You are on page 1of 408

DEFINITION OF PHARMACOLOGY;

DRUGS; CLASSIFICATION AND NAMING

Dr. Yousef Al-saraireh


Associate Professor
Faculty of Medicine
1
CONTENTS

 DEFINATION AND DIVISIONS

 DRUG SOURCES

 DRUG CLASSIFICATION

 DRUG NAMES

 DOSE FORMS OF DRUGS

2
DEFINITION AND DIVISIONS
 PHARMACOLOGY : It is the science that deals with
interaction of drugs with living systems.

Drugs : These are chemical substances that shows


biological activity (treatment or sometimes diagnosis).

 Divisions of Pharmacology:

1. Pharmacodynamics :

(What the drug does to the body)

 This deals with the action of drugs on living tissues ,


namely the type or quality of action, its quantitative 3
aspect , as well as the mechanism of action .
 Adverse effects and safety of drugs on body tissues or
systems are also included

 The main organ or tissue on which the drug acts , and for
which it is used therapeutically, is called the target
organ or tissue of drug action

2. Pharmacokinetics :

(What the body does to drug)

 This includes administration and absorption of drugs,


their distribution inside body, and their elimination by
metabolism or excretion

4
OTHER TOPICS LINKED WITH PHARMACOLOGY
1. Pharmacotherapeutics: It is concerned with the proper
use of drugs in treatment of disease in man

2. Clinical Pharmacology: This includes :-


A. Drug pharmacology
B. Clinical evaluation of drugs in treating disease
in man. This is done by :
a. Clinical trials b. Surveillance studies

3.Chemotherapy : It is used to imply the use of drugs to


inhibit growth or kill either :
a. Microbes (i.e. anti-microbial agents)
b. Cancer cells ( Cyto-toxic anti-cancer drugs) 5
4. Pharmacy : It is the science and profession that is
concerned with the preparation, storage, dispensing, and
proper utilization of drug products

5. Toxicology : It is the science that deals with the


harmful effects of chemicals (including drugs) .

6
DRUG SOURCES
These may be either :-

I. Synthetic sources : common at present


- these drugs are prepared by the labs or factories of the
pharmaceutical industry. Nowadays, computers
greatly assist in discovery of new drugs

II. Semi-synthetic drugs :


- these are obtained from natural sources, but are
modified by pharmaceutical industry in order to
improve their physical or chemical properties or
pharmacological activity. 7
III. Natural sources : These are less used now . They
may be either :
1. Organic :
A. Plants : Any part of the plant (stem, leaves, flowers,
seeds, roots) may be used to extract active ingredients for
drugs; same plant may contain more than one active
principle. All of this is dealt with in PHARMACOGNOSY

Examples of drugs from plants are : alkaloids, steroids,


some vitamins, tannins, volatile oils, gums

Note :
Alkaloids are small organic molecules containing
nitrogen . e.g. atropine, morphine, caffeine, theophylline,
quinine
8
B. Animals : these may include either proteins , oils,
enzymes from exocrine glands, hormones, vaccines and
anti-sera, and some vitamins

C. Microbes : like fungi, and sometimes bacteria which are


sources of antibiotics

2. Non-Organic sources :
- metals : Platinum, Zinc
- non-metals : Sodium chloride , magnesium sulfate

9
Rational drug design:

 This implies the ability to predict the chemical structure


of drug molecule on basis of 3-dimensional structure of
its receptor, employing at present suitable computer
programs. Only few drugs in clinical use at present
were developed in this rational way.

 Most drugs were in the past developed through random


testing of chemicals , or modified molecules of known
drugs that are known to have some pharmacological
effect.

However, as more becomes known about


detailed structure of receptors, rational drug
design with the aid of computers would become 10
more feasible
DRUG CLASSIFICATION
There is no fixed rule; classification is usually done
according to their :

1. Therapeutic use : e.g. anti-hypertensive drugs ;


anti-microbial drugs ; anaesthetics; hypoglycemic drugs;
anticoagulants;

2. Type of pharmacological action :


This should be precise. e.g. local or general
anaesthetics; vasodilators; anticoagulants OR
according to molecular or cellular site of action in
target cells e.g. enzyme inhibitors, receptor blockers ,
ion channel blockers, inhibitors of transporters,
antimicobials acting on cell wall, DNA, or ribosomes 11
3. Physiological systems on which they act : Drugs
acting on cardio-vascular system; drugs acting on GIT or
CNS or respiratory system
4. Chemical nature or Source :
Common chemical groups or structures can be used to
classify drugs that have similarity in their
pharmacological profile e.g. benzodiazepines, steroids.

