throbber
Summary
`
`Clinical Pharmacokinetics 9: 1-25 (1984)
`0312-5963/84/0001-0001/$12.50/0
`© ADIS Press Limited
`All rights reserved.
`
`First-Pass Elimination
`Basic Concepts and Clinical Consequences
`
`Susan M_ Pond and Thomas N. Tozer
`Clinical Pharmacology Division of the Medical Service, San Francisco General
`Hospital Medical Center, and the School of Medicine and the School of Pharmacy,
`University of California, San Francisco
`
`First-pass elimination takes place when a drug is metabolised between its site of admin(cid:173)
`istration and the site of sampling for measurement of drug concentration. Clinically, first(cid:173)
`pass metabolism is important when the fraction of the dose administered that escapes
`metabolism is small and variable. The liver is usually assumed to be the major site of
`first-pass metabolism of a drug administered orally, but other potential sites are the gas(cid:173)
`trointestinal tract, blood, vascular endothelium, lungs, and the arm from which venous
`samples are taken. Bioavailability, defined as the ratio of the areas under the blood con(cid:173)
`centration-time curves, after extra- and intravascular drug administration (corrected for
`dosage if necessary), is often used as a measure of the extent of first-pass metabolism.
`When several sites of first-pass metabolism are in series, the bioavailability is the product
`of the fractions of drug entering the tissue that escape loss at each site.
`The extent of first-pass metabolism in the liver and intestinal wall depends on a number
`of physiological factors. The major factors are enzyme activity, plasma protein and blood
`cell binding, and gastrointestinal motility. Models that describe the dependence of biG(cid:173)
`availability on changes in these physiological variables have been developed for drugs
`subject to first-pass metabolism only in the liver. Two that have been applied widely are
`the 'well-stirred' and 'parallel tube' models. Discrimination between the 2 models may be
`performed under linear conditions in which all pharmacokinetic parameters are inde(cid:173)
`pendent of concentration and time. The predictions of the models are similar when bio(cid:173)
`availability is large but differ dramatically when bioavailability is small. The 'parallel
`tube' model always predicts a much greater change in bioavailability than the 'well-stirred'
`model for a given change in drug-metabolising enzyme activity, blood flow, or fraction of
`drug unbound.
`Many clinically important drugs undergo considerable first-pass metabolism after an
`oral dose. Drugs in this category include alprenolol, amitriptyline, dihydroergotamine,
`5-fluorouracil, hydralazine, isoprenaline (isoproterenol), lignocaine (lidocaine), lorcainide,
`pethidine (meperidine), mercaptopurine, metoprolol, morphine, neostigmine, nifedipine,
`pentazocine and propranolol. One major therapeutic implication of extensive first-pass
`metabolism is that much larger oral doses than intravenous doses are required to achieve
`equivalent plasma concentrations. For some drugs, extensive first-pass metabolism pre(cid:173)
`cludes their use as oral agents (e.g. lignocaine, naloxone and glyceryl trinitrate). Inhalation
`or buccal, rectal or transdermal administration may, in part, obviate the problems of
`extensive first-pass metabolism of an oral dose.
`Drugs that undergo extensive first-pass metabolism may produce different plasma me(cid:173)
`tabolite concentration-time profiles after oral and parenteral administration. After an oral
`
`1
`
`

`
`First-Pass Elimination
`
`2
`
`dose, the concentration of the metabolite may reach a peak earlier than after a parenteral
`dose. Sometimes, metabolites have only been detected in plasma after an oral dose. Drugs
`in this category include alprenolol, amitriptyline, lorcainide, pethidine, m/edipine and
`propranolol. Although the plasma concentration-time profiles of metabolites may differ
`after oral and parenteral doses, the fraction of a dose eventually converted to a metabolite
`should be the same after each route of administration provided that the ingested drug is
`completely absorbed, is eliminated solely by metabolism in the liver, and has linear ki(cid:173)
`netics. Otherwise, the fraction of a dose administered that is converted to a metabolite
`may vary with route of administration (e.g. with isoprenaline and salbutamol). Variation
`in the concentration ratios between parent drug and metabolite may produce route(cid:173)
`dependent differences in pharmacological and toxicological responses to a given concen(cid:173)
`tration of the parent drug (e.g. with encainide, lorcainide. quinidine and verapamil).
