Pharmacokinetics and Pharmacodynamics for Medical … · Greenblatt and Abourjaily 1185 Table 2....

16
Core Entrustables in Clinical Pharmacology: Pearls for Clinical Practice The Journal of Clinical Pharmacology 2016, 56(10) 1180–1195 C 2016, The American College of Clinical Pharmacology DOI: 10.1002/jcph.732 Pharmacokinetics and Pharmacodynamics for Medical Students: A Proposed Course Outline David J. Greenblatt, MD, 1 and Paul N. Abourjaily, PharmD 2 Keywords pharmacokinetics, pharmacodynamics, drug interactions, medical The discipline of pharmacokinetics (PK) applies math- ematical models to describe and predict the time course of drug concentrations and drug amounts in body fluids. 1–3 Pharmacodynamics (PD) applies sim- ilar models to understand the time course of drug actions on the body. Clinicians are most concerned with pharmacodynamics—they want to know how drug dosage, route of administration, and frequency of ad- ministration can be chosen to maximize the probability of therapeutic success while minimizing the likelihood of unwanted drug effects. However the path to pharma- codynamics comes via pharmacokinetics. Because drug effects are related to drug concentrations, understand- ing and predicting the time course of concentrations can be used to help optimize therapy. The link of drug dosage to drug effect involves a sequence of events (Figure 1). Even when a drug is administered directly into the vascular system, the drug diffuses to both its pharmacologic target receptor and to other peripheral distribution sites where it does not have the desired activity but may exert toxic ef- fects. Simultaneously, the drug undergoes clearance by metabolism and excretion. After oral administration, the situation is more complex, since the drug must undergo dissolution and absorption, then survive first- pass metabolism in the liver, before reaching the sys- temic circulation. Pharmacokinetics provides a ratio- nal mathematical framework for understanding these concurrent processes, and facilitates achieving optimal clinical pharmacodynamic effects more efficiently than trial and error alone. The following 3 clinical vignettes illustrate how familiarity with principles of pharmacokinetics and pharmacodynamics can facilitate optimal understand- ing and prediction of drug effects in human subjects and patients. Clinical Vignettes Case 1 A 30-year-old man has been extensively evaluated for recurrent supraventricular tachycardia (SVT), which is associated with palpitations and dizziness. No iden- tifiable cardiac disease is evident, and other medical diseases have been excluded. The treating physician elects to start therapy with digitoxin, 0.1 mg daily. One week later the patient is seen again, and states that episodes of SVT are reduced in number. The plasma digitoxin level is 8 ng/mL (usual therapeutic range, 10–20 ng/mL). The dose is increased to 0.2 mg/day. At a follow-up visit 7 days later, the patient claims that symptoms attributable to SVT have disappeared completely. The plasma digitoxin level is 17.4 ng/mL. The patient continues on 0.2 mg/day of digitoxin. One month later the patient sees the physician on an urgent basis. He has diminished appetite and waves 1 Program in Pharmacology and Experimental Therapeutics, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA 2 Departments of Pharmacy and Medicine, Tufts Medical Center, Boston, MA, USA Submitted for publication 26 February 2016; accepted 3 March 2016. Corresponding Author: David J. Greenblatt, MD, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 Email:[email protected] This proposal was prepared under the guidance of the Special Com- mittee for Medical School Curriculum of the American College of Clinical Pharmacology. The proposal is intended as a resource for the Association of American Medical Colleges as it revises its Core Entrustable Professional Activities for Entering Residency, Curriculum Developer’s Guide.

Transcript of Pharmacokinetics and Pharmacodynamics for Medical … · Greenblatt and Abourjaily 1185 Table 2....

Page 1: Pharmacokinetics and Pharmacodynamics for Medical … · Greenblatt and Abourjaily 1185 Table 2. Continued Section 6.Clearanceofdrugsandmechanismsofelimination25 A.Model-independentdefinition(Figure6):

Core Entrustables in Clinical Pharmacology: Pearls for Clinical Practice

The Journal of Clinical Pharmacology2016, 56(10) 1180–1195C© 2016, The American College ofClinical PharmacologyDOI: 10.1002/jcph.732

Pharmacokinetics and Pharmacodynamicsfor Medical Students: A ProposedCourse Outline

David J. Greenblatt,MD,1 and Paul N.Abourjaily, PharmD2

Keywords

pharmacokinetics, pharmacodynamics, drug interactions, medical

The discipline of pharmacokinetics (PK) applies math-ematical models to describe and predict the timecourse of drug concentrations and drug amounts inbody fluids.1–3 Pharmacodynamics (PD) applies sim-ilar models to understand the time course of drugactions on the body. Clinicians aremost concerned withpharmacodynamics—they want to know how drugdosage, route of administration, and frequency of ad-ministration can be chosen to maximize the probabilityof therapeutic success while minimizing the likelihoodof unwanted drug effects. However the path to pharma-codynamics comes via pharmacokinetics. Because drugeffects are related to drug concentrations, understand-ing and predicting the time course of concentrationscan be used to help optimize therapy.

The link of drug dosage to drug effect involvesa sequence of events (Figure 1). Even when a drugis administered directly into the vascular system, thedrug diffuses to both its pharmacologic target receptorand to other peripheral distribution sites where it doesnot have the desired activity but may exert toxic ef-fects. Simultaneously, the drug undergoes clearance bymetabolism and excretion. After oral administration,the situation is more complex, since the drug mustundergo dissolution and absorption, then survive first-pass metabolism in the liver, before reaching the sys-temic circulation. Pharmacokinetics provides a ratio-nal mathematical framework for understanding theseconcurrent processes, and facilitates achieving optimalclinical pharmacodynamic effects more efficiently thantrial and error alone.

The following 3 clinical vignettes illustrate howfamiliarity with principles of pharmacokinetics andpharmacodynamics can facilitate optimal understand-ing and prediction of drug effects in human subjectsand patients.

Clinical VignettesCase 1A 30-year-old man has been extensively evaluated forrecurrent supraventricular tachycardia (SVT), which isassociated with palpitations and dizziness. No iden-tifiable cardiac disease is evident, and other medicaldiseases have been excluded.

The treating physician elects to start therapy withdigitoxin, 0.1mg daily. Oneweek later the patient is seenagain, and states that episodes of SVT are reduced innumber. The plasma digitoxin level is 8 ng/mL (usualtherapeutic range, 10–20 ng/mL). The dose is increasedto 0.2 mg/day. At a follow-up visit 7 days later, thepatient claims that symptoms attributable to SVT havedisappeared completely. The plasma digitoxin level is17.4 ng/mL. The patient continues on 0.2 mg/day ofdigitoxin.

