fact, been increased in proportion to dose. Therefore, free drug measurement of drugs such as valproic acid and disopyramide which exhibit variable binding can avoid the misleading information provided by total drug measurement. Moreover, the usefulness of monitoring total disopyramide is even more questionable in view of the protein binding properties of the deakylmetabolite. This metabolite accumulates to levels higher than those of the parent drug in some patients, exhibits concentration dependent binding and competes with disopyramide for binding to protein. Recent studies on the protein binding of cationic drugs suggest that in some acute clinical conditions drug therapy may be more effectively monitored by free drug levels. This may be particularly relevant for cationic drugs which bind to alpha-l acid glycoprotein (AAG) and lipoproteins. The rapidly changing plasma AAG levels after acute tissue injury, such as myocardial infarction and surgery, and the poorly controlled, diet-dependent plasma lipoprotein Ievels, mean that total drug levels should not be the best guide for drug therapy during these critical and unstable periods. Thus free drug monitoring of the cardiac active drugs such as the beta
207
blockers and the calcium channel blockers in the post-infarction period should prove to be important for critical care. Clinical documentation of this is awaiting the results of studies currently under way. Another area where future studies may point to the usefulness of free drug monitoring is perinatal patient care. The physiological changes during labor, delivery and in the immediate post-partum period are accompanied by rapid changes in plasma protein and fatty acid levels. Those medications used during this period which are highly protein bound, such as meperidine, bupivacaine and Valium, will have fluctuating free fractions and free drug concentrations. Obstetricians and anesthesiologists should find free drug monitoring helpful to perinatal care. Laboratory
considerations
Free drug is preferable to total drug monitoring because of its better correlation with pharmacological effects in all patients. Monitoring of total drug, however, is still the standard approach in the clinical laboratories. Free drug monitoring could assume a more important role if there was refinement in the technology to make free drug measurement easier and faster. Routine total drug measurement is technically and operationally simpler than free drug measurement by either equilibrium dialysis or ultrafiltration. Even though equilibrium dialysis has been employed most frequently and has accounted for most of the published information on drug binding, it has the major disadvantage of being laborious and may require many hours or even days to reach equilibrium. Ultrafiltration has been an attractive alternative to equilibrium dialysis. It is even more so now, since previous concerns about the effect of ultrafiltration on binding equilibrium have now been invalidated. Thus, it is not necessary to restrict the ultrafiltration process to give only a small volume of ultrafiltrate, and the analytical problem of measuring low concentration of drug in a small volume of ultrafiltrate is eliminated. Furthermore, centrifugation to obtain ultrafiltrates from multiple specimens is technically simpler and more rapid than equilibrium dialysis, making ultrafiltration more suitable for free drug measurement in the clinical laboratory environment. Conclusion
The indications for free drug monito~ng are understood for a limited number of drugs and clinical situations. Until new and easier techniques for free drug measurement are devised, therapeutic drug monitoring will continue to be based on total drug which will suffice for the management of many but not all patients. The need for free drug monitoring, however, will rise as we advance our understanding of protein binding as well as of the effects of altered binding on drug metabolism and clearance. As clinical experience is gained on the relationship between free drug concentration and drug effects, therapeutic ranges based on free drug rather than total drug concentrations will be established, and informed. clinicians will find free drug m~surement valuable, especially in the management of clinical situations where altered binding can lead to unpredictable free drug concentration, This clinical demand will become the impetus for technological innovations that are
needed activity
to make in clinical
free drug nl~?n~toring laboratories.
the routine
therapeutic
drug
rn~~nlt[~riilg
Acknowledgements I would like to thank Dr. Leslie Shaw for helpful discussions and Ms. Catherine Seneca1 and Ms. Sandra Hengstler for their patience during the preparation of this manuscript. Work in the author’s laboratory was supported by NIH Grant HL-33541. References 1 Koch-Weser 311-316. 2 Koch-Weser 526-531,
J, Sellers EM. Binding
of drugs to serum aibumin-Pari
J, Seller EM. Binding of drugs to serum albumin-Part
I. New Engl J Med 1976; 294: II. New Engl J Med 1976: 294:
3 Jusko W, Gretch M. Plasma and tissue protein binding of drugs in pharmacokinetics.
Drug
Metab
Rev 1976: 5: 43-140. 4 Rowland M, Tozer TN. In: Clinical and Febiger, 1980:
Pharmacokinetics:
Concepts
and Applications.