`Drugs that undergo extensive first-pass elimination exhibit pronounced interindividual
`variation in plasma concentrations or drug concentration-time curves after oral admin(cid:173)
`istration. This variation. often reflected in variability in drug response. poses one of the
`major problems in the clinical use of these drugs. Variability in first-pass metabolism is
`accounted for by differences in metabolising enzyme activity produced either by enzyme
`induction. inhibition. or by genetic polymorphism. Liver disease affects bioavailability by
`changing metabolising enzyme activity and plasma protein binding. and creating intra(cid:173)
`and extrahepatic portacaval shunts. In addition, food. by causing transient increases in
`splanchnic-hepatic blood flow, may also decrease the first-pass metabolism of certain drugs.
`The bioavailability of some drugs is dose- and time-dependent. The bioavailability of
`a single oral dose of 5-fluorouracil. hydralazine. lorcainide, phenacetin (acetophenetidin).
`propranolol and salicylamide increases as dose increases. When lorcainide, metoprolol.
`propranolol. dextropropoxyphene (propoxyphene) and verapamil are given repeatedly. their
`bioavailability increases. This time dependency may not be observed when the drugs are
`administered intravenously.
`The liver has been most extensively studied with respect to first-pass metabolism. Rela(cid:173)
`tively little information is available in humans on intestinal or pulmonary metabolism or
`on the effects of altered organ blood flow and plasma protein binding on first-pass me(cid:173)
`tabolism. These potentially important areas require further exploration to broaden our
`understanding of the clinically important phenomenon of first-pass metabolism.
`
`In clinical practice, drug concentrations are
`measured in peripheral venous blood or plasma.
`When drug is eliminated between the site of
`administration and the site of sampling for meas(cid:173)
`urement, first-pass elimination of the drug has oc(cid:173)
`curred. This elimination is usually assumed to
`occur via metabolism. First-pass metabolism in the
`gastrointestinal tract and liver of drugs admini(cid:173)
`stered orally has been studied most extensively and
`is of importance because most drugs are admini(cid:173)
`stered orally. First-pass metabolism is important
`clinically when the fraction of the dose that escapes
`loss at these metabolic sites is small and variable.
`In this review, concepts basic to the process of first(cid:173)
`pass metabolism are examined. The implications
`of first-pass metabolism on the therapeutic use of
`
`drugs are emphasised and illustrated by clinically
`important examples whenever possible.
`
`1. Basic Concepts
`l.l Organs Involved
`
`The liver is usually assumed to be the major site
`of first-pass metabolism of a drug administered
`orally. Other potential sites after oral administra(cid:173)
`tion are the gastrointestinal flora and mucosa,
`blood, vascular endothelium, lungs, and the limb
`in which venous samples are taken. It is difficult
`to establish the exact site of drug metabolism after
`oral administration by sampling only blood. Po(cid:173)
`tential sites can be identified in vitro in isolated
`whole organs, tissue slices, homogenates, or blood
`
`2
`
`

`
`First-Pass Elimination
`
`3
`
`enzyme preparations. In animals, identification of
`involvement of the intestine and liver can be
`achieved by comparing the areas under the plasma
`concentration-time curve (AVC) after intravenous,
`oral and intraportal drug administration (Iwamoto
`and Klaassen, 1977a,b; Rheingold et aI., 1982;
`Rowland, 1972). Similar manipulations of route of
`administration can identify other potential sites of
`metabolism, such as the lungs (Brazzell et aI., 1982).
`In humans, the portal vein has been catheter(cid:173)
`ised to study the first-pass metabolism in the in(cid:173)
`testine of oral doses of flurazepam (Mahon et aI.,
`1977) and proscillaridin (Andersson et aI., 1977).
`The kinetic behaviour of intraperitoneal, intraven(cid:173)
`ous (Collins et aI., 1980), or intrahepatic arterial
`doses (Ensminger et aI., 1978) of 5-fluorouracil has
`also been examined. Shand and Rangno (1972), by
`studying a patient who had a surgical portacaval
`anastamosis, showed that the intestine does not
`metaboIise propranolol. In other studies, the first(cid:173)
`pass uptake and elimination of compounds by the
`lungs have been studied in patients undergoing
`routine cardiac catheterisation (Geddes et aI., 1979;
`Jose et aI., 1976). Woodcock and co-workers (1981)
`directly measured the hepatic extraction of vera(cid:173)
`pamil in cardiac patients.
