throbber
Second Edition
`
`APPLIED
`PHARMACOKINETICS
`Principles of Therapeutic Drug Monitoring
`
`Edited by
`
`William E. Evans, Pharm.D.
`Professor of Clinical Pharmacy & Pharmaceutics
`Chairman, Department of Clinical Pharmacy
`University of Tennessee
`Center for the Health Sciences
`Memphis
`and
`Director, Pharmaceutical Division
`St. Jude Children's Research Hospital
`
`Jerome J. Schentag, Pharm.D.
`Director, Clinical Pharmacokinetics Laboratory
`Millard Fillmore Hospital
`and
`Associate Professor of Pharmaceutics and Pharmacy
`State University of New York at Buffalo
`Buffalo
`
`William J. Jusko, Ph.D.
`Frofessor of Pharmaceutics and Pharmacy
`State University of New York at Buffalo
`Buffalo
`
`Editorial Assistant
`Hayes Harrison, M.A.
`Pharmaceutical Division
`Si. Jude Children's Research Hospital
`Memphis
`
`Applied Therapeutics, Inc.
`Spokane, WA
`1986
`
`2
`Guidelines for Collection
`and Analysis
`of Pharmacokinetic Data
`
`William J. Jusko, Ph.D.
`
`Efforts in both theoretical and applied pharmacokinetics over the
`past decade have emphasized the utilization of the principles of phys(cid:173)
`iological pharmacokinetics and the use of noncompartmental
`approaches to analysis of drug disposition data. Physiological phar(cid:173)
`macokinetics involves the deployment of pharmacokinetic models and
`equations based on anatomical constructions and functions such as
`tissue masses, blood flow, organ metabolism and clearance, specific
`drug input rates and sites, and processes of partitioning, binding, and
`transport. While the complete applications of physiologic systems
`analysis may require extensive models, 1 even the simplest of phar(cid:173)
`macokinetic treatments should have a physiologic basis for interpre(cid:173)
`tation. Noncompartmental techniques in pharmacokinetics can serve
`in this regard. This term applies to curve analysis methods of data
`treatment which do not require a specific model and which yield the
`prime pharmacokinetic parameters such as systemic clearance (CL)
`and steady-state volume of distribution (V8,) which summarize the
`major elimination and distribution properties.
`This chapter is intended to provide an overview of major components
`of experimentally-applied pharmacokinetics. A summary is provided
`of the most relevant concepts, models, equations, and caveats which
`may be useful in the design, analysis, and interpretation of pharma(cid:173)
`cokinetic studies. References are provided for more complete details
`of the assumptions, derivations, and applications of these guidelines
`and relationships. This material may be helpful as a checklist in de(cid:173)
`signing animal and (or) human experiments in pharmacokinetics and
`in reviewing drug disposition reports; with greater elaboration, it has
`served as a basis for a graduate course in physiological
`pharmacokinetics.
`
`9
`
`
`
`Apotex v. Novartis
`IPR2017-00854
`NOVARTIS 2041
`
`