Philadelphia:
Lea
5 Wandell M, Wilcox-Thole. Protein binding and free drug concentrations. In: Mungall, ed. Applied clinical pharmacokinetics. New York: Raven Press, 1983. 6 Piafsky KM. Disease-induced changes in the plasma binding of basic drugs. Clin Pharmacokinet 1980; 5: 246-262. 7 Vallner JJ. Binding of drugs by albumin and plasma protein. J Pharm Sci 1977; 66: 447-465. 8 Bickel MH. Binding of chlo~romazine and imipramine to red cefls, albumin. lipoproteins and other blood components. J Pharm Pharmacol 1975: 27: 733-738. 9 Kwong TC, Sparks JD, Sparks CE. Lipoprotein and protein binding of the calcium channel blocker diltiazem. Proc Sot Exp Biol Med 1985; 178: 313 -316. 10 Tamir I, Rifkind BM, Levy RI. Measurements of lipids and evaluation of lipid disorders, In: Henry. ed. Clinical diagnosis and management by laboratory methods. Vol. I, 16th Ed. Philadelphia: WB Saunders, 1979: 189-227. 11 Gazzard BG. Ford-Hutchinson AW. Smith MJH, Williams R. The binding of paracetamol to plasma of man and pig. .i Pharm Pharmacol 1973: 25: 964-967. 12 Reidenberg MM, Drayer DE. Levy M. Warner H. Polymorphic acetylation of procamamide in man. Clin Pharmacol Ther 1975; 17: 722-730. 13 Clark JT. Libke RD. Regamey C, Kirby WMM. Comparative pharmacokinetica of amikacin and kanamycin. Clin Pharmacol Ther 1974; 15: 610-616. 14 Pike E, Kierulf P. Skuterud B, Bredesen JE, Lunde PKM. Drug binding in sera deficient in lipoproteins, albumin or orosomucoid. Br J Clin Pharmacol 1983; 16: 233-239. 15 Hooper WD, Dubetz DK, Bochner F. Cother LM, Smith GA, Eadie MJ, Tyrer JH. Plasma protein binding of carbamazepine. Clin Pharmacol Ther 1975; 17: 4333440. 16 Di Salle E, Pacifici G. Morselli P. Studies on plasma protein binding of carbamazepine. Pharmacol Res Commun 1974; 6: 193-202. 17 Koup JR, Lau AH, Brodsky B, Slaughter RL. Chloramphenicol pharmacokinetics in hospitalized patients. Antimicrob Agents Chemother 1979: 15: 651-657. 18 Roberts RK, Wilkinson GR. Branch RA, Schenker S. Effect of age and cirrhosis on the disposttion and ehmination of chlordiazepoxide (librium). Gastroenterology 1978; 75: 479-485. 19 Verbeeck RK, Cardinal JA, Hill AG, Midha KK. Binding of phenothiazine neurofeptics to plasma proteins. Eiochem Pharmacol 1983; 32: 2565-2570. 20 Borga 0, Azarnoff DL, Plym Forshell G. Sjoqvist F. Plasma protein binding of tricychc antidepressants in man, Biochem Pharmacol 1969; 18: 2135-2143.
209 21 Abel JG, Sellers EM, Narango CA, Shaw J, Kadar D, Romach MK. Inter- and intrasubject variation in diazepam free fraction. Clin Pharmacol Ther 1979; 26: 247-255. 22 Shoeman DW, Azarnoff DL. The alterations of plasma proteins in uremia as reflected in their ability to bind digitoxin and diphenylhydantoin. Pharmacology 1972; 7: 169-177. 23 Storstein L. Protein binding of cardiac glycosides in disease states. Clin Pharmacokinet 1977; 2: 220-233. 24 Lima JJ, Boudoulas H, Blanford M. Concentration-dependence of disopyramide binding to plasma protein and its influence on kinetics and dynamics. J Pharmacol Exper Therapeut 1981; 219: 741-747. 25 Bredesen JE, Pike E, Lunde PKM. Plasma binding of disopyramide and mono-n-dealkyldisopyramide. Br J Clin Pharmacol 1982; 14: 673-676. 26 Glazko AJ, Chang T. Ethosuximide absorption, distribution and excretion. In: Woodbury D. Penry J and Pippenger C, eds. Antiepileptic Drugs. New York: Raven Press. 1982: 623-629. 27 Christopher TG, Blair AD, Forrey AW, Cutler RE. Hemodialyzer clearances of gentamycin. kanamycin, tobramycin, amikacin, ethambutal. procainamide and flucytosine. with a technique for planning therapy. J Pharmacokinet Biopharm 1976; 4: 427-441. 28 Piafsky KM, Borga 0. Plasma protein binding of basic drugs. II. Importance of cy,-acid glycoprotein for inter-individual variation. Clin Pharmacol Ther 1977; 22: 545-549. 29 Kornguth M, Hutchins, L, Eichelman B. Binding of psychotropic drugs to isolated a,-acid glycoprotein. Biochem Pharmacol 1981; 30: 2435-2441. 30 Routledge PA, Barchowsky A, Bjornsson TD, Kitchell BB, Shand DG. Lidocaine plasma protein binding. Clin Pharmacol Ther 1980; 27: 347-351. 31 Thomsen K, Shou M. Renal lithium excretion in man. Am J Physiol 1968; 215: 823-827. 32 Wan SH, Hoffman DH, Azarnoff DL, Stephens R, Hoogstraten B. Effect of route of administration and effusions on methotrexate pharmacokinetics. Cancer Res 1974; 34: 3487-3491. 33 Pike E, Skuterud B. Plasma binding variations of amitriptyline and nortriptyline. Clin Pharmacol Ther 1982; 32: 228-234. 34 Ehrnebo M, Agurell S, Jailing B, Boreus LO. Age differences in drug binding by plasma protems: studies on human foetuses, neonates and adults, Eur J Clin Pharmacol 1971; 3: 189-193. 35 Hooper WD, Bochner F, Eadie MJ, Tyrer JH. Plasma protein binding of diphenylhydantoin. Effects of sex hormones, renal and hepatic disease. Clin Pharmacol Ther 1974; 15: 276-282. 36 Schotteliusd D. Primidone, absorption, distribution and excretion. In: Woodbury D, Penry J and Pippenger C, eds. Antiepileptic Drugs. New York: Raven Press, 1982: 405-413. 37 Pippenger, C, Garlock C, Desaulmiers C, Sternberg S. A rapid technique for the determination of antiepileptic drug concentrations in plasma. Epilepsia 1980; 21: 187. 38 Sager G, Nilsen 0, Jacobsen S. Variable binding of propranolol in human serum. Biochem Pharmacol 1979; 28: 905-911. 39 Kates RE, Sokoloski TD, Comstock TJ. Binding of quinidine to plasma proteins in normal subjects and in patients with hyperlipoproteinemias. Clin Pharmacol Ther 1978; 23: 30-35. 40 Pike E, Skuterud B, Kierulf P, Lunde P. Significance of lipoproteins in serum binding variations of amitriptyline, nortriptyline and quinidine. Clin Pharmacol Ther 1982; 32: 599-606. 41 Borga 0, Odar-Cederlof I, Ringberger V, Norlin A. Protein binding of salicylate in uremic and normal plasma. Clin Pharmacol Ther 1976; 20: 464-475. 42 Shaw L, Fields L, Maycock R. Factors influencing theophylline serum protein binding. Clin Pharmacol Ther 1982; 32: 490-496. 43 Brors 0, Sagar G, Sandnes D, Jacobson S. Binding of theophylline in human serum determined by ultrafiltration and equilibrium dialysis. Br J Clin Pharm 1983; 15: 393-397. 44 Williams RL, Blaschke TF, Meffin PJ, Melmon KL, Rowland M. Influence of acute viral hepatitis on disposition and plasma binding of tolbutamide. Clin Pharmacol Ther 1977; 21: 301-309. 45 BowdIe TA, PateI IH, Levy RH, Wilensky AJ. Dose-dependent valproate clearance (Abstracts). Acad Pharmaceut Sci 1979; 9: 98. 46 Bachmann K. Rapid determination of the concentration of unbound warfarin in human plasma. Res Commun Chem Pathol Pharmacol 1974; 9: 379-382. 47 Powell JR, Vozeh S, Hopewell P, Costello J, Sheiner LB, Riegelman S. Theophylline disposition in acutely iII hospitalized patients. Am Rev Respir Dis 1975; 11: 229-237.