`
`1.2 Concept of Bioavailability
`
`BioavaiiabiJity - defined as the ratio of the AVCs
`(corrected for dose if necessary) after extra- and
`intravascular drug administration (Tozer, 1979) -
`is often used as a measure of the extent of first(cid:173)
`pass metabolism. When several sites of first-pass
`metabolism are in series, the availability of the drug
`can be viewed as the product of the fractions of
`drug entering the tissue that escape loss in each
`successive tissue. Thus, the product (Foral) for an
`orally administered drug that is metabolised in the
`gastrointestinal lumen (G), the intestinal wall (I),
`liver (L), lung (Lu), arm (A), and blood and endo(cid:173)
`thelial linings or the vascular system (B) is
`
`(Eq. 1)
`
`arm and sampled in the other, the product is
`Fiv = FLu • FA • FB
`
`(Eq. 2).
`
`Combining equations 1 and 2 results in
`
`(Eq. 3).
`
`By definition, Fiv is conventionally assigned a value
`of unity. Thus, the bioavailability of an orally ad(cid:173)
`ministered drug is then the product of the fractions
`entering the irttestinal wall and escaping loss in the
`intestine and liver. It should be clear from equa(cid:173)
`tion 2 that the assignment of unity to Fiv is arbi(cid:173)
`trary. Consequently, the application of the term
`'absolute bioavailability' to the value obtained from
`comparison with intravenous administration may
`be inappropriate when elimination occurs in the
`lungs, arm, blood or vascular endothelium.
`
`1.3 Pharmacokinetic Considerations
`
`When the liver is the sole site of loss,
`
`Foral = 1 - E
`
`(Eq.4),
`
`where E is the extraction ratio, i.e. the fraction of
`drug entering the liver that is eliminated by the
`organ. Vnder steady-state conditions, the rate of
`extraction of drug by the liver [the product of the
`hepatic (H) blood clearance (CLHB) and the con(cid:173)
`centration of drug in blood (CB)] equals the rate of
`presentation of the drug to the liver [the product
`of liver blood flow (em) and CB]. Consequently,
`the extraction r!itio is, by definition:
`E = CLHB' CB
`QH' CB
`
`(Eq. 5).
`
`Therefore,
`Foral = 1 -
`
`(CLHB/QH)
`
`(Eq. 6).
`
`When intravenously (iv) administered in one
`
`Equation 6 indicates that the relative changes in
`the two pharmacokinetic parameters CLHB and F oral
`
`3
`
`

`
`First-Pass Elimination
`
`4
`
`will not be the same. For example, if clearance de(cid:173)
`creases from 99 to 97% of blood flow, a 2% de(cid:173)
`crease, the bioavailability increases from 0.01 to
`0.03, a 200% increase.
`Equations 5 and 6 use hepatic clearance calcu(cid:173)
`lated from drug concentrations in whole blood, CB.
`However, most clinical pharmacokinetic informa(cid:173)
`tion has been obtained from plasma drug concen(cid:173)
`trations. Plasma flow and plasma drug concentra(cid:173)
`tion of drug (Cp) cannot be substituted into
`equation 5 unless the drug is confined to plasma
`(Rowland, 1972), in which case Cp = CB • [1 -
`Haematocrit (Hct»). In all other cases the blood-to(cid:173)
`plasma concentration ratio must be determined.
`The relationship between blood and plasma drug
`concentrations depends on the haematocrit and the
`relative affinities of the drug for blood cells and
`plasma proteins. This conclusion can be derived
`from the principles of mass balance, in that
`
`Amount
`in blood
`
`==
`
`Amount
`in plasma
`
`V BC • CBC
`+ I
`i
`Amount in
`blood cells
`
`(Eq. 7),
`
`where VB, V p, and V Be are the volumes of blood,
`plasma, and red blood cells, respectively, and CB,
`Cp, and eBC are the concentrations of drug in these
`respective volumes; V Be/V B is the haematocrit and
`Vp/VB = I - Hct. Dividing by (VB· Cp) gives
`
`CB
`-
`Cp
`
`.