`

`10
`
`Chapter 2: Collection and Analysis
`
`Chapter 2: Collection and Analysis
`
`11
`
`CONTEXT OF PHARMACOKINETICS
`A pharmacokinetic analysis must be made in context of, be consis(cid:173)
`tent with, and explain the array of basic data regarding the properties
`and disposition characteristics of the drug.
`The tasks of model and equation selection and interpretation of data
`require a fundamental appreciation and integration of principles of
`physiology, pharmacology, biochemistry, physicochemistry, analytical
`methodology, mathematics, and statistics. Pharmacokinetics has de(cid:173)
`rived from these disciplines, and the relevant aspects of many of these
`areas must be considered in reaching any conclusions regarding a par(cid:173)
`ticular set of data. The physicochemical properties of a drug such as
`chemical form (salt,ester,complex), stability, partition coefficient, pKa,
`and molecular weight can affect drug absorption, distribution, and
`clearance. A drug disposition profile must be correlated with studies
`of structure-activity, disposition in alternative species, perfused organ
`experiments, tissue or microsomal metabolism, tissue drug residues,
`disease-state effects, and pharmacology and toxicology. For example,
`a much larger LD50 for oral doses of a drug compared with parenteral
`administration may be indicative of either poor gastrointestinal ab(cid:173)
`sorption Clow aqueous solubility?) or a substantial first-pass effect.
`Drug metabolism pathways may differ between species, but the bio(cid:173)
`transformation rate (V mox and Km) of microsomes, homogenates, or
`perfused organs can often be applied directly to whole-body disposition
`rates and often correlate between species. 1·3
`In general, the pharmacokinetic model and analysis should either
`conform to, or account for, the known properties and accumulated data
`related to the drug. One set of disposition data may misrepresent the
`characteristics of the drug because of any one or combination of rea(cid:173)
`sons. Experienced judgment is usually required in the final interpre(cid:173)
`tation of any experimental findings and analysis.
`
`ARRAY OF BASIC DATA
`
`Pharmacokinetic studies often serve to answer specific questions
`about the properties of a drug. For example, a limited experimental
`protocol can easily resolve the question of how renal impairment af(cid:173)
`fects the systemic clearance of an antibiotic. In the total design and
`implementation of pharmacokinetic studies, an ideal and complete
`array of experimental data should include a number of considerations:
`
`A. The dosage form should be pre-analyzed. All calculations stem
`from knowledge of the exact dose given [e.g., CL = dose/ AUC, (area
`under the plasma concentration-time curve)]. Most commercial dosage
`forms are inexact, and content uniformity should be examined. Vials
`
`or ampules of injectables typically contain some overage and require
`analysis or aliquoting for administration of a precise dose. Solid dosage
`forms are required to yield an average of the stated quantity of drug
`with limited variability, but both injectable and solid forms may be
`inaccurate for pharmacokinetic purposes. Manninen and Koriionen4
`provide an excellent example of both the variability and lack of stated
`quantity of digoxin in many commercial tablets. One product contained
`a range of 3_9% to 189% of the stated 0.25 mg dose of digoxin, while
`the most umform product, Lanoxin, exhibited a range of about 95% to
`106% for one batch of drug. To evaluate the potential uncertainty of
`the dose of drug used in disposition studies, it may be necessary to
`collect and analyze replicate doses of the product used. Poorly soluble
`drugs are susceptible to erratic formulation.
`B. Accuracy in administration of the dose should be confirmed. All
`doses should be timed exactly for starting time and duration of admin(cid:173)
`i~tration. For ease in subsequent calculations, pharmacokinetic equa(cid:173)
`tions can be used, to correct data from short-term infusion studies to
`the intercepts expected after bolus injection. The particular materials
`used in drug administration may cause loss of drug. In one of the most
`dramatic examples, MacKichan et al.5 found immediate loss of about
`50% of a dose of i.ntravenous diazepam by adsorption during passage
`through the plastic tubing of an infusion set. Inline filtration can also
`significantly reduce the potency of drugs administered intravenously
`in small doses. 6
`C. Attention to methods and sites of blood collection is needed. Ideally,
`blood sa~ples sho~ld be collected by direct venipuncture in clean glass
`t~bes without anticoagulant. Otherwise, the presence of possible ar(cid:173)
`tifacts should be tested. In the absence of any in vitro artifacts, serum
`and plasma concentrations are usually identical, and these terms are
`commonly used interchangeably. However, there are several reasons
`that they ~a~ not be identical. For example, the presence of heparin
`can result m mcreased free fatty acid concentrations causing altered
`plasma-protein binding. 7 Also, the type of blood collection tube or an(cid:173)
`ticoa~lant may be a factor.8 If protein binding is temperature depen(cid:173)
`den:, it may be. necessary to centrifuge the blood sample at 37°C to
`avoid changes m red cell-plasma distribution of some compounds.9
`The~e ~roble~s primarily pertain to weak bases such as propranolol
`and 1m1p~amme for which binding to cx1-acid glycoprotein is apprecia(cid:173)
`ble and displacement alters plasma-red cell drug distribution.
`Plasma or serum protein binding and red cell partitioning should
`be ~easured at 37°C over the expected range of plasma drug concen(cid:173)
`!rat1ons. Both _ra~e and degree of binding and uptake are theoretically
`1~portant. Th1_s m.formation may be especially needed for interpreta(cid:173)
`tion or normahzat1on of nonlinear disposition patterns.
`
`
`
`