4X CJ’Re~lly RA, Welling PG. Wagner JG. Pharanlacoklnctics of warfarm iatration in man. Thromb Diath Haemorrh 1971; 25: 17X-186. 49 Wilkinson GR. Shand 1975: 18: 377-390.
DG. A physiologtcal
50 Pond SM. ToLer TN. First Pharmacokinet 1984: 9: l--25
pass
approach
elimination.
to hepatic
basic
followtng
drug clearance.
concepts
and
climcal
51 Perucca E, Richens A. Reduction of oral btoavailahtlity of lignocaine metabolism in epileptic patients. Br J C‘lin Fharrnac~~l 1979: 7: 21 -31.
II~~I-~I~~I~~IL~\ adnun Clin Pharmacol
Ther
consequence,.
C’ftn
hy inductton
of ltrst pass
52 Evans GH. Shand DG. Disposition of propranolol. V. Drug accumulation and steady-state con centrations during chronic oral administration in man. Clin Pharmacol Ther 197.1: 14: 487.493. 53 Pardridge WM. Transport of protein-bound hormones into tissues in viva. Endocrine Revjiews 1981: 2(l): 103.-123. 54 Pardridge WM. Sakiyama R. Fierer G. Transport of propranolol blood-brain barrier. J Clin Invest 1983: 71: 900.-908. 55 Kun H. Trunk H, Weitz B. Evaluation of methods Arzneimittel-Forshung 1977: 27: 1373 1380. 56 Chignell CF. In: Garrett ER. Hirtz JL. eda. Protein binding Marcel Dekker, 1977; Vol. I: 188-228.
and lidocaine
to determtne
protein
through binding
in drug fate and metabolism.
57 Bowers WF, Fulton S. Thompson J. Ultrafiltration vs. equilibrium fractron. Clin Pharmacokinet 1984; 9(Suppl. 1): 49.-60.
dialysis
the rat of drugs.
New York.
for determination
of free
59 Nimmo IA. Atkins GL. Strange RL. Percy-Rot& JW. An evaluation of ways of using equilibrium and dialysis to quantify the binding of l&and to macromolecule. B&hem J 1977: 165: lO7- I 10. 59 Steinhardt J, Reynolds JA. Multiple equilibria in proteins. New York: Academrc Press. 1969; 34-53. 79Y83. 60 Behm HL, Wagner JC. Errors in interpretation of data from equilibrium dialyses protein hinding experiments. Res Commun Chem Pathol Pharmacol 1979: 26: 146- 160. nature of the errors 61 Builder SE. Segel IH. Equilibrium l&and binding assays using labeled substrates: introduced by radi~hemical impurities. Anal Biochem 1978; 85: 413-424. 62 Chadwick C. Phipps DA, Powell C. Potential method~~iogicai errors in the application of equilibrium dialysis to the determination of ‘free’ and ‘bound’ plasma tryptophan. Clin Chim Acta 1979: 99: 219-282. 63 Kinget R, Bontinck AM. Herbots H. Problems of dialysis techniques in the study of macromolecule binding of drugs. lnternat J Pharmaceut 1979; 3: 65-72. 64 McNamara PJ. Bogardus JB. Effect of initial conditions and drug-protein binding on the time to equilibrium in dialysis systems. J Pharmaceut Sci 19X2; 71: 1066-1068. 65 Selikson M, Gibson RE, Eckelman WC. Reba RC. Calculation of binding isotherms when l&and and receptor are in different volumes of distribution. Anal Biochem 1980; 108: 64- 71. 66 Smith JB, Jubiz W. A source of error in equilibrium dialysis. Steroids 1980; 36: 3933403. 67 Tanford C. Physical Chemistry of Macromolecules. New York: Wileg, 1961; 2255227. 68 Keen PM. The binding of penicillins to bovine serum albumin. Biochem Pharmacol 1966: 15: 443-463. 69 Suter P, Rosenbusch JP. Determination of the Donnan effect in equilibrium dialysis and a simple method for its correction. Anal Biochem 1977: 82: 109-114. 70 Monot C, Netter P, Stalars MC’, Martin J, Royer RJ. Gaucher A. Difficulties in applying the Scatchard model of ligand hinding to proteins proposal of new mathematical tools - application to salicylates. J Pharmaceut Sci 1983; 72; 35-41. 71 Woo E, Greenblatt DJ. Pharmacokinetic and clinical implications of quinidine protein binding. J Pharm Sci 1979; 4: 466-469. 72 Wilting J, van der Giesen WF, Janssen LHM, Weideman MM, Otagin M, Perrin JH. The effect of albumin conformation on the binding of warfarin to human serum albumin. J Biochem lY80; 255: 3032-3037. 73 Henry JA, Mitchell SN. Effect of pH on human plasma protein binding of a series of B-adrenoceptor antagonists. Br J Clin Pharmacol 1981; 11: 119-120. 74 Kristensen CB. Gram LF. Equilibrium dialysis for determination of protein binding of imipramine. Acta Pharm Tax 1982; 50: 130--l 36.