`
`CBC
`= (I-Hct) + Hct· -
`Cp
`
`(Eq. 8).
`
`The ratio ofbloodlplasma drug concentrations, CBI
`Cp, lies between the limits of(l - Hct) and a larger
`value determined by the relative affinities of drug
`for blood cells and for plasma proteins.
`Clearances based on drug in whole blood (CLB)
`and in plasma (CLp) are related by
`
`(Eq. 9).
`
`This relationship is obtained from the definition
`of clearance values, that is, rate of elimination =
`CLp • Cp = CLB • CB'
`Whether or not a drug undergoes extensive first(cid:173)
`pass elimination can be anticipated from plasma
`
`data when 4 parameters are known: the ratio of
`blood-to-plasma concentrations; plasma clearance
`(CLp); fraction of drug in the body that is excreted
`unchanged in urine (fe); and liver blood flow. Blood
`clearance (CLB) is calculated first using equation 9.
`The fraction undergoing non-renal elimination
`is (1 -
`fe)' If the liver is assumed to be the sole
`non-renal organ of elimination, the hepatic blood
`clearance (CLBB) is then
`
`(Eq. 10).
`
`By substituting equations 9 and 10 into equation
`6, bioavailability becomes
`
`Foral = I -
`
`(Eq. 11).
`
`(l-fe)· CLp
`QB • CB/Cp
`The importance of knowing the blood-to-plasma
`drug concentration ratio is illustrated by the. fol(cid:173)
`lowing example. Consider 2 drugs with plasma
`clearance values of 1500 ml/min, negligible excre(cid:173)
`tion in urine, and blood-to-plasma concentration
`ratios of 1.1/1 and 100/1. Assuming an hepatic
`blood flow of 1500 ml/min, the oral bioavailabil(cid:173)
`ities of the 2 compounds calculated from equation
`11 are 0.09 and 0.99, respectively. These values are
`greatly ditTerent even though the plasma clearances
`are identical.
`
`1.4 Non-Linear First-Pass Metabolism
`
`The bioavailability of somt! compounds changes
`with dose. For example, bioavailabilities of ribo(cid:173)
`flavine (vitamin B2) [Levy and Jusko, 1966] and
`cyanocobalamin (vitamin Bd (Diem, 1962) de(cid:173)
`crease with increasing doses because of capacity(cid:173)
`limited transport. Conversely, the bioavailability
`of propranolol increases as dose increases (Shand
`and Rangno, 1972) because of limited capacity of
`the metabolising hepatic enzymes (Walle et aI.,
`1981). Theoretically, saturable binding of drug to
`plasma proteins could also produce non-linear first(cid:173)
`pass metabolism.
`First-pass metabolism after oral doses may be
`non-linear when metabolism after intravenous
`doses, administered on separate occasions, is not.
`
`4
`
`

`
`First-Pass Elimination
`
`5
`
`The reason lies in the difference between the con(cid:173)
`centration of drug entering the liver after each route
`of administration. Consider the oral administra(cid:173)
`tion of a drug eliminated only in the liver and with
`I-compartment characteristics. Assuming that the
`absorption into the portal vein is first-order, the
`initial rate of input of drug is ka • dose, where ka
`is the absorption rate constant. The initial rate of
`entry of drug into the liver is Q,v • Cpv, where
`Q,v is the portal blood flow and Cpv is the drug
`concentration in the portal vein blood as it enters
`the liver. Anatomically, the hepatic artery adds to
`portal blood flow. So, the rate of entry of drug may
`be expressed as ~ • CA where ~ is the total liver
`blood flow and CA is the concentration of drug
`measured after dilution by hepatic arterial blood.
`The initial rate of presentation of drug to the liver
`after a single oral dose is
`
`(Eq. 12).
`
`On rearrangement, the initial concentration enter(cid:173)
`ing the liver is
`
`(Eq. 13).
`
`After a single intravenous dose (iv), the corre(cid:173)
`sponding concentration is
`
`(Eq. 14),
`
`where V d is the apparent volume of distribution
`of the drug. If the oral and intravenous doses are
`the same, the ratio of these resulting concentra(cid:173)
`tions is
`
`tration ratio is large only if absorption is fast. Thus,
`the potential for non-linear first-pass metabolism
`exists when absorption is fast and/or the apparent
`volume of distribution is large.