`

`12
`
`Chapter 2: Collection and Analysis
`
`Chapter 2: Collecti.on and Analysis
`
`13
`
`Sometimes the site of blood collection and the presence of a tourni(cid:173)
`quet can alter the composition of the blood sample: serum proteins,
`calcium, and magnesium concentrations rise by 5% to 13% during ven(cid:173)
`ous stasis.10
`One of the major assumptions employed in most pharmacokinetic
`studies is that venous blood collected from one site adequately reflects
`circulating arterial blood concentrations. For practical purposes, ven(cid:173)
`ous blood samples are usually collected. The pharmacokinetic analysis
`may need to be somewhat qualified, because arterial and capillary
`blood concentrations may differ markedly from venous blood concen(cid:173)
`trations of many drugs.11 The AUC of arterial versus venous blood is
`expected to be identical for a non-clearing organ, and thus the prin(cid:173)
`cipal difference expected is in distribution volumes. Physiologically,
`organ uptake of drugs occur from the arterial blood, and clearance
`organ models are based on arterial-venous extraction principles.
`D. Serum (or blood) concentration data following intravenous in(cid:173)
`jection (bolus or infusion) provides partial characterization of drug
`disposition properties. Accurate assessment of volumes of distribution,
`distribution clearance (CL0 ), and systemic clearance (CL) can best be
`attained with intravenous washout data.
`E. Serum (or blood) concentration data following oral doses of the
`drug in solution and common dosage forms provides additional phar(cid:173)
`macokinetic parameters related to absorption and intrinsic clearance.
`The doses (or resultant serum or blood concentrations of drug) should
`be comparable to those from the intravenous dose. These data permit
`assessment of either oral clearance (CL0 • 01) or bioavailability (F), and
`of the transit time for absorption (t0 ). If relevant, other routes of
`administration should be studied. For these, the FDA guidelines for
`bioavailability studies should be consulted.12
`F. Three dosage levels (both oral and intravenous) should be admin(cid:173)
`istered to span the usual therapeutic range of the drug to permit as(cid:173)
`sessment of possible dose-dependence (nonlinearity) in absorption, dis(cid:173)
`tribution, and elimination.
`G. Urinary excretion rates of drug (as a function of time, dose and
`route of administration) should be measured to accompany the above
`studies. Urinary excretion is often a major route of drug elimination,
`and analyses permit quantitation of renal clearance (CLR). Collection
`of other excreta or body fluids (feces, bile, milk, saliva) may permit
`determination of other relevant elimination or distributional
`pathways.
`H. Many drug metabolites are either pharmacologically active or
`otherwise of pharmacokinetic interest. Phase I products such as hy(cid:173)
`droxylated or demethylated metabolites are most commonly either ac(cid:173)
`tive or toxic. 13 Their measurement will allow evaluation of AUC and
`
`transit time and perhaps permit quantitation of metabolite formation
`and disposition clearances.
`I. Multiple-dose and steady-state experiments are necessary if ther(cid:173)
`apeutic use of the drug relies on steady-state concentrations. The du(cid:173)
`ration of multiple-dosing in relation to the terminal half-life is crucial
`for ascertaining applicability to steady-state conditions. Comparative
`single- and multiple-dose studies permit further assessment of line(cid:173)
`arity and (or) allow determination of chronic or time-dependent drug
`effects such as enzyme induction, 14 unusual accumulation, 15 or drug(cid:173)
`induced alterations in disposition. For example, aminoglycoside up(cid:173)
`take into tissues is extremely slow and difficult to assess from single(cid:173)
`dose studies. Multiple-dose washout measures (Figure 2-1) led to ob(cid:173)
`servation of a slow disposition phase which was the result of tissue
`accumulation and release.16
`J. Tissue analyses add reality and specificity to drug distribution
`characteristics. Comprehensive studies in animals permit detection of
`unusual tissue affinities while generating partition coefficients (K ;)
`for individual tissues (Vi;). This can lead to complete physiologic
`models for the drug in each species studied. 1•2 Autopsy or biopsy stud-
`
`Cenlral
`
`Dose
`
`Comp1r1meen1 (
`
`Tisoue
`
`Uplake Q Compar1menl
`
`_, .
`
`Release
`
`•
`
`-
`i Renal Clearance
`
`r-
`
`.
`
`5.0
`
`I :i.
`1
`~
`E t; 1.0
`
`C
`0
`(.)
`
`C ·o ·e ~ 0.4
`C .. C,
`
`I
`I
`
`0.2
`
`E
`:3
`Qi
`en
`
`t 80 mg
`t
`Doses
`0.1 ~-!--f-~"---~--:~~--L-..........;L.::-...:.i........:.:..:1---:.1....:.::::~
`4
`12
`0
`2
`6
`8
`10
`14
`16
`20
`22
`
`Time, Days
`
`~IGURE ~-1. Pla~ma_ concentration-time profile for gentamicin disposition dur(cid:173)
`ing multipl~ dosm~ 1~ a patient showing the prolonged terminal phase caused
`by strong tissue bmdmg. These data were characterized with a two-compart(cid:173)
`ment m_odel (inset) which included prediction of drug remaining in the body
`at the time of death of the patients. Data from reference 15.
`
`
`
`