211
75 Brors 0, Nilsen OG, Sager G, Sandnes D, Jacobsen S. Influence of pH and buffer type on drug binding in human serum. Clin Pharmacokinet 1984; 9 (Suppl. 1): 85-86. 76 Cronin M, Hammad N. Effect of buffer composition of free drug concentrations by ~uilibrium dialysis. Clin Pharmacol Ther 1982; 31: 214. 77 Guentert TW, Oie S. Factors influencing the apparent protein binding of quinidine. J Pharm Sci 1982; 71: 325-328. 78 Nilsen OG. Serum albumin and lipoprotein as the quinidine binding molecules in normal human sera. Biochem Pharmacol 1976: 25: 1007-1012. 79 Rudman D, Bixler TJ, Del Rio AE. Effect of free fatty acids on binding of drugs by bovine serum albumin, by human serum albumin and by rabbit serum. J Pharmacol Exp Ther 1971; 176: 261-272. 80 Craig WA, Evenson MA, Ramgopal V. In: Benet LZ, ed. The effect of disease states on drug pharmacokinetics. Washington, D.C.: APhA Academy of Pharmaceutical Sciences, 1976: 125-136. 81 Sampson D, Hensley WJ. A rapid gas chromatographic method for the quantitation of underivatized individual free fatty acids in plasma. Clin Chim Acta 1975; 61: 1-8. 82 Nilsen OG, Storstein L. Jacobsen S. Effect of heparin and fatty acids on the binding of quinidine and warfarin in plasma. Biochem Pharmacol 1977: 26: 229-235. 83 Whitlam JB, Brown KF. Ultrafiltration in serum protein binding determinations. J Pharmaceut Sci 1981; 70: 146-150. 84 Feldmann K. New devices for flow dialysis and ultrafiltration for the study of protein-ligand interactions. Anal Biochem 1978; 88: 225-235. 85 Sophianopoulos JA, Durham SJ, Sophianopoulos AJ, Ragsdale HL, Cropper WP. Ultrafiltration is theoretically equivalent to equilibrium dialysis but much simpler to carry out. Arch Biochem Biophys 1978; 187: 132-137. 86 Spector R, Vernick R. Lorenzo AV. Effect of pressure on the plasma binding of digoxin and ouabain in an ultrafiltration apparatus. Biochem Pharmacol 1973; 22: 2483-2487. 87 Wilson K. Sex-related difference in drug disposition in man. Clin Pharmacokinet 9: 189-202. 88 Kristensen CB. Imipramine serum protein binding in healthy subjects. Clin Pharmacol Ther 1983; 34: 689-694. 89 Roberts, RK, Desmond PV, Wilkinson GR, Schenker S. Disposition of chlordiazepoxide: sex differences and effects of oral contraceptives. Clin Pharmacol Ther 1979; 25: 826-831. 90 Jochemsen R, Van der Graaff M, Boeijinga JK, Breimer DD. Influence of sex, menstrual cycle and oral contraceptives on the disposition of nitrazepam. Br J Clin Pharmacol 1982; 13: 319-324. 91 Abels JG, Narabhi CA, Shaw J, Kadar D, Romach MK. Inter- and intra-subject variation in diazepam free fraction. Clin Pharmacol Ther 1979; 26: 247-255. 92 Greenblatt DJ, Harmatz JS, Shader RI. Sex differences in diazepam protein binding in patients with renal insufficiency. Pharmacology 1978; 16: 26-29. 93 Routledge PA, Stargel WW, Kitchell BB, Barchowski A, Shand DC. Sex-related differences in plasma protein binding of lidocaine and diazepam. Br J Clin Pharmacol 1981; 11: 245-250. 94 Giles HG, Sellers EM, Naranjo CA, Frecker RC, Greenblatt DJ. Disposition of intravenous diazepam in young men and women. EurJ Clin Pharmacol 1981; 20: 207-213. 95 Ochs HR, Greenblatt DJ, Divoll M, Abernathy DR, Feycrabend H, Dengler HJ. Diazepam kinetics in relation to age and sex. Pharmacology 1981; 23: 24-30. 96 Wood M, Wood AJJ. Changes in plasma binding and u,-acid glycoprotein in mother and newborn infant. Clin Pharmacol Ther 1981; 29: 522-526. 97 Divoll M, Greenblatt DJ. Effects of age and sex on lorazepam protein binding. J Pharm Pharmacol 1982; 34: 122-123. 98 Greenblatt DJ, Divoll M, Harmatz JS, Shader RI. Oxazepam’kinetics: effects of age and sex. J Pharmacol Exp Ther 1980; 215: 86-91. 99 Laurel1 C, Kullander S, Thorell J. Plasma protein changes induced by sequential type of contraceptive steroid pills. Clin Chim Acta 1969; 25: 294-296. 100 Joseph JC, Baker C, Sprang M, Bermes EW. Changes in plasma proteins during pregnancy. Ann Clin Lab Sci 1978; 8: 130-141. 101 Kramer B, Kramer F, Hytten FE. Drug disposition and pharmacokinetics in the maternal-placentalfetal unit. Pharmacol Ther 1980; 10: 301-308.
102 Perucca F.. Crema A. Plasma protein binding of drug\ III prcgnancv. Clin Pharmncok~no~ I 4x2; 7, 336 352. 103 Janz I). Antiepileptrc drugs and pregnancy: altered utilization patterns and teratogmrxis. t:pliepsia 19x2: 23(Suppl. 1): S53%S63. 104 Chen SS. Perucca E. Lee J-N. Richens A. Serum protein binding and free concentratmn of phen>toin and phenobarbitone in pregnancy. Br J Clin Pharmacol 1982; 13: 547-554. 105 Dean M. Stock B, Patterson RJ, Levy G. Serum protein binding of drugs during and after prqnancy in humans. Clin Pharmacol Ther 19X0; 2X: 253-261. 106 Prrucca ti. Ruprah M. Richena A. Altered drug binding to serum proteins in pregnant women: therapeutic relevance. J R Sot Med 19X1: 74: 422-426. 707 Perucca E. Ruprah M, Richens A. Decreased serum protein bindmg of diazepam and valproic acid in pregnant women. Br J Clin Pharmacol 19X1; 12: 276P. 108 Lovecchio JL, Krnsncr J. Yal’fe SJ. Serum protein hinding of sallcylate during pregnancy and the puerperium. Dev Pharmacol Ther 19X1: 2: 172-179. 109 Reboud I’. postpartum 110 Leonard R. 111 Greenbiatt 10X1-IOXX.