One month later the patient sees the physician onan urgent basis. He has diminished appetite and waves

1Program in Pharmacology and Experimental Therapeutics, SacklerSchool of Graduate Biomedical Sciences, Tufts University School ofMedicine, Boston, MA, USA2Departments of Pharmacy and Medicine, Tufts Medical Center, Boston,MA, USA

Submitted for publication 26 February 2016; accepted 3 March 2016.

Corresponding Author:David J. Greenblatt, MD, Tufts University School of Medicine, 136Harrison Avenue, Boston, MA 02111Email: [email protected]

This proposal was prepared under the guidance of the Special Com-mittee for Medical School Curriculum of the American College ofClinical Pharmacology. The proposal is intended as a resource forthe Association of American Medical Colleges as it revises its CoreEntrustable Professional Activities for Entering Residency, CurriculumDeveloper’s Guide.

Page 2: Pharmacokinetics and Pharmacodynamics for Medical … · Greenblatt and Abourjaily 1185 Table 2. Continued Section 6.Clearanceofdrugsandmechanismsofelimination25 A.Model-independentdefinition(Figure6):

Greenblatt and Abourjaily 1181

Figure 1. Sequence of events between intravenous or oral adminis-tration of a drug and the drug’s interaction with the target receptormediating pharmacologic action.This type of schematic diagram has beenattributed to Dr. Leslie Z. Benet. The segment above the dashed line isthe pharmacokinetic component—“what the body does to the drug.”Below the dashed line is the pharmacodynamic component—“what thedrug does to the body” (from reference 2, with permission).

of nausea. The electrocardiogram shows T-wave abnor-malities, and the plasma digitoxin level is 31.3 ng/mL.

Discussion—Case 1Digitoxin is seldom used in contemporary therapeutics,but case 1 nonetheless illustrates 2 important principles:dose proportionality, and attainment of steady-state.The rate of attainment of the steady-state conditionafter initiation of multiple-dose treatment is dependenton the half-life of the particular drug. In the case ofdigitoxin, the half-life is about 7 days, implying that 3–4weeks of continuous treatment (without a loading dose)is necessary for steady-state to be reached.4 In the exam-ple given, the increase in dosage from 0.1 to 0.2 mg/dayis anticipated to proportionally increase the steady-state concentration (Figure 2). At the daily dosage of0.1 mg (without a loading dose) and a half-life of 7days, the plasma digitoxin concentration of 8 ng/mLafter 7 days of treatment represents 50% of the eventualsteady-state concentration of 16 ng/mL. If the physi-cian had stayed with the 0.1mg/day dosage, that steady-state concentration—attained after several weeks oftreatment (4 to 5 times the half-life)—would have beenin the therapeutic range. However the dosage was in-creased to 0.2 mg/day, yielding a corresponding steady-state concentration of 32 ng/mL, exceeding the thera-peutic range and producing adverse effects (Figure 2).

Case 2A 56-year-old man has a history of grand mal seizures,which have been completely suppressed for the last 12years by phenytoin, 300mg daily. His plasma phenytoinlevel consistently falls in the range of 12–16μg/mL. Thepatient is able to lead a normal life, and is an excellenttennis player. During a particularly competitive tennismatch, the patient injures his shoulder. That night heexperiences severe pain, tenderness, and limitation of

Figure 2. Hypothetical mean plasma concentrations of digitoxin, cor-responding to case 1 in the text. Digitoxin is initially given at a dosageof 0.1 mg per day for 1 week, after which the plasma concentrationis 8 ng/mL (point a). The treating physician wants to increase theplasma concentration to a value within the usual therapeutic range(10–20 ng/mL). If the physician stayed with the 0.1-mg-per-day dosage,the eventual steady-state concentration would have been 16 ng/mL(dashed line)—within the desired therapeutic range. Instead, the dosageis increased to 0.2 mg per day on day 7. The next measured plasmaconcentration is 17.4 ng/mL 1 week later (point b), but 1 month laterit has reached 31.3 ng/mL (point c), close to the eventual steady-statevalue of 32 ng/mL. This is well above the therapeutic range and may beassociated with toxicity.

motion. He contacts his physician. An x-ray is negative.The physician prescribes rest, topical heat, and aspirin,650 mg 4 times daily.

At a return visit to the physician 2 days later, thepatient is greatly improved. The physician uses thatopportunity to do a routine check of the plasmaphenytoin level, which is reported as 5 μg/mL. Thereis no evidence of recurrent seizure activity, and thepatient insists that he is continuing to take phenytoinas directed (300 mg/day). The physician increases thedose to 500 mg/day.

One week later the patient returns complaining ofdifficulty with balance and with fixing his eyes onobjects. The plasma phenytoin level is 14 μg/mL.

Discussion—Case 2In case 2, plasma protein binding of phenytoin isreduced by coadministration of aspirin because ofdisplacement of phenytoin from plasma-binding sitesby salicylate.5–7 This is evident as an increase in the freefraction (Table 1). However, the clearance of unbound(free) drug, and the steady-state concentration ofunbound drug, are unchanged.8,9 As such, no changein clinical effect would be anticipated, and the correctclinical course would have been to leave the daily dosageat 300 mg/day. Because salicylate reduces plasmaprotein binding of phenytoin (higher free fraction inplasma), this has the effect of reducing total (free +

Page 3: Pharmacokinetics and Pharmacodynamics for Medical … · Greenblatt and Abourjaily 1185 Table 2. Continued Section 6.Clearanceofdrugsandmechanismsofelimination25 A.Model-independentdefinition(Figure6):

1182 The Journal of Clinical Pharmacology / Vol 56 No 10 2016

Table 1. Total and Free (Unbound) Plasma Phenytoin Concentrationsin Case 2

Phenytoin DailyDosage, andCotreatment

Total Phenytoin(μg/mL) Fraction Free

Free Phenytoin(μg/mL)

300 mg/day (nocotreatment)

12–16 0.1 1.2–1.6

300 mg/day +aspirin

5 0.3 1.5

500 mg/day +aspirin

14 0.3 4.2

bound) concentrations of phenytoin as well as the in-terpretation of these measured total concentrations.5–7

Increasing the daily dosage of phenytoin to 500mg/day increases the free (unbound) concentration to4.2 μg/mL, which is associated with adverse effectsdespite the total concentration of 14 μg/mL.

