01.10.2015 Views

Extent of Absorption

Extent of Absorption

Extent of Absorption

SHOW MORE
SHOW LESS
  • No tags were found...

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>Extent</strong> <strong>of</strong> <strong>Absorption</strong><br />

The extent <strong>of</strong> absorption is important in determining the total<br />

body exposure or internal dose, and therefore is an important<br />

variable during chronic toxicity studies and/or chronic human<br />

exposure. The extent <strong>of</strong> absorption depends on the extent to<br />

which the chemical is transferred from the site <strong>of</strong><br />

administration into the local tissue, and the extent to which it<br />

is metabolized or broken down by local tissues prior to<br />

reaching the general circulation. An additional variable<br />

affecting the extent <strong>of</strong> absorption is the rate <strong>of</strong> removal from<br />

the site <strong>of</strong> administration by other processes compared with the<br />

rate <strong>of</strong> absorption<br />

١٨


Chemicals given via the gastrointestinal tract may be<br />

subject to a wide range <strong>of</strong> pH values and<br />

metabolizing enzymes in the gut lumen, gut wall, and<br />

liver before they reach the general circulation. The<br />

initial loss <strong>of</strong> chemical prior to it ever entering the<br />

blood is termed first-pass metabolism or pre-<br />

systemic metabolism; ; it may in some cases remove<br />

up to 100% <strong>of</strong> the administered dose so that none <strong>of</strong><br />

the parent chemical reaches the general circulation.<br />

The intestinal lumen contains a range <strong>of</strong> hydrolytic<br />

enzymes involved in the digestion <strong>of</strong> nutrients. The<br />

gut wall can perform similar hydrolytic reactions and<br />

contains enzymes that can oxidize many drugs<br />

١٩


٢٠


<strong>Absorption</strong> and Bioavailability<br />

Irrespective <strong>of</strong> the reason that is responsible for the incomplete<br />

absorption <strong>of</strong> the chemical as the parent compound, it is<br />

essential that there is a parameter which defines the extent <strong>of</strong><br />

transfer <strong>of</strong> the intact chemical from the site <strong>of</strong> administration<br />

into the general circulation. This parameter is the<br />

bioavailability, , which is simply the fraction <strong>of</strong> the dose<br />

administered that reaches the general circulation as the parent<br />

compound. (The term bioavailability is perhaps the most<br />

misused <strong>of</strong> all kinetic parameters and is sometimes used<br />

incorrectly in a general sense as the amount <strong>of</strong> drug available<br />

specifically to the site <strong>of</strong> toxicity.)<br />

٢١


Calculation <strong>of</strong> Bioavailability<br />

The fraction absorbed as the intact compound or bioavailability (F)) is<br />

determined by comparison with intravenous (i.v(<br />

i.v.) .) dosing (where F = 1 by<br />

definition). The bioavailability can be determined from the area under the<br />

plasma concentration–time time curve (AUC) <strong>of</strong> the parent compound , or the<br />

percentage dose excreted in urine as the parent compound, i.e. for an oral<br />

dose:<br />

٢٢


٢٣


2. Distribution<br />

Distribution is the reversible transfer <strong>of</strong> the<br />

chemical between the general circulation and<br />

the tissues. Irreversible processes such as<br />

excretion, metabolism, or covalent binding are<br />

part <strong>of</strong> elimination and do not contribute to<br />

distribution parameters. The important<br />

distribution parameters relate to the rate and<br />

extent <strong>of</strong> distribution.<br />

٢٤


Rate <strong>of</strong> Distribution<br />

The rate at which a chemical may enter or leave a tissue may be<br />

limited by two factors:<br />

(i) the ability <strong>of</strong> the compound to cross cell membranes and<br />

(ii) the blood flow to the tissues in which the chemical<br />

accumulates.<br />

The rate <strong>of</strong> distribution <strong>of</strong> highly water-soluble compounds may<br />

be slow due to their slow transfer from plasma into body<br />

tissues such as liver and muscle; water-soluble compounds do<br />

not accumulate in adipose tissue. In contrast, very lipid-soluble<br />

chemicals may rapidly cross cell membranes but the rate <strong>of</strong><br />

distribution may be slow because they accumulate in adipose<br />

tissue, and their overall distribution rate may be limited by<br />

blood flow to adipose tissue<br />

٢٥


The rate <strong>of</strong> distribution is indicated by the distribution<br />

rate constant, which is determined from the decrease<br />

in plasma concentrations in early time points after an<br />

intravenous dose. The rate constants refer to a mean<br />

rate <strong>of</strong> removal from the circulation and may not<br />

correlate with uptake into a specific tissue. Once an<br />

equilibrium has been reached between the general<br />

circulation and a tissue, any process which lowers the<br />

blood (plasma) concentration will cause a parallel<br />

decrease in the tissue concentration.<br />

٢٦


Factors affecting distribution<br />

1. Blood flow<br />

Drugs are readily distributed to highly perfused tissue<br />

like brain, liver, and kidneys<br />

2. Permeability limitations<br />

Many drugs do not readily enter the brain due to the<br />

blood brain barrier<br />

3. Protein binding<br />

Acidic drugs are bound to the most abundant plasma<br />

protein (albumin); while basic drugs bind to -1-<br />

acid glycoprotein.<br />

٢٧


4. Effect <strong>of</strong> pH<br />

The pH <strong>of</strong> the blood or tissue affect the<br />

ionization <strong>of</strong> the drug and thus its distribution<br />

5. Age<br />

In old people, Protein binding and body water<br />

will decrease, thus increasing the concentration<br />

<strong>of</strong> the drug per unit time<br />

6. Existence <strong>of</strong> storage sites:<br />

These include: Adipose tissue, plasma proteins,<br />

liver and kidneys, and bone<br />

٢٨


<strong>Extent</strong> <strong>of</strong> Distribution<br />

The extent <strong>of</strong> tissue distribution <strong>of</strong> a chemical depends<br />

