Sunteți pe pagina 1din 20

International Journal of Pharmaceutics 201 (2000) 131 – 150

www.elsevier.com/locate/ijpharm

Review

Biopharmaceutics and pharmacokinetics in drug research


Ramesh Panchagnula *, Narisetty Sunil Thomas
Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar,
Punjab 160 062, India

Received 7 September 1999; received in revised form 6 January 2000; accepted 6 January 2000

Abstract

With the synergistic and multiplicative interactions of rational drug design, recombinant biotechnology, combina-
torial chemistry and high-throughput screening, millions of compounds are being synthesized by chemists. However,
development of these drug candidates has often been impeded, if not terminated, due to biopharmaceutic and/or
pharmacokinetic constraints. This has resulted in delays in development time and escalation of cost in the drug
research programmes. So, the present emphasis is to reduce development time and cost, which is analogous to added
patent life besides the enormous reduction in human suffering. In this compilation the important biopharmaceutic
and pharmacokinetic approaches are discussed, which will help in the development of safe and more efficacious drugs
with reduced development time and cost. © 2000 Elsevier Science B.V. All rights reserved.

Keywords: New drug development; Biopharmaceutics; Pharmacokinetics; Solubility; Permeability; Lipophilicity

1. Introduction lead molecules, screening of new lead molecules


for in vitro and/or in vivo biological activities,
Drug research is a unique multi-disciplinary and physicochemical characterization of leads.
process heading towards the development of novel Drug development focuses on evaluation of
therapeutic agents in areas of currently unmet safety/toxicity and efficacy of new drug molecules.
medical need. The drug research can be divided The key objective of drug development is the
functionally into two stages: discovery/design and generation of a scientific database that supports
development (Fig. 1). Drug discovery/design con- the effectiveness and safety profile of the dosage
sists of identification and characterization of new regimen(s) intended for marketing. On an average
targets (enzymes or receptors), synthesis of new every new drug molecule requires 12–15 years to
reach the patient and costs a staggering amount
of US$ 400–650 million (Collins et al., 1999).
* Corresponding author. Tel: + 91-172-673848; fax: +91-
172-677185. Although new methods, such as pharma-
E-mail address: niper@chd.nic.in (R. Panchagnula) cophore theory and molecular modeling, are be-

0378-5173/00/$ - see front matter © 2000 Elsevier Science B.V. All rights reserved.
PII: S 0 3 7 8 - 5 1 7 3 ( 0 0 ) 0 0 3 4 4 - 6
132 R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150

Fig. 1. Various stages of drug research.

ing used to assist rational drug design, the classi- Some data on reasons for withdrawal of
cal method of choosing potential drugs for drug candidate drugs from development has been
development in the pharmaceutical industry still published by the Center for Medicines
relies on an evaluation of pharmacodynamic ac- Research (Prentis et al., 1988), which says that
tivity in vitro and/or in animal models. However, nearly 40% of the drugs (78 of 198 compounds)
majority of the drug molecules fail in subsequent were terminated from further development due
drug development program because the efficacy to unsatisfactory pharmacokinetics (Table 1).
and safety are not governed by its pharmacody-
namic characteristics alone. It also depends to a
Table 1
large degree on the biopharmaceutical (e.g. solu- Reasons for failure in drug development (data obtained from
bility, stability, permeability and first pass effect) Prentis et al., 1988) (%)
and pharmacokinetic (clearance rate, biological
half-life, extent of protein binding and volume of Poor biopharmaceutical properties 40
Lack of efficacy 30
distribution) properties of the drug, since these Toxicity 21
properties control the rate and the extent to which Commercial reasons 8
the drug can reach its site of action, i.e. biophase.
R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150 133

Table 2 involved to elicit the response (Ritschel, 1992a).


Interdisciplinary scheme of LADMER system
One can subdivide this approach according to the
scheme shown in Table 2 (Ritschel, 1992b ).

3. Biopharmaceutics in drug development

Biopharmaceutics is a major branch of the


pharmaceutical sciences concerned with the rela-
tionship between the physicochemical properties
of a drug in dosage form and the pharmacologic,
toxicologic, or clinical response observed after its
administration (Gibaldi, 1991). In short, biophar-
maceutics mainly involves drug absorption pro-
cess. Systemic absorption of most drug products
consists of a succession of rate processes viz.: (i)
disintegration of the product; (ii) dissolution of
the drug in an aqueous environment; and (iii)
absorption across cell membranes into the sys-
temic circulation and, ultimately, to its site of
action. Several key hurdles such as aqueous solu-
Whereas poor pharmacokinetics were solely re-
bility, stability, permeability, and first pass effect,
sponsible for nearly all (90%) terminations of
must be overcome so as to get a drug from a
anti-infective drugs. Hence understanding of these
dosage form to its site of action (Fig. 2). These
properties is essential to all scientists involved in
hurdles vary in how easily they can be overcome
drug discovery and development.
by formulation efforts (Lipper, 1999). When it
becomes evident that a potential drug candidate
has poor biopharmaceutic properties, due consid-
2. Events that lead to in vivo drug action eration must be given to its developability.
Several mathematical models have been pro-
To elicit in vivo response, the drug must reach posed (Goodacre and Murray, 1981; Ho et al.,
the biophase, crossing several biomembranes. 1983; Dressman et al., 1985; Dressman and
Many factors are responsible for the entry of a Fleisher, 1986; Macheras and Symillides, 1989;
drug into the body and then into biophase. These Sinko et al., 1991; Oh et al., 1993) to estimate oral
factors include the route of administration; the absorption/bioavailability. A simple dimensionless
dosage form; the liberation rate of the drug from number, absorption potential (AP) proposed by
the dosage form; dissolution; penetration and per- Dressman et al. (1985) appears to correlate quan-
meation of the drug through biomembranes; its titatively with fraction absorbed (Macheras and
distribution within the body fluids and tissues; the Symillides, 1989) as well as identify the critical
type, amount and rate of biotransformation; and limiting physicochemical property of poorly ab-
recycling processes and elimination. In addition to sorbed compounds. However, as the AP is mainly
these factors, pharmacogenetics and certain concerned with the physicochemical properties of
pathophysiological conditions also affect the the drug it cannot be used as the sole indicator of
above process. The entire process can be de- bioavailability. Whereas, the mathematical models
scribed as the LADMER (liberation, absorption, of Sinko et al. (1991) and Oh et al. (1993) demon-
distribution, metabolism, elimination and re- strated good correlations between in vitro dissolu-
sponse) system showing that liberation, absorp- tion and in vivo bioavailability. These models
tion, distribution, metabolism and elimination are point out that the key parameters controlling
134 R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150

drug absorption are three dimensionless numbers; consuming much of time and resources, one may
an absorption number (An), a dissolution number go ahead with formulation modifications. Chemi-
(Dn) and dose number (Do); representing the fun- cal modification of structure of the lead com-
damental processes of membrane permeation, pound is the other strategy to overcome
drug dissolution and dose, respectively (Amidon biopharmaceutical hurdles.
et al., 1995). By calculating all these dimensionless As a result of combinatorial chemistry and high
numbers for a new molecule and by referencing in throughput screening millions of compounds are
contour plots (Oh et al., 1993), an estimate of the getting added to the library. In selecting the right
extent of drug absorption can be made. However, candidates from the database, for further develop-
all these numbers are related to two important ment, many major pharmaceutical companies
parameters controlling drug absorption, i.e. solu- have taken novel approaches that consider bio-
bility and permeability (Amidon et al., 1995). pharmaceutic properties like solubility, permeabil-
Based on these two parameters Biopharmaceutic ity and other physicochemical parameters. In this
Drug Classification Scheme (BCS) has been pro- section more emphasis is given to important bio-
posed by Amidon et al. (1995), which happens to pharmaceutic parameters that affect its absorp-
be an important tool in developing a suitable tion and/or bioavailability. The various
strategy for improving the bioavailability of new biopharmaceutic factors affecting dissolution, ab-
chemical entities (Fig. 2). If formulation efforts sorption and/or bioavailability of drugs are listed
can overcome the biopharmaceutic hurdle without in Table 3.