A second important point is the nonlinear kineticprofile of phenytoin.10 At daily doses in a rangeexceeding 300 mg/day, steady-state plasma concentra-tions increase disproportionately with an increase indosage. The free phenytoin concentration is 1.5 μg/mLat 300 mg/day, but increases to 4.2 μg/mL with anincrease in dosage to 500 mg/day. This property ofphenytoin makes it difficult to titrate dosage at thishigher dosage range.

Case 3A fourth-year dental student is doing a clerkship in adental surgeon’s practice. A healthy 34-year-old womanis scheduled to undergo procedures estimated to lastapproximately 2 hours. Following instillation of localanesthesia and prior to the start of the procedure,the surgeon notices that the patient still is extremelyagitated and fearful. The surgeon administers 0.5mg/kgof propofol intravenously over a 2-minute period. Thepatient becomes calm, relaxed, and falls into a lightsleep from which she is easily roused. The surgicalprocedure is initiated and proceeds without incident forabout 45 minutes. At this time, the patient becomesalert, and again is fearful and agitated. The surgeon ad-ministers another 0.5 mg/kg of propofol intravenously,the patient again becomes calm, and the surgical proce-dure proceeds to completion without incident.

The student is confused. He/she asks the surgeon,“Why did the patient wake up after only 45 minutes?The half-life of propofol usually is at least 8 hours.”

Discussion—Case 3Case 3 is an example of how the pharmacodynamiceffects of lipophilic psychotropic drugs after singleintravenous doses are dependent more on the rapidprocess of peripheral distribution than on clearance

Figure 3. Hypothetical plasma concentrations of propofol correspond-ing to case 3 in the text. A 0.5 mg/kg intravenous dose of propofol isinitially given at time zero. The plasma propofol concentration declinesrapidly, falling below the hypothetical minimum effective concentration(MEC) of 200 ng/mL at 0.75 hours, at which time the patient emergesfrom the sedated condition. The rate of drug disappearance in thepostdistributive phase would be slower (dashed line), but an additional0.5 mg/kg dose of propofol is required at the 0.75-hour time to maintainplasma concentrations above the MEC and maintain the patient in asedated condition for the remainder of the procedure.

or elimination. The pharmacokinetics of propofol aredescribed by a 2- or 3-compartment model, in whichthe initial “distribution” phase represents rapid distri-bution from systemic circulation to peripheral tissues,followed by the terminal “elimination” phase whichmainly reflects clearance.11–14 Although the propofolhas not been eliminated from the body, its distributionfrom systemic circulation into peripheral tissue resultsin lower concentrations in plasma and brain, which ishighly vascular and rapidly equilibrates with systemiccirculation (Figure 3). As such, the patient becomesmore alert, and a second dose is required.

Components of MedicalEducation in Pharmacokineticsand PharmacodynamicsAn outline of key elements of content for the teachingof clinical pharmacokinetics and pharmacodynamicsto first- or second-year medical students, along withpertinent literature references,15–54 are presented inTable 2. The same or similar content can be applied tothe teaching of graduate students at both the PhD andmasters levels, or to postdoctoral education programsfor house staff or practicing physicians. The materialcan be reasonably presented in 3 or 4 total lecturecontact hours. The didactic presentations should bereinforced through problem sets and review sessions

Page 4: Pharmacokinetics and Pharmacodynamics for Medical … · Greenblatt and Abourjaily 1185 Table 2. Continued Section 6.Clearanceofdrugsandmechanismsofelimination25 A.Model-independentdefinition(Figure6):

Greenblatt and Abourjaily 1183

aimed at supporting conceptual understanding, as wellas ensuring proficiency with pharmacokinetic calcula-tions and construction of graphics.

Individual instructors can adapt the outline andaccompanying graphics as needed, to construct specificlecture content consistent with their own style andinstitutional needs. An ongoing point of discussion isthe extent to which formulas, equations, andmathemat-ics are needed for the teaching of pharmacokinetics.Student backgrounds in mathematics and their comfortwith quantitative content vary widely. Some dislikeand resist the mathematical content, whereas otherswelcome it. “Equation-free” pharmacokinetics is notrealistic, but the density of equations can be managedsuch that the mathematical framework enhances con-

ceptual understanding. The outline in Table 2 has thatobjective. The need for memorization of formulas andequations is minimal.

Table 3 lists biomedical journals that have a focuson clinical pharmacokinetics and pharmacodynamics.Students are encouraged to consult original researchsources when seeking information on pharmacoki-netic/pharmacodynamic properties or drug interactionsinvolving specific drugs or drug classes. Review articlesand secondary sources do have a role in the educationalprocess, in that large amounts of data are collated andsummarized.However, secondary sources are inevitably“filtered” and interpreted by their authors. Studentsneed to consider the benefits and drawbacks of availableinformation sources.

Page 5: Pharmacokinetics and Pharmacodynamics for Medical … · Greenblatt and Abourjaily 1185 Table 2. Continued Section 6.Clearanceofdrugsandmechanismsofelimination25 A.Model-independentdefinition(Figure6):

1184 The Journal of Clinical Pharmacology / Vol 56 No 10 2016

Table 2. Course Outline: Pharmacokinetics and Pharmacodynamics for a Medical School Curriculum

Section 1.Definition and scopeA. Pharmacokinetics: concentration versus timeB. Pharmacodynamics: effect versus timeC. Kinetic-dynamic modeling: effect versus concentration

Section 2.Value of pharmacokinetic principles in medical science15–19

A. Choice of loading and maintenance doseB. Choice of frequency and route of administrationC. Predicting the rate and extent of drug accumulationD. Predicting the effect of dose changesE. Identifying and anticipating drug interactionsF. Identifying patient and disease factors that could alter clinical responseG. Interpreting drug concentrations in serum or plasma19

Section 3. Fundamental assumptionsA. Proportionality cascade (Figure 4):–Intravascular free drug–Extracellular water–Receptor occupancy–Quantitative pharmacodynamic effect

B. Concentration ranges–Subtherapeutic–Therapeutic–Potentially toxic

Section 4. Body compartments and volumes of distribution15–17,20–22

A. Definition of a compartmentB. Calculation of volume of distribution derived from definition of concentration:

Concentration = AmountVolume

Volume of distribution Vd = Amount of drug in bodyConcentration in reference compartment

C. The value and ambiguity of compartment models (hydraulic analogues)1. One-compartment model: Vd is unique2. Two-compartment model: Vd is not unique23