`

`14
`
`Chapter 2: Collection and Analysis
`
`Chapter 2: Collection and Analysis
`
`15
`
`ies in man may extend or complement pharmacokinetic expectations.
`This approach was found to be extremely helpful (Figure 2-2) in con(cid:173)
`firming the strong tissue binding of aminoglycosides in man which
`was anticipated on the basis of serum concentration profiles (Figure
`2-1).16
`K. Suitable drug disposition studies in patients with various dis(cid:173)
`eases and ages or given secondary drugs form the basis of clinical
`pharmacokinetics. Perturbations in organ function, blood flow, or re(cid:173)
`sponse will often alter drug disposition in a way that may warrant
`quantitative characterization. General principles may not always ap(cid:173)
`ply, and each drug needs individualized study. For example, while
`hepatic dysfunction may diminish the rate of oxidation of many drugs,
`some compounds such as oxazepam and lorazepam are predominantly
`metabolized by glucuronide conjugation, a process largely unaffected
`by liver diseases such as cirrhosis.17 Each disease state may require
`evaluation of direct effects on pharmacokinetic processes such as
`changes in renal clearance caused by kidney disease. However, indirect
`changes also require attention such as the effects on both distribution
`and clearance caused by altered plasma protein binding. 18 Finally,
`commonly encountered patient factors such as smoking habit19 and
`
`30()~
`
`CJ)
`E
`:,.-"
`
`._
`
`8 CD
`~ 200 ~
`.....
`z
`
`•• •
`•
`•
`
`•••• •
`
`~ i ,oo~
`
`0
`UJ
`
`t3
`15
`w a:
`
`CL
`
`-
`
`_._.
`• o• 0
`
`100
`
`200
`
`I
`300
`
`'
`MEASURED AMOUNT IN Tl-IE BODY, mg
`
`FIGURE 2-2. Correlation of gentamicin accumulation in the body determined
`by pharmacokinetic analysis of serum concentration data (see Figure 2-1) and
`by direct analysis of body tissues obtained at autopsy from the same patients
`who were evaluated pharmacokinetically prior to death. Dotted line indicates
`perfect correlation. Data from references 15 and 16.
`
`obesity may cause unusual changes in drug disposition and require
`specific study and notation in patient surveys.
`L. Many questions of drug disposition can be resolved from selected,
`carefully designed studies, and alternative types of information may
`be sufficient to validate various assumptions and permit some experi(cid:173)
`mental procedures to be omitted. However, it is the investigator's ob(cid:173)
`ligation to adequately assess the literature, to avoid unwarranted as(cid:173)
`sumptions, and to satisfy the demands of experimental strategies that
`would resolve a proposed hypothesis.
`
`DRUG ASSAYS
`
`Certainty in measurement of drugs and their metabolites is a sine
`qua non in pharmacokinetics and deserves considerable attention.
`Guidelines for quality assurance in laboratory analyses have been con(cid:173)
`cisely sumITJarized by the American Chemical Society.20 It is now com(cid:173)
`monplace to report the linearity, the coefficient of variation of the
`assay at low and high drug concentrations, the minimum level of de(cid:173)
`tection, and the procedures used to assure specificity and stability,
`especially in the presence of metabolites, secondary drugs, and in spec(cid:173)
`imens from diseased patients. Microbiological assays are notoriously
`unreliable. Other antibiotics often interfere in detection of the main
`drug. Active metabolites such as the desacetyl form of some
`cephalosporins21 may be included in the measurements unless prior
`separation is made of the two active compounds. An extreme case of
`metabolite inclusion is in the use of radioisotopic tracers; total radio(cid:173)
`isotope counts generally yield total drug and metabolite activity and
`possibly the products of radiolysis. Separation of parent drug and in(cid:173)
`dividual metabolites is required for pharmacokinetic specificity. Mi(cid:173)
`crobiologic, enzymatic, and radioimmunoassays are often of uncertain
`specificity, and matrix effects may require preparation of standards
`in each patient's pretreatment plasma. Most drug companies provide
`analytical-grade samples of their drugs (and sometimes metabolites)
`to qualified investigators upon written request.
`Sample Handling. Coupled with assay reliability is concern for the
`stability of drug in biological specimens, even in the frozen state. Am(cid:173)
`picillin is unusual in that it is less stable frozen than when refriger(cid:173)
`ated. 22 Some drug esters such as hetacillin (a prodrug of ampicillin)
`continue hydrolyzing in blood and during the bioassay, and imprudent
`handling of the blood specimens can confound the true disposition
`profiles of both prodrug and drug. Penicillamine is unstable in the
`presence of plasma proteins, and immediate deproteination after blood
`sample collection is necessary to avoid loss of reduced penicillamine
`prior to analysis.23 Measurement of drug stability in blood will com(cid:173)
`plement the pharmacokinetic characterization of drug by revealing
`
`
`
`

