anesthesia pharmacology compiled

106
ANESTHESIA: Pharmacologic Principles Raisa Cagurangan, M.D. Mustafah Mahboobian, M.D. John Saquilayan, M.D. Jovelyn Tan, M.D. 1

Upload: jovy-tan

Post on 09-Aug-2015

51 views

Category:

Documents


1 download

DESCRIPTION

pharmacokinetics, pharmacodynamics,etc

TRANSCRIPT

Page 1: Anesthesia Pharmacology Compiled

1

ANESTHESIA:Pharmacologic Principles

Raisa Cagurangan, M.D.Mustafah Mahboobian, M.D.

John Saquilayan, M.D.Jovelyn Tan, M.D.

Page 2: Anesthesia Pharmacology Compiled

2

Key Points

1. Most drugs must pass through cell membranes to reach their sites of action. Consequently, drugs tend to be relatively lipophilic, rather than hydrophilic.

2. The highly lipophilic anesthetic drugs have a rapid onset of action because they rapidly diffuse into the highly perfused brain tissue. They have a very short duration of action because of redistribution of drug from the central nervous system to the blood.

3. The cytochrome P450 (CYP) superfamily is the most important group of enzymes involved in drug metabolism. It and other drug-metabolizing enzymes exhibit genetic polymorphism.

Page 3: Anesthesia Pharmacology Compiled

3

Key Points

4. The kidneys eliminate hydrophilic drugs and relatively hydrophilic metabolites of lipophilic drugs. Renal elimination of lipophilic compounds is negligible.

5. The liver is the most important organ for metabolism of drugs. Hepatic drug clearance depends on three factors: the intrinsic ability of the liver to metabolize a drug, hepatic blood flow, and the extent of binding of the drug to blood components.

6. The volume of distribution quantifies the extent of drug distribution. The greater the affinity of tissues for a drug relative to blood, the greater its volume of distribution (i.e., lipophilic drugs have greater volumes of distribution).

Page 4: Anesthesia Pharmacology Compiled

4

Key Points

7. Elimination clearance is the parameter that characterizes the ability of drug-eliminating organs to irreversibly remove drugs from the body. The efficiency of the body to remove a drug from the body is proportional to the elimination clearance.

8. All else being equal, an increase in the volume of distribution of a drug will increase its elimination half-life; an increase in elimination clearance will decrease elimination half-life.

9. Most drugs bring about a pharmacologic effect by binding to a specific receptor that brings about a change in cellular function to produce the pharmacologic effect.

Page 5: Anesthesia Pharmacology Compiled

5

Key Points

10. Although most pharmacologic effects can be characterized by both dose-response curves and concentration-response curves, the dose-response curves are unable to determine whether variations in pharmacologic response are caused by differences in pharmacokinetics, pharmacodynamics, or both.

11. Integrated pharmacokinetic – pharmacodynamic models allow temporal characterization of the relationship between dose, plasma concentration, and pharmacologic effect.

Page 6: Anesthesia Pharmacology Compiled

6

Key Points

12. Simulations of multicompartmental pharmacokinetic models that describe intravenous anesthetics demonstrate that for most anesthetic dosing regimens, the distribution of drug from the plasma to the pharmacologically inert peripheral tissues has a greater influence on the plasma concentration profile of the drug than the elimination of drug from the body.

Page 7: Anesthesia Pharmacology Compiled

7

Key Points

13. Target-controlled infusions are achieved with computer-controlled infusion pumps worldwide (not yet approved by the Food and Drug Administration [FDA] in the United States) and permit clinicians to make use of the drug concentration–effect relationship, optimally accounting for pharmacokinetics and predicting the offset of drug effect.

14. By understanding the interactions between the opioids and the sedative-hypnotics (e.g., response surface models), it is possible to select target concentration pairs of the two drugs that produce the desired clinical effect while minimizing unwanted side effects associated with high concentrations of a single drug.

Page 8: Anesthesia Pharmacology Compiled

8

Key Points

15. The time until a patient regains responsiveness from a single drug anesthetic is determined by the pharmacokinetics of the individual drug, the concentration-effect relationship, and the duration of administration of the drug (context-sensitive decrement time). For two-drug anesthetics, the time to awakening not only depends on the individual drug pharmacokinetics and the duration of administration of the anesthetics, but it also depends on the pharmacodynamic interactions of the two drugs.