`Figure 1 was simulated when the oral and intra(cid:173)
`venous doses were given on separate occasions. It
`is important to note that if an intravenous radio(cid:173)
`labelled tracer dose is given concurrently with an
`oral dose, the intravenous kinetics will reflect the
`non-linear first-pass metabolism during absorp(cid:173)
`tion, but may be non-linear over too short a time
`to be measurable.
`
`Influence of Dosage Form
`The dosage form of a drug can have an impact
`on non-linear first-pass metabolism. From the ar(cid:173)
`gument above, the concentration entering the liver
`depends on the rate of entry of drug into the portal
`vein. The slower the rate of release from a dosage
`form, the lower the concentration entering the liver.
`Thus, slow absorption from a sustained release
`
`\
`
`Vd(L)
`
`~
`
`CA.oral
`
`CA.iv
`
`(Eq. 15).
`
`Absorption half-life (minutes)
`
`Figure 1, derived from equation 15, illustrates
`how this ratio varies with the absorption half-life
`(O.693/ka) and the apparent volume of distribution
`(V d). If V d is large (> 200L) the initial concentra(cid:173)
`tion entering the liver after an oral dose is much
`greater than that after an equivalent intravenous
`dose at all of the absorption half-lives shown. On
`the other hand, if V d is small « SOL) the concen-
`
`Fig. 1. The ratio of the initial drug concentrations entering the
`liver after an oral dose and after the same intravenous dose
`varies with the absorption half· life and the apparent volume of
`distribution (Vd). The shorter the absorption half-life and the larger
`the apparent volume of distribution, the greater the ratiO. This
`figure was obtained by simulation of equation 15; the absorption
`half-life = O.693/ka . The following assumptions were made: dis(cid:173)
`tribution is instantaneous, absorption and elimination are first(cid:173)
`order processes, and the liver is the sole site of elimination.
`
`5
`
`

`
`First-Pass Elimination
`
`6
`
`dosage form may not show non-linear bioavaila(cid:173)
`bility, whereas rapid absorption of the same dose
`given in solution or a rapidly dissolving dosage
`form may show non-linear bioavailability. This
`probably explains the much greater apparent bio(cid:173)
`availability of salicylamide given as a suspension
`than as a solid dosage form (fleckenstein et ai.,
`1976).
`
`1.5 Physiological Variables Involved in
`First-Pass Metabolism
`
`The extent of first-pass metabolism in the liver
`and intestinal wall depends on a number of phys(cid:173)
`iological factors. The major ones are enzyme ac(cid:173)
`tivity, plasma protein and blood cell binding, blood
`flow, and gastrointestinal motility.
`
`1.5.1 Hepatic First-Pass Metabolism
`
`Enzyme Activity
`The inherent enzyme activity is usually the most
`important determinant of the extent of first-pass
`metabolism. The activity is commonly expressed
`in terms of the Michaelis-Menten parameters, Vm
`(the maximum rate of metabolism) and Km (the
`concentration at which the rate of metabolism is
`Vm/2). The value of Vm is related to amount of
`enzyme present and the capacity per mole of en(cid:173)
`zyme. In classic enzyme kinetics
`Vm· S
`Km+S
`
`Rate of metabolism =
`
`(Eq. 16),
`
`models used to predict the dependence of F on flow
`are discussed in section 1.6.
`
`Plasma Protein Binding
`Increasing plasma protein binding increases
`bioavailability; the converse is also true. However,
`the extent of the changes is difficult to predict
`quantitatively. Two models used to predict the de(cid:173)
`pendence of bioavailability on protein binding are
`discussed in section 1.6; these models differ mark(cid:173)
`edly in their estimates of the degree of change. The
`theoretical predictions are difficult to verify in hu(cid:173)
`mans because factors that alter plasma protein
`binding usually also alter other physiological vari(cid:173)
`ables such as blood flow and hepatic enzyme ac(cid:173)
`tivity. For example, phenobarbitone increases the
`plasma protein binding of propranolol in the dog
`but also enhances hepatic metabolising enzyme ac(cid:173)
`tivity and probably increases liver blood flow (Bai
`and Abramson, 1982; Vu et ai., 1983).