`

`16
`
`Chapter 2: Collection and Analysis
`
`Chapter 2: Collectwn and Analysis
`
`L1
`
`whether hydrolysis can occur in blood or whether exposure to other
`body organs is required. Additional concerns in handling samples from
`a pharmacokinetic study include labeling and record-keeping proce(cid:173)
`dures and documentation of specimen storage conditions.
`Sample Timing. Appropriate pharmacokinetic evaluation requires
`properly timed specimens. The simplest and least ambiguous experi(cid:173)
`ment is the determination of systemic plasma clearance during con(cid:173)
`tinuous infusion at steady state:
`CL = k0
`(Eq. 2-1)
`/ C._,
`where k0 is the infusion rate and c •• is the steady-state plasma or
`serum concentration. For this equation to apply, the infusion period
`must be sufficiently long (about five terminal disposition half-lives) to
`allow steady state to be attained. Alternatively, a loading dose or
`short-term infusion may be administered to more rapidly achieve
`equilibrium. 24
`Practical and cost-effective methods are available for designing op(cid:173)
`timal sampling strategies for kinetic experiments where, such as in
`the clinic, the number of specimens is limited.25 Optimal designs
`largely depend on the likely "true" model parameter values, the struc(cid:173)
`ture of the model, and the measurement error. A sequential approach
`has been advocated with pilot studies and a sampling schedule which
`distributes time points over the major phases of drug disposition as
`the first step. Subsequent experiments can then be designed to resolve
`a specific hypothesis.
`A common and severe problem in applied pharmacokinetics is the
`inadequate or incomplete measurement of drug washout from the sys(cid:173)
`tem, either because of premature termination of sample collection or
`because of analytical limitations. The "true" terminal disposition
`phase must be examined in order for most aspects of data treatment
`and interpretation to be accurate. For example, the early distributive
`phase of aminoglycoside disposition measured by bioassay had long
`been accepted as the only phase, yet more sensitive radioimmunoas(cid:173)
`says, lengthier sample collection, and evaluation of multiple-dose
`washout revealed the slower phase of prolonged drug release from
`tissues (Figure 2-1).
`The two summary physiologic parameters in pharmacokinetics,
`namely systemic clearance and steady-state volume of distribution,
`can be most easily calculated by use of the area under the plasma
`concentration-time curve (AUC) and the area under the moment curve
`(AUMC). Both area values require extrapolation of plasma concentra(cid:173)
`tions to time infinity, and the AUMC is, in particular, prone to exag(cid:173)
`gerated error from an inaccurate terminal slope.26 If analytical or
`ethical constraints limit blood sample availability, extended saliva or
`urine collection may aid in defining the terminal disposition slope
`
`earance =
`Cl
`
`(Eq. 2-2)
`
`clearance is linear), as the sample volume is large and u:~ (if renal
`
`.
`while adding one or two other pharmacokinetic par
`analysis. Urine may be particularly useful in this r:;e~r~ to the