Page 9: Anesthesia Pharmacology Compiled

9

Pharmacokinetics

• Passive Diffusion• Active Transport–CNS, hepatocytes, renal tubular cells,

pulmonary capillary endothelium

Page 10: Anesthesia Pharmacology Compiled

10

Routes of Administration

Intravenous• Results in rapid increases in drug concentration

thus caution on drugs with low therapeutic index• Pulmonary endothelium – Can slow the rate at which intravenously

administered drugs reach the systemic circulation (e.g. fentanyl)

– Contains enzymes that may metabolize intravenously administered drugs (e.g. propofol) on first pass and reduce their absolute bioavailability

Page 11: Anesthesia Pharmacology Compiled

11

Routes of Administration

Oral• Safest and most convenient• Main determinant of the timing of absorption is gastric

emptying into the small intestines• NOT used significantly in anesthetic practice because of

the limited and variable rate of bioavailability• Nasal/Sublingual administration – limited by:

– Formulation– Surface area– Require a rapid onset of drug action (e.g. nitroglycerin, fentanyl)

Page 12: Anesthesia Pharmacology Compiled

12

Routes of Administration

Transcutaneous• “Drug patches” (e.g. scopolamine, nitroglycerin,

opioids, clonidine)• Extended amount of time to achieve an effective

therapeutic concentration limits practical application except for maintenance therapy

• Attempts to speed up passive diffusion using an electric current has been described for fentanyl

Page 13: Anesthesia Pharmacology Compiled

13

Routes of Administration

Intramuscular and Subcutaneous • Directly dependent on the drug formulation

and the blood flow to the depot• Subcutaneous route of drug absorption is

more variable in its onset because of the variability of subcutaneous blood flow during varying physiologic states

Page 14: Anesthesia Pharmacology Compiled

14

Routes of Administration

Intrathecal, Epidural, and Perineural Injection• Spinal cord – primary site of action of many anesthetic

agents• Direct injection of local anesthetics and opioids into the

intrathecal space bypasses the limitations of drug absorption and drug distribution of any other route of administration

• Epidural and perineural administration of local anesthetics necessitates that the drug be absorbed through the dura or nerve sheath

• Relative expertise required to perform

Page 15: Anesthesia Pharmacology Compiled

15

Routes of Administration

Inhalational• Large surface area of the pulmonary alveoli

available for exchange with the large volumetric flow of blood found in the pulmonary capillaries makes inhalational administration an extremely attractive method

• New technologies (currently in phase 2 FDA trials) that can rapidly and predictably aerosolize a wide range of drugs and thus approximate intravenous administration

Page 16: Anesthesia Pharmacology Compiled

16

Drug Distribution

• Relative distribution of cardiac output among organ vascular beds determines the speed at which organs are exposed to the drug

• Flow-limited drug uptake• Rate of drug equilibration by the tissue

depends on the ratio of blood flow to tissue content

Page 17: Anesthesia Pharmacology Compiled

17

Redistribution

• Single, moderate doses of highly lipophilic drugs have a very short CNS duration of action because of redistribution to the blood and other, less well-perfused tissues

• Repeated injections allows the rapid establishment of significant peripheral tissue concentrations

• When the tissue concentrations of a drug are high enough, the decrease in plasma drug concentration below therapeutic threshold becomes solely dependent on drug elimination

Page 18: Anesthesia Pharmacology Compiled

18

Drug Elimination

• Excretion• Biotransformation

• Elimination clearance (drug clearance) –theoretical volume of blood from which drug is completely and irreversibly removed in a unit of time [volume per time]

Page 19: Anesthesia Pharmacology Compiled

19

Biotransformation

• Phase I reactions – transform a drug into one or more polar, and hence potentially excretable compounds

• Phase II reactions – conjugating a variety of endogenous compounds to a polar functional group of the drug, making the metabolite even more hydrophilic

Often drugs will undergo a phase I reaction to produce a new compound with a polar functional group that will then undergo a phase II reaction. However, it is possible for a drug to undergo either a phase I reaction alone or a phase II reaction alone.

Page 20: Anesthesia Pharmacology Compiled

Cytochrome P450 (CYP)

20

Page 21: Anesthesia Pharmacology Compiled

21

Variations in Drug Metabolism

• Genetic• Chronologic

Page 22: Anesthesia Pharmacology Compiled

22

Renal Drug Clearance

• Primary role of the kidneys: excrete into urine the unchanged, hydrophilic drugs, and the hepatic-derived metabolites from phase I and phase II reactions of lipophilic drugs.

• Passive glomerular filtration is a very inefficient process. In order to make renal elimination more efficient, discrete active transporters of organic acids and bases exist in the proximal renal tubular cells

Page 23: Anesthesia Pharmacology Compiled

Renal Drug Clearance

23

Page 24: Anesthesia Pharmacology Compiled

24

Hepatic Drug Clearance

• Drug elimination by the liver depends on:– intrinsic clearance– hepatic blood flow

Changes in either will change hepatic clearance. However, the extent of the change depends on the initial relationship between intrinsic clearance and hepatic blood flow

Page 25: Anesthesia Pharmacology Compiled

Hepatic Drug Clearance

28

Page 26: Anesthesia Pharmacology Compiled

29

Page 27: Anesthesia Pharmacology Compiled

30

Pharmacokinetic Models

Page 28: Anesthesia Pharmacology Compiled

31

• concentration of drug at its site or sites of action

– fundamental determinant of a drug's pharmacologic effects

Page 29: Anesthesia Pharmacology Compiled

32

• tissue drug concentration of an organ

– blood flow to the organ– concentration of drug in the arterial inflow of the organ– capacity of the organ to take up drug– diffusivity of the drug between the blood and the

organ

Page 30: Anesthesia Pharmacology Compiled

33

Physiologic versus Compartment Models

Page 31: Anesthesia Pharmacology Compiled

34

Physiologic or Perfusion models

– body tissues grouped according to distribution of cardiac output and capacity for drug uptake. • vessel-rich group• lean tissue group• vessel-poor group

Page 32: Anesthesia Pharmacology Compiled

35

• vessel-rich group– Highly perfused tissues with a

large amount of blood flow per volume of tissue

– brain, lungs, kidneys, and a subset of muscle

Page 33: Anesthesia Pharmacology Compiled

36

• lean tissue group or fast tissue group– tissues with a balanced

amount of blood flow per volume of tissue

– majority of muscle and some of the splanchnic bed (liver)

Page 34: Anesthesia Pharmacology Compiled

37

• vessel-poor group (slow tissue group) – composed of tissues that

had a large capacity for drug uptake but a limited tissue perfusion

– majority of the adipose tissue and the remainder of the splanchnic organs

Page 35: Anesthesia Pharmacology Compiled

38

• (Price and Price et al) – awakening after a single dose of thiopental• primarily a result of redistribution of thiopental from the brain to the muscle with little contribution by distribution to less well-perfused tissues or drug metabolism

Page 36: Anesthesia Pharmacology Compiled

39

• fundamental concept of redistribution

– applies to all lipophilic drugs

Page 37: Anesthesia Pharmacology Compiled

40

Perfusion-based physiologic pharmacokinetic models

– significant insights into how physiologic, pharmacologic, and pathologic distribution of cardiac output can effect drug distribution and elimination.

– verification of the predictions of these models requires measurement of drug concentrations in many different tissues• experimentally inefficient and destructive to the

system.

Page 38: Anesthesia Pharmacology Compiled

41

Compartmental Pharmacokinetic Models

• simpler mathematical models

• the body is composed of one or more compartments.

• compartments are theoretical entities that are used to mathematically characterize the blood concentration profile of a drug.

Page 39: Anesthesia Pharmacology Compiled

42

Compartmental Pharmacokinetic Models

– cardiac output is not a parameter

– cannot be used to predict directly the effect of cardiac failure on drug disposition

– can still quantify the effects of reduced cardiac output on the disposition of a drug

Page 40: Anesthesia Pharmacology Compiled

Pharmacokinetic Concepts

43

Page 41: Anesthesia Pharmacology Compiled

44

Rate Constants and Half-Lives• first-order kinetics

– one in which a constant fraction of the drug is removed during a finite period of time regardless of the drug amount or concentration.

= rate constant of the process

– apply not only to elimination, but also to absorption and distribution.

Page 42: Anesthesia Pharmacology Compiled

45

Rate constants

• absolute amount of drug removed is proportional to the concentration of the drug

• rate of change of the amount of drug at any given time is proportional to the concentration present at that time.

• concentration is high, more drugs will be removed

Page 43: Anesthesia Pharmacology Compiled

46

– k – units of “inverse time,”

(min-1 or h-1)• If 10% of the drug is eliminated per minute, then the rate

constant is 0.1 min-1.

Rate constants

Page 44: Anesthesia Pharmacology Compiled

47

– the rapidity of pharmacokinetic processes– the time required for the concentration to change

by a factor of 2. • calculated directly from the corresponding rate

constants with this simple equation:

Half-lives

Page 45: Anesthesia Pharmacology Compiled

48

Rate Constants and Half-Lives

• First-order processes approach completion• – a constant fraction of the drug– not an absolute amount, is removed per unit of

time.

Page 46: Anesthesia Pharmacology Compiled

49

• after five half-lives, • the process will be almost 97% complete

Page 47: Anesthesia Pharmacology Compiled

50

Volume of Distribution

• quantifies the extent of drug distribution

• Overall tissue capacity for uptake of a drug

– a function of the total mass of the tissues into which a drug distributes and their average affinity for the drug.

Page 48: Anesthesia Pharmacology Compiled

51

• an “apparent” volume

• size of hypothetical compartments– the concentration of drug in a reference

compartment (central or plasma compartment)

Volume of Distribution

Page 49: Anesthesia Pharmacology Compiled

52

• Vd

• relates the total amount of drug present to the concentration observed in the central compartment:

a total of 10 mg of drug Concentration:2 mg/L

Apparent volume of distribution

= 5 L

Volume of Distribution

Page 50: Anesthesia Pharmacology Compiled

53

• If drug extensively distributed– the concentration will be lower relative to the

amount of drug present– equates to a larger volume of distribution.

• lipophilic drugs– larger volumes of distribution than hydrophilic

drugs.

Volume of Distribution

Page 51: Anesthesia Pharmacology Compiled

54

• a numeric index of the extent of drug distribution

• does not have any relationship to the actual

volume of any tissue or group of tissues.

Volume of Distribution

Page 52: Anesthesia Pharmacology Compiled

55

• achieve a target plasma concentration:

***any change in state because of changes in physiologic and pathologic conditions can alter the volume of distribution, necessitating therapeutic adjustments.

Page 53: Anesthesia Pharmacology Compiled

56

Total Drug Clearance (Elimination)

• Cl, calculated from the declining blood levels:

Page 54: Anesthesia Pharmacology Compiled

57

• If a drug is rapidly removed from the plasma– its concentration will fall more quickly than the

concentration of a drug that is less readily eliminated.

Total Drug Clearance (Elimination)

Page 55: Anesthesia Pharmacology Compiled

58

• results in a smaller area under the concentration vs time curve, which equates to greater clearance

Total Drug Clearance (Elimination)

Page 56: Anesthesia Pharmacology Compiled

59

• estimation of drug clearance with these models has made important contributions to clinical pharmacology

– provided a great deal of clinically useful information regarding altered drug elimination in various pathologic conditions.

Page 57: Anesthesia Pharmacology Compiled

60

Elimination Half-Life

• pharmacokinetic parameter that best describes the physiologic process of drug elimination

• the time during which the amount of drug in the body decreases by 50%.

Page 58: Anesthesia Pharmacology Compiled

61

• when a physiologic or pathologic perturbation changes the elimination half-life of a drug – it is not a simple reflection of the change in the

body's ability to metabolize or eliminate the drug.

Elimination Half-Life

Page 59: Anesthesia Pharmacology Compiled

62

• Example:– the elimination half-life of thiopental is prolonged

in the elderly

– the elimination clearance is unchanged and the volume of distribution is increased.

– elderly patients need dosing strategies that accommodate for the change in the distribution of the drug rather than a decreased metabolism of the drug

Elimination Half-Life

Page 60: Anesthesia Pharmacology Compiled

63

• unable to give insight into the time that it takes for a single or a series of repeated drug doses to terminate its effect.

• elimination of drug from the body – begins the moment the drug is delivered to the organs of

elimination

• the rapid termination of effect of a bolus of an IV agent – due to redistribution of drug from the brain to the blood

and subsequently other tissue (e.g., muscle).

Elimination Half-Life

Page 61: Anesthesia Pharmacology Compiled

64

• effects of most anesthetics have waned long before even one elimination half-life has been completed,

• incapable of providing useful information regarding the duration of action following administration of intravenous agents.

• limited utility in anesthetic practice.

Elimination Half-Life

Page 62: Anesthesia Pharmacology Compiled

65

Effect of Hepatic or Renal Disease on Pharmacokinetic Parameters

• Diverse pathophysiologic changes – precise prediction of the pharmacokinetics of a given

drug in individual patients with hepatic or renal disease.

• liver function tests (transaminases) – unreliable predictors of the degree of liver function

and the remaining metabolic capacity for drug elimination.

Page 63: Anesthesia Pharmacology Compiled

66

Effect of Hepatic or Renal Disease on Pharmacokinetic Parameters

• In px with hepatic disease

– the elimination half-life of drugs metabolized or excreted by the liver is often increased

– decreased clearance

– poss increased volume of distribution caused by ascites and altered protein binding.

Page 64: Anesthesia Pharmacology Compiled

67

• hepatic drug clearance reduced,

– repeated bolus dosing or continuous infusion of such drugs as benzodiazepines, opioids, and barbiturates

– may result in excessive accumulation of drug – excessive and prolonged pharmacologic effects.

Effect of Hepatic or Renal Disease on Pharmacokinetic Parameters

Page 65: Anesthesia Pharmacology Compiled

68

• Drug concentration at steady-state is inversely proportional to elimination clearance.

• recovery from small doses of drugs(thiopental & fentanyl) – largely the result of redistribution– recovery from conservative doses will be minimally

affected by reductions in elimination clearance.

Effect of Hepatic or Renal Disease on Pharmacokinetic Parameters

Page 66: Anesthesia Pharmacology Compiled

69

• In patients with renal failure– almost always better to underestimate a patient's

dose requirement– observe the response– give additional drug if necessary

Effect of Hepatic or Renal Disease on Pharmacokinetic Parameters

Page 67: Anesthesia Pharmacology Compiled

70

Nonlinear Pharmacokinetics

• drug distribution and elimination are first-order processes

• parameters, such as clearance and elimination half-life, are independent of the dose or concentration of the drug

• the rate of elimination of a few drugs is dose-dependent, or nonlinear.

Page 68: Anesthesia Pharmacology Compiled

71

• Phenytoin – exhibits nonlinear elimination at therapeutic

concentrations

Nonlinear Pharmacokinetics

Page 69: Anesthesia Pharmacology Compiled

72

• in anesthetic practice, – extremely high doses of thiopental used for cerebral

protection can demonstrate zero-order elimination

• In theory– all drugs are cleared in a nonlinear fashion.

• In practice– the capacity to eliminate most drugs is so great that this

is usually not evident, even with toxic concentrations.

Nonlinear Pharmacokinetics

Page 70: Anesthesia Pharmacology Compiled

73

• Elimination of drugs – involves interactions with either enzymes

catalyzing biotransformation reactions or carrier proteins for transmembrane transport

Nonlinear Pharmacokinetics

Page 71: Anesthesia Pharmacology Compiled

74

Compartmental Pharmacokinetic Models

• One-Compartment Model

• illustrate the basic relationships among clearance, volume of distribution, and the elimination half-life.

• the body is seen as a single homogeneous compartment.

• Drug distribution after injection assumed instantaneous

• no concentration gradients within the compartment

• concentration can decrease only by elimination of drug from the system.

Page 72: Anesthesia Pharmacology Compiled

75

• All else being equal, the greater the clearance, the shorter the elimination half-life;

• the larger the volume of distribution, the longer the elimination half-life.

• the elimination half-life depends on clearance and volume of distribution– characterize the extent of drug distribution and efficiency of

drug elimination respectively.

One-Compartment Model

Page 73: Anesthesia Pharmacology Compiled

76

Two-Compartment Model• two discrete phases in the decline of the plasma

concentration:1. “distribution phase”

- first phase after injection - very rapid decrease in concentration. - rapid decrease in concentration

largely caused by passage of drug from the plasma into tissues.

2. a slower decline of the concentration owing to drug elimination.

Page 74: Anesthesia Pharmacology Compiled

77

• Elimination – begins immediately after injection– its contribution to the drop in plasma

concentration is initially much smaller than the fall in concentration because of drug distribution.

Two-Compartment Model

Page 75: Anesthesia Pharmacology Compiled

78

• central compartment– To which the drug is injected – from which the blood samples for measurement of

concentration are obtained,– that drug is eliminated only from the central compartment.

Page 76: Anesthesia Pharmacology Compiled

79

• plasma concentration of drugs with two-compartment kinetics is equal to the sum of two exponential terms:

• t = time• Cp(t) = plasma concentration at time t,• A = y-axis intercept of the distribution phase line• α = hybrid rate constant of the distribution phase• B = y-axis intercept of the elimination phase line• β = hybrid rate constant of the elimination phase.

Page 77: Anesthesia Pharmacology Compiled

80

• Immediately after injection, the first term represents a much larger fraction of the total plasma concentration than the second term.

• After several distribution half-lives, the value of the first term approaches zero, and the plasma concentration is essentially equal to the value of the second

Page 78: Anesthesia Pharmacology Compiled

81

Multicompartmental Models

• drug is initially distributed only within the central compartment.

• the initial apparent volume of distribution is the volume of the central compartment.

• Immediately after injection– the amount of drug present is the dose– the concentration is the extrapolated concentration at time t

= 0, which is equal to the sum of the intercepts of the distribution and elimination lines.

Page 79: Anesthesia Pharmacology Compiled

82

Multicompartmental Models

• volume of the central compartment – is important in clinical anesthesiology

– it is the pharmacokinetic parameter that determines the peak plasma concentration after an intravenous bolus injection

– Hypovolemia, for example, reduces the volume of the central compartment.

– If doses are not correspondingly reduced, the higher plasma

concentrations will increase the incidence of adverse pharmacologic effects.

Page 80: Anesthesia Pharmacology Compiled

83

Multicompartmental Models

• Immediately after intravenous injection– all of the drug is in the central compartment.

• Simultaneously, three processes begin. – Drug moves from the central to the peripheral

compartment, which also has a volume, V2. – As soon as drug appears in the peripheral

compartment, some passes back to the central compartment

Page 81: Anesthesia Pharmacology Compiled

84

• third process – begins immediately after administration of the drug – irreversible removal of drug from the system via the

central compartment.

• rapidity of the decrease in the central compartment concentration after intravenous injection – the magnitude of the compartmental volumes, – the intercompartmental clearance, – the elimination clearance.

Page 82: Anesthesia Pharmacology Compiled

85

• At equilibrium, the drug is distributed between the central and the peripheral compartments

• volume of distribution at steady-state (Vss )– ultimate volume of distribution– sum of V1 and V2. – Extensive tissue uptake of a drug is reflected by a large volume of

the peripheral compartment, which, in turn, results in a large Vss.

• • the elimination clearance is equal to the dose divided by

the area under the concentration versus time curve.

Page 83: Anesthesia Pharmacology Compiled

86

Three-Compartment Model

• After intravenous injection of some drugs;– the initial, rapid distribution phase is followed by a second, slower distribution

phase before the elimination phase becomes evident.• plasma concentration is the sum of three exponential terms:

• t = time• Cp(t) = plasma concentration at time t,• A = intercept of the rapid distribution phase line• α = hybrid rate constant of the rapid distribution phase• B = intercept of the slower distribution phase line• β = hybrid rate constant of the slower distribution phase• G = intercept of the elimination phase line• γ = hybrid rate constant of the elimination phase.

Page 84: Anesthesia Pharmacology Compiled

87

• the drug is injected into and eliminated from the central compartment.

• Drug is reversibly transferred between the central compartment and two peripheral compartments, which accounts for two distribution phases.

• three compartmental volumes: V1, V2, and V3, whose sum equals Vss; • three clearances: the rapid intercompartmental clearance, the slow intercompartmental

clearance, and elimination clearance

Page 85: Anesthesia Pharmacology Compiled

88

• Currently, most pharmacokinetic models are developed using population pharmacokinetic modeling

• pharmacokinetic parameters– concentration vs time data from

the entire group of subjects in a single stage, using nonlinear regression methods.

Page 86: Anesthesia Pharmacology Compiled

89

Noncompartmental (Stochastic) Pharmacokinetic Models

• employ mathematical techniques• • to characterize a pharmacokinetic data set that attempt to

avoid any preconceived notion of structure• • yield the pharmacokinetic parameters that summarize drug

distribution and elimination.

Page 87: Anesthesia Pharmacology Compiled

90

• basic assumptions – all of the elimination clearance occurs directly

from the plasma– the distribution and elimination of drug is a linear

and first-order process– the pharmacokinetics of the system does not vary

over the time of the data collection (time-invariant).

Noncompartmental (Stochastic) Pharmacokinetic Models

Page 88: Anesthesia Pharmacology Compiled

91

• main advantage – a general description of drug absorption,

distribution, and elimination can be made without resorting to more complex mathematical modeling techniques.

Noncompartmental (Stochastic) Pharmacokinetic Models

Page 89: Anesthesia Pharmacology Compiled

92

• mean residence time (MRT)

– main unique parameter of noncompartmental analysis

– the average time a drug molecule spends in the body before being eliminated.

Noncompartmental (Stochastic) Pharmacokinetic Models

Page 90: Anesthesia Pharmacology Compiled

93

MRT – fail to capture the contribution of extensive

distribution versus limited elimination to allow a drug to linger in the body

– fail to describe the situation in which the drug effect can dissipate by redistribution of drug from the site of action back into blood and then into other, less well-perfused tissues

Noncompartmental (Stochastic) Pharmacokinetic Models

Page 91: Anesthesia Pharmacology Compiled

94

Pharmacodynamics

Page 92: Anesthesia Pharmacology Compiled

95

Pharmacodynamics

• deals with the effects of drugs on biologic systems

• Receptor interactions• Dose response phenomena• Mechanism of action

Page 93: Anesthesia Pharmacology Compiled

96

Drug receptor Interactions

• Drug drug specific receptor change in cellular function– Cell membrane-bound proteins– Cytoplasmic/Nucleoplastic

Page 94: Anesthesia Pharmacology Compiled

97

Desensitization and Down-Regulation of Receptors

• Prolonged exposure desensitization lower maximal effects

Page 95: Anesthesia Pharmacology Compiled

98

Agonists, Partial Agonists, and Antagonists

Page 96: Anesthesia Pharmacology Compiled

99

Dose response relationship

• determine the relationship between increasing doses of a drug and the ensuing changes in pharmacologic effects– Potency– Drug receptor affinity– Efficacy– Population pharmacodynamic variability

Page 97: Anesthesia Pharmacology Compiled

100

• The potency of the drug—the dose required to produce a given

• The maximal effect indicates the rate of increase in effect as the dose is increased and is a reflection of the affinity of the receptor for the drug.

• The maximum effect is referred to as the efficacy of the drug.

Page 98: Anesthesia Pharmacology Compiled

101

Concentration-Response Relationships

• The magnitude of the pharmacologic effect is a function of the amount of drug present at the site of action, so increasing the dose increases the peak effect.

Page 99: Anesthesia Pharmacology Compiled

102

Concentration-Response Relationships

• The perfusion of the organ on which the drug acts• The tissue:blood partition coefficient of the drug• The rate of diffusion or transport of the drug from

the blood to the cellular site of action• The rate and affinity of drug–receptor binding• The time required for processes initiated by the

drug-receptor interaction to produce changes in cellular function

Page 100: Anesthesia Pharmacology Compiled

103

Context-Sensitive Decrement Times

The context-sensitive half-time is defined as the time required for the drug concentration of the plasma to decrease by 50%, where the context is the duration of the infusion.

During an infusion, drug is taken up by the inert, peripheral tissues.18 Once drug delivery is terminated, recovery occurs when the effect site concentration decreases below a threshold concentration for producing a pharmacologic effect (e.g., MACAWAKE—the concentration where 50% of patients follow commands

rate of elimination of the drug from the body can give some indication for the time required to reach a subtherapeutic effect site drug concentration

For drugs with multicompartmental kinetics, the elimination half-life will always overestimate the time to recovery from anesthetic drugs. .

Page 101: Anesthesia Pharmacology Compiled

104

here redistribution describes the drug returning from all tissue compartments, not just from the brain, into plasma as opposed to the previous usage that described redistribution of drug from brain to blood and into inert tissues.

To characterize the contribution of redistribution to the time required to reach a subtherapeutic drug concentration, the duration of infusion must be taken into account.

The time required for the drug concentration of the plasma to decrease by 50% increases as the duration of infusion increases.

Page 102: Anesthesia Pharmacology Compiled

105

Target-Controlled Infusions

Target controlled infusion(TCI) is an infusion system which allows the anesthetist to select the target blood concentration required for a particular effect and then to control the depth of anaesthesia by adjusting the requested target concentration, which includes the instantaneous calculation of the infusion rate necessary to obtain and maintain a given therapeutic blood concentration of drug based on average pharmacokinetic parameters.

the amount of drug to be administered into the central compartment to maintain the target concentration = (drug eliminated from the central compartment + plus drug distributed from the central compartment to peripheral compartments) - drug returning to the central compartment from peripheral compartments

Page 103: Anesthesia Pharmacology Compiled

106

Time to Maximum Effect Compartment Concentration

the delay between attaining a plasma concentration and an effect-site concentration is presumed to be a result of transfer of drug between the plasma compartment, VC, and an effect compartment, Ve, as well as the time required for a cellular response.

Page 104: Anesthesia Pharmacology Compiled

107

Volume of Distribution at Peak Effect 

Although the plasma concentration can be brought rapidly to the targeted drug concentration by administering a bolus dose to the central compartment (C × VC) and then held there by a computer-controlled infusion,the time for the effect site to reach the target concentration will be much longer than TMAX (4 minutes for propofol effect-site concentration to reach 95% of that targeted)

Volume of distribution over time is calculated by dividing the total amount of drug remaining in the body by the plasma drug concentration at each time, t. The time-dependent volume at the time of peak effect (or TMAX) is VDPE

The product of the targeted effect-site concentration and VDPE plus the amount lost to elimination in the time to TMAX becomes the proper bolus dose that will attain the target concentration at the effect site as rapidly as possible without overshoot

Page 105: Anesthesia Pharmacology Compiled

108

Front-End Pharmacokinetics

The term front-end pharmacokinetics refers to the intravascular mixing, pulmonary uptake, and recirculation events that occur in the first few minutes during and after intravenous drug administration.39

These kinetic events and the drug concentration versus time profile that results are important because the peak effect of rapidly acting drugs occurs during this temporal window.

Page 106: Anesthesia Pharmacology Compiled

Response Surface Models of Drug-Drug Interactions 

 During the course of an operation, the level of anesthetic drug administered is adjusted to ensure amnesia to ongoing events, provide immobility to noxious stimulation, and blunt the sympathetic response to noxious stimulation

it is possible to achieve an adequate anesthetic state with a high dose of a sedative-hypnotic alone (i.e., a volatile anesthetic or propofol), the effect-site drug concentration necessary is often associated with excessive hemodynamic depression58 and excessively deep plane of hypnosis that may be associated with long-standing morbidity or mortality

administration of two volatile anesthetics or a volatile anesthetic and propofol produce a net-additive effect, the combination of an opioid and a sedative-hypnotic are synergistic for most pharmacologic effects

109