`
`Gastrointestinal Motility
`Gastric emptying and intestinal motility affect
`the rate of absorption of drugs and may also influ(cid:173)
`ence the extent of first-pass metabolism in the liver
`if the metabolism is non-linear. Increasing gas(cid:173)
`trointestinal motility may increase the rate of de(cid:173)
`livery of drug to the liver. If metabolism is non(cid:173)
`linear, this increased delivery will result in in(cid:173)
`creased bioavailability. The converse is anticipated
`when gastric emptying is delayed or intestinal mo(cid:173)
`tility is diminished.
`
`where S is the substrate concentration. Saturability
`or non-linearity occurs when S approaches and ex(cid:173)
`ceeds the value of Km; the rate then approaches
`Vm. The relationship in equation 16 often ade(cid:173)
`quately explains the kinetics of drug metabolism
`in vitro, but there are many additional factors in
`vivo that must be considered, including blood flow
`and the supply of co-factors.
`
`Blood flow
`Increasing blood flow increases bioavailability;
`the converse is also true. The extent of the change
`depends on the extraction ratio of the drug. Two
`
`1.5.2 First-Pass Metabolism in the
`Gastrointestinal Mucosa
`When the primary site of first-pass metabolism
`is the intestinal mucosa, the expected direction of
`the effects of altered enzyme activity, blood flow,
`and plasma protein binding on bioavailability are
`similar to those for the liver. However, the reasons
`for the relationships are different. In the intestinal
`mucosa, drug diffuses to the enzymes, and drug
`and metabolites are removed by the blood. One
`might speculate that blood acts as a sink. As a con(cid:173)
`sequence, when blood flow is increased, drug may
`be removed from the mucosa to a greater extent,
`
`6
`
`

`
`First-Pass Elimination
`
`7
`
`'Well-stirred' model
`
`'Parallel tube' model
`
`0.20
`
`0.16
`
`~ 0.12
`is
`~ 0.08
`
`> '" iij 0.04
`
`0.00
`
`a
`
`0.20
`
`0.16
`
`~ 0.12
`is
`.!!!
`'iii
`
`> '" 0
`CD
`
`Intrinsic clearance (L/min)
`
`C
`
`Blood flow (L/min)
`
`0.20
`
`0,16
`
`~0,12
`
`is '" ~ 0.08
`> '" o
`iii 0.04
`
`~"
`
`0.06
`
`Intrinsic clearance (L/min)
`
`Blood flow (L/min)
`
`0.20
`
`0.16
`
`0.12
`
`0.08
`
`0.04
`
`0.00
`
`b
`
`0.20
`
`0.16
`
`0.12
`
`0.08
`
`0.04
`
`0.00
`
`d
`
`0.20
`
`0.12
`
`0.04
`
`e
`
`Fraction unbound
`
`o.oo-~=-""'~~~""'"":~~
`f
`
`Fraction unbound
`
`Fig.2. The 'well-stirred' and 'parallel tube' models (see text) of hepatic extraction of drugs differ in their prediction of the effects
`on bioavailability of changes in intrinsic clearance (figs a and b) at different fractions unbound (fu); blood flow (figs c and d) at different
`fractions unbound; and fraction unbound in blood (figs e and f) at different blood flows (0). Whet! not specified, intrinsic clearance
`and blood flow were given the values of 60 and 1.5 Llmin, respectively.
`
`7
`
`

`
`First-Pass Elimination
`
`8
`
`resulting in increased bioavailability. Decreased
`plasma protein binding may reduce the ability of
`blood to pick up the drug, thus reducing bioavail(cid:173)
`ability. The potential effects of altering gastroin(cid:173)
`testinal motility are unpredictable.
`
`1.6 Models of First-Pass Metabolism
`in the Liver
`
`Models that describe dependence of bioavaila(cid:173)
`bility on changes in enzyme activity, blood flow,
`and plasma protein and blood cell binding have
`been developed. Two that have been applied widely
`are the 'well-stirred' model (Rowland et aI., 1973;
`Wilkinson and Shand, 1975) and the 'parallel tube'
`model (Brauer, 1963; Winkler et aI., 1973, 1974).