on the relative affinity <strong>of</strong> the blood or plasma<br />

compared with the tissues. Highly water-soluble<br />

compounds that are unable to cross cell membranes<br />

readily are largely restricted to extracellular fluid<br />

(about 13 L per 70 kg body weight). Water-soluble<br />

compounds capable <strong>of</strong> crossing cell membranes (e.g.(<br />

caffeine, ethanol) are largely present in total body<br />

water (about 41 L per 70 kg body weight).<br />

٢٩


Lipid-soluble compounds frequently show extensive<br />

uptake into tissues and may be present in the lipids <strong>of</strong><br />

cell membranes, adipose tissue.<br />

The partitioning between circulating lipoproteins and<br />

tissue constituents is complex and may result in<br />

extremely low plasma concentrations. A factor which<br />

may further complicate the plasma/tissue partitioning<br />

is that some chemicals bind reversibly to circulating<br />

proteins such as albumin (for acid molecules) and<br />

acid glycoprotein (for basic molecules).<br />

٣٠


The extent and pattern <strong>of</strong> tissue distribution can<br />

be investigated by direct measurement <strong>of</strong><br />

tissue concentrations in animals. Tissue<br />

concentrations cannot be measured in human<br />

studies and, therefore, the extent <strong>of</strong> distribution<br />

in humans has to be determined based solely<br />

on the concentrations remaining in plasma or<br />

blood after distribution is complete.<br />

٣١


The parameter used to reflect the extent <strong>of</strong> distribution is the<br />

apparent volume <strong>of</strong> distribution (V),(<br />

which relates the total<br />

amount <strong>of</strong> the chemical in the body (Ab(<br />

Ab) ) to the circulating<br />

concentration (C)(<br />

) at any time after distribution is complete:<br />

The volumes <strong>of</strong> distribution <strong>of</strong> tubocurarine and caffeine are<br />

about 13 and 41 L per 70 kg because <strong>of</strong> their restricted<br />

distribution<br />

٣٢


Caffeine<br />

tubocurarine<br />

٣٣


when a chemical shows a more extensive reversible<br />

uptake into one or more tissues the plasma<br />

concentration will be lowered and the value <strong>of</strong> V will<br />

increase. For highly lipid-soluble chemicals, such as<br />

organochlorine pesticides, which accumulate in<br />

adipose tissue, the plasma concentration may be so<br />

low that the value <strong>of</strong> V may be many litres for each<br />

kilogram <strong>of</strong> body weight. This is not a real volume <strong>of</strong><br />

plasma and therefore V is called the apparent volume<br />

<strong>of</strong> distribution.<br />

٣٤


It is an important parameter because extensive<br />

reversible distribution into tissues, which will<br />

give a high value <strong>of</strong> V, , is associated with a low<br />

elimination rate and a long half-life life . It must be<br />

emphasized that the apparent volume <strong>of</strong><br />

distribution simply reflects the extent to which<br />

the chemical has moved out <strong>of</strong> the site <strong>of</strong><br />

measurement (the general circulation) into<br />

tissues, and it does not reflect uptake into any<br />

specific tissue(s).<br />

٣٥


Elimination<br />

The parameter most commonly used to describe<br />

the rate <strong>of</strong> elimination <strong>of</strong> a chemical is the<br />

half-life life . Most toxicokinetic processes are<br />

first-order reactions, i.e. the rate at which the<br />

process occurs is proportional to the amount <strong>of</strong><br />

chemical present. High rates (expressed as<br />

mass/time) occur at high concentrations and<br />

the rate decreases as the concentration<br />

decreases; in consequence the decrease is an<br />

exponential curve.<br />

٣٦


The usual way to analyze exponential changes is to use<br />

logarithmically transformed data which converts an<br />

exponential into a straight line. The slope <strong>of</strong> the line<br />

is the rate constant (k)(<br />

) for the process and the half-life<br />

life<br />

for the process is calculated as 0.693/k. . Rate<br />

constants and half-lives lives can be determined for<br />

absorption, distribution, and elimination processes.<br />

The clearance <strong>of</strong> a chemical is determined by the<br />

ability <strong>of</strong> the organs <strong>of</strong> elimination (e.g.(<br />

the liver,<br />

kidney, or lungs) to extract the chemical from the<br />

plasma or blood and permanently remove it by<br />

metabolism or excretion.<br />

٣٧


The mechanisms <strong>of</strong> elimination depend on the<br />

chemical characteristics <strong>of</strong> the compound:<br />

• volatile chemicals are exhaled,<br />

• water-soluble chemicals are eliminated in the<br />

urine and/or bile and<br />

• lipid-soluble chemicals are eliminated by<br />

metabolism to more water-soluble molecules,<br />

which are then eliminated in the urine and/or<br />

bile.<br />

٣٨


٣٩


If a chemical undergoes metabolic activation then<br />

toxicokinetic studies should measure both the parent<br />

chemical and the active metabolite. If the metabolite<br />

is so reactive that it does not leave the tissue in which<br />

it is produced (e.g.(<br />

alkylating metabolites <strong>of</strong> chemical<br />

carcinogens), then toxicokinetic studies should define<br />

the delivery <strong>of</strong> the parent chemical to the tissues, and<br />

the process <strong>of</strong> local activation should be regarded as<br />

part <strong>of</strong> tissue sensitivity (toxicodynamics(<br />

toxicodynamics) ) because it<br />

is not part <strong>of</strong> toxicokinetics, i.e. the movement <strong>of</strong> the<br />