D. Anatomic correlates of volume of distributionE. Physiochemical correlates of volume of distribution24

Section 5. Exponential behavior and the meaning of half-lifeA. First-order processes:1. Rate is proportional to concentration

dCdt

= −kC

2. Rate is not constant, even though k is called a “rate constant”B. Calculation of half-life

t1/2 = In 2k

= 0.693k

C. k and t1/2 are independent of route of administrationD. Logarithmic versus linear graphs (Figure 5)E. Interpretation and implications of half-life

Time elapsed (multiples of t1/2 ) Fractional completion of process

1 0.52 0.753 0.8754 >0.90

F. Implications of first-order behavior–t1/2, Vd, and clearance are fixed–After single doses, AUC (area under the curve from time = zero to “infinity”) is proportional to dose–At steady state, Css (steady-state concentration) is proportional to infusion rate (or dosing rate)

Page 6: Pharmacokinetics and Pharmacodynamics for Medical … · Greenblatt and Abourjaily 1185 Table 2. Continued Section 6.Clearanceofdrugsandmechanismsofelimination25 A.Model-independentdefinition(Figure6):

Greenblatt and Abourjaily 1185

Table 2. Continued

Section 6.Clearance of drugs and mechanisms of elimination25

A. Model-independent definition (Figure 6):

Clearance = DoseAUC

B. Units of clearance: volume/timeClearance cannot exceed blood flow to clearing organ

C. Routes of clearance1. Hepatic clearance: chemical modification (biotransformation)2. Renal clearance: excretion of intact drug3. All other: pulmonary, biliary, fecal, intravascular (plasma enzymes)

D. Biologic dependence of t1/2 on both Vd and clearance

t1/2 = 0.693 x Vd

clearance

E. The meaning of “linear” or “dose proportional” kinetics (see also section 5F)

Section 7.Rapid single-dose intravenous injectionA.One-compartment model

1.dCdt

= −kC

Boundary condition: at t = 0, C= C0

C = C0 e-kt satisfies differential equation and boundary condition

Total AUC = C0

k

2. If dose = D, then

Vd = DoseC0

Note: For a 1-compartment model, Vd is unique. In practice, the best time to measure Vd is at t = 0.3. Monoexponential decline (Figure 5)

t1/2 = 0.693k

4.Clearance = DoseAUC

= k . Vd = k .DoseC0

5. Correct biologic relation among t1/2 , Vd, and clearance:

t1/2 = 0.693 x V d

C learance

6. Graphical approach to problem solving

B. Two-compartment model16,17(Figure 7)1. Simultaneous differential equations with boundary condition:

C = C0 at t = 0

Solution: C = Ae-αt + Be-βt

A, B, α, β are HYBRID

Total AUC = Aα

+ Bβ

2. Volumes of distribution20–22

a. “Central” compartment

V1 = DoseC0

= DoseA + B

b. “Total” volume of distribution

Vd = Doseβ × AUC

= Clearanceβ

3. Half-livesa. Distribution half-life (cannot be derived from graph)

t1/2α = 0.693α

b. Elimination half-life (can be derived from graph)

t1/2β = 0.693β

4. Clearance = dose/AUC = Vd · β

Page 7: Pharmacokinetics and Pharmacodynamics for Medical … · Greenblatt and Abourjaily 1185 Table 2. Continued Section 6.Clearanceofdrugsandmechanismsofelimination25 A.Model-independentdefinition(Figure6):

1186 The Journal of Clinical Pharmacology / Vol 56 No 10 2016

Table 2. Continued

C.One-compartment approximation of the 2-compartment modelFundamental assumption:A and α (and the corresponding component of AUC) are ignoredConsequences:B becomes C0

β becomes kt1/2β becomes t1/2

Quantity 2-Compartment 1-Compartment

Equation for concentration versus time C = Ae-αt + Be-βt C = C0e−kt

(B analogous to C0,β analogous to k)

Total AUCAα

+ Bβ

C0

k

(analogous to

)

Total V dDose

β .AUCDoseC0

(analogous to

DoseB

)

ClearanceDoseAUC

= Vd . βDoseAUC

= Vd . k

Elimination half-life doseIn 2β

In 2k

D. Graphical approach to problem-solvingE. Distribution versus elimination as the determinant of duration of action (Figures 3 and 7)

Section 8.Multiple-dose kinetics: continuous intravenous infusion (without a loading dose) (Figure 8)A. Rate of attainment of steady state:Depends almost entirely on t1/2 (and k) as would occur with a single dose.Does not depend on infusion rate (abbreviation: Q).Inverted exponential function:C = Css (1 − e-kt). This is the same k as after a single dose.

B. Determinants of the extent of accumulation (absolute steady-state concentration) (Figures 8 and 9)

Css = Infusion rateClearance

C. Predictable consequences of changing infusion rate or stopping infusion (Figure 10)1. Rate of attainment of new steady state2. New steady-state concentration

Section 9.Drug absorption and bioavailability (Figure 11)A. Rate of drug absorption1. Lag time (due to dissolution, gastric emptying, etc.)2. First-order absorption3. Factors influencing absorption rate4. Implications of absorption rate5. Slow-release preparations

B. Completeness of absorption (fractional absorption or absolute systemic availability)1. Methods of assessment—intravenous data needed (Figure 12)

F = AUC (P.O.)AUC (I.V.)

AUC must be dose-normalized and extrapolated to infinity.2. Mechanisms of incomplete bioavailability25–29

a. Incomplete absorption- Intrinsic properties of the chemical- Properties of the dosage form

b. Efflux transportc. Presystemic extraction (first-pass metabolism)- Hepatic- Enteric (CYP3A)

C. Bioequivalence and the pharmacopolitics of generic substitution30,31

1. Competing political and economic influences2. Fundamental premise:Bioequivalence implies therapeutic equivalence (not the reverse)

3. Methods of determining bioequivalence: statistical “equivalence” of Cmax, Tmax, and AUC

Page 8: Pharmacokinetics and Pharmacodynamics for Medical … · Greenblatt and Abourjaily 1185 Table 2. Continued Section 6.Clearanceofdrugsandmechanismsofelimination25 A.Model-independentdefinition(Figure6):

Greenblatt and Abourjaily 1187

Table 2. Continued

4. Areas of uncertaintya. Patients versus volunteersb. Limits of statistical tolerancec. Interpretation of anecdotesd. Voluntary versus forced generic substitutione. Sustained-release preparations

5. Special considerations for biologic therapies (biosimilars)32,33

Section 10.Multiple-dose kinetics: discrete doses (Figure 13)A. There is not a unique steady-state concentration, only a mean:

Css = AUCover dose intervellength of dose intervel

B. Rate of attainment of steady-state: identical to section 8AC. Extent of accumulation: if each dose (D) is 100% available and given at fixed intervals (T),

Css = D/TClearance

D. Interdose fluctuation (Figure 14)1. Estimating the degree of fluctuationCmax = maximum plasma concentration over the dosage intervalCmin = Minimum plasma concentration over the dosage intervalCmax/Cmin ratio is interdose fluctuation

2. Css stays the same as long as D/T is unchanged, but changing both D and T influences interdose fluctuation (Figure 15)3. Clinical benefits of “slow-release” preparations

E. Termination of multiple dosage: the same value of k (and t1/2 ) is applicableF. Loading doses (DL)1. Determinants of when needed; benefits and disadvantages2. Approximate calculations:Given a desired Css and assuming Vd is known, DL should cause C0 for a single dose (see section 7A) to equal the target Css during continuous infusion(see section 8B) or Css during multiple discrete doses (see section 10C).

3. Complications with 2-compartment model: overshoot and undershootG. Relative drug accumulation: depends on the interval between doses relative to the elimination half-life

Section 11.Nonlinear (zero-order) kinetics10,35,37 (Figure 16)A. Linear (first-order) kinetics (as in section 5A):

dCdt

= −kC

Solution: C = Coe-kt

B. Nonlinear (zero-order) kinetics

dCdt

= −k

Solution: C = Co − ktC. For most affected drugs, first-order kinetic profile transitions to zero order as the concentration increases (Figure 17)D. Implications of transition to zero-order kinetics

Section 12.Drug interactions involving altered drug clearance2,3,36–43

A. Epidemiology of drug interactionsB. Mechanisms of inhibition versus inductionC.Quantitative outcome of drug interactions (Figure 18)D. Clinical consequences of drug interactions: depends on the quantitative magnitude of the drug interaction and the therapeutic index of the affected drug.

Statistical significance does not imply clinical importance.E. In vitro prediction of clinical drug interactions: relation of inhibitor “exposure” to inhibitory “potency”

Section 13.Drug therapy in vulnerable populations: the elderly44–48

A. Epidemiology of agingB. Mechanisms of altered drug response in the elderly1. Kinetic2. Dynamic

C. Consequences of impaired clearance in the elderly.

Section 14.Other vulnerable or special populationsA. Renal insufficiencyB. Hepatic insufficiencyC.ObesityD. ChildrenE. Pregnancy

Page 9: Pharmacokinetics and Pharmacodynamics for Medical … · Greenblatt and Abourjaily 1185 Table 2. Continued Section 6.Clearanceofdrugsandmechanismsofelimination25 A.Model-independentdefinition(Figure6):

1188 The Journal of Clinical Pharmacology / Vol 56 No 10 2016

Table 2. Continued

Section 15. Pharmacodynamics3,51–53

A. Definition: time course of drug effectB. Approaches to measuring drug effect1. Fully objective (blood pressure, QT interval, serum cholesterol, etc.)2. Partially objective (memory tests, reaction times, pain threshold, etc.)3. Subjective (psychiatric endpoints)

C. Surrogate measures of drug effect (glycated hemoglobin, T-cell count, bone mineral density, intraocular pressure, etc.)D. Problems with pharmacodynamic measures1. Effects of practice, adaptation, and time2. Acute and chronic tolerance3. Fatigue4.Weak connection to clinical endpoints

E. Kinetic-dynamic modeling49–54 (Figure 19)1. Link between concentration and effect (linear, exponential, or sigmoid-Emax)2. Sources of bias3. Modification by effect-site entry delay4. Interpretation of outcome

Table 3. Medical and Scientific Journals Having a Focus on ClinicalPharmacokinetics, Drug Metabolism, and Drug Interactions

Biopharmaceutics and Drug DispositionBritish Journal of Clinical PharmacologyClinical PharmacokineticsClinical Pharmacology and TherapeuticsClinical Pharmacology in Drug DevelopmentDrug Metabolism and DispositionEuropean Journal of Clinical PharmacologyInternational Journal of Clinical PharmacologyJournal of Clinical PharmacologyJournal of Clinical PsychopharmacologyJournal of Pharmaceutical SciencesJournal of Pharmacology and Experimental TherapeuticsTherapeutic Drug MonitoringXenobiotica

Figure 4. Schematic relation between drug concentrations (greentriangles) in circulating blood, concentrations in extracellular fluid(ECF) surrounding the receptor site, the extent of occupancy ofcellular receptor sites, and subsequent pharmacologic action. If receptoroccupancy proportionally reflects blood and ECF concentrations, thenblood concentration may serve as a surrogate measure of drug effect.

Page 10: Pharmacokinetics and Pharmacodynamics for Medical … · Greenblatt and Abourjaily 1185 Table 2. Continued Section 6.Clearanceofdrugsandmechanismsofelimination25 A.Model-independentdefinition(Figure6):

Greenblatt and Abourjaily 1189

Figure 5. Plasma concentrations versus time after dosage of a drug given by rapid intravenous injection, assuming an underlying 1-compartmentpharmacokinetic model. Each time an interval equal to the half-life elapses, the concentration falls to 50% of the value at the start of the interval.Left: linear concentration scale; right: logarithmic concentration scale. The same declining exponential function is applicable to both graphs (see Table2, section 5A–E and section 7A1–3), but the logarithmic scale transforms the exponential curve into a straight line, which can be used for graphicalcalculations. Note that a logarithmic concentration scale never goes to zero.

Figure 6. Plasma concentrations of the same dose of the same drug given to different individuals. The area under the plasma concentration curve(AUC) is shown as the shaded region. Since clearance can be calculated as administered dose divided by AUC, the subject in the left graph has lowerclearance (higher AUC) than the subject in the right graph (proviso: in the calculation of clearance, the “dose” represents the systemically-availabledose, and “AUC” represents the total area under the curve from time zero to “infinity”).

Page 11: Pharmacokinetics and Pharmacodynamics for Medical … · Greenblatt and Abourjaily 1185 Table 2. Continued Section 6.Clearanceofdrugsandmechanismsofelimination25 A.Model-independentdefinition(Figure6):

1190 The Journal of Clinical Pharmacology / Vol 56 No 10 2016

Figure 7. Left: Plasma concentrations of a drug, consistent with a 2-compartment model, after a single intravenous dose. Right: Corresponding drugbehavior in the 2-compartment model schematic. Point I: Immediately after the dose, the entire dose is confined to the central compartment, and theplasma concentration is maximal (C0 = 60). Point II: During the initial distribution phase (the “alpha” phase), plasma concentrations fall rapidly andextensively, due mainly to drug distribution from central to peripheral compartments.Clearance (irreversible elimination) contributes minimally to thisinitial decline. If the concentration falls below the minimal effective concentration (MEC) during the distribution phase, the distribution process maylimit the duration of clinical action.Point III:This is the point termed distribution equilibrium.The distribution process is complete, and the ratio of centralto peripheral compartment concentrations from this time forward will remain approximately constant. Point IV: The decline in plasma concentrationsduring the elimination phase (the “beta” phase) is relatively slow, and is due mainly to clearance (irreversible elimination). (see Table 2, section 7B)

Figure 8. A continuous zero-order (fixed-rate) intravenous infusion ofa drug obeying 1-compartment kinetics is started at time zero withouta loading dose. The concentration rises in exponential fashion until thesteady-state condition is reached (dashed lines). The actual steady-stateconcentration (Css) at an infusion rate of Q is 3 units. If the infusionrate were 2 × Q,Css would be 6 units. However the rate of attainmentof steady state is the same in both cases, being dependent only on thehalf-life (from reference 2, with permission).

Figure 9. Relation between zero-order infusion rate (Q, X axis) andsteady-state plasma concentration (Css, Y axis) for a drug having linear(first-order) kinetic properties, as shown in Figure 8. Css has a directlinear relation to Q (see Table 2, sections 5F and 8B). The slope of theline is 1/clearance.

Page 12: Pharmacokinetics and Pharmacodynamics for Medical … · Greenblatt and Abourjaily 1185 Table 2. Continued Section 6.Clearanceofdrugsandmechanismsofelimination25 A.Model-independentdefinition(Figure6):

Greenblatt and Abourjaily 1191

Figure 10. Left: A drug is given by continuous zero-order intravenous infusion (fixed rate of Q) starting at time zero. The plasma concentrationascends to Css (4 units) in exponential fashion, with attainment of steady state more than 90% complete after 4 × t1/2. This is similar to Figure 8.Right: After steady state is reached, the infusion rate is changed (arrow), and the time scale “resets” to zero. The rate is either increased to 2 × Q,decreased to 0.5 × Q, or stopped altogether. The new Css correspondingly changes to 8 units, 2 units, or 0, respectively. However, another 4 or moremultiples of t1/2 are needed for the new steady state to be reached.

Figure 11. Plasma concentrations of a drug after oral dosage attime zero. After a lag time (Tlag), plasma concentrations begin to rise.During this “absorption” phase, rates of drug entry into blood due toabsorption exceed rates of elimination due to clearance. The maximumconcentration (Cmax = 7 units) is reached at time Tmax (1 hour afterdosage); at this point, instantaneous absorption and elimination rates areequal. Concentrations then fall, indicating that elimination rates exceedabsorption rates.The area under the plasma concentration curve (AUC)is used as a surrogate for the extent of absorption (from reference 2,withpermission).

Figure 12. Mean plasma concentrations of midazolam after singleintravenous and oral doses administered to healthy volunteers29 (*con-centrations were normalized to a 2-mg dose). Based on the relationshipin section 9B1, in Table 2, the absolute bioavailability of oral midazolam(F) was calculated as 0.29, indicating that only 29% of an oral dose actuallyreaches the systemic circulation (from reference 2, with permission).

Page 13: Pharmacokinetics and Pharmacodynamics for Medical … · Greenblatt and Abourjaily 1185 Table 2. Continued Section 6.Clearanceofdrugsandmechanismsofelimination25 A.Model-independentdefinition(Figure6):

1192 The Journal of Clinical Pharmacology / Vol 56 No 10 2016

Figure 13. A drug is administered as discrete oral doses at intervals equal to the half-life. Six consecutive doses were given, after which the drugwas discontinued. The dashed line is the hypothetical curve if the same dosing rate were administered by continuous zero-order intravenous infusion.Note that the rate of attainment of steady state is the same in both cases.

Figure 14. A dosage interval at steady state. The elimination half-lifeis assumed to be 24 hours, and the dosage interval is also 24 hours(as in Figure 13). The plasma concentration starts at Cmin, increases toCmax, then falls again to Cmin.The mean steady-state concentration (Css,dashed line) is the area under the curve for 1 dose interval divided bythe length of the interval. The interdose fluctuation is calculated as theCmax/Cmin ratio. (see Table 2, section 10A–D).

Figure 15. Serum concentrations of a drug at steady state if the drugis given using 3 different dosing schedules: either 500 mg every 24 hours,250 mg every 12 hours, or 125 mg every 6 hours. In each case the meansteady-state concentration (Css) is the same, since the [(dose)/(doseinterval)] ratio does not change. However, the interdose fluctuationbecomes smaller as the dose interval becomes smaller. With the once-daily dosing schedule, the interdose fluctuation is large, and the plasmaconcentration falls outside the therapeutic range just after and justbefore each dose (in part from reference 16, with permission).

Page 14: Pharmacokinetics and Pharmacodynamics for Medical … · Greenblatt and Abourjaily 1185 Table 2. Continued Section 6.Clearanceofdrugsandmechanismsofelimination25 A.Model-independentdefinition(Figure6):

Greenblatt and Abourjaily 1193

Figure 16. Plasma concentrations (1-compartment model) of one drugwith first-order elimination (solid line), and another drug with zero-order elimination (dashed line). Note that the concentration axis islinear.

Figure 17. Schematic relation of daily phenytoin dosage (X axis) tophenytoin steady-state plasma concentrations (Y axis). In the lowerdosage range, the relation is linear, and Css increases in propor-tion to dosage. As the daily dosage increases to higher ranges, thekinetic pattern transitions toward zero-order, and Css increases dispro-portionately with dosage (dashed line). At doses above 350 mg per day,Css falls above the therapeutic range of 10–20 μg/mL) (horizontal dottedlines).

Figure 18. Pharmacokinetic consequences of a drug–drug interaction that either decreases clearance or increases clearance of the substrate drug(inhibition or induction, respectively). Left: After single doses of the substrate (victim) drug, area under the plasma concentration curve (AUC) isincreased by inhibition of clearance,or decreased by induction of clearance.Right:The substrate drug is given as a continuous regimen such that steadystate is reached (interdose fluctuation not shown). At the arrow, an inhibiting or inducing drug is coadministered, causing a new steady state to bereached. Note that the onset of action of the inducer is slower than that of the inhibitor.

Page 15: Pharmacokinetics and Pharmacodynamics for Medical … · Greenblatt and Abourjaily 1185 Table 2. Continued Section 6.Clearanceofdrugsandmechanismsofelimination25 A.Model-independentdefinition(Figure6):

1194 The Journal of Clinical Pharmacology / Vol 56 No 10 2016

HOURS

0 1 2 3 4 5 6

SE

RU

M P

RO

PR

AN

OL

OL

(n

g/m

L)

0

50

100

150

200

250

300

350

PO

ST

ER

IOR

WA

LL

VE

LO

CIT

Y R

ED

UC

TIO

N(c

han

ge

ove

r b

asel

ine,

cm

/sec

)

0

4

8

12

16

20

SERUM PROPRANOLOLVELOCITY REDUCTION

SERUM PROPRANOLOL (ng/mL)

0 50 100 150 200 250 300 350

0

4

8

12

16

20

Figure 19. Example of a kinetic-dynamic study. A single 80-mg oral dose of propranolol was administered to a series of healthy volunteers.54

Serum propranolol concentrations were measured at multiple points after the dose, and posterior ventricular wall velocity was determined byechocardiography as a measure of pharmacodynamic effect — higher numbers indicate greater decrements in wall velocity, consistent with reducedvelocity of cardiac contraction.Left:Time-course of the 2 measures plotted on the same graph.Points represent the mean values at corresponding times.After the serum Cmax is reached, the contractility measure returns close to zero in advance of the serum concentration. This might be explainedby acute tolerance. Right: A kinetic-dynamic plot, in which pharmacodynamic effect is shown in relation to serum concentration at correspondingtimes. Arrows indicate the direction of increasing time. The pattern is termed “clockwise hysteresis,” and can be observed as a consequence of acutetolerance.

AcknowledgmentsWe are grateful for the collaboration and assistance of Mr.Jerold S. Harmatz,Ms. Yanli Zhao,Ms. Smaro Panagiotidou,and Dr. Tianmeng Chen. The authors thank the AmericanCollege of Clinical Pharmacology for the opportunity toparticipate in this project.

References1. Greenblatt DJ, Shader RI.Pharmacokinetics in Clinical Practice.

Philadelphia: WB Saunders; 1985.2. Greenblatt DJ, von Moltke LL. Pharmacokinetics and drug in-

teractions. In: Sadock BJ, Sadock VA, eds. Comprehensive Text-book of Psychiatry. 8th ed. Philadelphia: Lippincott Williams &Williams; 2005:2699–2706.

3. Greenblatt DJ, von Moltke LL, Harmatz JS, Shader RI. Phar-macokinetics, pharmacodynamics, and drug disposition. In:Davis KL, Charney D, Coyle JT, Nemeroff C, eds. Neuropsy-chopharmacology: The Fifth Generation of Progress. Philadel-phia: Lippincott Williams & Williams; 2002:507–524.

4. Ochs HR, Pabst J, Greenblatt DJ, Hartlapp J. Digitoxin accu-mulation. Br J Clin Pharmacol. 1982;14:225–229.

5. Odar-Cederlof I, Borga O. Impaired plasma protein binding ofphenytoin in uremia and displacement effect of salicylic acid.Clin Pharmacol Ther. 1976;20(1):36–47.

6. Leonard RF, Knott PJ, Rankin GO, Robinson DS, Mel-nick DE. Phenytoin-salicylate interaction. Clin Pharmacol Ther.1981;29(1):56–60.

7. Fraser DG, Ludden TM, Evens RP, Sutherland EW. Displace-ment of phenytoin from plasma binding sites by salicylate. ClinPharmacol Ther. 1980;27(2):165–169.

8. GreenblattDJ, Sellers EM,Koch-Weser J. Importance of proteinbinding for the interpretation of serum or plasma drug concen-trations. J Clin Pharmacol. 1982;22:259–263.

9. Koch-Weser J, Sellers EM. Binding of drugs to serum albumin.N Engl J Med. 1976;294;311–316, 526–531.

10. Martin E, Tozer TN, Sheiner LB, Riegelman S. The clinicalpharmacokinetics of phenytoin. J Pharmacokinet Biopharm.1977;5(6):579–596.

11. Kanto J, Gepts E. Pharmacokinetic implications for the clinicaluse of propofol. Clin Pharmacokinet. 1989;17(5):308–326.

12. Morgan DJ, Campbell GA, Crankshaw DP. Pharmacokineticsof propofol when given by intravenous infusion. Br J ClinPharmacol. 1990;30(1):144–148.

13. Han T, Kaneda K, Greenblatt DJ, Martyn JAJ. Propofol clear-ance and volume of distribution are increased in patients withmajor burns. J Clin Pharmacol. 2009;49:768–772.

14. Eleveld DJ, Proost JH, Cortinez LI, Absalom AR, Struys MM.A general purpose pharmacokinetic model for propofol. AnesthAnalg. 2014;118(6):1221–1237.

15. Gibaldi M, Perrier D. Pharmacokinetics. New York: MarcelDekker Inc.; 1975.

16. GreenblattDJ,Koch-Weser J. Clinical pharmacokinetics.NEnglJ Med. 1975;293:702–705, 964–970.

17. HugCC. Pharmacokinetics of drugs administered intravenously.Anesth Analg. 1978;57(6):704–723.

18. Greenblatt DJ, von Moltke LL. Pharmacokinetics and drug in-teractions. In: Sadock BJ, Sadock VA, eds. Comprehensive Text-book of Psychiatry. 8th ed. Philadelphia: Lippincott Williams &Williams; 2005:2699–2706.

19. FriedmanH, Greenblatt DJ. Rational therapeutic drug monitor-ing. JAMA. 1986;256:2227–2233.

20. Greenblatt DJ, Abernethy DR, Divoll M. Is volume of dis-tribution at steady-state a meaningful kinetic variable? J ClinPharmacol. 1983;23:391–400.

21. Greenblatt DJ. Volume of distribution - again. Clin PharmacolDrug Dev. 2014;3:419–420.

22. Benet LZ, Ronfeld RA. Volume terms in pharmacokinetics. JPharm Sci. 1969;58(5):639–641.

23. Niazi S. Volume of distribution as a function of time. J PharmSci. 1976;65(3):452–454.

24. Ochs HR, Greenblatt DJ, Abernethy DR, et al. Cerebrospinalfluid uptake and peripheral distribution of centrally acting drugs:relation to lipid solubility. J PharmPharmacol. 1985;25:204–209.

Page 16: Pharmacokinetics and Pharmacodynamics for Medical … · Greenblatt and Abourjaily 1185 Table 2. Continued Section 6.Clearanceofdrugsandmechanismsofelimination25 A.Model-independentdefinition(Figure6):

Greenblatt and Abourjaily 1195

25. Wilkinson GR. Clearance approaches in pharmacology. Phar-macol Rev. 1987;39:1–47.

26. Greenblatt DJ. Presystemic extraction: mechanisms and conse-quences. J Clin Pharmacol. 1993;33:650–656.

27. Greenblatt DJ, von Moltke LL, Shader RI. The importance ofpresystemic extraction in clinical psychopharmacology. J ClinPsychopharmacol. 1996;16:417–419.

28. Pond SM, Tozer TN. First-pass elimination: basic concepts andclinical consequences. Clin Pharmacokinet. 1984;9:1–25.

29. Tsunoda SM, Velez RL, von Moltke LL, Greenblatt DJ. Differ-entiation of intestinal and hepatic cytochrome P450 3A activitywith use of midazolam as an in vivo probe: effect of ketocona-zole. Clin Pharmacol Ther. 1999;66:461–471.

30. Koch-Weser J. Bioavailability of drugs. N Engl J Med.1974;291:233–237, 503–506.

31. Greenblatt DJ, Smith TW, Koch-Weser J. Bioavailability ofdrugs: the digoxin dilemma. Clin Pharmacokinet. 1976;1(1):36–51.

32. Weise M, Bielsky MC, De Smet K, et al. Biosimilars: whatclinicians should know. Blood. 2012;120(26):5111–5117.

33. Chandra A, Vanderpuye-Orgle J. Competition in the age ofbiosimilars. JAMA. 2015;314(3):225–226.

34. Ludden TM.Nonlinear pharmacokinetics: clinical Implications.Clin Pharmacokinet. 1991;20(6):429–446.

35. Jusko WJ. Pharmacokinetics of capacity-limited systems. J ClinPharmacol. 1989;29(6):488–493.

36. Greenblatt DJ. Introduction to drug-drug interactions. In:Piscitelli SC, Rodvold KA, Pai MP, eds. Drug Interactions inInfectious Diseases. 3rd ed. New York: Human Press; 2011:1–10.

37. Greenblatt DJ, von Moltke LL. Clinical studies of drug-druginteractions: design and interpretation. In: Pang KS, RodriguesAD, Peter RM, eds. Enzyme and Transporter-Based Drug-Drug Interactions: Progress and Future Challenges. New York:Springer; 2010:625–649.

38. vonMoltke LL, Greenblatt DJ. Clinical drug interactions due tometabolic inhibition: Prediction, assessment, and interpretation.In: Lu C, Li AP, eds. Enzyme Inhibition in Drug Discoveryand Development. Hoboken, NJ: John Wiley & Sons; 2010:533–547.

39. Greenblatt DJ, He P, von Moltke LL, Court MH. The CYP3family. In: Ioannides C, ed. Cytochrome P450: Role in theMetabolism and Toxicology of Drugs and Other Xenobiotics.Cambridge, UK: Royal Society of Chemistry; 2008:354–383.

40. Farkas D, Shader RI, von Moltke LL, Greenblatt DJ. Mecha-nisms and consequences of drug-drug interactions. In: Gad SC,

ed. Preclinical Development Handbook: ADME and Biopharma-ceutical Properties. Philadelphia: Wiley-Interscience; 2008:879–917.

41. Greenblatt DJ, vonMoltke LL. Drug-drug interactions: Clinicalperspectives. In: Rodrigues AD, ed.Drug-Drug Interactions. 2nded. New York: Informa Healthcare; 2008:643–664.

42. Volak LP, Greenblatt DJ, von Moltke LL. In vitro approachesto anticipating clinical drug interactions. In: Li AP, ed. Drug-Drug Interactions in Pharmaceutical Development. Philadelphia:Wiley-Interscience; 2008:31–74.

43. Venkatakrishnan K, von Moltke LL, Obach RS, Greenblatt DJ.Drug metabolism and drug interactions: application and clinicalvalue of in vitro models. Curr Drug Metab. 2003;4(5):423–459.

44. Cotreau MM, von Moltke LL, Greenblatt DJ. The influence ofage and sex on the clearance of cytochrome P450 3A substrates.Clin Pharmacokinet. 2005;44(1):33–60.

45. von Moltke LL, Abernethy DR, Greenblatt DJ. Kinetics anddynamics of psychotropic drugs in the elderly. In: Salzman C,ed. Clinical Geriatric Psychopharmacology. 4th ed. Philadelphia:Lippincott Williams & Wilkins; 2005:87–114.

46. Greenblatt DJ, Sellers EM, Shader RI. Drug disposition in oldage. N Engl J Med. 1982;306:1081–1088.

47. Schwartz JB. The current state of knowledge on age, sex, andtheir interactions on clinical pharmacology. Clin PharmacolTher. 2007;82(1):87–96.

48. Greenblatt DJ. Sleep and geriatric psychopharmacology. In:Pandi-Perumal SR, Verster JC, Monti JM, Lader MH, LangerSZ, eds. Sleep Disorders: Diagnosis and Therapeutics. London:Informa Healthcare; 2008:163–173.

49. HolfordNHG, Sheiner LB.Kinetics of pharmacologic response.Pharmacol Ther. 1982;16:143–166.

50. Derendorf H, Meibohm B. Modeling of pharmacoki-netic/pharmacodynamic (PK/PD) relationships: conceptsand perspectives. Pharm Res. 1999;16(2):176–185.

51. Campbell DB. The use of kinetic-dynamic interactions in theevaluation of drugs. Psychopharmacology. 1990;100(4):433–450.

52. Greenblatt DJ, Harmatz JS. Kinetic-dynamic modeling in clini-cal psychopharmacology. J Clin Psychopharmacol. 1993;13:231–234.

53. Laurijssens BE, Greenblatt DJ. Pharmacokinetic-pharmacodynamic relationships for benzodiazepines. ClinPharmacokinet. 1996;30:52–76.

54. Ochs HR, Grube E, Greenblatt DJ, Knuchel M, Bodem G.Kinetics and cardiac effects of propranolol in humans. KlinWochenschr. 1982;60(10):521–525.