`In a 'well-stirred' model, the liver is treated
`as a well-stirred container in which the unbound
`drug concentration in hepatic venous blood is the
`concentration available to the metabolising en(cid:173)
`zyme(s). In this model, the fraction of incoming
`drug that escapes metabolism in the liver (F d is
`QH
`QH + CLint' fUB
`where ~ is hepatic blood flow, CLint is the in(cid:173)
`trinsic clearance, which relates the rate of metab(cid:173)
`olism to the unbound drug concentration in he(cid:173)
`patic venous blood, and fUB is the unbound fraction
`of drug in blood.
`In the 'parallel tube' model, the liver is treated
`as a series of equivalent parallel tubes that have
`constant enzyme activity along the length of each
`tube. Drug concentration in perfusing blood de(cid:173)
`creases along the tube. The fraction escaping loss
`in this model is
`
`(Eq. 17),
`
`(Eq. 18),
`where fUB and Q; are as defined above, and CLint
`relates the rate of metabolism to the average un(cid:173)
`bound concentration within the tube.
`These models are similar in their predictions
`when bioavailability is large but differ dramatically
`when bioavailability is small. Figure 2 shows how
`bioavailability changes with enzyme activity, blood
`
`flow, and plasma protein binding. Note that the
`'parallel tube' model always predicts a much greater
`change in F than the 'well-stirred' model for a given
`change in intrinsic clearance, blood flow, or frac(cid:173)
`tion unbound.
`Because these two models summarise quanti(cid:173)
`tatively the expected relationships between bio(cid:173)
`availability and changes in enzyme activity, blood
`flow and protein binding, they may be useful.
`However, they must be tested for thdr validity, and
`drugs with low bioavailability due to extensive first(cid:173)
`pass hepatic metabolism provide tools for doing
`so. Similarities and differences between the two
`models have been examined (Ahmad et aI., 1983;
`Pang and Rowland, I 977a,b,c). The 'well-stirred'
`model describes the hepatic elimination of pro(cid:173)
`pranolol (Branch and Shand, 1976) and lignocaine
`(Pang and Rowland, 1977b,c). The 'parallel tube'
`model describes the elimination of galactose in the
`perfused pig liver (Keiding et aI., 1976; Keiding and
`Chiarantini, 1978).
`Without modification, these models are ex(cid:173)
`pected to be inadequate to predict bioavailability
`in vivo when metabolism is non-linear, hepatic
`blood flow is shunted, drug undergoes enterohe(cid:173)
`patic recycling, or metabolism occurs in other first(cid:173)
`pass organs.
`
`2. Clinical Consequences of First-Pass
`Elimination
`2.1 Route of Administration and
`Bioavailability
`
`2.1.1 Oral Administration
`Many clinically important drugs undergo con(cid:173)
`siderable first-pass metabolism after an oral dose.
`The characteristics of selected drugs that undergo
`50% or more first-pass metabolism after an oral
`dose are presented in table I. Although many of
`the drugs listed in the table undergo first-pass me(cid:173)
`tabolism in the liver, some are metabolised by in(cid:173)
`testinal flora or mucosa. Drugs metabolised in the
`intestine include levodopa, flurazepam, isoprena(cid:173)
`line, ~-methyldigoxin, oestrogen, phenacetin and
`salicylamide (George, 1981), and proscillaridin
`(Andersson et ai., 1977).
`
`8
`
`

`
`First-Pass Elimination
`
`9
`
`The major therapeutic implication of extensive
`first-pass metabolism is that much larger oral than
`intravenous doses are required to achieve equiv(cid:173)
`alent plasma concentrations. For some of these
`drugs, the extent of first-pass metabolism, at least
`in part, precludes their use as oral agents. Com(cid:173)
`pounds in this category include lignocaine, nalox(cid:173)
`one, glyceryl trinitrate (nitroglycerin), and dihy(cid:173)
`droergotamine. Despite its low bioavailability,
`dihydroergotamine can be used orally to treat some
`patients with orthostatic hypotension, probably be(cid:173)
`cause they have greater sensitivity to the drug's
`vasoconstricting effects (Bobik et aI., 1981).