`f
`ne concen-
`trations o ten exceed plasma values by one or more ord
`magnitude.
`ers of
`The "midpoint" (C,.v) is generally the most desirable time to collect
`blood samples to match an excretion interval in order to assess a time(cid:173)
`dependent clearance process:
`Excretion Rate Amount Excreted
`= - ----,---c=--
`-
`Cav
`AUC
`The arithmetic mean time is acceptable for slow processes, but errors
`will be incurred if the kinetic process produces rapid changes in
`plasma concentrations.27 It is common to miss an early exponential
`phase of drug disposition because of infrequent blood sampling. For a
`polynomial curve with intercepts C; and slopes >-.;, the total AUC is:
`AUC = lC;lt..;
`(Eq.2-3)
`If the _initial distributive phase is missing (area = C1 / >,.1), then the
`error mcurred in calculation of a clearance parameter (CL = dose/
`AUC) is
`
`% of CL error = 100 x (C 1 / A1) / AUC
`
`(Eq.2-4)
`
`BASIC PHYSIOLOGIC PARAMETERS
`The evolution of complete physiologic models1 and clearance con(cid:173)
`cepts applied to perfused organ systems,28•29 with the restrictions in(cid:173)
`curred by the limited in vivo visibility offered by most blood or plasma
`dru~ disposition profiles, has led to the employment of partial physi(cid:173)
`ologic models for description of pharmacokinetic data. One such model
`is shown in Figure 2-3. Its construction and use should be viewed with
`some conceptual flexibility, and this material will apply to linear pro(cid:173)
`cesses unless stated otherwise.
`Volumes. The drug in blood or plasma (Cp) is considered to be part
`of the central comp.artment (Ve). The minimum value of Ve is plasma
`volume (Vp), but, either because drug diffuses rapidly out of plasma
`
`V' the number of early time data are limited, the Ve value often exceeds
`
`p·
`. Drug which is located outside of VP or Ve is, of course, present in
`tiss~es. The ~pparent volume of the tissue compartment (VT) has two
`basic determinants: physiologic weight or volume of each tissue (V .)
`and pa~tition or distribution factors (Kp;). In analysis of plasma co~(cid:173)
`c.entrat_1on-time profiles, tissues must commonly be clustered together
`(mcludmg the clearing organs) thus:
`
`(Eq.2-5)
`
`
`
`

`

`18
`
`Chapter 2: Collection and Analysis
`
`Chapter 2: Collection and Analysis
`
`19
`
`This equation leads to definition of one of the primary pharmacoki(cid:173)
`netic parameters with a physiologic basis, volume of distribution at
`steady state (V55):
`
`v •• = V, + VT
`If plasma and tissue binding are the sole determinants of nonhomo(cid:173)
`geneous distribution of drug in the body, then one definition of v •• is
`fup V
`v .. =VP+ f
`' T
`ut
`where fup and fut are the fractions of drug unbound in plasma and
`tissue.30 Other factors may also contribute to the apparent partition
`coefficient of drugs between tissues and plasma (Table 2-1). Since, by
`definition, VP and ~Vti comprise total body weight (TBWt)
`TBWt = V., + l Vt1
`
`(Eq. 2-6)
`
`(Eq.2-7)
`
`(Eq.2-8)
`
`then the quotient of
`
`K 0 = V., / TBWt
`(Eq.2-9)
`defines the distribution coefficient (K0 ), a physicochemical and phys(cid:173)
`iological measure of the average tissue:plasma ratio of the drug
`throughout the body. Approximate values of K0 and the primary ra(cid:173)
`tionalization of the size of K0 are provided in Table 2-2 for several
`common drugs. Normalization of v •• for TBWt is thus of value for
`
`generating the K0 and for making interindividual interspecies2 com(cid:173)
`parisons of this parameter.
`One qualification of Vss is needed. Drug equilibration between
`plasma and tissue of a clearing organ is affected by blood flow (QH)
`and intrinsic clearance (CLint).31 For hepatic tissue, this yields the
`following relationship between the true partition coefficient (Kph) and
`the lower, apparent value which would be experimentally measured
`at steady state (K~tP):
`
`K = Kcxp (1 + CL;"')
`
`ph
`
`ph
`
`QH
`
`(Eq. 2-10)
`
`Distribution Clearance. The least developed and appreciated ele(cid:173)
`ment of the basic pharmacokinetic properties of drugs is the distri(cid:173)
`bution clearance (CL0 ) or intercompartmental clearance. This term
`reflects the flow or permeability property of drugs between plasma
`and tissue spaces. The simplest assumption made in constructing a
`generalized model is that distribution clearance is equal in both di(cid:173)
`rections in and out of tissues.
`Renkin has characterized distribution clearance in terms of trans(cid:173)
`capillary movement of small molecular weight substances.32 The
`model proposed is depicted in Figure 2-4. Drug transfer from blood to
`tissues is represented by flow down a cylindrical tube (Q) with perme(cid:173)
`ability (P) determined by diffusion across the capillary. Distribution
`clearance is thus defined by flow and permeability according to the
`following relationship:
`
`(Eq. 2-11)
`
`Compounds with high tissue permeability will exhibit a limiting CL0
`of Q, while those with low permeability are limited by P. These con(cid:173)
`cepts have been applied by Stec and Atkinson33 to a multicompartment
`model of procainamide and NAPA disposition and used to predict the
`extent of hemodynamic changes caused by hemodialysis. The flow or
`
`TABLE 2-1. PHYSIOLOGICAL DETERMINANTS OF DRUG PARTITION OR
`DISTRIBUTION RATIOS BETWEEN TISSUES AND PLASMA
`
`Active transport
`Donno-n ion effect
`pH differences
`Plasma protein binding
`Tissue binding
`Lipid partitioning
`
`
`
`Dpo
`I
`I
`I
`
`....
`
`!
`
`OH
`
`Div
`I
`I
`
`'
`
`\l
`
`Q
`CL0
`
`·o rn
`
`I
`I
`-.!L
`Kp I
`Vt
`C ~
`I
`I
`
`Clint
`
`I -1,.,
`I~
`I 'V
`
`I
`
`1 r
`I
`
`cvh
`
`Cvt
`
`FIGURE 2-3. Basic physiologic pharmacokinetic model for drug distribution
`and elimination. Symbols are defined in the text. The clearance organ is phar(cid:173)
`macokinetically perceived as separate from other compartment s for drugs with
`high intrinsic clearances (CL;n,) allowing characterization of the first-pass
`input.
`
`