Fig. 2. LADMER system, biopharmaceutic hurdles in drug development, approaches to overcome them.
R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150 135

Table 3 The aqueous solubility of solid compounds is


Biopharmaceutic factors affecting dissolution and/or absorp-
governed by interactions between molecules in the
tion
crystal lattice, intermolecular interactions in the
Physicochemical characteristics of the drug solution, and the entropy changes accompanying
Solubility fusion and dissolution. The observed mole frac-
Partition coefficient, pKa tion solubility of any solute Xw is related to its
Dissolution rate
ideal mole fraction solubility, Xi, and the activity
Salt formation
Prodrugs coefficient in water, gw, as shown in Eq. (1).
Particle size, surface area and shape log Xw = log Xi − log gw (1)
Crystallinity, polymorphism Eq. (1) clearly says that both crystalline structure
Stereochemical factors as well as the activity coefficient can contribute to
Pharmaceutical factors the insolubility of a solute (Yalkowsky, 1981).
Factors related to formulation (excipients) Therefore, solubility of poorly soluble solute can
Galenical/dosage form be increased by two approaches. One is by in-
Manufacturing process
creasing its ideal solubility by chemical modifica-
Stability and storage of drug and drug product
tions or solid state manipulations, and the second
Physiological factors affecting bioa6ailability
approach is to decrease its activity coefficient to
Permeability
GI transit/motility
unity by formulation changes. First approach in-
Site specific absorption cludes introduction of polar or ionizable groups
Preabsorptive metabolism or introduction of groups that decrease melting
Hepatic metabolism point (Amidon, 1981) or polymorphic alterations.
Biliary excretion
The second approach includes micellar solubiliza-
Renal excretion
Protein and tissue binding tion, cosolvency, hydrotrophy, and formation of
inclusion complexes.
The development of various peptidomimetics
3.1. Solubility like HIV protease inhibitors and fibrinogen recep-
tor (GP IIb/IIIa) antagonists are good examples
Solubility is an important determinant in drug that illustrate the concept and importance of drug
liberation and absorption and hence plays a key solubility in drug selection and drug development.
role in its bioavailability. For a drug to be ab- A variety of compounds that display excellent
sorbed it must be present in the form of an inhibition of HIV-protease have been designed by
aqueous solution at the site of absorption. This is molecular modeling. However, most of these re-
true regardless of the mechanism of absorption, tain peptidic nature (Bohacek et al., 1996) and
whether it be passive diffusion, convective trans- hence, have poor biopharmaceutic and pharma-
port, active transport, facilitated transport or ion cokinetic properties such as low aqueous solubil-
pair transport, except in the case of pinocytosis, ity/high lipophilicity, high molecular weight,
which is very rare and applies only to the fat-solu- susceptibility to proteolytic degradation, high
ble vitamins A, D, E, K, and some lipids. If the hepatic metabolism and biliary excretion, charac-
drug is insoluble or very poorly soluble it poses a terized by low oral absorption and rapid elimina-
problem of dissolution and/or absorption, since tion (Kempf et al., 1995). The identification of
the flux of drug across an intestinal membrane is HIV protease inhibitors with optimal oral
proportional to its concentration gradient be- bioavailability, high potency and selectivity there-
tween the apical side and basolateral sides of GI fore represents a critical milestone. In order to
lumen. Hence, the aqueous solubility of drugs is improve oral bioavailability, it may be necessary
of central importance in the process of drug dis- to change physicochemical properties such as sol-
covery and development, from molecular design ubility by designing modifications in non-pharma-
to pharmaceutical formulation and bio- cophore regions (first approach). The following
pharmaceutics. examples best illustrate this point.
136 R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150

Vacca et al. (1991) initially developed a series of creased aqueous solubility (Fig. 3). Indinavir sul-
hydroxyethylene dipeptide isosteres, represented fate is the clinical formulation, because of
by L-685,434, and were found to be highly potent improved aqueous solubility (\ 450 mg/ml) and
and selective HIV protease inhibitors. Although consistent bioavailability (Dorsey et al., 1994).
they are highly potent and selective, the main Development of ritonavir is another such example
drawback was that they lacked aqueous solubility where solubility approach had been applied in the
and an acceptable pharmacokinetic profile, result- drug development (Kempf et al., 1991, 1995).
ing in poor bioavailability (Thompson et al., Another illustrative example was reported by El-
1992). The efforts made to increase the solubility dred et al. (1994). They synthesized a series of
by incorporating a basic amine (replacement of benzamidine containing fibrinogen receptor (GP
tert-butyl carbamate and Phe moieties with dec- IIb/IIIa) antagonists. But the highly potent com-
ahydroisoquinoline tert-butylamide) into the pounds when dosed orally to marmoset showed
backbone of this series, led to the development of very low oral activity and was assumed to be due
a novel class of hydroxylamine pentanamide to poor absorption that in turn was claimed to be
(HAPA) isosteres, represented by L-704,486, with the result of their poor aqueous solubility. When
a favorable oral pharmacokinetics but its efficacy the solubility of these compounds was increased
was diminished. When decahydroisoquinoline by introducing piperazine ring, the oral activity
tert-butylamide group was replaced with two tert- was improved attesting the importance of good
butyl carboxamide 4-substituted piperazines, the aqueous solubility for drug absorption.
basic amine improved aqueous solubility and N4 One of the techniques commonly used to over-
gave a chance for further modifications that could come the problems of poor and erratic
balance hydrophilic and hydrophobic require- bioavailability is the prodrug approach, wherein
ments. The 3-pyridyl methyl substitution at N4 the physicochemical properties of the drug are
(lead to the discovery of L-735,524/Indinavir) improved by bioreversible chemical alteration.
provided both lipophilicity for binding to the The most common prodrug strategy involves the
target and a weakly basic nitrogen further in- incorporation of a polar or ionizable moiety into

Fig. 3. Chemical structures of L-685,434, L-704,486, and indinavir.


R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150 137

the parent compound to improve aqueous solubil- be investigated. Often a useful approach to in-
ity (Stella et al., 1998). In ‘ad hoc prodrug ap- crease the aqueous solubility of an ionizable drug
proach’ the target activity is optimized first is pH adjustment. The next approach most fre-
irrespective of pharmacokinetic properties and quently tried is the use of water-miscible cosol-
once the target activity is demonstrated, the phar- vents. Other approaches include the use of
macokinetic properties are optimized by prodrug surface-active agents, complexing agents, lipo-
design without altering the pharmacophore in the somes, salting-in, salting-out, clathrate formation.
active chemical (Testa and Caldwell, 1996). Such Complexation of water insoluble drugs usually
an approach may well prove most useful or even involves the incorporation of the drug within the
indispensable in the development of therapeutic inner core of the complexing agent so that the
peptides and antisense drugs. Though the applica- outer hydrophilic groups of the complexing agent
tion of this approach in drug research is uncom- interacts with water, rendering the drug more
mon, it was successfully applied in the soluble. Cyclodextrins are the commonly used
development of enalapril, an angiotensin convert- complexing agents that form inclusion complexes
ing enzyme inhibitor. The ‘post hoc prodrug ap- in both the solution and solid states. For example,
proach’ (prodrugs of established drugs) has been the solubility of miconazole in water increased to
successfully used to improve water solubility of nine- to 55-fold in the presence of cyclodextrins
the corticosteroids, vitamins and benzodiazepins. leading to increased dissolution rate (increased to
Phenytoin is another example of a drug where the 28–255-fold) (Tenjarla et al., 1998). In the same
prodrug approach has been considered to increase study oral bioavailability of miconazole in rats
water solubility (Stella et al., 1998). A series of increased to 2.3-fold by complexation with hy-
prodrugs of phenytoin with improved aqueous droxypropyl b-cyclodextrins and transdermal dif-
solubilities have been evaluated. The disodium fusivity increased to two- to eight-fold (Tenjarla
salt of the phosphate ester of 3-hydroxymethyl et al., 1998). Solubility of many other poorly
phenytoin was found to be 4500 times more solu- soluble drugs such as hydrocortisone, digitoxin,
ble than phenytoin and the parent compound was diazepam, indomethacin, iotraconazole, kynos-
generated rapidly in in vivo. It should be empha- tatin, pilocarpine, cinnarizine, naproxen, and thi-
sized, however, that an increase in aqueous solu- abendazole has been improved by natural and
bility does not necessarily result in an improved substituted cyclodextrins (Muller and Brauns,
bioavailability. In order to be able to permeate 1985; Putteman et al., 1997; Uekama et al., 1998).
the lipophilic epithelial cell membranes lining the
gastrointestinal tract, a drug molecule must posses 3.2. Permeability and partition coefficient
lipophilic properties. Thus for highly polar com-
pounds, administration of less polar, more The ability of a molecule to cross the biological
lipophilic prodrugs may improve absorption. Ex- membranes (permeability) is a very important bio-
amples are bacampicillin, pivampicillin, and ta- pharmaceutic parameter that governs the absorp-
lampicillin, which are prodrugs of highly polar tion, distribution, metabolism and excretion
ampicillin. (pharmacokinetics) of a drug.
If chemical modification is not feasible, formu- Enroute to its biophase, the drug has to parti-
lation approaches can be used to improve oral tion between the lipid biomembranes and the
absorption of poorly soluble drugs. Usually the aqueous biological fluids. Although constituents
initial approach used to improve solubility of an vary from one membrane to the other, major
insoluble drug in water is to form water-soluble constituents of biomembranes are phospholipids,
salts. The classic review on pharmaceutical salts cholesterol, sphingolipids, and glycolipids. All of
by Berge et al. (1977) needs a special mention. If these lipids are amphipathic in nature. Therefore,
salt formation is not possible, e.g. too unstable, or to successfully cross the various biomembranes
does not render the molecule sufficiently water and to reach its site of action, any drug molecule
soluble, a series of formulation approaches may should have a balance between hydrophilic and
138 R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150