chemical and/or metabolites around the body.<br />

٤٠


• Clearance = a ratio relating the rate <strong>of</strong><br />

elimination <strong>of</strong> a chemical from an appropriate<br />

reference fluid (usually plasma) to its<br />

concentration in the same reference fluid.<br />

Clearance has the units <strong>of</strong> flow rate in<br />

milliliters per minutes (mL(<br />

mL/min).<br />

• A clearance <strong>of</strong> 100 mL/minute <strong>of</strong> a chemical<br />

means that 100 mL <strong>of</strong> blood/plasma is<br />

completely cleared <strong>of</strong> the compound in each<br />

minute.<br />

٤١


The best measure <strong>of</strong> the ability <strong>of</strong> the organs <strong>of</strong><br />

elimination to remove the compound from the<br />

body is the clearance (CL(<br />

CL):<br />

Because the rate <strong>of</strong> elimination is proportional to the concentration,<br />

clearance is a constant for first-order processes and is independent <strong>of</strong><br />

dose. It can be regarded as the volume <strong>of</strong> plasma (or blood) cleared <strong>of</strong><br />

compound within a unit <strong>of</strong> time (e.g. mL/ min).<br />

٤٢


Renal clearance depends on the extent <strong>of</strong> protein<br />

binding, tubular secretion and passive reabsorption in<br />

the renal tubule; it can be measured directly from the<br />

concentrations present in plasma and urine:<br />

The total clearance or plasma clearance (which is the<br />

sum <strong>of</strong> all elimination processes, i.e. renal metabolic,<br />

etc.) ) is possibly the most important toxicokinetic<br />

parameter.<br />

٤٣


It is measured from the total amount <strong>of</strong> compound available for<br />

removal (i.e.(<br />

an intravenous dose) and the total area under the<br />

plasma concentration–time time curve (AUC) extrapolated to<br />

infinity.<br />

Plasma clearance reflects the overall ability <strong>of</strong> the body to remove<br />

permanently the chemical from the plasma. Plasma clearance is the<br />

parameter that is altered by factors such as enzyme induction, liver<br />

disease, kidney disease, inter-individual or inter-species differences in<br />

hepatic enzymes or in some cases organ blood flow.<br />

٤٤


Once the chemical is in the general circulation, the same volume<br />

<strong>of</strong> plasma will be cleared <strong>of</strong> chemical per minute (i.e.(<br />

the<br />

clearance value) applies irrespective <strong>of</strong> the route <strong>of</strong> delivery <strong>of</strong> o<br />

chemical into the circulation. However, the bioavailability (F)(<br />

will determine the proportion <strong>of</strong> the dose reaching the general<br />

circulation. Therefore, bioavailability has to be taken into<br />

account if clearance is calculated from data from a non-<br />

intravenous route (e.g.(<br />

oral):<br />

٤٥


Measurement <strong>of</strong> dose/AUC for an oral dose determines CL/F, , which contains<br />

two potentially independent variables – the amount <strong>of</strong> chemical delivered<br />

to the blood from the site <strong>of</strong> administration and the clearance <strong>of</strong> chemical<br />

present in the blood. The overall rate <strong>of</strong> elimination, as indicated by the<br />

terminal half-life life (t(<br />

), is dependent on two physiologically related and<br />

independent variables:<br />

where CL is the ability to extract and remove irreversibly the compound from f<br />

the general circulation, and V the extent to which the compound has left the<br />

general circulation in a reversible equilibrium with tissues.<br />

٤٦


Chemicals that are extremely lipid-soluble and<br />

are sequestered in adipose tissue are<br />

eliminated slowly. Lipid soluble<br />

organochlorine compounds, which are not<br />

substrates for P450 oxidation, due to the<br />

blocking <strong>of</strong> possible sites <strong>of</strong> oxidation by<br />

chloro-substituents<br />

substituents, , are eliminated extremely<br />

slowly: for example, the half-life life <strong>of</strong> 2,3,7,8-<br />

tetrachlorodibenzodioxin (TCDD) is about 8<br />

years in humans.<br />

٤٧


Summary <strong>of</strong> Toxicokinetic<br />

Parameters<br />

• Apparent volume <strong>of</strong> distribution (V(<br />

d )<br />

• A way to express the apparent space in the body that a chemical<br />

occupies<br />

• Expressed as a proportionality constant (in units <strong>of</strong> volume [e.g.,<br />

.,<br />

liters <strong>of</strong> blood] or volume/body weight)<br />

• Relates the total amount <strong>of</strong> chemical in the body to the<br />

concentration in plasma<br />

• For a 70-kg human,<br />

• Plasma volume ≈ 3 L (~0.045 L/kg body weight)<br />

• Total blood volume ≈ 5 L (~0.07 L/kg body weight)<br />

• Total extracellular fluid volume ≈ 12 L (~0.2 L/kg body weight)<br />

• Total body water ≈ 42 L (~0.6L/kg body weight)<br />

٤٨


• Example – If you know that 3 mg <strong>of</strong> chemical has been<br />

injected into the body, and the concentration <strong>of</strong> the<br />

chemical in the plasma is 1 mg/L, then you can calculate<br />

that the apparent V d to be 3 L. This suggests that the<br />

chemical is confined to the plasma space, and is not<br />

significantly absorbed into the tissues or into the<br />

particulate components (i.e., blood cells) in blood.<br />

3 mg ÷ 1.0 mg/L = 3.0 L<br />

٤٩


• Example – If you know that 3 mg <strong>of</strong> chemical has been<br />

injected and the concentration <strong>of</strong> the chemical in the<br />

plasma is 0.55 mg/L, then the apparent V d = 5.5 L. This<br />

suggests that the chemical may be distributed evenly<br />

between the plasma and the blood cells. More likely,<br />

the reality is that the chemical is somewhat absorbed<br />

into the tissues.<br />

3 mg ÷ 0.55 mg/L = 5.5 L<br />

٥٠


• Example – If you know that 3 mg <strong>of</strong> chemical has been<br />

injected and the concentration <strong>of</strong> the chemical in the<br />

plasma is 0.25 mg/L, then the apparent V d = 12 L. This<br />

suggests that the chemical may be distributed evenly<br />

within the total extracellular water. More likely, the<br />

reality is that the chemical is somewhat absorbed into<br />

the tissues, and perhaps somewhat metabolized.<br />

3 mg ÷ 0.25 mg/L = 12 L<br />

• It is possible to have a V d greater than the total body<br />

water<br />

• A high V d is an indication that one or both <strong>of</strong> two things<br />

has occurred,<br />

• The chemical has been absorbed from the blood and<br />

concentrated in the tissues.<br />

• The chemical has been metabolized.<br />

٥١


• Clearance (Cl(<br />

Cl)<br />

• A ratio relating the rate <strong>of</strong> elimination <strong>of</strong> a chemical<br />

from an appropriate reference fluid (usually plasma) to<br />

its concentration in the same reference fluid.<br />

• Has the units <strong>of</strong> flow rate in volume cleared per unit<br />

time (e.g., mL/min)<br />

• A clearance <strong>of</strong> 100 mL/minute <strong>of</strong> a chemical means that<br />

100 mL <strong>of</strong> blood/plasma is completely cleared <strong>of</strong> the<br />

compound in each minute .<br />

٥٢


• Zero- versus first-order kinetics<br />

• For zero order kinetics, the rate <strong>of</strong> elimination <strong>of</strong> a compound is a<br />

constant, and is independent <strong>of</strong> the concentration <strong>of</strong> the chemical l in<br />

the blood. For example, the average human body is able to<br />

eliminate ~10-15<br />

15 mL <strong>of</strong> ethanol per hour, regardless <strong>of</strong> the amount<br />

<strong>of</strong> ethanol consumed. This may be higher or lower depending on<br />

the factors previously discussed (e.g., induction <strong>of</strong> alcohol<br />

dehydrogenase by repeated alcohol exposure).<br />

• For first-order kinetics, the rate <strong>of</strong> elimination <strong>of</strong> a compound is<br />

dependent on the concentration <strong>of</strong> the chemical in the blood. The<br />

higher the concentration, the more rapidly the chemical is<br />

eliminated, unless the elimination mechanisms have been saturated.<br />

At that point, the kinetics become zero-order. order. This is known as<br />

saturation kinetics.<br />

٥٣


• Half-life life (t 1/2<br />

1/2 )<br />

• The time required for the concentration <strong>of</strong> a chemical in<br />

the plasma to decrease by 50%.<br />

• This is a constant for all but zero-order order kinetics. For<br />

example, for a compound eliminated by first-order<br />

kinetics, if the concentration at time 0 is 4 mg/L, and the<br />

t 1/2 is 6 hours, then at the end <strong>of</strong> 6 hours, the plasma<br />

concentration will be (0.5 x 4 mg/mL<br />

mL =) 2 mg/L, and at<br />

the end <strong>of</strong> the next 6 hours, the concentration will be<br />

(0.5 x 2 mg/mL<br />

mL =) 1 mg/L, and so on.<br />

٥٤


• Area under the Curve (AUC)<br />

• A measure <strong>of</strong> the total amount <strong>of</strong> chemical present in<br />

the body over a defined period. This is defined as the<br />

area under the concentration vs time curve for the<br />

defined period.<br />

• Example: The shaded area in the figure below<br />

represents the AUC(0-6hr). You can express the AUC<br />

for any given time period. It is <strong>of</strong>ten expressed as the<br />

AUC(0-∞).<br />

•<br />

Concentration<br />

in<br />

plasma<br />

0<br />

Time (hrs)<br />

6<br />

٥٥


• Classical toxicokinetics<br />

• One compartment model<br />

• The elimination <strong>of</strong> a chemical is said to follow a one-<br />

compartment model when the elimination phase <strong>of</strong> the<br />

log concentration vs time curve is a straight line.<br />

Conceptually, this is as though the chemical were<br />

evenly distributed throughout a single body<br />

compartment (e.g., total body water), and is eliminated<br />

at a constant rate over time. That is, a constant<br />

percentage <strong>of</strong> the chemical present is eliminated over<br />

any given time period.<br />

٥٦


• The rate at which a chemical is eliminated at any time is directly<br />

proportional to the amount <strong>of</strong> that chemical in the body at that time.<br />

• A one-compartment model indicates that no one tissue has a high affinity<br />

for the chemical. Changes in the plasma reflect changes in the tissue.<br />

Plasma<br />

Concentratio<br />

n<br />

Log <strong>of</strong><br />

Plasma<br />

Concentratio<br />

n<br />

Time<br />

Time<br />

٥٧


• Two compartment model<br />

• The elimination <strong>of</strong> a chemical is said to follow a two-<br />

compartment model when the elimination phase <strong>of</strong> the<br />

log concentration vs time curve is not a straight line.<br />

Conceptually, this is as though the chemical were<br />

unevenly distributed throughout two body<br />

compartments (e.g., highly concentrated in tissues and a<br />

lower concentration in the plasma). The net elimination<br />

is a sum <strong>of</strong> the rates <strong>of</strong> elimination from the various<br />

compartments.<br />

• The appearance <strong>of</strong> a two-compartment curve suggests<br />

that the chemical is distributed out <strong>of</strong> the blood into<br />

tissues.<br />

٥٨


Alpha distribution<br />

Log <strong>of</strong> Plasma<br />

Concentration<br />

Beta distribution<br />

Time<br />

٥٩


Chronic Administration<br />

The kinetic concepts and parameters <strong>of</strong> a single dose (as<br />

discussed above) apply to chronic administration, but<br />

the exposure has to allow for the fact that not all <strong>of</strong><br />

the previous dose(s) ) may have been eliminated when<br />

the subsequent dose is given. Therefore, there may be<br />

an increase in plasma concentration (and body load)<br />

until an equilibrium is reached in which the rate <strong>of</strong><br />

elimination balances out the rate <strong>of</strong> input, in other<br />

words, the daily dose is eliminated each day.<br />

٦٠


٦١


٦٢


The equations above assume that CL is not<br />

altered by repeated exposure; the assumption is<br />

not correct if the chemical induces or inhibits<br />

its own elimination because clearance would<br />

be increased or decreased, respectively, after<br />

the period <strong>of</strong> chronic intake.<br />

٦٣


Because the plasma and therefore tissue concentrations<br />

increase during chronic intake until an equilibrium is<br />

reached , the amount in the body (Ab(<br />

Ab) ) will also<br />

increase to reach a steady state. The time taken to<br />

reach steady state is 4–54<br />

5 times the elimination half-<br />

life and, therefore, the true duration <strong>of</strong> steady-state<br />

state<br />

exposure in a toxicity study is the study duration<br />

minus 4–54<br />

5 half-lives lives <strong>of</strong> the chemical (needed to reach<br />

the steady state). This is particularly important for<br />

chemicals that have a very long half-life; life; for example<br />

in rodents the steady-state state body load <strong>of</strong> TCDD, which<br />

has a half-life life in rats <strong>of</strong> about 1 month, will not be<br />

reached until after about 4–54<br />

5 months <strong>of</strong> continuous<br />

treatment.<br />

٦٤


Saturation Kinetics<br />

All the parameters described above relate to first-order<br />

processes and, therefore, are independent <strong>of</strong> dose at<br />

low doses. However, at high doses and/or during<br />

chronic studies it is possible to overload or saturate<br />

compound–protein protein interactions. Under such<br />

circumstances any increase in the concentration <strong>of</strong> the<br />

compound cannot give a proportional (first-order)<br />

increase in the rate <strong>of</strong> the process. When a process is<br />

saturated the rate is at the maximum possible and is<br />

essentially independent <strong>of</strong> concentration.<br />

٦٥


In simple mathematical terms this means that the reaction<br />

changes from first to zero order. This is best described by<br />

Michaelis–Menten<br />

Menten kinetics, i.e.<br />

٦٦


3. Metabolism<br />

• Many xenobiotics undergo chemical transformation<br />

(biotransformation; metabolism) when introduced<br />

into biologic systems like the human body.<br />

• Biotransformation is <strong>of</strong>ten mediated by enzymes<br />

• End result <strong>of</strong> biotransformation is either alteration <strong>of</strong><br />

the parent molecule, or conjugation <strong>of</strong> the parent<br />

molecule (or its metabolites) with endogenous<br />

substances in the body.<br />

• Enzymes involved in biotransformation can act on<br />

either endogenous or xenobiotic compounds,<br />

especially if the xenobiotics are structurally similar to<br />

endogenous compounds<br />

٦٧


• Example: Cholinesterase is an enzyme that normally metabolizes<br />

acetylcholine, a neurotransmitter found in the synapse.<br />

Cholinesterase can also metabolize the local anesthetic agent<br />

procaine and the muscle-paralyzing agent succinylcholine. . If a<br />

person has high levels <strong>of</strong> cholinesterase activity, the effectiveness<br />

<strong>of</strong> these drugs can be compromised. If a person is exposed to<br />

anything that inhibits cholinesterase activity, the pharmacologic<br />

effects <strong>of</strong> the drugs can be enhanced, resulting in toxicity.<br />

• Example: Monoamine oxidase (MAO) is an enzyme that normally<br />

metabolizes biologic amines like epinephrine. MAO can also<br />

oxidize a variety <strong>of</strong> drugs. If a person is taking a drug that inhibits i<br />

MAO activity (like many blood pressure medications), it can be<br />

dangerous for that person to take other drugs that can be<br />

metabolized by MAO.<br />

٦٨


• The products <strong>of</strong> biotransformation can be either less toxic<br />

than, more toxic than, or about as toxic as the parent<br />

molecules.<br />

• Enzymes involved in biotransformation are sometimes called<br />

“drug metabolizing enzymes”. . Although strictly speaking this<br />

is a misnomer because many <strong>of</strong> the substrates are not drugs,<br />

the term is still commonly used.<br />

• Location <strong>of</strong> metabolic enzymes<br />

• Species – found in virtually every species, although the type and<br />

amount vary tremendously.<br />

• Organs – present in many tissues. Many enzymes are particularly<br />

abundant in the liver.<br />

• Subcellular – many <strong>of</strong> the drug-metabolizing enzymes are located in<br />

the smooth endoplasmic reticulum (SER). These are called<br />

“microsomal” enzymes.<br />

٦٩


• Types <strong>of</strong> Biotransformation Reactions<br />

• Basically, two types <strong>of</strong> reactions, nonsynthetic (Phase I) and<br />

synthetic (Phase II)<br />

• Phase I reactions<br />

• Involve modification <strong>of</strong> the basic structure <strong>of</strong> the substrate<br />

• Do not involve covalent binding <strong>of</strong> the substrate to an endogenous<br />

compound<br />

• Examples include hydrolysis, oxidation, and reduction reactions<br />

• Phase I enzymes are <strong>of</strong>ten membrane-bound bound (e.g.,<br />

microsomal). This is because they generally act on more lipid-<br />

soluble (nonpolar(<br />

nonpolar) ) substrates, and their purpose is to make the<br />

compounds MORE POLAR and therefore, MORE EASILY<br />

EXCRETABLE by the kidney and biliary tract. Think <strong>of</strong> the<br />

active transport mechanisms in the kidney.<br />

٧٠


• Oxidation<br />

• Uses molecular oxygen (O2). One atom <strong>of</strong> oxygen is<br />

combined with hydrogen to form water, and the other<br />

atom <strong>of</strong> oxygen is introduced into the substrate<br />

molecule.<br />

• Involves several enzymatic steps.<br />

• The oxidative system is <strong>of</strong>ten known as the “mixed<br />

function oxidase” system”. . These enzymes are some <strong>of</strong><br />

the most thoroughly researched enzymes in biological<br />

systems.<br />

• One subfamily <strong>of</strong> the mixed function oxidase system is the<br />

group <strong>of</strong> enzymes known as Cytochrome P-450<br />

enzymes.<br />

They are so called because <strong>of</strong> their absorbance characteristics<br />

at wavelengths <strong>of</strong> 448-450 450 nm.<br />

٧١


• Anything that affects the activity <strong>of</strong> any one <strong>of</strong> the steps can affect a<br />

the way the body reacts to a given drug or other xenobiotic.<br />

• Examples <strong>of</strong> the various types <strong>of</strong> oxidation reactions are in the<br />

textbook,<br />

• Deamination – replacement <strong>of</strong> an amine group (NH 2<br />

) with an oxygen<br />

(O) atom<br />

• N-, , O-, O , or S-DealkylationS<br />

– replacement <strong>of</strong> an alkyl group (e.g., CH 3<br />

)<br />

with a hydrogen atom. Typically, the alkyl group in the parent<br />

molecule is bonded to a N, O, or S atom.<br />

• Aliphatic or aromatic hydroxylation – addition <strong>of</strong> a hydroxyl group<br />

(OH) to a molecule<br />

• N-oxidation<br />

– replacement <strong>of</strong> a hydrogen atom on an amine with an<br />

oxygen<br />

• S-oxidation<br />

– addition <strong>of</strong> an oxygen atom to a sulfur atom<br />

• Conversion <strong>of</strong> a hydroxyl group (alcohol) to a carboxyl group (acid)<br />

٧٢


• Reduction<br />

• Azo reduction – reduction <strong>of</strong> an azo bond (N=N) to two<br />

amines (NH 2 )<br />

• Nitro reduction – reduction <strong>of</strong> a nitro group (NO 2 ) to an<br />

amine<br />

• Hydrolysis<br />

• Addition <strong>of</strong> water (H 2 O) to an ester bond (C-O-O-C) C) to<br />

form an alcohol (C-OH) and a carboxylic acid (COOH)<br />

R-C-O-O-C-R R + H-O-H H = R-C-OH + R-COOHR<br />

٧٣


• Phase II reactions<br />

• Involve addition <strong>of</strong> a c<strong>of</strong>actor to a substrate to form a new<br />

product. Therefore, the rate <strong>of</strong> these reactions can be<br />

limited by the availability <strong>of</strong> the c<strong>of</strong>actor.<br />

• Phase II enzymes may be either microsomal or cytosolic.<br />

This is because the primary purpose <strong>of</strong> the Phase II<br />

reactions is not so much to increase the polarity <strong>of</strong> the<br />

parent compound (although that is part <strong>of</strong> what they<br />

accomplish). The primary purpose is to increase the<br />

molecular weight <strong>of</strong> the parent compound to make it a<br />

better substrate for active transport mechanisms in the<br />

biliary tract.<br />

• Various factors can affect the availability <strong>of</strong> c<strong>of</strong>actors. For<br />

example, fasting markedly reduces the amount <strong>of</strong><br />

glutathione available in the liver.<br />

٧٤


• Sulfation<br />

• Replacement <strong>of</strong> a hydrogen atom (H) with a sulfonate (SO3)<br />

• Uses the enzyme sulfotransferase<br />

• Uses the c<strong>of</strong>actor called PAPS (phosphoadenosine(<br />

phosphosulfate)<br />

• Produces a highly water-soluble sulfuric acid ester<br />

• Glucuronidation<br />

• Replacement <strong>of</strong> a hydrogen atom with a glucuronic acid<br />

• Uses the enzyme UDP-glucuronosyl<br />

transferase (UDP-GT)<br />

• Uses the c<strong>of</strong>actor called UDPGA (uridine(<br />

diphosphate glucuronic<br />

acid)<br />

• One <strong>of</strong> the major Phase II enzymatic pathways<br />

٧٥


• Acetylation<br />

• Replacement <strong>of</strong> a hydrogen atom with an acetyl group<br />

• Uses the enzyme acetyltransferase<br />

• Uses the c<strong>of</strong>actor called acetyl CoA (acetyl coenzyme A)<br />

• Sometimes results in a less water-soluble product<br />

• Remember the discussion <strong>of</strong> slow versus fast acetylators.<br />

• Methylation<br />

• Replacement <strong>of</strong> a hydrogen atom with a methyl group<br />

• Uses the enzyme methyltransferase<br />

• Uses the c<strong>of</strong>actor called SAM (S-adenosyl<br />

methionine)<br />

• Common but relatively minor pathway<br />

٧٦


• Glutathione conjugation<br />

• Adds a glutathione molecule to the parent compound, either by<br />

direct addition or by replacement <strong>of</strong> an electrophilic substituent<br />

(e.g., a halogen atom)<br />

• Uses the enzyme glutathione transferase (GST)<br />

• Uses the c<strong>of</strong>actor called glutathione (a tripeptide made up <strong>of</strong><br />

glycine, cysteine, , and glutamic acid<br />

• One <strong>of</strong> the major Phase II enzymatic pathways<br />

• Amino acid conjugation<br />

• Adds an amino acid to the parent compound.<br />

• Mercapturic acid formation<br />

• Formed by cleavage <strong>of</strong> the glycine and glutamic acid substituents<br />

from a glutathione conjugate, followed by N-acetylationN<br />

<strong>of</strong> the<br />

resulting product<br />

٧٧


• Significance <strong>of</strong> Biotransformation Reactions<br />

in Toxicology<br />

• Biotransformation is a major part <strong>of</strong> the<br />

pathway for elimination <strong>of</strong> many xenobiotic<br />

compounds.<br />

• Biotransformation can result in either a<br />

decrease or an increase (or no change) in<br />

toxicity.<br />

• Biotransformation can result in the formation<br />

<strong>of</strong> reactive metabolites.<br />

٧٨


• Good example – metabolism <strong>of</strong> acetaminophen<br />

• Acetaminophen is ordinarily metabolized in the liver by sulfation and<br />

glucuronidation to form non-toxic conjugates<br />

• These are low capacity pathways, in that the c<strong>of</strong>actors are available able in<br />

only limited concentrations, so these are rate-limiting.<br />

• As long as the amount <strong>of</strong> acetaminophen in the liver is relatively y low,<br />

the Phase II pathways can handle the compound, and there is no<br />

toxicity.<br />

• If the concentration <strong>of</strong> acetaminophen becomes high enough to<br />

overwhelm the capacity <strong>of</strong> the Phase II pathways, an alternate<br />

metabolic pathway, involving Phase I enzymes, becomes active.<br />

• The product <strong>of</strong> the Phase I reaction is a highly reactive quinoneimine,<br />

which can form adducts with (bind covalently to) cellular<br />

macromolecules, especially proteins.<br />

• The binding <strong>of</strong> the reactive intermediate to cellular macromolecules<br />

les<br />

destroys the activity <strong>of</strong> those molecules, and can lead to compromised<br />

mised<br />

cell function and, ultimately, cell death.<br />

٧٩


• Another good example – metabolism <strong>of</strong> carbon<br />

tetrachloride<br />

• Carbon tetrachloride is metabolized by the<br />

cytochrome P-450 system in the liver by<br />

abstraction <strong>of</strong> one <strong>of</strong> the four chlorine atoms.<br />

• This results in formation <strong>of</strong> a highly reactive<br />

trichloromethane radical, which initiates a cascade<br />

<strong>of</strong> lipid peroxidation by removing a hydrogen atom<br />

from membrane phospholipids.<br />

• Damage to the cell membrane causes loss <strong>of</strong><br />

osmotic integrity, cell swelling and death.<br />

٨٠


• The activity <strong>of</strong> drug metabolizing enzymes is<br />

dependent on numerous factors<br />

• Species<br />

• Age (activity is generally lower in very young and aged<br />

animals)<br />

• Sex (activity is generally higher in males than in females)<br />

• Genetics (remember slow versus fast acetylators)<br />

• Organ (activity <strong>of</strong> many enzymes is highest in the liver)<br />

• General health status (e.g., hepatic injury decreases<br />

metabolic activity in the liver)<br />

• Diet (remember how fasting decreases the amount <strong>of</strong><br />

glutathione available for GST)<br />

• Previous exposure to other compounds<br />

٨١


• Induction – an increase in the activity <strong>of</strong> one or more<br />

enzymes as a result <strong>of</strong> previous exposure <strong>of</strong> the<br />

organism to compounds that serve as substrates for<br />

the enzyme(s)<br />

• Classic example <strong>of</strong> an inducer is phenobarbital, , which<br />

induces the activity <strong>of</strong> cytochrome P-450 enzymes<br />

• Induction may involve either increases in the synthesis <strong>of</strong><br />

enzymatic protein, or increases in activation <strong>of</strong><br />

proenzymes.<br />

• Induction may result in increases in the amount <strong>of</strong> cellular<br />

protein, hypertrophy (increases in size) <strong>of</strong> the cells, and<br />

increases in weight <strong>of</strong> the affected organs (especially liver).<br />

• One effect <strong>of</strong> induction <strong>of</strong> microsomal enzymes is an<br />

increase in the amount <strong>of</strong> smooth endoplasmic reticulum in<br />

a cell. This can be seen microscopically.<br />

٨٢


• Induction is usually temporary, and enzyme activity levels<br />

return to normal after several weeks.<br />

• Induction can result in tolerance to drugs, if the metabolism<br />

<strong>of</strong> the drugs results in a product with lower (or no)<br />

pharmacologic activity. This is why, for example, patients<br />

can develop tolerance to Phenobarbital anesthesia after<br />

repeated administration.<br />

• Induction may result in increases or decreases in toxicity,<br />

depending on whether the metabolite is more or less toxic<br />

than the parent compound. This is why, for example,<br />

alcoholics are more susceptible to acetaminophen toxicity,<br />

since alcohol induces the enzyme that is responsible for<br />

production <strong>of</strong> the reactive metabolite from acetaminophen.<br />

٨٣


• Inhibition – a decrease in the activity <strong>of</strong> one or more<br />

enzymes<br />

• Classic example <strong>of</strong> an inhibitor is SKF-525A, which<br />

inhibits microsomal enzymes<br />

• Inhibition may be either competitive or non-competitive.<br />

• Competitive inhibition<br />

• Occurs when an inhibitor binds to the same active site on the<br />

enzyme as the substrate. The higher the concentration <strong>of</strong> the<br />

inhibitor, the less likely it is that the substrate molecule will l be able<br />

to find and bind to an available enzyme molecule.<br />

• Reversible, since the binding <strong>of</strong> the inhibitor to the active site e is not<br />

covalent<br />

• Example: Omeprazole and diazepam are both metabolized by<br />

cytochrome P-450 2C19 (CYP2C19). Co-administration <strong>of</strong> these<br />

two drugs results in prolonged plasma half-life life for diazepam.<br />

٨٤


• Non-competitive inhibition<br />

• May occur when an inhibitor binds to the same active site on the enzyme<br />

as the substrate, but binds so tightly that it is effectively not t released. Thus,<br />

the binding site is permanently blocked.<br />

• May also occur when an inhibitor binds tightly (sometimes covalently) to a<br />

different site on the enzyme than the active site. This can result in<br />

conformational or affinity changes that effectively inactive the enzyme.<br />

• Non-competitive inhibition is generally not reversible. Therefore,<br />

recovery takes much longer because it requires the synthesis <strong>of</strong> new<br />

enzyme.<br />

• A special subset <strong>of</strong> non-competitive inhibition is “suicide inhibition”, , in<br />

which a compound binds to the active site <strong>of</strong> an enzyme and is<br />

metabolized, but the product then binds irreversibly to the active site. An<br />

example <strong>of</strong> this is the binding <strong>of</strong> organophosphate insecticides to the<br />

enzyme acetylcholinesterase (AchE). This results in a prolonged inhibition<br />

<strong>of</strong> AchE activity.<br />

• Inhibition may result in increases or decreases in toxicity, depending ending on<br />

whether the metabolite is more or less toxic than the parent compound.<br />

٨٥


Factors affecting metabolism<br />

1. Age<br />

The metabolizing enzymes in neonates are not fully<br />

developed, therefore those cannot efficiently<br />

metabolize drugs. Also in the elderly, enzymatic<br />

systems may not function well leading to same<br />

conclusion.<br />

2. Sex<br />

Males who are deficient in glucose -6-phosphate<br />

dehydrogenase are more prone to hemolysis when<br />

subjected to some drugs like sulfonamides<br />

٨٦


3. Pharmacogenetic factors<br />

Some individuals may be deficient in some enzymes, regardless<br />

<strong>of</strong> sex<br />

4. Pregnancy<br />

Hepatic metabolism <strong>of</strong> drugs is decreased in pregnancy<br />

5. Nutritional status/ liver dysfunction<br />

Malnutrition can cause a decreased level <strong>of</strong> some enzyme system<br />

and liver dysfunction can lead to decreased metabolism<br />

6. Bioactivation<br />

Some drugs may be transformed to more toxic metabolites<br />

7. Enzyme induction / inhibition<br />

A result <strong>of</strong> this is either an increase in the metabolism or a<br />

decrease in the drug metabolism<br />

8. Changes in the kinetic mechanism: depending on whether the<br />

concentration <strong>of</strong> drug is in the therapeutic or overdose range<br />

٨٧


4. Excretion<br />

• Excretion<br />

• The removal <strong>of</strong> materials from the body<br />

• Routes <strong>of</strong> excretion<br />

• Bile – through the liver<br />

• Urine – through the kidneys<br />

• Feces – through the intestines<br />

• Expired air – through the lungs<br />

• Sweat – through the skin<br />

• Saliva – through the mouth<br />

• Milk<br />

• Hair<br />

٨٨


Factors affecting excretion<br />

1. Age<br />

Many <strong>of</strong> the functions <strong>of</strong> the kidney are not well developed in<br />

neonates, toxic drugs will be slowly excreted. In the elderly,<br />

lower renal plasma flow will also decrease excretion<br />

2. Disease states<br />

Renal and gastrointestinal diseases can considerably slow the<br />

excretion<br />

3. Recirculation<br />

Some drugs that are excreted as their more water soluble<br />

metabolites may be back transformed to the parent<br />

compound by some bacteria<br />

4. Ion trapping<br />

Weakly acidic drugs trapped in the tubules are excreted at higher<br />

urinary pH as the ion form, thus preventing reabsorption.<br />

٨٩

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!