`The lack of pharmacological activity and bio(cid:173)
`availability of oral naloxone (Fishman et aI., 1973)
`has been utilised recently in a novel formulation
`of pentazocine tablets (Talwin® Nx, package insert,
`Winthrop Laboratories, New York) to prevent their
`injection intravenously by drug abusers (Poklis and
`Mackell, 1982). The combination of pentazocine
`and naloxone taken orally has the desired analgesic
`effect. In contrast, no effect is experienced when
`the tablets are crushed and injected intravenously
`because the naloxone, now fully bioavailable, blocks
`the pentazocine effect.
`
`2.1.2 Transdermal Delivery
`Clinicians apply drugs topically to treat various
`skin diseases but have only recently begun to use
`the skin to reach the general circulation. Topical
`dosage forms of glyceryl trinitrate provide an ex(cid:173)
`ample of this latter use. Glyceryl trinitrate has a
`short elimination half-life (1.3 to 3.8 minutes), a
`large apparent volume of distribution (1.7 to 5.2
`L/kg), and a high plasma clearance (0.3 to 1.0 L/
`min/kg) [McNitT et aI., 1981]. Transdermal deliv(cid:173)
`ery of glyceryl trinitrate has been developed, in part,
`to avoid hepatic first-pass metabolism of an oral
`dose. However, the transdermally delivered drug
`still undergoes some first-pass metabolism in the
`skin (Wester et aI., 1981), blood, and vascular
`endothelium (Armstrong et aI., 1980; Fung and
`Kamiya, 1981; Wu et aI., 1981). The volume of
`blood and surface area of the vascular endothelium
`to which the drug is exposed are among the factors
`that dictate the extent of metabolism between the
`
`site of administration and the site of blood sam(cid:173)
`pling. When 2% glyceryl trinitrate ointment was
`applied to the skin of the left wrist, contralateral
`antecubital venous blood concentrations of gly(cid:173)
`ceryl trinitrate did not exceed 0.1 ng/ml (AzarnotT
`et aI., 1983). In contrast, blood from ipsilateral veins
`had mean peak concentrations of 12 ng/ml. These
`site-related ditTerences in apparent bioavailability
`are reflected by significant differences in the re(cid:173)
`sponse of normal subjects to glyceryl trinitrate when
`the same doses are applied to ditTerent body sites
`(Hansen et aI., 1979).
`
`2.1.3 Inhalation and Buccal Administration
`Societies have known for hundreds of years that
`inhalation of nicotine or opium produces the de(cid:173)
`sired effect, whereas oral administration either does
`not or requires much larger doses. Drugs given by
`inhalation include sympathomimetic agents and
`corticosteroids used to treat asthma. These are given
`by inhalation to deliver the drug directly to their
`sites of action. Some do undergo extensive first(cid:173)
`pass metabolism after oral administration, e.g. sal(cid:173)
`butamol (Evans et aI., 1973) and isoprenaline
`(Blackwell et aI., 1973). Because the lung plays only
`a small role in the biotransformation of these com(cid:173)
`pounds, inhalation of these drugs might be ex(cid:173)
`pected to give the metabolite pattern expected of
`an intravenous dose. However, after administra(cid:173)
`tion of 3H-salbutamol by inhalation of an aerosol
`form, 55% of the radioactivity in urine over the
`first 24 hours was recovered as metabolite, similar
`to the 61 % after oral administration; in contrast
`only 27% was recovered as metabolite after intra(cid:173)
`venous administration (Evans et aI., 1973). Inhaled
`isoprenaline also behaves pharmacokinetically like
`an oral dose, with the majority of the drug under(cid:173)
`going sulphation - not O-demethylation, the fate of
`an intravenous dose (Blackwell et aI., 1973). These
`data indicate that most of the drug inhaled as an
`aerosol is eventually swallowed. Presumably, most
`of the aerosol is trapped in the upper airway be(cid:173)
`cause of the large droplet size.
`Sublingual or buccal administration of a drug
`may produce blood concentrations similar to those
`of an equivalent intravenous dose. This similarity
`
`9
`
`

`
`-o
`
`
`
`~: o
`S'
`t!l
`I:l
`;;l'
`~
`';!I.
`
`I:'
`
`(1979); Pond et al. (1980)
`Ehrnebo et al. (1977); Neal et al.
`
`with cancer
`F determined in patients Brunk and Delle (197

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket