You are on page 1of 13

On the Influence of Protein Binding on Pharmacological

Activity of Drugs

LEONID M. BEREZHKOVSKIY
Genetech Inc., 1 DNA Way, South San Francisco, California 94080

Received 6 July 2009; accepted 26 August 2009


Published online 13 October 2009 in Wiley InterScience (www.interscience.wiley.com). DOI 10.1002/jps.21958

ABSTRACT: The effect of variable protein binding (taken as independent parameter) on


pharmacological activity of drugs is considered in terms of the exposure or the steady
state concentration of unbound drug at targeted tissue. Based on the application of the
parallel tube or dispersion models it is shown that for the most common case of orally
administered drugs eliminated mainly by hepatic metabolism the increase of protein
binding may be beneficial for drug action. In contrary, consideration of this case using
the well-stirred model suggests that changes in protein binding do not influence drug
efficiency. The relatively simplistic well-stirred model appears not accurate enough to
reveal the influence of variation in protein binding on drug exposure. The conclusion in
favor of the predictions based on parallel tube or dispersion models is supported by
experimental data. In case of the oral dosing of drugs that are subjected to nonhepatic
elimination as well as for parenteral drug administration with arbitrary routes of
elimination the decrease in protein binding would lead to the increase of unbound drug
exposure and thus may enhance drug efficiency. An advanced approach to evaluation of
drug activity based on the assumption of the necessity to exceed certain minimal drug
concentration at action site is implied. Such a consideration leads to the conclusion that
there should be an optimal value of protein binding which provides maximum drug
activity. The case when drug action is determined by binding to targeted receptors is
discussed in terms of equilibrium binding and kinetics. ß 2009 Wiley-Liss, Inc. and the
American Pharmacists Association J Pharm Sci 99:2153–2165, 2010
Keywords: protein binding; drug exposure; drug efficiency; bioavailability; well-
stirred model; parallel tube model; dispersion model; tissue–plasma partitioning;
clearance; drug–receptor binding

INTRODUCTION tion as independent parameter), the mechanism of


drug action should be known. A relevant PK/
The influence of protein binding on drug pharma- PD model needs to be developed to study the
cokinetics and pharmacological effects is widely contribution of different factors (including dosing
recognized, and appears to be a common con- regiment) on achieving the optimal drug per-
sideration in drug discovery and development.1 In formance. A simplified consideration of reaching
general, to assess the impact of protein binding on greater exposure, AUCu,t, of the unbound drug in
drug activity (assuming the unbound drug frac- the targeted tissue for a given fixed drug dose may
be applied at the early screening stage of drug
development. The advanced models may also
Correspondence to: Leonid M. Berezhkovskiy (Telephone: consider the drug affinity to the target receptor
650-225-1798; Fax: 650-467-4836;
E-mail: berezhkovskiy.leo@gene.com) and possibly kinetics of drug–receptor binding.
Journal of Pharmaceutical Sciences, Vol. 99, 2153–2165 (2010) The assumption that drug transfer to the
ß 2009 Wiley-Liss, Inc. and the American Pharmacists Association targeted tissue (or organ) is provided by a passive

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 99, NO. 4, APRIL 2010 2153


2154 BEREZHKOVSKIY

diffusion allows reducing the evaluation of obtaining the higher value of AUCu due to the
the compound efficiency to consideration of the variation of the unbound drug fraction fu, may be
exposure, AUCu, of the unbound drug in plasma. assumed as a criterion for the evaluation of the
Indeed, for the passive diffusion from plasma to influence of protein binding on drug efficiency.
tissue, the quantity of drug in tissue, At(t), is Some limitations of this approach are considered
described as in the Discussion section. It is assumed further
dAt ðtÞ that the contribution of possible drug elimination
¼ PS½Cu ðtÞ  Cu;t ðtÞ (1) by the targeted tissue to the total body clearance is
dt
negligible.
where Cu(t) and Cu,t(t) are the concentrations of Drug protein binding depends on the concen-
unbound drug in plasma and tissue extracellular tration of plasma proteins, which can be altered
water, respectively, PS is the product of per- due to various diseases, or, not that often, due to
meability coefficient, P, and the plasma–tissue possible protein binding displacement by another
contact surface area, S. Integrating Eq. (1) over drug (competitive protein binding),4 or due to
time from t ¼ 0 to infinity with the account that relatively high drug concentration.5 It was pub-
At(t ¼ 0) ¼ At(t ! 1) ¼ 0 yields licized (based on the application of well-stirred
AUCu;t ¼ AUCu (2) model) that for the most common case of the orally
administered drugs subjected mainly to hepatic
The common measurements of the total drug elimination, that AUCu is independent of protein
plasma concentration, C(t), and the unbound binding.6 Thus for such a consideration of this
drug fraction in plasma, fu, yield the exposure case, changes in plasma protein binding may have
of unbound drug in plasma little clinical relevance.
AUCu ¼ fu AUC (3) The goal of this article is to show that a more
detailed consideration actually suggests that the
where AUC is the exposure of the total drug increase of protein binding can be beneficial for
in plasma. Since AUCu,t ¼ AUCu (Eq. 2), Eq. (3) drug action (in terms of increasing AUCu) in the
eventually provides the exposure of the unbound particular case of the orally administered drugs,
drug concentration in tissue. Protein binding which are eliminated by hepatic metabolism
equilibration almost always occurs virtually only. It is also shown that for the case of oral
instantaneously, so that Cu(t) ¼ fuC(t) is a usual drug administration and nonhepatic elimination,
assumption.2 Though if binding kinetics is linear and for the case of parenteral administration
and protein bound drug is not eliminated, Eq. (3) and both hepatic or nonhepatic elimination, the
holds even if there is no instantaneous equilibra- decrease in protein binding may enhance the
tion, that is, Cu(t) 6¼ fuC(t) during the time course drug pharmacological activity. It is pointed out
of drug in plasma.3 In case the target tissue that the same conclusions about the influence of
eliminates a drug, or there is efflux transport, an protein binding on drug effectiveness are valid
extra term kVtCu,t is added into the right-hand when a continuous drug administration is con-
side of Eq. (1), where k is the rate constant sidered and the value of the steady state unbound
of the process and Vt is the tissue volume. The drug concentration in plasma, Cu,ss, is taken a
term kVt can be considered as intrinsic tissue marker of drug activity. In addition, the use of
clearance, Clint,t ¼ kVt, corresponding either to apparently more accurate criteria of drug effi-
drug elimination or efflux. Thus Eq. (1) becomes ciency, rather than AUCu, is considered, which
dAt ðtÞ leads to the conclusion that there should be an
¼ PS½Cu ðtÞ  Cu;t ðtÞ  Clint;t Cu;t ðtÞ (4) optimal value of protein binding resulting in
dt
highest drug activity.
Integration of Eq. (4) (similarly as done with Eq. 1)
results in
AUCu DETERMINATION OF DRUG EXPOSURE
AUCu;t ¼ ¼ AUCu ð1  Et Þ (5) IN PLASMA
1 þ Clint;t =ðPSÞ
where Et ¼ Clint,t/(Clint,t þ PS) is the tissue extrac- The total body clearance (calculated with respect
tion ratio. Therefore the exposure of unbound to drug plasma concentration) is defined as
drug in tissue is proportional to the exposure Cl ¼ FD/AUC, where FD is the quantity of drug
AUCu of the unbound drug in plasma. Thus that entered the circulation, D is the drug dose

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 99, NO. 4, APRIL 2010 DOI 10.1002/jps
PHARMACOLOGICAL ACTIVITY OF DRUGS 2155

and F is bioavailability. For the intravenous Hepatic clearance, calculated with respect to drug
administration of dose Div (assuming the absence plasma concentration, is provided as
of the first-pass lung metabolism), D ¼ Div and Clh ¼ rQEh ¼ rQð1  Fh Þ (9)
F ¼ 1, and D ¼ Dpo for the oral administration of
dose Dpo. Consequently the total exposure can be where r is the equilibrium blood–plasma concen-
calculated as AUC ¼ FD/Cl, and according to tration ratio, Q is the rate of liver blood flow, and
Eq. (3) the exposure of unbound drug in plasma is Eh ¼ 1  Fh is the drug hepatic extraction ratio.
For the case of only hepatic elimination, taking
FD
AUCu ¼ fu (6) Cl ¼ Clh in Eq. (8) and applying Eq. (7) for F yields
Cl
Fa Fg Fh Dpo
In general, the total clearance is the sum of AUCu ¼ fu (10)
rQEh
clearances provided by each elimination route
(organ or tissue), and the model of elimination Let us apply the two commonly used models for
organ is applied to obtain its clearance (commonly the calculation of Clh by Eq. (9). According to the
considering linear models and using the steady well-stirred model
state approach7). The clearance value obtained Clint fu
from the steady state condition of eliminating Eh ¼ (11)
rQ þ Clint fu
organs is actually the same as the mean value
D/AUC obtained from the plasma concentration and according to the parallel tube model
time course after intravenous input of dose D. This  
Clint fu
appears to be an intrinsic property of linear Eh ¼ 1  exp  (12)
rQ
pharmacokinetic system. Indeed, at steady state
of the linear system corresponding to iv drug where Clint is the intrinsic hepatic clearance.8
infusion at constant rate R, the steady state Substitution of Eq. (11) for Eh and Fh ¼ 1  Eh in
plasma concentration, Css, is provided by the Eq. (10) yields for a well-stirred model
general equation Css(D/AUC) ¼P R. The rate of Fa Fg Dpo
elimination at steady state, Ess ¼ Ess,i is equal to AUCu ¼ (13)
Clint
the rate of infusion, that is, Ess ¼ R, where Ess,i are
the steady state elimination rates of individual Thus Eq. (13), which does not contain the term fu,
organs. Thus Ess/Css ¼ R/Css ¼ D/AUC, which leads to the conclusion that AUCu is independent
denotes the equality of the mean D/AUC and of protein binding. Therefore the changes in
the
P sum of the steady state organ clearances protein binding have no clinical relevance in this
Ess,i/Css ¼ Ess/Css. case, that is, no dose adjustment is needed to
The oral bioavailability (assuming negligible maintain the same exposure AUCu (and even-
lung first pass effect) is calculated as tually sustain the drug action) to account in
F ¼ Fa Fg Fh (7) regard to the variation of protein binding.6 As
considered in the Discussion section, the inde-
where Fa is the fraction of administered drug that pendence of AUCu on fu is a mathematical artifact
is absorbed into the gut wall, Fg is the fraction of of the equation for clearance given by the well-
the absorbed drug that gets through the gut wall stirred model.
unchanged, and the hepatic bioavailability Fh is On the other hand, if hepatic clearance is
the fraction of drug which subsequently passes obtained from Eq. (12) for parallel tube model, we
unchanged through the liver into the systemic get
circulation. Fa Fg Dpo exp½Clint fu =ðrQÞ
AUCu ¼ fu (14)
rQf1  exp½Clint fu =ðrQÞg
Oral Drug Dosing, Hepatic Elimination For such consideration, Eq. (14) shows a strong
dependence of the exposure AUCu on the value
Let us first consider the common case of oral drug
of unbound drug fraction fu. This is shown in
administration, and hepatic clearance as being
Figure 1A for AUCu calculated using well-stirred
the only route of drug elimination. For oral dosing,
and parallel tube models. AUCu in Figure 1A is
according to Eq. (6)
normalized by the factor N ¼ FaFgDpo/Clint to
FDpo bring data to the same scale, so that according
AUCu ¼ fu (8)
Cl to Eq. (10) AUCu/N ¼ (Clint/rQ)( fuFh/Eh) ! 1 when

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 99, NO. 4, APRIL 2010
2156 BEREZHKOVSKIY

fu ! 0 for both models. Only in the limiting case of


low extraction ratio drug Clintfu << rQ Eq. (14) for
parallel tube model yields the same value of AUCu
as that calculated using well-stirred model
equation (13). For moderate and especially high
extraction ratio drug the change of AUCu pre-
dicted by the parallel tube model is quite
significant. For instance for the drug with
Clint/(rQ) ¼ 10 the decrease of AUCu is 156-fold
when the unbound fraction changes from 0.01
(99% protein binding) to 0.7 (30% protein binding).
Of course, there is a strong dependence of AUCu
on drug intrinsic clearance, which is shown in
Figure 1B, where AUCu/( FaFgDpo/rQ) ¼ fuFh/Eh
(Eq. 10) is plotted. The higher stability (smaller
value of Clint/(rQ)) results in substantial increase
of AUCu.
What is really important, is that AUCu
increases with the increase of protein binding
(decrease of fu). Thus the consideration of parallel
tube model results in the conclusion that for orally
administered drugs which are mainly eliminated
by liver, the increase of protein binding is
beneficial for drug action (in terms of getting
higher exposure AUCu). It means that in drug
development high protein binding should not be a
concern in the case of oral drug administration
and elimination by hepatic metabolism only.
Though (depending on the mechanism of drug
action) a very high protein binding could become
problematic issue, which is considered in the
Figure 1. Case of oral drug administration and Discussion section. In terms of clinical relevance,
elimination by hepatic metabolism. Comparison of the decrease of protein binding due to the
AUCu dependence on the unbound drug fraction fu for reduction of protein content of plasma (related
calculations based on well-stirred (Eq. 13) and parallel to disease) or due to drug–drug interaction would
tube (Eq. 14) models at different values of intrinsic
require to increase the drug dose compared to that
clearance. (Panel A) AUCu is normalized by the
given to subjects with normal levels of fu.
factor N ¼ FaFgDpo/Clint. - - -, Calculation by well-stirred
model (does not depend on Clint/rQ); —, calculation by There is obviously a huge difference in the
parallel tube model: (1) Clint/(rQ) ¼ 0.2, (2) Clint/ prediction of AUCu by the well-stirred and parallel
(rQ) ¼ 0.5, (3) Clint/(rQ) ¼ 1, (4) Clint/(rQ) ¼ 2, (5) Clint/ tube models. The well-stirred model yields
(rQ) ¼ 5, (6) Clint/(rQ) ¼ 10. (Panel B) AUCu is normal- the highest value of AUCu (independent of fu),
ized by the factor K ¼ FaFgDpo/(rQ). - - -, Calculation by which coincides with the prediction of parallel
well-stirred model, —, calculation by parallel tube tube model in two limiting cases fu ! 0 and
model: (1) Clint/(rQ) ¼ 0.2, (2) Clint/(rQ) ¼ 0.5, (3) Clint/ Clint/(rQ) ! 0 (as seen in Fig. 1). The concentra-
(rQ) ¼ 1, (4) Clint/(rQ) ¼ 5. tion of drug is rather changing along the organ (as
given by parallel tube model) than being constant
due to instantaneous uniformed distribution
(well-stirring). Both models are the extreme cases
of a general dispersion model,7 which gives

4a
Fh ¼ (15)
ð1 þ aÞ exp½ða  1Þ=2DN   ð1  aÞ2 exp½ða þ 1Þ=2DN 
2

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 99, NO. 4, APRIL 2010 DOI 10.1002/jps
PHARMACOLOGICAL ACTIVITY OF DRUGS 2157

where RN ¼ fuClint/(rQ) is the efficiency number,


DN is the axial dispersion number, and a ¼
(1 þ 4RNDN)1/2. A well-stirred model corresponds
to DN ! 1 and the parallel tube model corre-
sponds to DN ! 0. Calculation of AUCu by Eq. (10)
using the dispersion model (Eq. 15) with
DN ¼ 0.17 9 is shown in Figure 2. As expected,
the dispersion model provides a significant
dependence of AUCu on the unbound drug
fraction, though less strong than that calculated
based on parallel tube model.
The direct experimental data of Diaz-Garcia
et al.10 on the kinetics of diazepam hepatic
elimination by the isolated perfused rat liver
allowed us to figure out the relevance of the
predictions made based on the well-stirred
and parallel tube models. The values of hepatic Figure 3. Comparison of the experimental data for
bioavailability measured at different levels of the dependence of fuFh/Eh ¼ AUCu/K, K ¼ FaFgDpo/(rQ)
unbound drug fraction fu in perfusate were (Eq. 10) on the unbound drug fraction fu with fuFh/Eh
provided in Table 1 of the article.10 We used calculated by dispersion and parallel tube models. &,
Experimental data of Diaz-Garcia et al.10 —, Calcula-
this data to calculate the factor fuFh/Eh,
tion by dispersion model with Clint/(rQ) ¼ 7.4, DN ¼ 0.18,
which according to Eq. (10) provides AUCu as - - -, calculation by parallel tube model with Clint/
fuFh/Eh ¼ AUCu/[FaFgDpo/(rQ)] (as plotted in (rQ) ¼ 6.7.
Fig. 1B). The experimental results10 shown in
Figure 3 are fitted using the dispersion model agreement with predictions of dispersion model
with Clint/(rQ) ¼ 7.4, DN ¼ 0.18 and the parallel and definitely do not confirm that fuFh/Eh is
tube model with Clint/(rQ) ¼ 6.7. These experi- constant independent of fu as suggested by the
mental data are obviously in reasonably good well-stirred model. The predictions of parallel
tube model are not as accurate as that provided
by the dispersion model, but exhibit the right
tendency in the dependence of fuFh/Eh on fu.
The use of a well-stirred model (which does not
seem quite physiological) both for eliminating
and noneliminating organs in the simulations of
drug pharmacokinetics is dictated by a relative
simplicity of this approach. Each tissue (organ) is
characterized by a single drug concentration in
this case, while for a parallel tube model there
should be a concentration profile along the organ
(assuming one-dimensional consideration, that is,
the existence of instantaneous radial equilibra-
tion8). The current profile is determined by the
concentration of drug in blood that entered
the organ at earlier instants of time. Just a
simple two-compartment model of a well-stirred
Figure 2. Case of oral drug administration and plasma and the eliminating organ considered in
elimination by hepatic metabolism. Comparison of a parallel tube mode yields quite complicated
AUCu dependence on the unbound drug fraction fu for
solution for the plasma concentration–time
calculations based on parallel tube (Eq. 14) and disper-
course.11
sion (Eqs. 10 and 15) models at different values of
intrinsic clearance. AUCu is normalized by the factor
N ¼ FaFgDpo/Clint. - - -, Calculation by dispersion model, Oral Drug Dosing, Nonhepatic Elimination
—, calculation by parallel tube model: (1) Clint/
(rQ) ¼ 0.2, (2) Clint/(rQ) ¼ 1, (3) Clint/(rQ) ¼ 3, (4) Clint/ Let us consider the orally dosed drugs that are not
(rQ) ¼ 10. eliminated by liver. In this case Fh ¼ 1 in Eq. (7)

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 99, NO. 4, APRIL 2010
2158 BEREZHKOVSKIY

for oral bioavailability, so that F ¼ FaFg. We drugs application of the parallel tube model
consider that drug elimination is provided by a (Eq. 23) results in the same equations for the
single organ (most likely kidneys) and the organ exposure (Eqs. 21 and 22) as that obtained using
clearance, Clo, is described either by well-stirred the well-stirred model. The graphs, which show
or parallel tube model, which incorporates plasma the dependence of AUCu on protein binding
protein binding calculated using well-stirred and parallel tube
Clo ¼ rQo Eo (16) models, are shown in Figure 4. Both models
exhibit the same tendency of increasing AUCu
where Qo is the blood flow to the organ and Eo is with decrease of protein binding (slightly more
the organ extraction ratio. For a well-stirred pronounced according to well-stirred model).
model
Clint;o fu
Eo ¼ (17) Parenteral Drug Administration
rQo þ Clint;o fu
To obtain the exposure of a drug administered
and for parallel tube model
  intravenously as bolus dose or as infusion,
Clint;o fu assuming that the first-pass lung bioavailability
Eo ¼ 1  exp  (18)
rQo Flung ¼ 1, according to Eq. (6) we get
where Clint,o is the organ intrinsic clearance. Div
Applying the general equation (8) for AUCu with AUCu ¼ fu (24)
Cl
F ¼ FaFg and Cl ¼ Clo given by Eq. (16), results in
For a drug given by other parenteral route
Fa Fg Dpo
AUCu ¼ fu (19) Div should just be replaced by FparDpar in
rQo Eo the equations of this section, where Dpar is the
If a well-stirred model for elimination organ is parenteral drug dose and Fpar is parenteral
applied, then according to Eqs. (17) and (19), we bioavailability. Considering the elimination by
get either nonhepatic or hepatic route (Eq. 17 or
Eq. 18 depending on model choice) yields the
Fa Fg Dpo ðrQo þ Clint;o fu Þ
AUCu ¼ (20) same equation for AUCu as for the case of
rQo Clint;o
oral administration and nonhepatic elimination
Eq. (20) indicates that the increase of protein (Eqs. 20 and 23), but with FaFgDpo replaced by Div.
binding (smaller fu) leads to the decrease of the Thus for the well-stirred model
exposure AUCu. This is especially pronounce for
Div ðrQo þ Clint;o fu Þ
the high extraction ratio drugs (Clint,ofu rQo), so AUCu ¼ (25)
that AUCu is directly proportional to fu rQo Clint;o

Fa Fg Dpo and for the parallel tube model


AUCu ’ fu (21)
rQo
Div fu
For low extraction ratio drugs (Clint,ofu  rQo) AUCu ¼ (26)
rQo f1  exp½Clint;o fu =ðrQo Þg
Eq. (20) provides
Fa Fg Dpo To calculate AUCu for the case of a drug which
AUCu ’ (22) is eliminated by hepatic metabolism, the para-
Clint;o
meters Qo and Clint,o should be, respectively,
so that AUCu does not depend on protein binding replaced by Q and Clint in Eqs. (25) and (26). Same
in this case. equations for the asymptotic limits of high and low
For consideration of elimination organ by the extraction ratios (Eqs. 21 and 22 with FaFgDpo
parallel tube model Eqs. (18) and (19) yield replaced by Div) are valid for the intravenous
Fa Fg Dpo fu administration.
AUCu ¼ (23) Eventually the conclusion is that the larger
rQo f1  exp½Clint;o fu =ðrQo Þg
value of AUCu is obtained when protein binding
Eq. (23) provides that the increase of protein is lower (larger fu). Figure 4 (for consideration
binding leads to smaller values of the exposure of oral drug administration with nonhepatic
AUCu (same as for well-stirred model). For the elimination) also illustrates the dependence of
limiting cases of high and low extraction ratio AUCu on protein binding for the case of parenteral

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 99, NO. 4, APRIL 2010 DOI 10.1002/jps
PHARMACOLOGICAL ACTIVITY OF DRUGS 2159

more realistic models (dispersion and parallel


tube) show a strong influence of fu on AUCu value.
Interestingly, the well-stirred and parallel tube
models are in a reasonably good agreement in
calculation of hepatic clearance (within 1–1.3-fold
greater values provided by parallel tube model),
though lead to contradictory conclusions on the
prediction of AUCu for the case of orally adminis-
tered drugs which are eliminated mainly by
hepatic metabolism. Such a singularity of well-
stirred model in prediction of AUCu after oral drug
administration and hepatic metabolic elimination
only is actually embedded in the form of Eq. (11)
for the extraction ratio Eh. According to the
general equation (10), for this case AUCu would be
independent of fu if the factor fuFh/Eh were
Figure 4. Case of oral drug administration and non- constant. Taking
hepatic elimination. Comparison of AUCu dependence
on the unbound drug fraction fu for calculations fu Fh fu ð1  Eh Þ
¼ ¼B
based on well-stirred (Eq. 20) and parallel tube Eh Eh
(Eq. 23) models at different values of intrinsic clearance. where B is an arbitrary constant yields
AUCu is normalized by the factor K ¼ FaFgDpo/(rQ).
(1) Clint,o/(rQo) ¼ 0.2, (2) Clint,o/(rQo) ¼ 0.5, (3) Clint,o/ fu =B
(rQo) ¼ 2, (4) Clint,o/(rQo) ¼ 10; – – –, calculation by Eh ¼
1 þ fu =B
well-stirred model, —, calculation by parallel tube
model. When B is taken as rQ/Clint, the equation above
exactly coincides with the equation for Eh
provided by the well-stirred model (Eq. 11), which
drug administration (taking Div instead of can be written as
FaFgDpo in normalization factor).
fu =ðrQ=Clint Þ
Eh ¼
1 þ fu =ðrQ=Clint Þ
DISCUSSION Thus the form of Eq. (11) given by the well-
stirred model for the hepatic extraction ratio Eh
According to the consideration of the previous underlies the independence of AUCu on fu for
section both scenarios that are the increase and a drug given orally and eliminated by liver
decrease of the exposure AUCu with the increase metabolism only.
of fu, are possible depending on the routes of drug If orally administered drug is eliminated mainly
administration and elimination. For oral dosing by nonhepatic routes, the increase of the exposure
AUCu is determined by the quantity of drug that AUCu is expected with the reduction of protein
was absorbed from intestine and got through binding. This is also valid for parenteral drug
the first-pass into systemic circulation, and by the administration regardless the route of elimina-
following elimination. Considering liver as the tion. Both the well-stirred and parallel tube modes
only eliminating organ, the increase of fu leads to are in agreement in prediction of the dependence
the decrease of the first-pass unchanged drug of AUCu on fu in this cases, though a little stronger
quantity, but to the increase of AUCu conse- influence of the increase of fu on the increase of
quently generated by this quantity (as finally AUCu is expected according to well-stirred model
reflected in Eq. 10 by the factor fuFh/Eh). (Fig. 4).
Eventually, as supported by experimental data, The same conclusions about the influence of
the combination of the parameters fu, Fh, and Eh protein binding on drug effectiveness are valid
(as fuFh/Eh) results in the increase of AUCu after when a continuous drug administration is con-
oral administration with the increase of protein sidered and the value of the steady state unbound
binding (smaller fu). The simplest well-stirred drug concentration in plasma, Cu,ss, is taken a
model is not accurate enough to exhibit the change marker of drug activity. Indeed the steady
of AUCu with the change of fu, while apparently state unbound drug concentration in tissue

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 99, NO. 4, APRIL 2010
2160 BEREZHKOVSKIY

Cu,t,ss ¼ Cu,ss (as follows from Eq. 1), or according According to the provided analysis, for the most
to Eq. (4) Cu,t,ss ¼ Cu,ss(1  Et) if the targeted common case of orally administered drugs that
tissue eliminates a drug. In any case the are eliminated mainly due to hepatic metabolism,
steady state unbound drug concentration in the increase of protein binding leads to larger
tissue is proportional to Cu,ss in plasma. Thus to AUCu and thus can be considered beneficial for
maximize Cu,ss could be a goal in developing better drug activity. The consideration of AUCu as the
drug, so that the same concentration level Cu,ss determinant of drug action may not be quite
would be reached with less drug dose. The steady adequate. Most likely there is a certain minimal
state drug plasma concentration, Css, correspond- concentration of unbound drug in the targeted
ing to a continuous constant rate drug adminis- tissue Cu,t,min (and the corresponding value for
tration (considering linear phamacokinetics) is plasma Cu,min) below which the drug activity can
calculated as be assumed negligible. Thus it would be relevant
to consider that the area under the curve,
FI AUCu,t,eff, in the time interval (t1, t2) when Cu,t(t)
Css ¼ (27)
Cl is above Cu,t,min (as shown in Fig. 5A), determines
where I is the rate of continuous drug adminis-
tration (infusion for iv dosing). Same as for a
single dose F ¼ 1 for iv administration (assuming
Flung ¼ 1) and F ¼ FaFgFh for oral dosing. Since
Cu,ss ¼ fuCss, from Eq. (27) we get
FI
Cu;ss ¼ fu (28)
Cl
The right-hand side of Eq. (28) differs from that of
Eq. (6) by just having the infusion rate I instead of
dose D. Therefore the analysis of the factor Ffu/Cl
for the dependence of Cu,ss on fu should be
performed, which is exactly the same as for the
analysis of the dependence of AUCu on fu. Thus
the appropriate equations above will be valid
when AUCu is replaced by Cu,ss and the dose D is
replaced by the rate of continuous administration
I (Div ! Iiv, Dpo ! Ipo).
In theory the parameter fu may be considered as
independent variable. This approach is relevant
for the estimation of the influence of protein
binding changes (due to decrease of plasma
protein content, or interaction with other drug)
on the efficiency of the given drug, that is, for
assessing the clinical relevance of the variation in
protein binding.6 For drug development, though fu
may be considered as independent parameter, this
is not technically feasible. If some modification of
the potentially active compound is done to alter its
protein binding, most likely it will be reflected in Figure 5. Same unbound drug exposure in tissue
the changes of other parameters that determine (or plasma) may or may not be sufficient for drug
physiological activity. (Panel A) Exposure of the
AUCu (or Cu,ss), which are Fa, Fg, Clint,o and r.
unbound drug in tissue only at concentrations Cu,t(t),
The biological activity of the compound can
which are above certain minimal value Cu,t,min, may
possibly change as well. Eventually the advantage determine the drug action. (Panel B) Unbound drug
provided by improved protein binding can be concentration in tissue Cu,t(t) does not exceed the mini-
cancelled out by the changes in other parameters mal value Cu,t,min, so that the drug may not be active
(most likely Fg and Clint,o). This makes the process regardless the same exposure in tissue AUCu,t (or in
of optimizing protein binding very complicated. plasma AUCu) as in Panel A.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 99, NO. 4, APRIL 2010 DOI 10.1002/jps
PHARMACOLOGICAL ACTIVITY OF DRUGS 2161

the drug action fu  VpCu,t,min/(FDpo)), the drug unbound concen-


tration in tissue Cu,t(t) will be below Cu,t,min value,
Zt2
as shown in Figure 5B. This will be the case when
AUCu;t;eff ¼ Cu;t ðtÞ dt (29) a strong protein binding makes a drug inefficient
t1 (AUCu,t,eff ¼ 0 in Eq. 29 or in Eq. 30), though AUCu
will be very large. In other words protein binding
Depending on mechanism of drug activity it could
limits the drug distribution to the site of activity,
be more adequate to define AUCu,t,eff as
though sustain the low concentration there for a
Zt2 long time. Eventually a large value of AUCu will
AUCu;t;eff ¼ ðCu;t ðtÞ  Cu;t;min Þ dt (30) not be sufficient for drug action. In terms of
t1
continuous administration of drug in this case,
the high steady state concentration Cu,t,ss will be
achieved (assuming that linear pharmacokinetics
The case when during the drug time course approach is not violated, so that Eqs. 27 and 28 are
Cu,t(t) < Cu,t,min indicates the lack of drug pharma- valid). Though it will take a very long time to
cological activity. AUCu,t,eff is nonexistent in this reach the steady state concentration in plasma
case (i.e., t1 and t2 are not found), and can be and possibly even much longer in tissue.12
accepted as AUCu,t,eff ¼ 0. Using this criterion Practically this is not very convenient, and such
(Eq. 29 or Eq. 30) for evaluation of drug efficiency property of a drug is not desirable. In general, the
makes the analysis quite complicated. It would calculation of drug concentration–time course in
require the knowledge of the drug time course tissue is needed for an advanced consideration of
Cu,t(t) in the targeted tissue, so that a detailed drug efficacy (i.e., using AUCu,t,eff), which is
pharmacokinetic model needs to be developed to discussed further.
obtain the concentration time curves, rather than The assumption of the necessity of reaching
using the integrated values of areas under the certain minimal concentration of drug at action
curves. An adequate determination of the minimal site leads to the conclusion that the rate of drug
concentration Cu,t,min (based on the mechanism of absorption after oral dosing may be crucial for
drug action) seems also quite complicated. This drug action. Indeed, if absorption is relatively
parameter may strongly influence the outcome of slow, the concentration of drug in plasma C(t) may
AUCu,t,eff calculations, especially when the max- not be high enough to provide the tissue concen-
imum of Cu,t(t) and Cu,t,min are of the same order of tration Cu,t(t) above Cu,t,min (especially if drug
magnitude. Apparently AUCu,t,eff would charac- elimination is relatively fast). The area under the
terize the dependence of drug activity on the given curve AUCu could be relatively large due to a long
dose more accurately than AUCu,t (which is just drug time course (dictated by slow absorption),
dose proportion for a linear pharmacokinetics). but not efficient for drug action since the condition
AUCu,t,eff would be especially dose sensitive at Cu,t(t) > Cu,t,min will not be met. The total absorbed
relatively low levels of administered dose when quantity of drug is taken into account by Eq. (10)
the highest values of Cu,t(t) 0 Cu,t,min. through the integrated value of AUCu (which is
In case of oral dosing of a highly protein bound reflected by oral bioavailability F ¼ FaFgFh).
drug ( fu ! 0) which is stable in the blood stream, Eq. (10) though does not provide the information
the elimination and tissue distribution are quite on the kinetics of absorption in terms of the
slow, so that (assuming relatively fast absorption) plasma concentration levels C(t) during the drug
the concentration of drug in plasma may be close time course. In other words the same value of
to maximum possible value of Cmax ¼ FDpo/Vp, AUCu may be achieved through different concen-
where Vp is the plasma volume. If this drug is tration profiles C(t), which depend on drug
eliminated by hepatic metabolism only, the absorption kinetics (though having not only the
value of AUCu will be reaching the maximum same value of protein binding fu, but even the
value (Fig. 1). The unbound drug concentration in same values of all parameters in the right-
plasma Cu(t) will be limited by the maximum hand side of Eq. 10). Eventually the condition
possible value fuCmax. According to Eq. (1) the Cu,t(t) > Cu,t,min may or may not be met. Thus the
unbound drug concentration in tissue Cu,t(t)  advanced consideration of possible criteria of
Cu(t), so that Cu,t(t)  fuCmax ¼ fuFDpo/Vp. Then for drug pharmacological activity (Eq. 29 or Eq. 30)
the values of fu that at any instant yield indicates that the rate of drug absorption followed
Cu,t(t)  Cu,t,min, that is, fuFDpo/Vp  Cu,t,min (or by the oral dose should be taken into account and

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 99, NO. 4, APRIL 2010
2162 BEREZHKOVSKIY

may become a limiting step in developing better time that a drug molecule would spend in
drug. the tissue extracellular water in the unbound
In terms of protein binding, for an orally state. Similarly, reaching the largest value of
administered drug that is eliminated by liver AUCu,t,eff (Eq. 29) for best pharmacological effect
metabolism only, a strong protein binding corresponds to the achieving the longest average
could be beneficial for obtaining higher levels of time that a drug molecule would spend as
AUCu (Fig. 1), but results in low concentration unbound in the tissue extracellular water when
levels in tissue (Cu,t(t) < Cu,t,min when fu ! 0). This concentrations Cu,t(t) are above Cu,t,min.
is the case when a strong protein binding The action of drug may occur due to the
limits the delivery of a drug to the site of activity. interaction of the free drug in tissue with the
On the other hand a week protein binding targeted receptors. This interaction is often
would lead to smaller AUCu but higher levels of described in terms of the equilibrium of a simple
drug in tissue (possibly above Cu,t,min, so that binding reaction
a drug would be pharmacologically active).
Therefore there should be an optimal value or D þ T ¼ DT (31)
interval of fu (considering it as independent
where D denotes the unbound drug in tissue
parameter) that would result in the best drug
(which has the concentration Cu,t(t)), T denotes
performance.
the targeted receptor, and DT is the drug–
For drugs administered intravenously a weak
target complex. The equilibrium dissociation
protein binding is beneficial for obtaining larger
constant Kd determines the concentration of
values of AUCu (Eqs. 25 and 26). In this case a
bound receptor at equilibrium. According to the
drug is eliminated relatively fast (compared to the
mass action law
case of strong protein binding). Thus it may turn
out that reaching the targeted tissue may be Cu;t ðTo  Tb Þ
limited by a slow drug transfer (combined with Kd ¼ (32)
Tb
fast elimination), so that in the worst case Cu,t(t)
may not reach Cu,t,min (though AUCu, as well as where To is the total receptor concentration, Tb is
AUCu,t, would be relatively large, as shown in the concentration of drug–receptor complexes
Fig. 5B). On the other hand a strong protein DT (bound receptors), so that To  Tb is the
binding leads to smaller values of AUCu, but concentration of free receptors. Thus for the
prolongs the duration of drug time course, target occupancy Tb/To (which is time dependent)
which eventually may yield higher concentration Eq. (32) yields
levels in targeted tissue. Therefore the unbound
drug fraction fu could have an optimal value (or Tb ðtÞ 1
¼ (33)
interval) that provides the highest drug efficiency. To 1 þ Kd =Cu;t ðtÞ
The average time a drug molecule spends
as unbound in the tissue extracellular water, For example, it can be assumed, based on
MRTt,u, is calculated as13 the drug action mechanism, that the drug is
Z1 Z1 physiologically active when at least half of
1 Vt;w the targeted receptors are occupied (50% occu-
MRTt;u ¼ Au;t ðtÞ dt ¼ Cu;t ðtÞ dt
D D pancy), that is, when Tb/To  0.5. According
0 0 to the equation above this takes place when
Vt;w AUCu;t Cu,t(t)  Kd. Thus for such a consideration
¼
D the border line Cu,t,min in Figure 5 will be
where Au,t(t) is the quantity of unbound to drug in Cu,t,min ¼ Kd.
tissue extracellular water at instant t, and Vt,w is In general, to find the time interval (t1, t2)
the volume of tissue extracellular water. As in Figure 5A and perform further integration to
considered previously AUCu,t ¼ AUCu (Eq. 2) or find AUCu,t,eff (Eq. 29 or Eq. 30) requires the
AUCu,t is proportional to AUCu (Eq. 5). This leads knowledge of the drug time course Cu,t(t) in tissue.
to the conclusion that AUCu is proportional to the This can be obtained from Eq. (4) taking
average time MRTt,u that a drug molecule spends At(t) ¼ Cu,t(t)Vt/fu,t, where Vt is the tissue volume
in the tissue extracellular water as unbound. and fu,t is the unbound drug fraction in tissue,
Thus the requirement to maximize AUCu actually which can be calculated based on physico-
corresponds to the achieving the longest average chemical properties of a drug.14 Then Eq. (4)

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 99, NO. 4, APRIL 2010 DOI 10.1002/jps
PHARMACOLOGICAL ACTIVITY OF DRUGS 2163

becomes where the concentration profile Cu,t(t) may be


dCu;t ðtÞ ðPS þ Clint;t Þfu;t provided by Eq. (34). Depending on the mechan-
þ Cu;t ðtÞ ism of drug action, it could be more appropriate to
dt Vt
perform integration in Eqs. (35) and (36) in the
PSfu;t time interval (t1, t2) when Cu,t(t)  Cu,t,min. The
¼ Cu ðtÞ (34)
Vt larger value of Cu,t(t)/Kd in Eqs. (33) and (36)
This type of equations has a general solution for results in a larger value of receptor occupancy
Cu,t(t) for the known (measured) plasma time- Tb(t)/To. This allows to simplify the criterion of
concentration profile Cu(t).15 For instance, if the drug effectiveness and instead of Eq. (36) use the
unbound drug concentration in plasma is given following expression
as Cu(t) ¼ a exp(at) þ b exp(bt), the solution of Z1
Eq. (34) (with initial condition Cu,t(t ¼ 0) ¼ 0, that Cu;t ðtÞ dt AUCu;t
¼
is, a drug is not administered directly into the Kd Kd
0
tissue) is
 at With the account of Eq. (5), we obtain from the
ae b ebt equation above AUCu,t/Kd ¼ AUCu(1  Et)/Kd. The
Cu;t ðtÞ ¼ k1 þ advantage of this simplification is that we can just
k2  a k2  b
  use the integrated value of exposure in plasma
a b AUCu for drug evaluation. Eq. (36) would be more
 þ ek2 t 
k2  a k 2  b relevant for the estimation of drug activity, but it
requires the knowledge of drug time course in
where k1 ¼ PSfu,t/Vt and k2 ¼ (PS þ Clint,t)fu,t/Vt. tissue, which is not routinely available. In most
Such in vivo based pharmacokinetic model was cases the targeted tissue does not eliminate a drug
successfully applied to interpret the drug time (i.e., Et ¼ 0), so that the calculation of
course of compounds in brain and find the rate AUCu AUCu;t
constants of blood–brain transfer.12 Thus having ¼ (37)
Kd Kd
the drug plasma concentration–time curve
Cu(t) ¼ fuC(t), the time course of drug in tissue as a key factor of drug action, which accounts for
can be obtained by Eq. (34) and used to calculate both the exposure and the affinity for receptor,
AUCu,t,eff in the interval Cu,t(t)  Cu,t,min (Eq. 29 or appears to be pertinent for the case when drug
Eq. 30). In case when the targeted tissue does not action is provided by the binding to the targeted
eliminate a drug, Clint,t ¼ 0 should be used in receptors. To apply Eq. (37) for the evaluation
Eq. (34). The problem is that the parameter PS/Vt of drug action rather than just considering AUCu
is not readily available except for the tissue that is obviously more relevant. In other words, if
can be considered in terms of well-stirred perfu- two compounds were compared and had the
sion limited physiologically based pharmacoki- same exposure AUCu, the one with higher affinity
netic model. to receptor (smaller Kd) would appear to have
In case of drug action being determined by drug better potential since it had larger value of
binding to the targeted receptors in tissue, it AUCu/Kd.
appears that the integrated occupancy level, L, Obviously using AUCu ¼ fuAUC as a criterion
defined as for the evaluation of drug action in terms of the
exposure at activity site is very convenient as it
Z1
Tb ðtÞ requires just a commonly measured drug plasma
L¼ dt (35)
To concentration–time course and its protein bind-
0 ing. A more detailed consideration (Eq. 36 or
could be a more adequate criterion of drug action Eq. 37, or possibly using some other equations
than AUCu. The larger value of L would be an based on pharmacodynamic model of drug action)
indication of better drug efficacy. According to could be more appropriate for the late stage
Eq. (33) the integrated occupancy level can be of drug development. Drug binding to the
expressed as receptor may not necessarily be very fast, so that
binding kinetics (rather than instantaneous
Z1
dt equilibrium, Eq. 33) could be considered for the
L¼ (36) calculation of target occupancy in this more
1 þ Kd =Cu;t ðtÞ
0 intricate case.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 99, NO. 4, APRIL 2010
2164 BEREZHKOVSKIY

The occupancy of the receptors at equilibrium is REFERENCES


determined by the affinity Kd. It is suggested that
a longer average time interval, tb, that a drug 1. Trainor GL. 2007. The importance of plasma pro-
molecule spends bound to the receptor may be tein binding in drug discovery. Expert Opin Drug
beneficial for drug action.16 Compounds may have Metab Toxicol 2:51–64.
the same values of affinity to the receptor, but 2. Berezhkovskiy LM. 2008. Some features of the
different values of tb. The affinity is the ratio of kinetics and equilibrium of drug binding to plasma
proteins. Expert Opin Drug Metab Toxicol 4:1479–
the reaction rate constants, that is, Kd ¼ koff/kon,
1498.
where and kon and koff are respectively the on- and
3. Berezhkovskiy LM. 2004. Determination of
off-rate (dissociation rate) constants. The average volume of distribution at steady state with complete
bound time is determined by the off-rate constant consideration of the kinetics of protein and tissue
and is calculated as binding in linear pharmacokinetics. J Pharm Sci
93:364–374.
1 4. Christensen H, Baker M, Tucker GT, Rostami-
tb ¼ (38)
koff Hodjegan A. 2006. Prediction of plasma protein
binding displacement and its implications for
At equilibrium the average time, tu, between each quantitative assessment of metabolic drug-drug
binding to the receptor (‘‘unbound time’’) can be interactions from in vitro data. J Pharm Sci 95:
found using the interpretation of unbound and 2778–2787.
bound drug fractions in terms of kinetics, so that 5. Berezhkovskiy LM. 2009. Consideration of the
tb/tu ¼ Tb/Cu,t. Using Eqs. (33) and (38) we get linear concentration increase of the unbound drug
fraction in plasma. J Pharm Sci 98:383–393.
6. Benet LZ, Hoener B. 2002. Changes in plasma
ð1 þ Cu;t =Kd Þ
tu ¼ protein binding have little clinical relevance. Clin
kon To Pharm Ther 71:115–121.
7. Roberts MS, Rowland M. 1986. A dispersion model
of hepatic elimination: 2. Steady-state considera-
In terms of achieving longer bound time tb for the tions–influence of hepatic blood flow, binding
same value of affinity Kd ¼ koff/kon, the better within blood, and hepatocellular enzyme activity.
compound would have smaller value of koff (as Pharmacokinet Biopharm 14:261–288.
provided by Eq. 38), as well as smaller value of 8. Roberts MS, Rowland M. 1986. A dispersion model
the on-rate constant kon. Thus to find the best of hepatic elimination: 1. Formulation of the
compound it terms of optimal equilibrium and model and bolus considerations. Pharmacokinet
kinetic features would include both the reaching Biopharm 14:227–260.
of higher exposure–affinity ratio AUCu/Kd 9. Roberts MS, Rowland M. 1986. Correlation between
(or integrated occupancy, Eq. (36), for a more in-vitro microsomal enzyme activity and whole
organ hepatic elimination kinetics: Analysis with
elaborate consideration) along with possibly
a dispersion model. J Pharm Pharmacol 38:177–
smaller value of dissociation rate constant of
181.
drug–receptor complex. 10. Diaz-Garcia JM, Evans AM, Rowland M. 1992.
It appears that there is also a problem in finding Application of the axial dispersion model of hepatic
the quantitative criteria that could adequately drug elimination to the kinetics of diazepam in the
reflect the influence of bound time tb on drug isolated perfused rat liver. J Pharmacokinet Bio-
action. As mentioned earlier, in theory it is pharm 20:171–193.
possible to consider the unbound drug fraction 11. Berezhkovskiy LM. 2009. Prediction of the pos-
(protein binding) as independent variable that can sibility of the secondary peaks of iv bolus drug
be optimized to achieve better drug action. plasma concentration time curve by the model
Though most likely that the change of fu would that directly takes into account the transit
time through the organ. J Pharm Sci, in Early
result in the change of bound time tb. Therefore
View.
there should be suggested a criterion that
12. Berezhkovskiy LM. 2007. On the determination of
adequately accounts for the influence on drug the time delay in reaching the steady state drug
performance of both the integrated receptor concentration in the organ compared to plasma.
occupancy and the average time a drug molecule J Pharm Sci 96:3432–3443.
resides on the receptor. This appears to be a 13. Berezhkovskiy LM. 2009. Determination of mean
formidable problem to approach in terms of both residence time of drug in plasma and the influence
physiological and mathematical considerations. of the initial drug elimination and distribution on

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 99, NO. 4, APRIL 2010 DOI 10.1002/jps
PHARMACOLOGICAL ACTIVITY OF DRUGS 2165

the calculation of pharmacokinetic parameters. 15. Korn GA, Korn TM. 1961. Mathematical handbook
J Pharm Sci 98:748–762. for scientists and engineers. New York: McGraw-
14. Rodgers T, Rowland M. 2006. Physiologically Hill, Inc.
based pharmacokinetic modelling 2: Predicting 16. Copeland RA, Pompliano DL, Meek TD. 2006.
the tissue distribution of acids, very weak bases, Drug-target residence time and its implications
neutrals and zwitterions. J Pharm Sci 95:1238– for lead optimization. Nat Rev Drug Discov 5:
1257. 730–739.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 99, NO. 4, APRIL 2010

You might also like