Groulade J. Groalamhert P, Colombo M. The influence of normal pregnancy and the state on plasma proteins and lipids. Am .I Obstet Gyn 1963: X6: 820-X2X. Phenytoin binding in pregnancy. Lancet 19X0: 2(X207): 1312-1313. DJ. Sellers EM. Shader RI. Drug disposition in old age. N Engl J Med 1982: 306:
112 Morselli PL, Franc+Morhelli R. and Bossi L. Clinical pharmacokinetics in newhorns and infants. Clin Pharmacokmet 1980: 5: 4X5--527. 113 Schmucker DL. Age-related changes in drug disposition. Pharmacol Rev 1979: 30~4): 445-456. 114 Piafsky KM. Woolner EA. The hinding of basic drugs to oc,-acid glycoprotein in cord serum. J Ped 1982; 100: X20-821. 115 Peterson MC. Moore RG. Nation RL, McMeniman W. Relationship between the transplacental gradients of bupiva~aine and n,-acid glycoprotein. Br J Clin Pharmacol 1981: 12: X59-862. 116 Natton RL. Meperidine binding in maternal and fetal plasma. Clin Pharmacol Ther 1981; 29: 472-419. 117 Ridd MJ. Brown KF, Nation RL. Collier CB. Differential transplacental binding of diazepam: causes and implications. Eur J Clin Pharma~ol 1983: 24: 595-601. 1 I8 Nau H. Luck W. Kuhnz W. Decreased serum protein binding of diazepam and its major metabolite in the neonate during the first postnatal week related to increased free fatty acid levels. Br J Clin Pharmacol 1984: 17: 92-98. 119 Nau H, Rating D. Koch S. Hauser I. Helge H. Vaiproic acid and its metabolites: placental transfer. neonatal pharmacokinetics. transfer via mother’s milk and clinical status in neonates of epileptic mothers. J Pharmncol Exp Ther 19X1; 219: 768-777. 120 Albani E’. Riva R, Contin M. Barurri A. Altomare M, Merlin G. Perucca. Differential transplacental hinding of valproic acid: influence of free fatty acids. Br J Clin Pharmacol 1984; 17: 759-762. 121 Paclfici GM, Taddeucce-Brunelli G. Kane A. Clonazepam serum binding during development. Clin Pharmacol Ther 1984; 35: 354-359. 122 Hamar C. Levy G. Serum binding of drugs and bilirubin in newborn infants and their mothers. Clin Pharmacol Thkr 19X0: 2X: 5X-63. 123 Wallace S. Altered plasma albumin in the newborn infant. Br J Clin Pharmacol 1977: 4: X2-85. 124 Frledmnn LA. Lewis PJ. The effect of semisynthetic penicillins on the binding of bilirubm by neonatal serum. Br .I Clin Pharmacol 1980: 9: 61-65. 125 Krasner J. Giacoiu GP. Yaffe SJ. Drug hinding in the newborn infant. Ann NY Acad SCI 1973: 226: 101-114. 126 Loughnam PM, Greenwald A, Purton WW. Aranda JV. Walters G, Neims AH. Pharmacokinetic observations of phenytoin disposition in the newhorn and young infant. Arch Dis Child 1977; 52: 302-304. 127 Fredholm BB, Rane A, Persson B. Diphenylhydantoin binding to protein in plasma and its dependence on free fatty acid and bilirubin concentration in dogs and newborn infants. Ped Res 1975; 9: 26-30. 12X Greenhlatt DJ. Reduced serum albumin concentration in the elderly: a report from the Boston Collaborative Drug Surveillance Program. J Am Geriatr SOC 1979: 27: 20-22.
213 129 Dybkaer R, Lauritzen M, Krakauer R. Relative reference values for clinical chemical and hematological quantities in ‘healthy’ elderly people. Acta Med Stand 1981; 209: l-9. 130 MacLennan WJ, Martin P, Mason BJ. Protein intake and serum albumin levels in the elderly. GerontoIogy 1977; 23: 360-367. 131 Tillement JP, Lhoste F, Giudecelli 3: 144-154. 132 Blaschke TF. Protein 32-44. 133 Williams
RL, Mamelok
binding
JF. Diseases and drug protein
and kinetics
RD. Hepatic
of drugs
binding.
in liver disease.
disease and drug pharmacokinetics.
Clin Pharmacokinet
Clin Pharmacokinet Clin Pharmacokinet
1978; 1977: 2: 1980:
5: 528-547. 134 Olsen CD, Bennet WM, Porter GA. Morphine and phenytoin binding to plasma proteins in renal and hepatic failure. Clin Pharmacol Ther 1975; 17: 677-684. 135 Affirme M, Reidenberg MM. Protein binding of some drugs in plasma from patients with alcoholic liver disease. Eur J Clin Pharmacol 1975; 8: 267-269. 136 Wood AJ, Kornhauser DM, Wilkinson GR, Shand DG, Branch RA. The influence of cirrhosis on steady-state blood concentrations of unbound propranolol after oral administration. Clin Pharmacokinet 1978; 3: 478-487. 137 Klotz U, Avant GR, Hoyumpa A, Schenker S, Wilkinson GR. The effects of age and liver disease on the disposition and elimination of diazepam in adult man. J Clin Invest 1975; 55: 347-359. 138 Roberts RK, Wilkinson GR, Branch RA, Schenker S. Effects of age and parenchymal liver disease on the disposition and elimination of chlordiazepoxide. Gastroenterology 1978; 75: 479-483. 139 Kraus JW, Desmond PV, Marshall JP, Johnson RF, Schenker S, Wilkinson GR. Effects of aging and liver disease on disposition of lorazepam. Clin Pharmacol Ther 1978; 24: 411-419. 140 Giacomini KM, Massoud N, Wong FM, Giacomini. Decreased binding of verapamil to plasma proteins in patients with liver disease. J Card Pharmacol 1984; 6: 924-928. 141 Blaschke TF, Meffin PJ, Melmon KL, Rowland M. Influence of acute viral hepatitis on phenytoin kinetics and protein binding. Clin Pharmacol Ther 1975; 17: 685-691. 142 Rane A, Lunde, PKM, Jalling B, Yaffe SJ, Sjoqvist F. Plasma binding of diphenylhydantoin in normal and hyperbilirubinemic infants. J Pediatr 1971; 78: 877-882. 143 Odar-Cederlof I, Lunde P, Sjoqvist F. Abnormal pharmacokinetics of phenytoin in a patient with uraemia. Lancet 1970; 2: 831-832. 144 Reidenberg MM, Odar-Cederlof I, von Bahr C, Borga 0, Sjoqvist F. Protein binding of diphenylhydantoin and desmethylimipramine in plasma from patients with poor renal function. N Engl J Med 1971; 285: 264-267. 145 Blum MR, Riegelman S. Altered protein binding of diphenylhydantoin in uremic plasma. New Engl J Med 1972; 286: 109. 146 Ehrnebo M, Odar-Cederlof I. Binding of amobarbital, pentobarbital, and diphenylhydantoin to blood cells and plasma proteins in healthy volunteers and uraemic patients. Eur J Clin Pharmacol 1975; 8: 445-453. 147 Gugler R, Mueller G. Plasma protein binding of valproic acid in healthy subjects and in patients with renal disease. Br J Clin Pharmacol 1978; 5: 441-446. 148 Brewster D, Muir NC. Valproate plasma protein binding in the uremic condition. Clin Pharmacol Ther 1980; 27: 76-82. 149 Bruni J, Wang LH, Marbury TC, Lee CS, Wilder BJ. Protein binding of valproic acid in uremic patients. Neurology 1980; 30: 557-559. 150 Farrell PC, Grib NL, Fry DL, Popovich RP, Broviac JW, Babb AL. A comparison of in vitro and in vivo solute protein binding interactions in normal and uremic subjects. Transac Amer Sot Artificial Internal Organs 1972; 18: 268-276. 151 Belpaire FM, Bogaert MG, Mussche MM. Influence of renal failure on the protein binding of drugs in animals and man. Eur J Clin Pharmacol 1977; 11: 27-32. 152 Borga 0, Odar-Cederlof I, Ringberger VA, Norlin A. Protein binding of salicylate in uremic and normal plasma. Clin Pharmacol Ther 1976; 20: 464-475. I53 Arango G. Mayberry WE, Hockert TJ, Elveback LR. Total and free human serum thyroxine in normal and abnormal thyroid states. Mayo Clinic Proc 1968; 43: 503-516.
214 154 Bachmann K, Shapiro R. Mackiewcz J. Influence of renal dysfunction on warfarin plasma protein binding. J Clin Pharmacol 1976; 16: 468-472. 155 Kober A, SJoholm I. Borga 0, Odar-Cederlof 1. Protein binding of diazepam and digitoxin in uremic and normal serum. Biochem Pharmacol 1979; 2X: 1037- 1042. 156 Odar-Cederlof 1. Vessman J. Alvan G. SJoqvist F. Oxazepam disposition in uremic patient\. ~cta Pharmacolog Toxicol 1977; 40 (Suppl. 1): 52-62. 157 Lowenthal DT. Pharmacokinetics of propranolol. renal disease. Am J Med 1977: 62: 5322538.
quinidme.
procainamide
and hdocaine
in chronic
158 Perez-Mate0 M. Erill S. Protein binding of salicylate and quinidine in plasma from patients with renal failure, chronic liver disease and chronic respiratory insufficiency. Em J Clin Pharmacol 1977: 11: 2255231. 159 Keefe DL. Yee YG. Kates RE. Verapamil Pharmacol Ther 1981: 29: 21-26. 160 Gugler R. Azarnoff 1976: 1: 25535.
DL. Drug
protein
161 Reidenberg MM. Drayer DE. Alteration cokinet 1984; 9(Suppl. I): 1X-26.
protein
binding
binding
in patients
and the nephrotic
of drug-protein
binding
and in normal
syndrome. in renal
subjects.
C‘lin
Clin Pharmacokinet disease.
Clin
Pharma-
162 Schmid K. a,-Acid glycoprotein. In: Putnam FM. ed.. The plasma proteins, Vol. 1. New York: Academic Press. 1975. 163 Routledge PA, Stargel WW, Wagner GS. Shand DG. Increased plasma propranolol binding in myocardial infarction. Br J Clin Pharmacol 1980; 9: 438-439. 164 Paxton JW. Norris RM. Propranolol disposition after acute myocardial infarction. CIin Pharmacol Ther 1984; 36: 3377342. 165 Routledge PA. Stargel WW, Wagner GS, Shand DG. Increased acid glycoprotein and lidocaine disposition in myocardial infarction. Ann Intern Med 1980; 93: 7OlI704. 166 Routledge PA, Shand DG, Barchowsky A. Wagner G, Stargel WW. Relationship between n,-actd glycoprotein and lidocaine disposition in myocardial infarction. Clin Pharmacol Ther 1981: 30: 154-157. 167 David BM, Ilett KF. Whitford EG, Stenhouse NS. Prolonged variability in plasma protein binding of disopyramide after acute myocardial infarction. Br J Clin Pharmacol 1983: 15: 435-441. 168 Freilich DI. Giardina EV. Imipramine binding to alpha-a-acid glycoprotein in normal subjects and cardiac patients. Clin Pharmacol Ther 1984: 35: 670-674. 169 Fremstad D. Bergerud K, Haffner JFW, Lunde PKM. Increased plasma binding of quinidine after surgery: a preliminary report. Eur J Clin Pharmacol 1976: 10: 441-444. 170 Feely J, Forrest A, Gunn A, Hamilton W. Stevenson I, Crooke J. Influence of surgery on plasma propranolol levels and protein binding. Clin Pharmacol Ther 1980: 28: 759-764. 171 Jackson PR, Tucker CT, Woods HF. Altered plasma drug binding in cancer: role of ol,-acid glycoprotein and albumin. Clin Pharmacol Ther 1982: 32: 2955302. 172 Abramson FP. Jenkins J. Ostchega Y. Effects of cancer and its treatments on plasma concentration of alpha,-acid glycoprotein and propranolol binding. Clin Pharmacol Ther 1982; 32: 6599663. 173 Abramson FP. Methadone plasma protein binding: alterations in cancer and displacement from n,-acid glycoprotein. Clin Pharmacol Ther 1982: 32: 652-658. 174 Edwards DJ. Lalka D. Cerra F. Slaughter RL. Alpha,-acid glycoprotein concentration and protem binding in trauma. Clin Pharmacol Ther 1982; 31: 62-67. 175 Martyn J. Abernethy DR, Greenblatt DJ. Plasma protein binding of drugs after severe burn injury. Clin Pharmacol Ther 1984; 35: 535539. 176 Piafsky KM, Borga 0, Ddar-Cederlof I. Johnansson C, Sjoqvist F. Increased plasma protein binding of propranolol and chlorpromazine mediated by disease-induced elevations of plasma a,-acid glycoprotein. N Engl J Med 1978; 299: 143551439. 177 Bowdle TA. Pate1 I. Levy RH. Wilensky AJ. Valproic actd dosage and plasma protein binding and clearance. Clin Pharmacol Ther 1980; 28: 486-492. 178 Rowland M. Plasma protein binding and therapeutic drug monitoring. Ther Drug Monitor 1980: 2: 29-31. 179 Furst D, Tozer TN, Melmon KL. Salicylate clearance, the result of protein binding and metabolism. Clin Pharmacol Ther 1979; 26: 380.
215 180 Giacomini KM, Blaschke TF. Effect of concentration-dependent binding to plasma proteins on the pharmacokinetics and pharmacodynamics of disopyramide. Clin Pharmacokinet 1984; 9(Suppl. 1): 42-48. 181 Alvan 182 183 184 185 186 187 188 189 190 191 192 193
G,
Bergman,
U, Gustafsson
H. High
unbound
fraction
of salicylate
in plasma
during
intoxication. Br J Clin Pharmacol 1981; 11: 625-626. Hinderling PH, Bres J, Garrett ER. Protein binding and erythrocyte partitioning of disopyramide and its monodealkylated metabolite. J Pharmaceut Sci 1974; 63: 1684-1690. Meffin, Koch-Weser J. Drug therapy, disopyramide. N Engl J Med 1979; 300: 957-962. Cunningham JL, Shen DD, Shudo I, Azarnoff DL. The effect of non-linear disposition kinetics on the systemic availability of disopyramide. Br J Clin Pharmacol 1977; 5: 343-346. Lima JJ, Boudoulas H, Blanford M. Concentration dependence of disopyramide binding to plasma protein and its influence on kinetics and dynamics. J Pharmacol Exp Ther 1981; 219: 741-747. Hinderling PH, Garrett ER. Pharmacokinetics of the antiarrhythmic disopyramide in healthy humans. J Pharmacokinet Biopharmaceut 1976; 4: 199-230. Whiting B, Holford NHG, Sheiner LB. Quantitative analysis of the disopyramide concentration-effect relationship. Br J Clin Pharmacol 1980; 9: 67-75. Aitio M-L. Enhanced metabolism and diminished efficacy of disopyramide by enzyme induction. Br J Clin Pharmacol 1980; 9: 149-152. Dahlqvist R, Borga 0, Rane A, Walsh 2, Sjoqvist F. Decreased plasma protein binding of phenytoin in patients on valproic acid. Br J Clin Pharmacol 1979; 8: 547-552. Monks A, Richens A. Effect of single doses of sodium valproate on serum phenytoin levels and protein binding in epileptic patients. Clin Pharmacol Ther 1980; 27: 89-95. Perucca E, Hebdige S, Frigo GM, Gatti G, Lecchini S, Crema A. Interaction between phenytoin and valproic acid: plasma protein binding and metabolic effects. Ciin Pharmacol Ther 1980; 28: 779-789. Rodin EA. DeSousa G, H~duke~ch D, Lodhi R, Berchou RC. Diss~iation between free and bound phenytoin levels in presence of sodium valproate. Arch Neurol 1981; 38; 240-242. Friel PN, Lea1 KW, Wilensky AJ. Valproic acid-phenytoin interaction. Ther Drug Monitor 1979; 1:
243-248. 194 Kober A, Olsson Y, Sjoholm I. Binding of drugs to human serum albumin XIV. The theoretical basis for the interaction between phenytoin and valprorte. Molec Pharmacol 1980; 18: 237-242. 195 Aggeler PM, O’Reilly RA, Leong L, Kowitz PE. Potentiation of anticoagulant effect of warfatin by phenylbut~one. N Engl J Med 1967; 276: 496-501. 196 McEinay JC, D’Arcy PF. Displacement of albumin-bound warfarin by anti-inflammatory agents in vitro. J Pharm Pharmacol 1980; 32: 709-711. 197 Levy RH, Koch KM. Drug interactions with valproic acid. Drugs 1982; 24: 543-556. 198 Tsanaclis LM, Allen J, Perucca E. Routledge PA, Richens A. Effects of valproate on free plasma phenytoin concentrations. Br J Clin Pharmacol 1984; 18: 17-20. 199 Levy RH, Lai AA. Valproate, absorption, distribution and excretion. In: Woodbury DM, Penry JK and Pippenger CE, eds. Antiepileptic drugs. New York: Raven Press, 1982. 200 Fleitman JS, Bruni J, Perrin JH, Wilder BJ. Albumin binding interactions of sodium valproate. J Clin Pharmacol 1980; 20: 314-317. 201 Orr JM, Abbott FS, Farrell K, Ferguson S, Sheppard I, Godolphin W. Interaction between valproic acid and aspirin in epileptic children: serum protein binding and metabolite effects, Chn Pharmacol Ther 1982; 31: 642-649. 202 Schobben F, Vree TB, Van der Kleijn E. Pharmaco~neti~s, metabolism and distribution of di-N-propylacetate (sodium valproate) and the influence of salicylate comedication. In: Meinardi and Rowan, eds. Advances in epileptology 1977, 1978. Amsterdam: Swets and Zeitlinger, 1978; 271-278. 203 D’Arcy PF, McElnay JC. Drug interactions involving the displacement of drugs from plasma protein and tissue binding sites. Pharmacol Ther 1982; 17: 211-220. 204 Powell-Jackson PR. Interaction between azapropazone and warfarin. Br Med J 1977; 1: 1193-1194. 205 MacKichan JJ. Pharmacokinetic consequences of drug displacement from blood and tissue proteins. Clin Pha~aco~net 1984; 9 (Suppl. 1): 32-41. 206 Demon between
DD, Myers JA, Coyle DE. The clinical relevance of the drug displacement interaction meperidine and bupivacaine. Res Comm Chem Path Pharmacol 1984; 45: 323-330.
216 207 Goolkasian glycoprotein
DL, Slaughter RL. Edwards DJ. Lalka D. I~ispIa~~m~nt of lidocaine sites by basic drugs. Eur J Clin Pharmacol 1983: 25: 413-417.
fritm serum n-acid
208 Hartrick CT, Dukes WE, Coyle DE. R~J PP. Demon DD. Influence of bupivacamr on mepivacatne protein binding. Clin Pharmacol Ther 1984: 36: 5466550. 209 Monks A, Richens A. Serum protein binding of vslproic acid and its displacement by palmitic acid in vitro. Br J Clin Pharmacol 1979; 8: 1X7718X. 210 Pate1 IH. Levy RH. Valproic acid binding to human serum albumin and determination ,)I free fraction in the presence of anti~onv~llsants and free fatty acids. Epilepsia 1979: 20: X5-90. 211 Zimmerman CL, Pate1 IH. Levy RH. Nelson SD, Hutchmson M. Protein binding of valproic acid in the presence of elevated free fatty acids in patient and normal human serum. Epilepsia 19X1: 22: 11-17. 212 Bowdle plasma 213 Gugler binding
TA, Pate1 IH, Levy RH, Wilensky AJ. The influence of free fatty acids on valproic acid protein binding durmg fastmg in normal humans. Eur J Clin Pharmacol 1982: 23: 343-347. R. Shoeman DW. Azarnoff DL. Effect of in viva elevation of free fatty acids on protein of drugs. Pharmacology 1974: 12: 160-165.
214 Tsutsumi E, inaha T. Mahon WA. K&w W. The displacing effect of fatty acid on the binding of diazepam to human serum. Biochem Pharmacol 1975; 24: 136ll1362. 215 Desmond PV, Roberta RK. Wood AJJ. Dunn GD. Wilkinson CR. Shenker S. Effects of heparin administration on plasma binding of benzodiazepines. Br J Clin Pharmacol 1980; 9: 171-175. 216 Grossman MI, Moeller HC. Palm L. Effect of lipemia and heparin on free fatty acid concentration of serum in humans. Proc See Exp Biol Med 1955: 90: 106-109. 217 Gia~omini KM. Swezey SE. Giacomini JC. Blasehke TF. Administration of heparin causes in vitro release of non-esterified fatty acids in human plasma. Life Sci 1980; 27: 771-780. 218 Giacomini KM, Giacomini JC, Blaschke TF. Absence of effect of heparin on the hinding of prazosin and phenytoin to plasma proteins. Biochem Pharmacol 1980; 29: 3337.--3340. 219 Schultz P, Giacomini KM, Luttrell S. Turner-Tamiyasu K. Blaschke TF. Effect of low doses of heparin on the plasma binding of phenytoin and prazosin in normal man. Eur J Clin Pharmacol 1983: 25: 211-214. 220 Rimmer EM. Buss DC. Routledge PA. Richens A. Should be routinely phenytoin concentration? Br J Clin Pharmacol 1984; 17: 999102.
measured
free plasma
221 Greenblatt DJ, Sellers EM, Koch-Weser J. Importance of protein binding for the interpretation of serum or plasma drug concentrations. J Clin Pharmacol 1982; 22: 259.-263. 222 Reidenberg MM, Affrime M. Influence of disease on binding of drugs to plasma proteins. Ann NY Acad Sci 1973: 226: 115-126. 223 Wilder BJ. Willmore IJ, Bruni J, Villareal HJ. Valproic acid: interaction with other anti~onvulsant drugs, Neurology 1978; 28: 892-896. 224 Windorfer A. Sauer W. Drug interactions during anticonvulsant therapy in childhood; diphenylhydantoin. primidone, phenoharbitone, clonazepam, nitrazepam, carbamazepine. and dipropylacetate. Neuropaediatrie 1977; 8: 29-41. 225 Bardy A, Hari R. Lehtovaara R. Majuri H. Valproate may lower serum phenytoin. Lancet 1976: 1: 125661257. 226 Windorfer A, Sauer W. Gadeke R. Elevation of diphenylhydantoin and primidone serum concentration by addition of dipropylacetate, a new anticonvulsant drug. Acta Paed Stand 1975: 64: 771-772.