`

`20
`
`Chapter 2: Collection and Analysis
`
`Chapter 2: Collection and Analysis
`
`21
`
`permeability coefficient can be calculated for drugs exhibiting polyex(cid:173)
`ponential disposition and is of more fundamental value than intercom(cid:173)
`partmental rate constants.
`Hepatic Clearance. The model shown in Figure 2-3 represents the
`common situation where drug must pass through a specific organ such
`as the liver or kidney for elimination. It does not apply to enzymatic
`hydrolysis in blood. This type of model reflects the dual role of blood
`flow (Q) and either biotransformation (V max, Km) or renal filtration
`(GFR) and transport (Tmnx, Tm) on removal of drug from the body and
`allows for some effects of route of administration (e.g., first-pass
`effect).
`Two types of clearing organ models are commonly used for hepatic
`TABLE 2-2. DISTRIBUTION COEFFICIENTS (Ko) FOR VARIOUS DRUGS AND PROB·
`ABLE PHYSIOLOGIC (PHYSICOCHEMICAL) CAUSE
`
`Drug
`
`Indocyanine Green
`
`Inulin
`
`Ampicillin
`
`Theophylline
`
`Antipyrine
`
`Gentamicin
`
`Tetracycline
`
`Diazepam
`
`Digoxin
`
`Ko=~
`TBWt
`
`0.06
`
`0.25
`
`0.25
`
`0.5
`
`0.6
`
`1.1
`
`1.6
`
`1.7
`
`8.0
`
`Imipramine
`
`10.0
`
`Explanation/indication
`
`Strong binding to plasma
`proteins and limited extra(cid:173)
`vascular permeability.
`Distribution limited to
`plasma and interstitial
`,fluid owing to large molecu(cid:173)
`lar weight (5,500) and lipid
`insolubility.
`Limited intracellular distri(cid:173)
`bution owing to poor lipid
`solubility (common to
`penicillins).
`
`Moderate plasma binding and
`distribution primarily into
`total body water.
`Slight plasma binding and
`fairly uniform distribution
`into total body water.
`Strong tissue binding
`(common to
`aminoglycosides).
`Strong tissue binding to cal(cid:173)
`cium in bone.
`Appreciable lipid
`partitioning.
`Strong binding to Na/K
`transport ATPase in cell
`membranes.
`Strong tissue binding
`(common to weak bases).
`
`elimination: The "Jar" or ''Venous Equilibrium" Model28 (Figure 2-5)
`and the "Tube" Model34 (Figure 2-6). Both include blood flow for sys(cid:173)
`temic drug access to the organ and, as shown in the figures, assume
`that free or unbound drug (fup) in plasma equilibrates with free drug
`in the tissue available to enzymes. The Jar Model involves the as(cid:173)
`sumption that drug in arterial blood (C 0 ) entering the clearing organ
`instantaneously equilibrates with that in the venous blood (CJ. The
`Tube Model assumes that a drug concentration gradient exists down
`the tube, with enzymes acting upon declining perfusate drug
`concentrations.
`The Jar Model yields the following relationship for hepatic clearance
`to account for the variables in the system:
`CL - QH . fup . CLuinl - Q
`QH + fup • CLuinl
`where intrinsic clearance is the ratio of V max / Km for linear biotrans(cid:173)
`formation and EH is the extraction ratio.
`The corresponding equation for CLH described by the Tube Model is
`(Eq. 2-13)
`A simulation is provided in Figure 2-7 to depict the dual effects of
`
`H -
`
`-
`
`. E
`H
`
`t!
`
`(Eq.2-12)
`
`) Q
`
`C >
`
`V
`
`Cone
`
`Distance
`
`FIGURE 2-4. Model for distribution clearance where blood flow (Q) along the
`cylindrical tube and capillary permeability (Pl are the primary determinants
`of drug loss from arterial blood (C0 ). Drug concentration in the tube will de(cid:173)
`cline monoexponentially according to distance (length) along the tube emerg(cid:173)
`ing at the venous concentration (Cvl·
`
`
`
`

`

`22
`
`Chapter 2: Collection and Analysis
`
`Chapter 2: Collectwn and Analysis
`
`23
`
`blood flow and intrinsic clearance on hepatic clearance for the two
`clearance organ models. Both models predict a lower CLH limit of fup
`· CLuint (or CLint in the absence of protein binding considerations) and
`an upper value of QH. Thus, the hepatic clearance of low clearance
`drugs is essentially equal to the product of intrinsic clearance and the
`fraction unbound in plasma.35 The maximum hepatic clearance will
`be organ blood flow. As seen by' the shape of the surfaces in Figure 2-
`7, the two models diverge somewhat in characterizing drugs with in(cid:173)
`termediate to high clearance. At the present time, it is uncertain
`which model is most generally appropriate for describing organ clear(cid:173)
`ance. The Jar Model has had most extensive use in physiological phar(cid:173)
`macokinetics.1 A spherical liver acinus model has also been proposed.36
`Radial diffusion considerations in this model produce plasma disposi(cid:173)
`tion curves of the power form C" = At·n, rather than polyexponentials,
`which can be fitted well to elimination of several drugs.
`The organ clearance models provide definitions for two types of gen(cid:173)
`eral clearance terms. Systemic clearance (CL) reflects any situation
`where

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