lipophilic properties. The octanol – water partition tral and positively charged form of amines. In
coefficient (P) is a physical property used exten- such cases membrane/water partition values log D
sively to describe a chemical’s lipophilic or hy- (membrane− water) and D log D [(octanol/wa-
drophilic properties. It is the ratio of unionized ter)−(membrane/water)] are good models for
compound in mutually saturated octanol and wa- permeability. In the case of peptide drugs log D
ter. Since, P values may range several orders of (octanol/water) values will not correlate well with
magnitude, the logarithm (log P) is commonly permeability data because, peptide drugs have
used for convenience. The best choice out of many polar functionalities that form hydrogen
various non-polar and slightly polar solvents bonds with hydroxyl groups in aqueous phase. As
available is n-octanol (Smith et al., 1975) because, octanol can also form hydrogen bonds with pep-
it mimics the biological membranes in several tides and amide drugs, it will give misleading
aspects: n-octanol has a saturated alkyl chain, it higher partition values that will not correlate with
has a hydroxyl group that can act as both hydro- permeability. However, correlations can be
gen bond donor as well as acceptor, it dissolves achieved between the permeabilities of peptides
water to the extent of 1.7 M, and its solubility and the number of potential hydrogen bonds that
parameter (doctanol =10) is close to that of biolog- peptides can make with water, suggesting that
ical membranes, for example skin (dskin = 10). desolvation of the polar bonds in the molecule is
This combination of lipophilic chains, hydrophilic a determinant of permeability. Hence, partition
groups, ability to take up water molecules and coefficients between heptane–ethylene glycol or
similar solubility parameter gives n-octanol prop- the differences in partition coefficients between
erties very close to those of natural membranes. octanol buffer and isooctane/cyclohexane buffer
As the physiological pH is 7.4 partition coeffi- (D log P), both of which are experimental esti-
cients are calculated using n-octanol buffer pH mates of hydrogen bond or desolvation potential
7.4 system and is more appropriate for drugs are good descriptors for permeability of peptides
ionizable at physiological pH. Partition coeffi- through intestinal membrane as well as blood–
cients that are measured at a given pH are known brain barrier (BBB) (Abraham et al., 1994;
as distribution coefficients (D) and is defined as Chikhale et al., 1994; Von Geldern et al., 1996).
the ratio of the concentration of compound in the Von Geldern and colleagues reported the im-
organic phase to the concentration of both ion- provement of oral absorption profile of azole-
ized and unionized species in the aqueous phase at based ETA-selective antagonists through rational
a given pH (Scherer and Howard, 1977). structural modifications suggested by D log P
Though the log P or log D values measured for [(octanol/water) − (cyclohexane/water)], which
various drugs and xenobiotics using n-octanol as tends to emphasize the hydrogen bonding capac-
non-aqueous solvent correlated well with perme- ity of molecule, relative to its hydrophobicity. The
ability, distribution and other pharmacokinetic hydrogen bonding capacity was reduced using a
parameters, these values had no correlation with series of urea modifications. The resulting com-
the same pharmacokinetic parameters of peptides pounds showed a good correlation between
(Burton et al., 1992), amide containing drugs D log P and small intestinal absorption in rat.
(Smith et al., 1996) and many basic amines Though there is no general rule that can be
(Austin et al., 1995; Barton et al., 1997). Many applied across the vastly diverse drug molecules,
basic amines show a much higher partition into some generalizations can be made within a ho-
membranes than one would expect considering mologous series of drug molecules. Within a ho-
their log D (octanol/water) values (Barton et al., mologous series, drug absorption usually
1997). This is because n-octanol can only support increases as lipophilicity rises and is maintained at
the efficient partitioning of the neutral form of the a plateau for a few units of log P after which
drug whereas biomembrane, as a consequence of there may be a steady decrease, giving a parabolic
having negatively charged phosphate head relation (Navia and Chaturvedi, 1996). Similar
groups, can support the partitioning of both neu- parabolic relationship has been found between
R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150 139

Table 4 mainly to the increased affinity of drugs for the


List of computer software available to calculate log P
enzymes (Martin and Hansch, 1971).
Software Method Reference If a lead compound has modest activity and has
program log P value below zero, it would be reasonable to
synthesize an analogue with a higher log P. At
Fragmental this stage predictive methods of log P are highly
approaches
valuable. Although, none of them give accurate
PROLOGP – cdr Original Rekker Rekker and
Mannhold, 1992 values as they did not include all the effects of
SYBYL Revised Rekker Rekker and molecular confirmation, proximity, tautomerism
Mannhold, 1992 and potential for hydrogen bonding. into the cal-
CLOGP Leo–Hansch Hansch and Leo, 1979 culation procedure, these methods are still useful
KLOGP Klopman Klopman et al., 1985
and practised commonly in pharmaceutical indus-
KOWWIN Meylan–Howard Meylan and Howard,
1995 try since experimental measurement can be
difficult, time-consuming, and/or expensive (Kristl
Atom-based
approaches
et al., 1999). The literature contains many meth-
PROLOGP Broto Broto et al., 1984 ods for estimating log P and can be mainly cate-
gorized into three major groups: the most
– atomic
PROLOGP Ghose–Crippen Ghose and Crippen, common fragment constant methods (Fujita et al.,
1986 1964; Hansch and Leo, 1979; Rekker and
– atomic5
MOLCAD Ghose–Crippen Ghose and Crippen,
Mannhold, 1992; Meylan and Howard, 1995),
1986 methods based on atomic contributions (Broto et
CHEMICALC2 Suzuki Suzuki and Kudo, al., 1984; Ghose and Crippen, 1986; Suzuki and
1990 Kudo, 1990) and those based on molecular prop-
Molecular property based approaches erties (Sasaki et al., 1991). To simplify computa-
BLOGP Bodor Buchwald and Bodor, tion calculations of these methods many computer
1998 programs are available and are listed in Table 4.
SciLogP Bodor Buchwald and Bodor,
The effect of lipophilicity on oral absorption is
1998
ASCLOGP van de Van de Waterbeemd best exemplified by the classical study of barbitu-
Waterbeemd et al., 1996 rates (Schanker, 1960), b-blockers (Taylor et al.,
HINT Abraham–Kellog Kellogg et al., 1991 1985; Saha et al., 1994), bisphosphonates (Lin,
1996) and 6-fluoroquinolones (Merino et al.,
1995). Development of orally bioavailable peptide
log P and biological activity. Saha et al. (1994), based renin inhibitor A-72 517 by Kleinert et al.
Merino et al. (1995) showed a sigmoidal relation- (1992) is a good example for improving oral ab-
ship between absorption rate and log P in a series sorption by altering the physicochemical proper-
of 6-fluoroquinolones and b-blockers respectively. ties like partition coefficient and solubility.
In general, log P values between 0 and 3 consti- Initially Kleinert and his colleagues developed
tutes an optimal window for passive drug absorp- A-64 662 (enalkiren) a first generation renin in-
tion. A log P value below 0 means that the hibitor that is effective intravenously but shown
compound is hydrophilic, and hence it will have a to lack oral bioavailability (Kleinert et al., 1992).
good solubility but it may have poor permeability. Then they devised A-72 517 a close analogue of
Whereas, a log P value far higher than 3 means A-64 662 to improve oral bioavailability by im-
that the compound is highly lipophilic, hence, proving oral absorption as well as metabolic sta-
tends to favour absorption, and renders the com- bility. The P2-site histidine and NH2-terminal
pounds more susceptible to metabolism and/or b-alanine residues of A-64 662 are more basic
biliary clearance (Toon and Rowland, 1983; than their counterparts in A-72 517 and contain
Humphrey, 1989). The influence of lipophilicity nitrogen-bound protons capable of forming hy-
on the metabolic clearance of drugs is attributed drogen bonds (Fig. 4). Consequently, A-72 517 is
140 R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150

the more lipophilic compound with a log P of 4.6 cokinetic data in drug development has gradually
(in octanol– water, pH 7.4), as compared with a increased. Today it is well recognized that success-
log P of 2.6 for A-64 662, and the aqueous solu- ful drug development programs include support-
bilities of the salts are 10 mg/ml and 100 mg/ml, ive pharmacokinetic data. Pharmacokinetics
respectively. These physicochemical properties of serves as a useful tool in the drug development
A-72 517 along with its proteolytic stability made process both in terms of therapeutics and in defin-
it orally bioavailable (53% in dog). The discovery ing drug disposition characteristics. In this section
of fluconazole (Richardson, 1993), a systemic an- critical pharmacokinetic parameters and high
tifungal, is another good example that substanti- throughput in vitro and in vivo screening tools to
ates the importance of lipophilicity in drug determine the pharmacokinetic parameters are
discovery and development. Pfizer’s tioconazole discussed.
was clinically effective against fungal infections of
the vagina and skin but failed to act systemically 4.1. Critical pharmacokinetic parameters in drug
when given intravenously or orally. Biopharma- de6elopment
ceutic and pharmacokinetic studies revealed that
although this drug was absorbed reasonably well Ten critical pharmacokinetic parameters; clear-
from GI lumen, it was subject to extensive first ance, effective concentration range, extent of
pass metabolism and high protein binding. Efforts availability, fraction of the available dose excreted
to decrease lipophilicity of the molecule lead to unchanged, blood/plasma concentration ratio,
the novel systemic antifungal agent, i.e. flucona- half-life, toxic concentration, extent of protein
zole. This clearly substantiates the fact that the binding, volume of distribution, and rate of
lipophilicity of a drug not only affects its absorp- availability should be determined for each new
tion but also its metabolism, protein binding and chemical molecule both in test animal and in man
distribution. Generally, the higher the lipophilicity (Benet, 1993). Role of pharmacokinetics in vari-
of a drug, the higher its metabolism, the stronger ous stages of drug development were given in
its binding to protein and greater its distribution Table 5. All the aforementioned pharmacokinetic
(Toon and Rowland, 1983). High protein binding parameters critical for drug candidate selection
may result in pharmacokinetic drug interactions can be explained from the following equations,
when concomitantly given with other drugs. which hold good at steady state during therapy.
FD/t= CssCL (2)
4. Pharmacokinetics in drug development CL= 0.693Vd/t1/2 (3)
Vd = Vp + (Vt fup/fut) (4)
As pharmacokinetics of a drug molecule dictate
its availability at its site of action, they are very where F is extent of bioavailability, D is dose, t is
important in drug selection and optimization. dosing interval, Css is steady state concentration
Over the past decade the application of pharma- of drug in plasma, CL is total clearance, Vd is

Fig. 4. Chemical structures of A-64 662 and A-72 517.


R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150 141

Table 5
Role of pharmacokinetics in various stages of drug development

Stage of development Role of pharmacokinetic studies

Selection of drug candidates for Consideration of the pharmacokinetic profile desired in connection with known
development biotransformation processes; explorative in vitro studies.
Preclinical development Design and interpretation of pharmacological and toxicological investigations also with
respect to species differences.
Clinical development: phase 1, Establishing dosage regimens, absolute/relative bioavailability, identification of metabolites
2 and 3 and evaluation of their contribution to the biological profile of the drug. Studies in special
patient groups at potential risk (age, disease, metabolic disorders, co-medications) to adjust
dose regimens.

volume of distribution, t1/2 is elimination half-life, drug development. Such drugs are prone to show
Vp is the volume of plasma, Vt is the tissue high first pass metabolism, dose dependent phar-
volume and fup and fut are the unbound fractions macokinetics and pharmacokinetic drug interac-
of drug in plasma and tissue respectively. tions. Hence, drugs with fu close to zero are not
CL, Css, and F are necessary to define the suitable for oral drug delivery. By decreasing the
appropriate dosing rate (amount/day) of a drug lipophilicity of such drug molecules either by ad-
by a particular route of administration (Benet and dition of polar groups or removal of non-polar
Williams, 1990). In vitro CL rates determined groups that are away from pharmacophore, one
using human systems (enzyme isoforms, micro- can make such molecules suitable for oral
somes, hepatocytes) are predictive of the in vivo delivery.
CL of many lipophilic drugs in man (Smith and Half-life is an important parameter in therapeu-
van de Waterbeemd, 1999).The in vitro CL rates tics, since this parameter defines the dosing inter-
can be determined from Vmax/KM (maximum ve- val at which drugs should be administered.
locity/Michaelis – Menten constant) determina- Half-life also describes the time required to attain
tions for the major metabolic pathway. steady state or to decay from steady-state condi-
Fraction of the available dose excreted un- tions after a change in the dosage regimen. Eq. (3)
changed ( fu) and blood/plasma concentration describes the half-life relationship for a drug that
(Css) are necessary to make one of the most appears to follow one-compartment body kinetics.
important judgements in drug development pro- It is always preferable to select a drug candidate
cess. That is, can this drug be successfully mar- with long elimination half life as it allows less
keted as an oral dosage form in man? (Benet, frequent dosing and hence, can improve patient
1993). Knowledge of fu allows one to estimate convenience and compliance.
non-renal clearance, which may be assumed to Once the drug is in systemic circulation it will
represent hepatic clearance. If the total drug avail- then distribute to all the tissues at a particular
able is excreted unchanged in urine, it implies that rate depending on its physicochemical characteris-
no first pass, no dose dependent pharmacokinet- tics such as lipophilicity and charge. To enter
ics, no enzyme induction and enzyme inhibition various organs and finally to cells the drug has to
by the drug. In such cases, which of course is rare, cross several rate limiting barriers such as cell
less attention may be paid to metabolic studies. membranes and some physiological barriers
But, in cases where most of the drug is eliminated (BBB, blood placental barrier). Hence, lipophilic-
in the form of one or more of its metabolites ity is a good descriptor for determination of dis-
( fu $ 0), enzymes responsible for metabolism, tribution characteristics such as volume of
metabolite profile and pharmacodynamics, toxi- distribution (Vd). Vd is a hypothetical volume of
cokinetics and pharmacokinetics of metabolites body fluid that would be required to dissolve the
must be studied extensively at the early stages of total amount of drug at the same concentration as
142 R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150

that found in the blood. Vd can be estimated from ity, charge, chemical structure and complementar-
partition coefficient (P) and the fraction of protein ily of the drug to binding sites. Only the free,
binding (p) as shown in Eq. (5) in which BW is non-protein-bound fraction of the drug can diffuse
body weight in grams (Ritschel and Kearns, 1998). across membranes that restrict distribution of
drug between vascular compartment and tissues.
Vd = (0.0955P +1.2232)(1 − p)BW (ml) (5)
And only the free drug can exert a pharmacologi-
In general, higher the lipophilicity higher is its cal action. Hence, drug–protein binding can affect
distribution. Hence lipophilic drugs have high Vd. the distribution, clearance and pharmacodynamics
Hydrophilic drugs and drugs ionizable at physio- of a drug. On an average an adult human has
logical pH will be confined mostly to blood com- approximately 0.18-kg plasma proteins and 10-kg
partment and therefore have low Vd. In case of macromolecular structures such as proteins and
basic drugs ionized at physiological pH such as nucleic acids in tissue (Ritschel and Kearns, 1998).
quaternary amines, Vd is high due to ion–pair Considering the apparent volume of distribution
interaction between the positively charged basic of a drug and its relationship to plasma and tissue
centre and negatively charged phosphate head distribution, the Vd can be characterized as de-
groups of biomembranes. In case of distribution to scribed in Eq. (4), from which it is clear that both
brain, the general rule, i.e. higher the lipophilicity plasma and tissue protein binding influence the
higher is the distribution, will hold good only volume of distribution. But the quantitative rela-
within a certain range. Drugs with extremely high tion of plasma to tissue proteins indicates that
lipophilicity cannot permeate easily through BBB. plasma protein binding should not be of signifi-
It is believed that P-glycoprotein, located on apical cant influence on the distribution equilibrium of
surface of the endothelial cells of the brain capil- drugs of extremely high lipid solubility unless they
laries, is responsible for the poor permeability of have an extremely high affinity for plasma
highly lipophilic compounds through BBB (Lin proteins. Plasma protein binding is of significant
and Lu, 1997). influence on the distribution equilibrium if the
For drugs whose site of action is located intra- drug is polar.
cellularly (such as antisense drugs) Vd should be
very high. Otherwise most of the administered 4.2. In 6itro studies
dose of the drug will be confined to blood com-
partment and sufficient amount may not reach the With the on-going emphasis to reduce develop-
actual site of action. In other words more dose ment time, it became imperative to introduce ap-
may be required to achieve the sufficient levels of proaches for evaluating absorption and
drug at the site of action at which it may show metabolism prior to selection of a development
some adverse effects. As Vd is high, i.e. more of the candidate. Advances in in vitro methodologies
drug is in tissues and hence, less amount of dose along with computational and physicochemical
administered is available for clearance leading to approaches for predicting drug absorption are
lower clearance rates. As a result longer elimina- required to meet the increasing demands. Advan-
tion half-lives (t1/2) can be achieved with high Vd. tages of these in vitro techniques include: rapid
The relationship between Vd, CL and t1/2 is shown evaluation of membrane permeability and
in Eq. (3). For drugs having low safety of margin metabolism, ability to identify and characterize
and those required only in circulatory system such mechanisms and pathways of drug transport and
as anti-arrythmatics and other cardio- metabolism, methods for enhancing drug perme-
vascular drugs, low Vd are preferable in order to ability and minimizing drug metabolism, mini-
prevent unnecessary exposure of the drug to the mization of the use of animal studies that are
tissues. resource intensive; and the potential to use human
Many drugs are bound to plasma and tissue rather than animal cells or tissues. A list of in vitro
proteins. The extent of protein binding of a drug methods used to estimate pharmacokinetic
is influenced by several factors such as lipophilic- parameters are given in Table 6.
R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150 143

4.3. High throughput in 6i6o screening in sette dosing can be efficiently used to create an in
preclinical drug de6elopment vivo pharmacokinetics database to use in develop-
ing structure–pharmacokinetic relationships,
The difficulties with the identification of com- which will help in defining molecular changes that
pounds that possess the desired pharmacokinetic do and that do not alter pharmacokinetics (Shaf-
(CL, Vd, t1/2, area under the curve (AUC)) profile fer et al., 1999). However, cassette dosing does
eventually became severe enough, with the high have some disadvantages such as drug–drug in-
productivity of combinatorial chemistry, forcing teractions that can lead to misleading pharma-
for a re-evaluation of the process used to conduct cokinetic results.
these studies. Increased throughput in in vivo An alternative to these cassette dosing studies
pharmacokinetic screening has recently been re- that obviates the problem of drug–drug interac-
ported by (a) cassette dosing, i.e. dosing multiple tions is the use of post-dose pooling of samples.
compounds to a single animal at one time, and (b) By pooling plasma samples collected at equally
by pooling samples from singularly administered spaced time points over a specific period, an
compounds prior to analysis (Berman et al., 1997; average plasma concentration can be determined.
Allen et al., 1998; Cox et al., 1999; Shaffer et al., Multiplication of this average concentration by
1999). the time period gives an estimated AUC, and
In vivo cassette dosing has been investigated as provides a means of ranking compounds based on
a way to increase throughput of the pharmacoki- early pharmacokinetic evaluation. Analysis of the
netic screening of large numbers of potential drug plasma collected at the last time point can give a
candidates (Berman et al., 1997; Shaffer et al., more accurate AUC determination and can also
1999). This technique was applied successfully to give some insight into the oral half-life of each
speed the progress of an a1A-adrenoceptor antag- compound. This rapid in vivo pharmacokinetic
onist to the clinic (Frick et al., 1998). This method screening method has been successfully used to
studies the concurrent dosing of multiple com- provide a rank order of over 200 compounds in a
pounds to a group of animals in an effort to specific therapeutic area (Cox et al., 1999).
reduce the number of animals, the total time and
the cost required to complete the pharmacokinetic
evaluations. A cassette may contain five to 20 5. Empirical rules for the selection of drug
potential drug compounds with similar character- candidates based on biopharmaceutics and
istics, as well as a structurally related internal pharmacokinetics
standard with well-characterized pharmacokinet-
ics. The internal standard is used to monitor Biopharmaceutic and pharmacokinetic data can
analytical performance and to screen for potential be used to select, from several thousand candi-
drug–drug interactions. Animal models will be dates, those drugs most likely to be orally
given an oral gavage or intravenous bolus injec- bioavailable, safe, and effective. A few empirical
tion of each cassette. Blood samples are collected rules for such an evaluation of drug candidates
at specific time points following cassette dosing. are summarized below.
Compounds are extracted from blood samples Only drugs with appropriate physical properties
and analyzed using LC – MS – MS methods specifi- for absorption should be used. A certain extent of
cally developed for the compounds in the cassette. water-solubility is essential for transport to the
The cassette dosing techniques have been success- absorptive site. On the other hand, optimum
fully applied to a group of 40 or more chemical lipophilicity is required for passive diffusion
entities in cancer drugs and GP IIb/IIIa antago- through membrane barriers. These are the two
nists (Pang, 1998). The advantages of a cassette basic requirements for any drug to have good
dosing are that more compounds can be dosed in pharmacokinetic parameters. Four parameters
a shorter time using fewer animals and that the namely molecular weight, log P, the number of
sample analysis is more efficient. Moreover cas- H-bond donors and acceptors are thought to be
144
Table 6
A partial list of in vitro methodologies

Model Description References

Absorption
Excised tissues

R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150


Everted sacs Extensively used to study transport of drugs and xenobiotics through Middleton, 1990
the intestine due to their ease of preparation and simplicity in the
set up required.
Intestinal rings Intestinal rings offer a simple and effective method to study the Porter et al., 1985
kinetics of uptake into the intestinal mucosa. But, one of their
disadvantages is that polarity of uptake cannot be determined.
Mucosal sheets The intestine is cut open into strips, the musculature is removed and Edmonds and Marriott, 1968; Smith et al., 1988
clamped between compartments of Ussing type diffusion chamber.
Can be used to evaluate the transport and metabolism of drugs and
other compounds.
Isolated cells Isolated enterocytes are prepared by treating the intestinal Webster and Harrison, 1969; Iglesias et al., 1988
preparations with proteolytic enzymes, chelating agents or by
mechanical methods. They are short lived. Useful to evaluate uptake,
site dependence and metabolism of various drugs.
Membrane vesicles Brush border membrane vesicles are prepared from either intestinal Hopfer et al., 1973; Osiecka et al., 1985; Sugawara et al., 1992
scrapings or isolated enterocytes by homogenization and divalent
cation precipitation. Useful only to study active carrier mediated
processes.
Cell culture models
Caco-2 cell line Human colon adenocarcinoma spontaneously differentiate in culture Hidalgo et al., 1989; Artursson et al., 1996; Delie and Rubas,
to form confluent polarized monolayers that structurally and 1997; Gan and Thakker, 1997; Stevenson et al., 1999
functionally resemble the small intestinal epithelium. They are
extensively used in permeability characterization and in the
evaluation of brush border metabolism. Good in vivo correlations
can be achieved.
HT-29 cell line HT-29 cells, when induced, will differentiate to form mucin Kreusel et al., 1991; Wikman et al., 1993
producing clones. Used to establish the role of the mucin layer in
intestinal drug absorption.
Metabolism
Subcellular fractions
Enzyme isoforms Useful to identify the isoenzymes responsible for metabolism of Parkinson, 1996; Guengerich and Shimada, 1993; Guengerich and
drugs and in understanding the binding site morphology and Parikh, 1997; Tarbit and Berman, 1998
structure–metabolism relationships.
S9 preparation Prepared by a 9000×g centrifugation of liver homogenates. Contains Alvares et al., 1973; Czygan et al., 1973
both microsomal and cytosolic enzymes and hence, suitable to
evaluate both phase I and phase II reactions. High reproducibility
can be achieved but show poor in vivo correlations.
R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150
Table 6 (Continued)

Model Description References

Microsomes Prepared by a 100 000×g centrifugation of liver homogenates. They Lin, 1996; Carlile et al., 1997; Chiba et al., 1997
mainly contain fragments of the endoplasmic reticulum and lack
cytosolic enzymes and cofactors. Hence, suitable to evaluate phase I
metabolic reactions only. In combination with selective inhibitors,
microsomes can be used to identify isoenzymes responsible for drug
metabolism. High reproducibility can be achieved but show poor in
vivo correlations.
Isolated hepatocytes Can be prepared by any one of various methods that are based Le Bigot et al., 1987; Padgham et al., 1992; Carlile et al., 1997;
upon collagenase perfusion. Though viable only for a few hours, Lave et al., 1997
suspended hepatocytes are often very successful for the investigation
of pathways of metabolism of novel compounds. However, they are
not suitable for the investigation of time dependent metabolism,
enzyme induction and the expression of many drug-induced
toxicities, as they are short lived. Cultured hepatocytes are good
alternatives for suspended hepatocytes to perform longer-term
investigations.
Liver slices Precision cut liver slices can be prepared by tissue slicer or Dogterom, 1993; Balani et al., 1995; Worboys et al., 1996
microtome. This model is suitable to evaluate both phase I and
phase II metabolic reactions. Cell to cell contact and absence of any
enzymatic treatment may be considered as advantages. Poor
reproducibility but high in vivo correlation can be achieved.

145
146 R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150

associated with solubility and permeability (Lipin- cokinetics and biopharmaceutics play an impor-
sky et al., 1997). The ‘rule of five’ proposed by tant role as determinants of in vivo drug action.
Lipinsky and his colleagues states that poor ab- Development of HIV protease inhibitors (indi-
sorption or permeation are more likely when: navir and ritonavir) GP IIb/IIIa antagonists, and
there are more than five H-bond donors; the the antifungal agent fluconazole are good exam-
molecular weight is over 500; the log P is over 5; ples of successfully incorporating pharmacoki-
there are more than ten H-bond acceptors. netic and biopharmaceutic information into drug
Compounds with a hepatic extraction ratio development.
close to unity should be avoided. Such drugs show The increased number of pharmacologically ac-
very high first pass metabolism and therefore tend tive compounds identified during the drug discov-
to show poor and variable bioavailability. Hence, ery process has created a great demand for rapid
such drugs are not suitable for oral administra- screening, selection and development of com-
tion, which is considered as the most convenient pounds with favorable human pharmacokinetics
and safe route. At higher doses saturation of the and safety credentials. New advances in develop-
metabolic activity is likely to occur and conse- ment processes like high sensitive and highly spe-
quently, all pharmacokinetic and pharmacody- cific analytical methods (HPLC–MS–MS), caco-2
namic responses may show high variability after cell techniques, in vitro metabolic studies, in vivo
oral administration of such drugs. cassette dosing and other biopharmaceutic ap-
Compounds that are bound excessively to proaches will definitely enhance the pace and
plasma proteins should be avoided. When the quality of drug development. A great develop-
fraction unbound is B 10%, variations up to sev- ment may be foreseen in the sphere of the use of
eral-fold in Css, Vd, CL and t1/2 can occur when computer techniques for evaluation and commu-
bound drug is competitively displaced by metabo- nication of laboratory data and clinical data on
lites, other drugs, or endogenous/exogenous com- the patient.
pounds from food or disease processes. In conclusion, the role of biopharmaceutics and
Depending on the therapeutic class, volume of pharmacokinetics is an indispensable part in ac-
distribution of the drug candidate should be complishing the goals of drug development, i.e.
optimized. getting safe and more efficacious drugs with re-
Compounds with higher half-life values are duced development time and cost.
preferable as they allow less frequent dosing and
hence patient convenience and compliance.
Drug molecules should lend themselves to effi- References
cient and predictable pathways of elimination (the
soft-drug concept). There are only a few processes Abraham, M., Chada, H.S., Mitchell, R.C., 1994. Hydrogen
with a virtually unlimited capacity for elimina- bonding 33. Factors that influence the distribution of
solutes between blood and brain. J. Pharm. Sci. 83, 1257–
tion. Among these are the metabolic processes of 1268.
hydrolysis and glucuronidation. Glomerular filtra- Allen, M.C., Shah, T.S., Day, W.W., 1998. Rapid determina-
tion is also unlimited, but it is not effective with tion of oral pharmacokinetics and plasma free fraction
lipophilic compounds as they are subsequently using cocktail approaches: methods and application.
reabsorbed. Pharm. Res. 15, 93 – 97.
Alvares, A.P., Bickens, D.R., Kappas, A., 1973. Polychlori-
nated biphenyls: a new type of inducer of cytochrome
P-448 in the liver. Proc. Natl. Acad. Sci. USA 70, 1321–
6. Conclusions 1325.
Amidon, G.L., Lennernäs, H., Shah, V.P., Crison, J.R., 1995.
Aim of drug development is identifying poten- A theoritical basis for a biopharmaceutic drug classifica-
tion: the correlation of in vitro drug product dissoultion
tial drug molecules among the hundreds of lead and in vivo bioavailability. Pharm. Res. 12, 413 – 420.
molecules, and getting a safe and more efficacious Amidon, G.L., 1981. Drug derivatization as a means of solubi-
drug molecule to the therapeutic arena. Pharma- lization: physical and biochemical strategies. In:
R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150 147

Yalkowsky, S.H. (Ed.), Techniques of Solubilization of Chikhale, E.G., Ng, K.Y., Burton, P.S., Borchardt, R.T.,
Drugs. Marcel Dekker, New York, pp. 183–211. 1994. Hydrogen bonding potential as a determinant of the
Artursson, P., Plam, K., Luthman, K., 1996. Caco-2 monolay- in vitro and in situ blood – brain barrier permeability of
ers in experimental and theoretical predictions of drug peptides. Pharm. Res. 11, 412 – 419.
transport. Adv. Drug Deliv. Rev. 22, 67–84. Collins, M.A., Shaw, I., Billington, D.C., 1999. Driving drug
Austin, R.P., Davis, A.M., Manners, C.N., 1995. Partitioning discovery and patent therapy via the encapsulation and
of ionizing molecules between aqueous buffers and physio- fusion of knowledge. Drug Des. Discov. 16, 181 – 194.
logical vesicles. J. Pharm. Sci. 84, 1180–1183. Cox, K.A., Dunn-Meynell, K., Korfmacher, W.A., Broske, L.,
Balani, S.K., Arison, B.H., Mathai, L., Kauffman, L.R., Nomeir, A.A., Lin, C., Cayen, M.N., Barr, W.H., 1999.
Miller, R.R., Stearns, R.A., Chen, I.W., Lin, J.H., 1995. Novel in vivo procedure for rapid pharmacokinetic screen-
Metabolites of L-735,524, a potent HIV-1 protease in- ing of discovery compounds in rats. Drug Discov. Today 4,
hibitor, in human urine. Drug Metab. Dispos. 23, 266 – 232 – 237.
270. Czygan, P., Greim, H., Garro, A.J., Hatterer, F., Schaffer, F.,
Barton, P., Davis, A.M., McCarthy, D.J., Webborn, J.H., Popper, H., Rosenthal, O., Cooper, D.Y., 1973. Microso-
1997. Drug–phospholipid interactions. 2. Predicting the mal metabolism of dimethyl nitrosamine and the cy-
sites of drug distribution using n-octanol/water distribu- tochrome P-450 dependency of its activation to a mutagen.
tion coefficients. J. Pharm. Sci. 86, 1034–1039. Cancer Res. 33, 2983 – 2986.
Benet, L.Z., 1993. The role of pharmacokinetics in the drug Delie, F., Rubas, W., 1997. A human colonic cell line sharing
development process. In: Yacobi, A., Skelly, J.P., Shah, similarities with enterocytes as a model to examine oral
V.P., Benet, L.Z. (Eds.), Integration of Pharmacokinetics, absorption advantages and limitations of the caco-2 model.
Pharmacodynamics, and Toxicokinetics in Rational Drug Crit. Rev. Ther. Drug Carrier Syst. 14, 221 – 286.
Development. Plenum, New York, pp. 115–124. Dogterom, P., 1993. Development of a simple incubation
Benet, L.Z., Williams, R.L., 1990. Design and optimization of system for metabolism studies with precision-cut liver
dosage regimens: pharmacokinetic data. In: Gilman, A.G., slices. Drug Metab. Dispos. 21, 699 – 704.
Rall, T.W., Nies, A.S., Taylor, P. (Eds.), The Pharmaco- Dorsey, B.D., Levin, R.B., McDaniel, S.L., Vacca, J.P.,
logical Basis of Therapeutics. Pergamon, New York, pp. Guare, J.P., Darke, P.L., Zugay, J.A., Emini, E.A., Schleif,
1650 – 1735. W.A., Quintero, J.C., Lin, J.H., Chen, I.W., Holloway,
Berge, S.M., Bighley, L.D., Monkhouse, D.C., 1977. Pharma- M.K., Fitzgerald, P.M.D., Axel, M.G., Ostovic, D., An-
ceutical salts. J. Pharm. Sci. 66, 1–19. derson, P.S., Huff, J.R., 1994. L-735,524: the design of a
Berman, J., Halm, K., Adikson, K., Shaffer, J., 1997. Simulta- potent and orally bioavailable HIV protease inhibitor. J.
neous pharmacokinetic screening of a mixture of com- Med. Chem. 37, 3443 – 3451.
pounds in the dog using API LC/MS/MS analysis for Dressman, J.B., Fleisher, D., 1986. Mixing-tank model for
increased throughput. J. Med. Chem. 40, 827–829. predicting dissolution rate control of oral absorption. J.
Bohacek, R.S., McMartin, C., Giuda, W.C., 1996. The art and Pharm. Sci. 75, 109 – 116.
practice of structure-based drug design: a molecular model- Dressman, J.B., Amidon, G.L., Fleisher, D., 1985. Absorption
ing perspective. Med. Res. Rev. 16, 3–50. potential: estimating the fraction absorbed for orally ad-
Broto, P., Moreau, G., Vandycke, C., 1984. Molecular struc- ministered compounds. J. Pharm. Sci. 74, 588 – 589.
tures perception, auto correlation descriptor and SAR Edmonds, J., Marriott, J., 1968. Factors influencing the elec-
studies — system of atomic contributions for the calcula- trical potential across the mucosa of rat colon. J. Physiol.
tion of the normal octanol water partition coefficients. Eur. 194, 457 – 478.
J. Med. Chem. 19, 71–78. Eldred, C.D., Evans, B., Hindley, S., Judkins, B.D., Kelly,
Buchwald, P., Bodor, N., 1998. Octanol–water partition: H.A., Kitchen, J., Lumley, P., Porter, B., Ross, B.C.,
searching for predictive models. Curr. Med. Chem. 5, Smith, K.J., Taylor, N.R., Wheatcroft, J.R., 1994. Orally
353 – 380. active non-peptide fibrinogen receptor (GP IIb/IIIa) antag-
Burton, P.S., Conradi, R.A., Hilgers, A.R., Ho, N.F.H., Mag- onists: identification of 4-[4-[4-(aminoiminomethyl)
giora, L.L., 1992. The relationship between peptide struc- phenyl]-1-piperazinyl]-1-piperidine acetic acid as a long
ture and transport across epithelial cell monolayers. J. acting, broad spectrum antithrombotic agent. J. Med.
Control. Release 19, 87–98. Chem. 37, 3882 – 3885.
Carlile, D.J., Zomorodi, K., Houston, J.B., 1997. Scaling Frick, L.W., Adikson, K.K., Wells-Knecht, K.J., Woollard,
factors to relate drug metabolic clearance in hepatic micro- P., Highton, D.M., 1998. Cassette dosing: rapid in vivo
somes, isolated hepatocytes, and the intact liver — studies assessment of pharmacokinetics. Pharm. Sci. Tech. Today
with induced livers involving diazepam. Drug Metab. Dis- 1, 12 – 18.
pos. 25, 903–911. Fujita, T., Iwasa, J., Hansch, C., 1964. A new substituent
Chiba, M., Hensleigh, M., Lin, J.H., 1997. Hepatic and intesti- constant, p, derived from partition coefficients. J. Am.
nal metabolism of indinavir, a potent HIV protease in- Chem. Soc. 86, 5175 – 5180.
hibitor, in rat and human microsomes. Biochem. Gan, L.S.L., Thakker, D.R., 1997. Applications of the caco-2
Pharmacol. 53, 1187–1195. model in the design and development of orally active
148 R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150

drugs: elucidation of biochemical and physical barriers immunodeficiency virus protease and has high oral
posed by the intestinal epithelium. Adv. Drug Deliv. Rev. bioavailability in humans. Proc. Natl. Acad. Sci. USA 92,
23, 77 – 98. 2484 – 2488.
Ghose, A.K., Crippen, G.M., 1986. Atomic physicochemical Kleinert, H.D., Rosenberg, S.H., Baker, W.R., Stein, H.H.,
parameters for 3-dimensional structure-directed qauantita- Klinghofer, V., Barlow, J., Spina, K., Polakowski, J.,
tive structure–activity relationships. 1. Partition coeffi- Kovar, P., Cohen, J., Denissen, J., 1992. Discovery of a
cients as a measure of hydrophobicity. J. Comput. Chem. peptide-based renin inhibitor with oral bioavailability and
7, 565 – 577. efficacy. Science 257, 1940 – 1943.
Gibaldi, M., 1991. Preface. In: Biopharmaceutics and Clinical Klopman, G., Namboodiri, K., Schochet, M., 1985. Simple
Pharmacokinetics. Lea and Febiger, Malvern, PA, p. vii. method of computing the partition coefficient. J. Comput.
Goodacre, B.C., Murray, R.J., 1981. A mathematical model of Chem. 6, 28 – 38.
drug absorption. J. Clin. Hosp. Pharm. 6, 117– 133. Kreusel, K.M., Fromm, M., Schulzke, J.D., Hagel, U., 1991.
Guengerich, F.P., Parikh, A., 1997. Expression of drug-metab- C4-secretion epithelial monolayers of mucus-forming hu-
olizing enzymes. Curr. Opin. Biotechnol. 8, 623 – 628. man colon cell (HT-29/B6). Am. J. Physio. 261, C574–582.
Guengerich, F.P., Shimada, T., 1993. Human cytochrome Kristl, A., Pecar, S., Kmetec, V., 1999. Are calculated log P
P-450 enzymes and chemical carcinogenesis. In: Jeffery, values for some guanine derivatives by different computer
E.H. (Ed.), Human Metabolism: from Molecular Biology programs reliable? Int. J. Pharm. 181, 219 – 226.
to Man. CRC Press, Boca Raton, FL, pp. 5–12. Lave, T., Dupin, S., Schmitt, C., Balles, B., Ubeaud, G.,
Hansch, C., Leo, A.J., 1979. Substituent constants for correla- Chou, R.C., Jaeck, D., Coassolo, P., 1997. The use of
tion analysis. In: Chemistry and Biology. Wiley, New human hepatocytes to select compounds based on their
York. expected hepatic extraction ratios in humans. Pharmacol.
Hidalgo, I.J., Raub, T.J., Borchardt, R.T., 1989. Characteriza- Res. 14, 152 – 155.
tion of human colon carcinoma cell line (caco-2) as a Le Bigot, J.D., Begue, J.M., Kiechel, J.R., Guillouzo, A.,
model system for intestinal epithelial permeability. Gas- 1987. Species differences in metabolism of ketotifen in rat,
troenterology 96, 736–749. rabbit and human: demonstration of similar pathways in
Ho, N.F.H., Merkle, H.P., Higuchi, W.I., 1983. Quantitative vivo and in cultured hepatocytes. Life Sci. 40, 883 – 890.
mechanistic and physiologically realistic approach to the Lin, J.H., 1996. Bisphosphonates: a review of their pharma-
biopharmaceutical design of oral drug delivery systems. cokinetic properties. Bone 18, 75 – 85.
Drug Dev. Ind. Pharm. 9, 1111–1184. Lin, J.H., Lu, A.H., 1997. Role of pharmacokinetics and
Hopfer, U., Nelson, K., Perrotto, J., Isselbacher, K.J., 1973. metabolism in drug discovery and development. Pharma-
Glucose transport in isolated brush border membrane from col. Rev. 49, 403 – 448.
rat small intestine. J. Biol. Chem. 248, 25–32. Lipinsky, C.A., Lombardo, F., Dominy, B.W., Feeney, P.J.,
Humphrey, M.J., 1989. Pharmacokinetic studies in the selec- 1997. Experimental and computational approaches to esti-
tion of new drugs: a case history on dihydropyridine mate solubility and permeability in drug discovery and
calcium channel blockers. In: Kato, R., Estahrook, R.W., development settings. Adv. Drug Deliv. Rev. 23, 3 –25.
Gayen, M.N. (Eds.), Xenobiotic Metabolism and Disposi- Lipper, R.A., 1999. E pluribus product. Modern Drug Discov.
tion. Taylor and Francis, London, pp. 245–253. 2, 55 – 60.
Iglesias, J., Gonzalez-Pacanowska, D., Caamano, G., Garcia- Macheras, P.E., Symillides, M.Y., 1989. Toward a quantitative
Peregrin, E., 1988. Mevalonate 5-pyrophosphate decar- approach for the prediction of the fraction of dose ab-
boxylase in isolated villus and crypt cells of chick intestine. sorbed using the absorption potential concept. Biopharm.
Lipids 23, 291–294. Drug Dispos. 10, 43 – 53.
Kellogg, G.E., Semus, S.F., Abraham, D.J., 1991. HINT: a Martin, Y.C., Hansch, C., 1971. Influence of hydrophobic
new method of empirical hydrophobic field calculation for character on the relative rate of oxidation of drugs by rat
CoMFA. J. Comput.-Aided Mol. Des. 5, 545–552. liver microsomes. J. Med. Chem. 14, 777 – 779.
Kempf, D.J., March, K.C., Paul, D.A., Knigge, M.K., Nor- Merino, V., Freixas, J., Bermejo, M.D.V., Garrigues, T.M.,
beck, D.W., Kohlbrenner, W.E., Codacovi, L., Vasa- Moreno, J., Pla-Delfina, J.M., 1995. Biophysical models as
vanonda, S., Bryant, P., Wang, X.C., Wideburg, N.E., an approach to study passive absorption in drug develop-
Clement, J.J., Plattner, J.J., Erickson, J., 1991. Anti viral ment: 6-fluoroquinolones. J. Pharm. Sci. 84, 777 – 782.
and pharmacokinetic properties of C2 symmetric inhibitors Meylan, W., Howard, P., 1995. Atom/fragment contribution
of the human immunodeficiency virus type I protease. method for estimating octanol – water partition coefficients.
Antimicrob. Agents Chemother. 35, 2209–2214. J. Pharm. Sci. 84, 83 – 92.
Kempf, D.J., March, K.C., Denissen, L.F., McDonald, E., Middleton, H.M., 1990. Uptake of riboflavin by rat intestinal
Vasavanonda, S., Flentge, C.A., Green, B.E., Fino, L., mucosa in vitro. J. Nutrition 120, 588 – 593.
Park, C.H., Long, X.P., Wideburg, N.E., Saldivar, A., Muller, B.W., Brauns, U., 1985. Solubilization of drugs by
Ruiz, L., Kati, W.M., Sham, H.L., Robins, T., Stewart, modified b-cyclodextrins. Int. J. Pharm. 26, 77 – 88.
K.D., Hsu, A., Plattner, J.J., Leonard, J.M., Norbeck, Navia, M.A., Chaturvedi, P.R., 1996. Design principles for
D.W., 1995. ABT-538 is a potent inhibitor of human orally biovailable drugs. Drug Discov. Today 1, 179–189.
R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150 149

Oh, D.M., Curl, R.L., Amidon, G.L., 1993. Estimating the Scherer, R.A., Howard, S.M., 1977. Use of distribution coeffi-
fraction dose absorbed from suspensions of poorly soluble cients in quantitative structure – activity relationships. J.
compounds in humans: a mathematical model. Pharm. Med. Chem. 20, 53 – 58.
Res. 10, 264–270. Shaffer, J.E., Adkison, K.K., Halm, K., Hedeen, K., Berman,
Osiecka, I., Porter, P.A., Borchardt, R.T., Fix, J.A., Gardner, J., 1999. Use of ‘N-in-one’ dosing to create an in vivo
C.R., 1985. In vitro drug absorption models. I. Brush pharmacokinetics database for use in developing struc-
border membrane vesicles, isolated mucosal cells and ev- ture – pharmacokinetic relationships. J. Pharm. Sci. 88,
erted intestinal rings: characterization and salicylate accu- 313 – 318.
mulation. Pharm. Res. 2, 284–292. Sinko, P.J., Leesman, G.D., Amidon, G.L., 1991. Predicting
Padgham, C.R.W., Paine, A.J., Phillips, I.R., Sheppard, E.A., fraction dose absorbed in humans using a macroscopic
1992. Maintenance of total cytochrome P-450 content in mass balance approach. Pharm. Res. 8, 979 – 988.
rat hepatocyte culture and the abundance of CYP 1A2 and Smith, D.A., van de Waterbeemd, H., 1999. Pharmacokinetics
CYP 2B 1/2 in RNAs. Biochem. J. 285, 929–932. and metabolism in early drug discovery. Curr. Opin.
Pang, D.C., 1998. Bridging gaps in drug discovery and devel- Chem. Biol. 3, 373 – 378.
opment. Pharm. Tech. 22, 82–94. Smith, R.N., Hansch, C., Ames, M.M., 1975. Selection of a
Parkinson, A., 1996. An overview of current cytochrome P-450 reference partitioning system for drug designing work. J.
technology for assessing the safety and efficacy of new Pharm. Sci. 64, 599 – 606.
materials. Toxicol. Pathol. 24, 45–57. Smith, P., Mirabelli, C., Fondacaro, J., Ryan, F., Dent, J.,
Porter, P.A., Osiecka, J., Brochardt, R.T., Fix, J.A., Frost, L., 1988. Intestinal 5-fluorouracil absorption: use of ussing
Gardner, C., 1985. In vitro drug absorption models. II. chambers to assess transport and metabolism. Pharm. Res.
salicylate, cefoxitin, a-methyldopa and theophylline uptake 5, 598 – 603.
in cells and rings: correlation with in vivo bioavailability. Smith, D.A., Jones, B.C., Walker, D.K., 1996. Design of drugs
Pharm. Res. 2, 293–298. involving the concepts and theories of drug metabolism
Prentis, R.A., Lis, Y., Walker, S.R., 1988. Pharmaceutical
and pharmacokinetics. Med. Res. Rev. 16, 243 – 266.
innovation by the seven UK-owned pharmaceutical com-
Stella, V.J., Martodihardjo, S., Terada, K., Rao, V.M., 1998.
panies (1964–1985). Br. J. Clin. Pharmacol. 25, 387 – 396.
Some relationships between the physical properties of vari-
Putteman, W., Caers, J., Mesens, J., Peeters, J., 1997. Ab-
ous 3-acyloxymethyl prodrugs of phenytoin to structure:
stracts of the International Pharmaceutical Applications of
potential in vivo performance implications. J. Pharm. Sci.
Cyclodextrins Conference, Lawrence, KS.
87, 1235 – 1241.
Rekker, R.F., Mannhold, R., 1992. Calculation of Drug
Stevenson, C.L., Augustijns, P.F., Hendren, R.W., 1999. Use
Lipophilicity: The Hydrophobic Fragmental Constant Ap-
of caco-2 cells and LC/MS/MS to screen peptide combina-
proach. VCH, Weinheim.
torial library for permeable structures. Int. J. Pharm. 177,
Richardson, K., 1993. The discovery of fluconazole. DN&P 6,
103 – 115.
299 – 303.
Sugawara, M., Toda, T., Iseki, K., Miyazaki, K., Shiroto, H.,
Ritschel, W.A., 1992a. The LADMER system: liberation, ab-
sorption, distribution, metabolism, elimination and re- Kondo, Y., Uchino, J., 1992. Transport characteristics of
sponse. In: Ritschel, W.A. (Ed.), Handbook of Basic cephalosporin antibiotics across intestinal brush-border
Pharmacokinetics Including Clinical Applications. Drug membrane in man, rat, and rabbit. J. Pharm. Pharmacol.
Intelligence, Hamilton, pp. 19–24. 44, 968 – 972.
Ritschel, W.A., 1992b. Preface. In: Ritschel, W.A. (Ed.), Suzuki, T., Kudo, Y., 1990. Automatic log P estimation based
Handbook of Basic Pharmacokinetics Including Clinical on combined additive modeling methods. J. Comput.-
Applications. Drug Intelligence, Hamilton, p. iv. Aided Mol. Des. 4, 155 – 198.
Ritschel, W.A., Kearns, G.L., 1998. Volume of distribution Tarbit, M.H., Berman, J., 1998. High-throughput approaches
and distribution coefficient. In: Ritschel, W.A., Kearns, for evaluating absorption, distribution, metabolism and
G.L. (Eds.), Handbook of Basic Pharmacokinetics Includ- excretion properties of lead compounds. Curr. Opin.
ing Clinical Applications. American Pharmaceutical Asso- Chem. Biol. 2, 411 – 416.
ciation, Washington, DC, pp. 178–188. Taylor, D.C., Pownall, R., Burke, W., 1985. Absorption of
Saha, P., Kim, K.J., Yamahara, H., Crandall, E.D., Lee, beta-adrenoceptor antagonists in rat in situ small intestine:
V.H.L., 1994. Influence of lipophilicity on b-blocker per- effect of lipophilicity. J. Pharm. Pharmacol. 37, 280 –283.
meation across rat alveolar epithelial cell monolayers. J. Tenjarla, S., Puranjoti, P., Kasina, R., Mandal, J., 1998.
Control. Release 32, 191–200. Preparation, characterization, and evaluation of micona-
Sasaki, Y., Kubodera, H., Matuszaki, T., Umeyama, H., 1991. zole-cyclodextrin complexes for improved oral and topical
Prediction of octonol/water partition coefficients using delivery. J. Pharm. Sci. 87, 425 – 429.
parameters derived from molecular structures. J. Pharma- Testa, B., Caldwell, J., 1996. Prodrugs revisited: the ‘ad hoc’
cobiodyn. 14, 207–214. approach as a complement to ligand design. Med. Res.
Schanker, L.S., 1960. On the mechanism of absorption from Rev. 16, 233 – 241.
the gastrointestinal tract. J. Med. Pharm. Chem. 2, 343 – Thompson, W.J., Fitzgerald, P.M.D., Holoway, M.K., Emini,
346. E.A., Darke, P.L., McKeever, B.M., Schleif, W.A., Quin-
150 R. Panchagnula, N.S. Thomas / International Journal of Pharmaceutics 201 (2000) 131–150

tero, J.C., Zugay, J.A., Tucker, T.J., Schwering, J.E., Von Geldern, T.W., Hoffman, D.J., Kester, J.A., Nellans,
Homnick, C.F., Nunberg, J., Springer, J.P., Huff, J.R., H.N., Dayton, B.D., Calzadilla, S.V., Marsh, K.C., Her-
1992. Synthesis and antiviral activity of a series of HIV-1 nandez, I., Chiou, W., Dixon, B.D., Wu-Wong, J.R.,
protease inhibitors with functionality tethered to the P1 or Opgenorth, T.J., 1996. Azole endothelin antagonists. 3.
P%1 phenyl substituents: X-ray crystal structure assisted Using delta log P as a tool to improve absorption. J. Med.
design. J. Med. Chem. 35, 1685–1701. Chem. 39, 982 – 991.
Toon, S., Rowland, M., 1983. Structure–pharmacokinetic re- Webster, H.L., Harrison, D.D., 1969. Enzymatic activities
lationships among the barbiturates in the rat. J. Pharma- during the transformation of crypt columnar intestinal
col. Exp. Ther. 225, 752–763. cells. Exp. Cell Res. 56, 245 – 253.
Uekama, K., Hirayama, F., Irie, T., 1998. Cyclodextrin drug
Wikman, A., Karlsson, J., Carlstedt, I., Artursson, P., 1993. A
carrier systems. Chem. Rev. 98, 2045–2076.
drug absorption model based on the mucus layer produc-
Vacca, J.P., Guare, J.P., deSolms, S.J., Sanders, W.M., Giu-
ing human intestinal goblet cell line HT-29-H. Pharm. Res.
liani, E.A., Young, S.D., Darke, P.L., Zugay, J., Sigal, I.S.,
10, 843 – 852.
Schleif, W.A., Quintero, J.C., Emini, E.A., Anderson, P.S.,
Huff, J.R., 1991. L-687,908, a potent hydroxyethylene-con- Worboys, P.D., Bradburty, A., Houston, B., 1996. Kinetics of
taining HIV protease inhibitor. J. Med. Chem. 34, 1225 – drug metabolism in rat liver slices: II, comparison of
1228. clearance by liver slices and freshly isolated hepatocytes.
Van de Waterbeemd, H., Karajiannis, H., Kansy, M., Drug Metab. Dispos. 24, 676 – 681.
Obrecht, D., Mueller, K., Lehmann, C., 1996. Conforma- Yalkowsky, S.H., 1981. Solubility and solubilization of
tion – lipophilicity relationships of peptides and peptide nonelectrolytes. In: Yalkowsky, S.H. (Ed.), Techniques of
mimetics. In: Sanz, F. (Ed.), Trends in QSAR and Molecu- Solubilization of Drugs. Marcel Dekker, New York, pp.
lar Modeling. Prous, Barcelona. 1 – 14.

S-ar putea să vă placă și