For drugs derived from nature, both the plant species or


genus and drug chemistry are included e.g. belladona
alkaloids from atropa belladona , digitalis glycosides
from Digitalis leaves .

12
DRUG NAMES
1. Chemical name :
 Because of its complexity , the chemical structure is not
usually used to name drugs.
 However, sometimes a shorthand name based on a simple
chemical structure is employed e.g. acetylsalisylic acid
(aspirin) , acetaminophen (parectamol)

13
2. Generic (non-proprietory ) name :
 This is a unique name that is given by official
pharmaceutical bodies;
 It is present in pharmacopeas (BP or USP) .
 It is the approved scientific name, and must be used in
scientific publications as well as in prescriptions esp. in
hospitals .
 Its use makes it easier for pharmacist to choose from
many available brands of same drug.
 Only few drugs show more than one generic name :
Noradrenaline & adrenaline in UK but are named Nor-
epinephrine and epinephrine, respectively, in USA &
WHO; salbutamol in UK while albuterol in USA

 Generic names of drugs in a classified group may


have common endings e.g. – olol for beta-
adrenoceptor blockers; -caine for local anaesthetic drugs.
These endings may give a hint about the drug 14
pharmacotherapeutic action
3. Commercial or trade or brand or proprietory name:
 This name is given by the specific pharmaceutical
company synthesizing and marketing the drug.
 Examples: Diclofenac Na (Voltaren, Inflaban, Diclogesic)
 A single drug can have many brand names (this may be
confusing) due to its manufacture and marketing by many
pharmaceutical companies.

15
DOSE FORMS OF DRUGS
 It is the physical form of drug product that is suitable for
administration to man. It contains specified dose or
amount of drug in a specified quantity or unit of the
formulation.

 Types of drug dose forms:


1. Oral
2. Inhalational
3. Parenteral
4. Topical
5. Suppository

16
1. Oral dose forms: It includes the following
A. Pill: Tablets and capsules
B. Liquid: Syrup or suspension
C. Powder
D. Herbal plants: seeds, leaves etc..
E. Pastes
2. Inhalational:
A. Aerosol
B. Inhaler
C. Vaporizer (Solutions)
3. Parenteral:
A. Intradermal (ID) B. Intramuscular (IM)
C. Intraperitoneal (IP) D. Intravenous (IV)
17
E. Subcutanous (SC) F. Intrathecal (IT)
4. Topical:
A. Cream, gel, ointment, lotion
B. Eye drops ( ophthalmic)
C. Ear drops (otic)
D. Skin patch (transdermal)

5. Suppository:
A. Vaginal
B. Rectal

18
THANKS

19
PRINCIPLES OF
PHARMACODYNAMICS

Dr. Yousef Al-saraireh


Assistant Professor
Faculty of Medicine
1
CONTENTS

 MECHANISMS OF DRUG ACTION

 TYPES OF LIGAND-RECEPTOR INTERACTIONS

 TYPES OF DRUG-RECEPTOR BONDING

 CLASSIFCATION OF RECEPTORS

2
MECHANISMS OF DRUG ACTION
 Drugs can act through:
1. Physical action:
Drug can produce a therapeutic response because of
it’s physical properties. e.g: Mannitol as diuretic
because it increase osmalerity, Radio-isotopes : emit
ionizing radiation
2. Simple chemical reaction:
Drug may act through a chemical reaction. e.g: Gastric
antacids work by neutralizing the stomach acidity
with a base, Chelating agents that bind heavy metals
in body.
3. Receptors:
A receptor is a specialized target macromolecule
mostly protein, present on the cell surface or
intracellular, that binds a drug and mediates it’s
pharmacological actions. 3
Receptors can either be enzymes,
nucleic acids or structural proteins to
which drugs may interact.
 A molecule that binds to a receptor
is called a ligand, and can be
a peptide or another small molecule
like a neurotransmitter, hormone, or
drug.
 Ligand binding changes
the conformation (three-dimensional
shape) of the receptor molecule. This
alters the shape at a different part of
the protein, changing the interaction
of the receptor molecule with
associated biochemicals, leading in
turn to a cellular response mediated
by the associated biochemical
pathway. 4
TYPES OF LIGAND-RECEPTOR INTERACTIONS

Agonist Hormone
e.g. important
Antagonist
binds β2 receptor in lung → control heart beat
therapy bronchial relaxation
in asthma
binds β2 receptor in heart muscle →
increased heart rate
5
TYPES OF LIGAND-RECEPTOR INTERACTIONS
Not every ligand that binds to a receptor also activates the
receptor. The following classes of ligands exist:
1. (Full) agonists are able to activate the receptor and
result in a maximal biological response. The
natural endogenous ligand with greatest efficacy for a
given receptor is by definition a full agonist (100%
efficacy).
2. Partial agonists do not activate receptors thoroughly,
causing responses which are partial compared to those of
full agonists (efficacy between 0 and 100%).
3. Antagonists bind to receptors but do not activate them.
This results in receptor blockage, inhibiting the binding of
agonists and inverse agonists.
4. Inverse agonists reduce the activity of receptors by 6
inhibiting their constitutive activity (negative efficacy).
TYPES OF DRUG-RECEPTOR BONDING
Drugs interact with receptors by means of chemical forces
or bonds. These are of three major types:

1. Covalent: It is very strong and in many cases not


reversible under biologic conditions. Thus, the duration of
drug action is frequently, but not necessarily, prolonged
(irreversible)

2. Electrostatic: is much more common than covalent


bonding in drug-receptor interactions. These vary from
relatively strong linkages between permanently charged
ionic molecules to weaker hydrogen bonds and very weak
induced dipole interactions such as van der Waals forces.
Electrostatic bonds are weaker than covalent bonds. 7
(reversible)
3. Hydrophobic: are usually quite weak and are probably
important in the interactions of highly lipid-soluble drugs
with the lipids of cell membranes and perhaps in the
interaction of drugs with the internal walls of receptor
"pockets.“

 Drugs which bind through weak bonds to their receptors


are generally more selective than drugs which bind
through very strong bonds.

 This is because weak bonds require a very precise fit of


the drug to its receptor if an interaction is to occur

8
DURATION OF DRUG ACTION
Termination of drug action at the receptor level results from
one of several processes:
1. The effect lasts only as long as the drug occupies the
receptor, so that dissociation of drug from the receptor
automatically terminates the effect.
2. The action may persist after the drug has dissociated,
because, for example, some coupling molecule is still
present in activated form.
3. Drugs that bind covalently to the receptor, the effect may
persist until the drug-receptor complex is destroyed and
new receptors are synthesized.
4. Many receptor-effector systems incorporate
desensitization mechanisms for preventing excessive
activation when agonist molecules continue to be present 9

for long periods


CLASSIFCATION OF RECEPTORS
This is based on the type of the transduction mechanism that
these receptors activate when stimulated by their agonists:

1. Transmembrane ligand-gated
ion channels: These receptors are
present in the walls of ion channels in
cell membranes. When activated by
their specific agonist, they open these
ion channels & lead to movement of
ions across cell membrane.

These mediate diverse functions,


including neurotransmission, cardiac
conduction, and muscle contraction.
10
Examples :
1. Nicotinic receptors for acetylcholine (Ach.) : when
stimulated, they open receptor-operated Na+ channels,
and thus increase influx of sodium ions across
membranes of neurons or NMJ(neuromuscular junction)
in skeletal muscle and therefore activation of contraction
in muscle.
2. γ-aminobutyric acid (GABA) receptors:
Benzodiazepines enhance the stimulation of the GABA
receptor by GABA, resulting in increased chloride influx
and hyperpolarization of the respective cell.

11
2. Transmembrane G protein–coupled receptors:

 When these receptors are stimulated by their specific


agonists, they will activate a regulatory G-protein in cell
membrane which in turn change activity of membrane
enzymes ( either adenyl cyclase or phospholipase C )
leading to a change in intracellular level of a second
messenger like cAMP (cyclic adenosine monophosphate),
or IP3 (inositol triphosphate), respectively, and this
would lead to cell response.

 Examples : e.g. Receptors for transmitters :


Stimulation of muscarinic receptors (M1 and M3) for
Ach will activate G and leads to increase intracellular
level of IP3 & DAG

12
13

guanosine triphosphate (GTP), guanosine diphosphate (GDP)


3. Enzyme-linked receptors:

 These membrane receptors have an extra-cellular site


that binds to specific agonists and an intra-cytoplasmic
domain which contains tyrosine and other amino acids.

 Binding to specific agonist and activation of these


receptors usually lead to phosphorylation of
tyrosine in intra-cellular domain which then
acquires kinase activity and leads to activation of
intracellular substrates or enzymes that finally leads to
cell response.

 Examples:
Receptors for insulin,
Receptors for growth factors like EGF or PDGF,
Receptors for immune cytokines 14
15
4. Intracellular receptors:

 These receptors are located in cytoplasm (e.g. steroid


receptors) or nucleus (receptors for thyroid hormones or
vitamin D3) .

 The specific agonist must cross cell membrane to inside of


cell, binds and activates these receptors, which will then
bind to DNA gene response elements in nucleus and lead to
change in gene transcription , and thus synthesis of new
proteins

16
17
THANKS

18
PHARMACODYNAMICS II

Dr. Yousef Al-saraireh


Associate Professor
Faculty of Medicine
1
RELATION BETWEEN DRUG DOSE & CLINICAL
RESPONSE
In order to make rational therapeutic decisions, the
prescriber must understand how drug-receptor
interactions underlie
1. The relations between dose and response in
patients

2. The nature and causes of variation in


pharmacologic responsiveness

3. The clinical implications of selectivity of drug


action. 2
1. The relations between dose and response in patients

These relations are exhibited as following:

A. Graded dose–response relationships ( individual):

The response is a graded effect, meaning that the response is


continuous and gradual

B. Quantal dose–response relationships (population)

describes an all-or-no response

3
A. GRADED DOSE–RESPONSE RELATIONSHIPS
➢The magnitude of the drug effect depends on the drug concentration at
the receptor site, which in turn is determined by the dose of drug
administered and by factors characteristic of the drug pharmacokinetic
profile, such as rate of absorption, distribution, and metabolism.

➢As the concentration of a drug increases, the magnitude of its


pharmacologic effect also increases.

➢Plotting the magnitude of the response against increasing doses of a drug


produces a graph, the graded dose–response curve, that has the general
shape described as a rectangular hyperbola.

➢ Two important properties of drugs, can be determined by graded dose–


response curves which are

1. Potency
4
2. Efficacy
1. POTENCY:
A measure of the amount of drug
necessary to produce an effect of a
given magnitude.

➢ The concentration of drug


producing an effect that is 50
percent of the maximum is used to
determine potency and is
commonly designated as the EC50
➢ Drug A is more potent than Drug
B, because a lesser amount of
Drug A is needed when compared
to Drug B to obtain 50-percent
effect. 5
➢ Potency is affected by:
1. Receptor concentration or density in tissue,
2. Efficiency of stimulus-response coupling mechanism in
tissue
3. Affinity: describes the strength of the interaction (binding)
between a ligand and its receptor
4. Efficacy

➢ Potent drugs are those which elicit a response by binding


to a critical number of a particular receptor type at low
concentrations (high affinity) compared with other drugs
acting on the same system and having lower affinity and
thus requiring more drug to bind to the same number of
receptors
6
2. EFFICACY
It is the ability of a drug to elicit a response when it
interacts with a receptor.
➢Efficacy is dependent on:

1. Number of drug–receptor complexes formed


2. the efficiency of the coupling of receptor
activation to cellular responses.

➢ Maximal efficacy (Emax) of a drug assumes that


all receptors are occupied by the drug, and no
increase in response will be observed if more drugs
are added

➢The height of maximal response is used to


measure maximal efficacy of agonist drug, and to
compare efficacy of similar acting agonists

7
EFFECT OF DRUG CONCENTRATION ON
RECEPTOR BINDING
The quantitative relationship between drug concentration
and receptor occupancy is expressed as follows:
Drug + Receptor ←→ Drug–receptor complex → Biologic effect

➢ As the concentration of free drug increases, the ratio of


the concentrations of bound receptors to total receptors
approaches unity

8
➢ the relationship between the percentage of bound receptors and the
drug concentration:

[D] = the concentration of free drug,


[DR] = the concentration of bound drug,
[Rt] = the total concentration of receptors and is equal to the sum of the
concentrations of unbound (free)receptors and bound receptors,
Kd = the equilibrium dissociation constant for the drug from the receptor.

➢ The value of Kd is used to determine the affinity of a drug for its


receptor.

Affinity describes the strength of the interaction (binding) between a


ligand and its receptor.
➢ The higher the Kd value, the weaker the interaction and the lower the
9

affinity.
ANTAGONISTS
➢ They are of 3 main types :
1. Chemical antagonist :
This combines with agonist and inactivates it away from
tissues or receptors
Examples:
a. Alkaline antacids neutralize HCl in stomach of peptic
ulcer patients;
b. protamine (basic) neutralizes the anti-coagulant heparin
(acidic) in plasma ;
c. chelating agents bind with higher affinity to heavy
metals (e.g. lead, mercury, arsenic ) in plasma and tissues,
preventing their tissue toxicity
10
2. Physiological antagonist :

➢This is actually an agonist on the same tissue but


produces opposite effect to that of the specific agonist; it
acts by mechanisms or receptors that are different
from those of the specific agonist .

➢Physiological antagonists quickly reverse the action of


the specific agonist on the same tissue.

Examples:

Adrenaline, given IM, is a quick acting physiologic


antagonist to histamine (that is released from mast cells or
basophils) in anaphylactic shock; it is a life-saving drug in
this condition
11
3. Pharmacological antagonist :

Pharmacological receptor antagonists have affinity for the


receptors but have no intrinsic activity or efficacy

There are three main types :

A. Competitive reversible antagonist :


This antagonist , because of similarity in its chemical
structure to agonist, competes with agonist for binding to
its specific receptors in tissue, and thus decreases or
prevents binding of agonist and its effect on tissue.

The antagonist molecules bind to the agonist


receptors with reversible ionic bonds, so that it can
be displaced competitively from receptors by
increasing the concentration or dose of agonist ,
and thus response of tissue to agonist is restored. 12
agonist (A) and antagonist (I)

➢ The DR curve of agonist is shifted to the right, and the


maximal response can be restored by increasing dose of
agonist. The more is the concentration of antagonist, the
greater is this shift of DR curve of agonist to the right.
Examples:
➢ atropine is a competitive reversible antagonist to Ach at
muscarinic receptors;
➢ Beta-blockers are competitive antagonists to adrenaline13
at beta –adrenergic receptors.
➢The affinity of competitive antagonist KI to its receptors is
calculated from :

C*/C = 1 + [I] / KI
where C* is concentration of agonist that restores response
in presence of antagonist concentration [I], and C is agonist
concentration giving this response in absence of antagonist.

B. Non-competitive antagonist :

There are two subtypes:

1. Irreversible antagonist :

Here, the antagonist molecules either bind to agonist


receptors by strong irreversible covalent bonds or
dissociate very slowly from the receptors, so that the effect
of antagonist can not be overcome fully by increasing 14
concentration of agonist.
➢ The dose response curve of agonist is shifted slightly
to the right , but the maximal height or response of
curve is depressed and can NOT be restored by
increasing the dose of agonist . This is due to decrease
in number of receptors remaining available to bind to
agonist.

➢ The more is the concentration of antagonist, the 15


more is depression of maximal response
2. Allosteric antagonism :
Here, the antagonist binds to allosteric site on receptor
that is different from the site that binds agonist molecules,
leading to change in receptor binding or affinity to agonist
with subsequent antagonism.
The dose response curve of antagonist is similar to that of
irreversible non-competitive antagonist.

Note : Allosteric enhancement : with some receptors, a


drug can bind to another allosteric site on agonist receptor
leading to increase in binding of agonist to its receptor and
thus allosteric enhancement of agonist effect . e.g. Binding of
benzodiazepines to GABA-A receptors can enhance the
depressant GABA effect on brain neurons.

16
17
C. Uncompetitive antagonism :
Here antagonist bind to a receptor different from that of
agonist, and is located more distally in the effector
mechanism so that the effect of agonist is blocked as well
as that of other agonists that produce similar effect by
acting on a different receptor. The dose-response curve is
similar to that of irreversible non-competitive antagonist.

A + RA Depolarization → Increases free


calcium
B + RU
Y
Uncompetitive antagonist
X
Competitive antagonist
18

Contraction
RECEPTOR REGULATION
1. Receptor up-regulation :
This means increase in number and/or affinity of specific
receptors ( receptor supersensitivity).
It may occur with :
A. Prolonged use of receptor antagonist : here,
there is lack of binding of receptor to agonist for long
period of time

B. Disease : e.g. hyperthyroidism : here excess


thyroxine hormone in blood stimulate proliferation of
beta-adrenergic receptors in heart which increases risk of
cardiac arrhythmia from adrenaline or use of beta-
adrenoceptor agonists .

19
B. Receptor down-regulation (Receptor tolerance):
This means a decrease in number and/or affinity of
available specific receptors due to their prolonged
occupation by agonist .

➢ It occurs with continued use (for days or weeks) of


receptor agonist , and is evident as decrease in response
to agonist .
➢ In order to restore the intensity of response, the dose of
agonist must be increased.

Tachyphylaxis : it is a rapidly developing tolerance


➢ It is not due to receptor downregulation

➢ It is associated with repeated use of large doses

of direct receptor agonist, usually at short dose intervals ,


20
OR with continuous IV infusion of agonist.
➢ It may be due to :
1. Desensitization of receptors :
Change in the receptor: where the agonist-induced
changes in receptor conformation result in receptor
phosphorylation, which diminishes the ability of the
receptor to interact with G proteins
2. Depletion of intra-cellular stores of transmitter
e.g. depletion of noradrenaline stores in vesicles inside
sympathetic nerve ending resulting from repeated use of
indirect sympathomimetic amphetamine

➢ In order to restore the response, the agonist drug


must be stopped for short time to allow for
recovery of receptors or stores of transmitter. 21
2. INDIVIDUALS USUALLY SHOW VARIATION IN
INTENSITY OF RESPONSE TO DRUGS DUE TO :

1. Variation in concentration of drug that reaches


the tissue receptors : due to pharmacokinetic factors
2. Abnormality in receptor number or function :
either genetically-determined or acquired due to up-
regulation or down-regulation
3. Post-receptor defect inside cells :
This is an important cause of response variation

4. Variation in Concentration of an Endogenous


Receptor Ligand
contributes greatly to variability in responses to
pharmacologic antagonists. 22
THANKS

23
PHARMACODYNAMICS III

Dr. Yousef Al-saraireh


Associate Professor
Faculty of Medicine
1
B. QUANTAL DOSE–RESPONSE RELATIONSHIPS

the influence of the magnitude of the dose on the proportion


of a population that responds.
 These responses are known as quantal responses,
because, for any individual, the effect either occurs or it
does not.

The desired response is either :

A. Specified in amount or magnitude :


e.g. increase in heart rate of 20 beats/min by a drug that
stimulates heart.
If the recorded response in any individual shows this
amount or more, then this is regarded as positive
response; otherwise, the response is negative 2
B. All-or-none response :
e.g. death; prevention of epileptic seizures; prevention of
cardiac arrhythmias

 For most drugs, the doses required to produce a specified


quantal effect in individuals are lognormally distributed; ie,
a frequency distribution of such responses plotted against
the log of the dose produces a gaussian normal curve of
variation

Determines minimum dose at which


each patient responded with the
desired outcome. The results have
been plotted as a histogram, and fit
with a gaussian curve. μ = mean
response; σ = standard deviation.
3
 When these responses are summated, the resulting
cumulative frequency distribution constitutes a quantal
dose-effect curve of the proportion or percentage of
individuals who exhibit the effect plotted as a function of
log dose

Example:

at 5mg/L, 2% respond, and


7mg/L 3% respond, then at
5mg/L plot 2%, at 7mg/L
plot 2+3 = 5% etc.)

4
 The quantal dose-effect curve is often characterized by:
1. median effective dose (ED50): the dose at which 50%
of individuals exhibit the specified quantal effect.

2. median toxic dose (TD50): the dose required to


produce a particular toxic effect in 50% of Animals.

3. Median lethal dose (LD50): the dose required to


produce a death in 50% of Animals.

5
SUMMATION AND POTENTIATION
Two common types of “agonistic” drug interactions are :
1. Summation: When two drugs with similar mechanisms
are given together, they typically produce additive
effects.
2. Potentiation or synergism : if the effect of two drugs
exceeds the sum of their individual effects.
Potentiation requires that the drugs act
at different receptors or effector systems.

Example of potentiation would be the


increase in beneficial effects noted in the
treatment of AIDS by combination
therapy with AZT (a nucleoside analog
that inhibits HIV reverse transcriptase)
and a protease inhibitor (protease activity
is important for viral replication). 6
PREDICTION OF DRUG SAFETY IN MAN
 This may be obtained from knowledge of Therapeutic
Index (TI) of drug.

the ratio of the dose that produces toxicity to the dose that
produces a clinically desired or effective response in a
population of individuals

TI = TD50 / ED50
where :
TD50 = the drug dose that produces a toxic effect in half the population
ED50 = the drug dose that produces a therapeutic effect in half the
population.

7
 A larger value indicates a wide margin between doses that
are effective and doses that are toxic.
 TI is determined by measuring the frequency of desired
response, and toxic response, at various doses of drug.

 In humans, the therapeutic index of a drug is determined


using drug trials and accumulated clinical experience.
These usually reveal a range of effective doses and a
different (sometimes overlapping) range of toxic doses.

 The concentration range over which a drug produces its


therapeutic effect is known as its therapeutic window

8
when the therapeutic index is low,
it is possible to have a range of
concentrations where the effective
and toxic responses overlap
Agents with a low therapeutic
index are those drugs for which
bioavailability critically alters the
therapeutic effects

When therapeutic index is large, it


is safe and common to give doses in
excess (often about ten-fold excess) of
that which is minimally required to
achieve a desired response. In this
case, bioavailability does not
critically alter the therapeutic 9
effects.
SPECIFICITY VS. SELECTIVITY
 Specificity : If a drug has one effect, and only one effect
on all biological systems it possesses the property of
specificity.
 In experience, the vast majority of drugs are selective
rather than specific. This is the case because most drugs
will act on more than one receptor site once they reach an
appropriately high concentration.

 Example: yohimbine a drug used


therapeutically as an α2-adrenoceptor blocker
but which also blocks 5-HT receptors, α1-
adrenoceptors, Na channels, monoamine
oxidase, and cholinesterase at higher
concentrations. An illustration of the
therapeutic window for selective blockade of
α2-adrenoceptors by yohimbine is shown in
Figure . 10
ADVERSE EFFECTS OF DRUGS
These are unwanted and/or harmful effects
I. Predictable or dose-related or type A effects :
A. Side effects : These occur at therapeutic doses of a
drug. They are usually minor, and decrease or disappear
on reducing dose or sometimes with continued use of drug

B. Toxic effects : These are due to large toxic doses .


They are usually serious, and need stopping drug use,
and sometimes supportive treatment to save life. They
may be :
1. Functional e.g. respiratory depression OR
2. Structural : causing tissue damage e.g.
damage to liver or kidney or heart or nerves 11
II. Unpredictable or Type B reactions :
A. Allergy : This is due to activation of immune
mechanisms by drug. Drug acts as hapten to
induce formation of antibodies by plasma cells or to
sensitize T-lymphocytes .
Usually, allergic reactions have no dose-response
relation ; they are of 4 main types :

Type 1 : Immediate type ; it is the commonest type ;


it is mediated by IgE antibodies that bind to
membrane of mast cells in tissues or basophils in
blood.
After re-exposure and binding to their specific antigen,
they trigger release of histamine and other mediators
from granules of these cells.

This causes urticaria or , in severe cases , anaphylactic


shock which is a life threatening emergency
12
Type 2 : Cyto-toxic reaction :
mediated by either IgM antibodies in plasma or IgG
antibodies that causes tissue damage by fixing
complement and activating complement cascade
e.g. hemolysis ; liver or kidney damage .

Type 3 : Immune complex mediated reaction :


Circulating immune complexes form between antigen
and IgG antibodies which become deposited in capillaries
of skin , joints , and kidney. Clinical features occur after
many days of exposure to drug e.g. serum sickness

Type 4 : Delayed cell-mediated reactions :


These are due to activation of sensitized T lymphocytes
which release their cytokines and attract macrophages to
site that also release tissue damaging cytokines
13
B. Idiosyncrasy :
abnormal drug reactions due usually to genetic
factors affecting tissue enzymes or receptors.
Examples:
a. Hemolysis by sulfonamides or the antimalarial drug
primaquin in patients with genetic deficiency of the
enzyme glucose-6-phosphate dehydrogenase (G-6-PD) in
their RBC

b. Resistance to vitamin D or to the oral anti-coagulant


warfarin

14
III. Special toxicity including
1. Genotoxicity leading to Mutagenicity :
Alkylating agents

2. Teratogenicity :
Congenital disorder : drugs taken in pregnancy

3. Carcinogenicity : may take about 2 years .


- may be related to mutagenicity but less than
is the case with teratogenicity

4. Reproductive toxicity recording pregnancy


rate, number of live or stillbirths, & postnatal growth

15
IV . Others

1. Delayed toxicity : occurs sometime after


stopping drug use e.g. idiosyncratic
aplastic anemia due to chloramphenicol

2. Chronic toxicity : occurs with prolonged use


of drug e.g. Cushing syndrome from
long-term use of steroids

3. Dependence : occurs with prolonged use


of CNS depressants e.g. alcohol ; opioids like morphine
16
Adverse effects may be caused by :

1. Over-extension of same mechanism of action on


same target tissue : e.g. sedative-hypnotics;
anticoagulants ; beta-adrenoceptor blockers
2. Effect on same receptor type but in another
tissue :
e.g. anti-muscarinic drugs ; beta-blockers
3. Effect on different receptor or by different
mechanism on target or other tissues

The following groups are more susceptible to adverse


drug reactions : foetus during pregnancy; elderly ;
patients receiving many drugs (polypharmacy); patients
with pre-existing disease ; patients with genetic enzyme
defects in liver (poor oxidizers or slow acetylators) or
tissues 17
THANKS

18
Pharmacokinetics (I)

Dr Mohammed Al-sbou
Professor of Clinical Pharmacology
Department of Pharmacology
Faculty of Medicine, Mutah University

1 1
 The goal of drug therapy is to prevent,
cure, or control diseases
 To achieve this goal, adequate drug doses
must be delivered to the target tissues so
that therapeutic yet nontoxic levels are
obtained

2
Pharmacokinetics (PK)

 Greek: Phrmaco= drug


Kinein = to move

 Definition PK: examines movement of a


drug over time through body

3 3
 Pathways of drug movement: (ADME)
 Absorption
 Distribution
 Metabolism,
 Elimination

4
Clinicians must recognize that:
 Speed of onset of drug action

 Intensity of drug’s effect

 Duration of drug action are controlled

by four fundamental pathways (ADME)

5
 Knowledge of these four processes
(ADME) influences clinician’s decision of:
 Route of administration for drug

 Amount and frequency of each dose, and

the dosing intervals

6
Pathways of PK

 Absorption:
- Is transfer of a drug from its site of
administration to bloodstream
 Distribution:

- Drug leaves bloodstream and distributes


into interstitial & intracellular fluids

7 7
 Metabolism:
- By liver, kidney, or other tissue
 Elimination:

- Removal of drug & its metabolites from


body in urine, bile, or feces

8
Absorption

 Rate & efficiency of absorption depend


on route of administration
 Routes of drug administration:
 Enteral: (by mouth) oral, subligual
 Parenteral: intravenous (IV),
intramuscular (IM), subcutaneous (Sc)
 Others: inhalation, intranasal, intrathecal,
topical, rectal
9 9
Routes of drug administration:
 IV absorption is complete
(total dose of drug reaches systemic
circulation)
 Drug delivery by other routes may result

in partial absorption and, thus, lower


bioavailability

11
 For example, oral route requires that a
drug dissolve in GI fluid and then pen-
etrate epithelial cells of intestinal mucosa,
disease states or presence of food may
affect this process

12
Absorption

A. Transport of drug from GI tract:


- Depending on chemical properties
- Drugs may be absorbed from GI tract
by either passive diffusion or active
transport

13 13
1. Passive diffusion:

- Drug move from high concentration to


one of lower concentration
- The vast majority of drugs gain access to
body by this mechanism
- Lipid-soluble drugs: readily move
across biological membranes due to their
solubility in membrane bilayers

14
 Water-soluble drugs: penetrate cell
membrane through aqueous channels or
pores
 Other drugs enter cell through specialised
transmembrane carrier proteins (large
molecules)

15 15
2. Active transport:

- Involves specific carrier proteins


- Is energy-dependent & is driven by
hydrolysis of ATP
- Moving drugs against concentration
gradient

17 17
3. Endocytosis & exocytosis

- Endocytosis involves engulfment of a


drug molecule by cell membrane
- For example, vitamin B12 is transported
across gut wall by endocytosis

18
 Exocytosis is reverse of endocytosis and
is used by cells to secrete many
substances
 Certain neurotransmitters (Norepinephrine)
are stored in membrane-vesicles in nerve
terminal & are released by exocytosis

19
20
B. Effect of pH on drug absorption:
- Most drugs are either weak acids or weak
bases
- Uncharged drugs passes through
membranes readily

21 21
Physical factors influencing absorption:

 Blood flow to absorption site


 Total surface area available for abs

 Contact time at abs surface

(in severe diarrhea, drug is not well absorbed)


 Presence of food in stomach: dilutes drug &

slow gastric emptying & delay abs in small


intestine

22 22
Bioavailability

 Is fraction of administered drug that


reaches systemic circulation in a chemically
unchanged form
 Bioavailability is determined by comparing
plasma levels of drug after particular route
of administration with plasma drug levels
after IV injection

23 23
 Bioavailability for drugs delivered IV is
100%
 When drug is given orally, only part of
the administered dose appears in
plasma, bioavailability is less 100%

24 24
 Bioavailability= AUC orally *100
AUC IV

25
 If 100 mg of drug are administered orally,
70 mg are absorbed unchanged,
bioavailability 70%

26
 Area under the curve (AUC): by
plotting plasma concentration of drug
versus time
 Bioavailability of orally administered drug
is ratio of AUC for oral admin compared
with AUC for IV injection

27
Determination of bioavailability
Factors influencing bioavailability

A- First-pass hepatic metabolism:


- When drug is absorbed across GI tract, it
enters portal circulation before entering
systemic circulation
- If drug is rapidly metabolised by liver,
amount of unchanged drug that enters
systemic circulation is decreased

29 29
 Propranolol, lidocaine undergo
significant metabolism during passage
through liver

30
First-pass metabolism occurs with orally
administered drugs
B- Solubility of drug:
- Hydrophilic drugs are poorly absorbed
because of their inability to cross lipid-rich
cell membrane

32
C- Chemical instability:
 Benzylpenicillin (Penicillin G) unstable

in pH of stomach, is given IV
 Phenoxymethylpenicillin (Penicillin V)

is used orally, it is acid-stable & is not


destroyed by gastric acid
 Insulin destroyed by enzymes in GI tract,

is given IV
33
Bioequivalence:

 Two related drugs are bioequivalent if


they show comparable bioavailability &
similar times to achieve peak blood
concentrations

34 34
Pharmacokinetics (II)

Dr Mohammed Alsbou
Professor of Clinical Pharmacology
Department of Pharmacology
Faculty of Medicine, Mutah University

1 1
Drug Distribution

 Is the process by which a drug leaves


blood stream & enters interstitium
(extracelullar fluids) and/or cells of the
tissues

2 2
Drug distribution depends on:

 Blood flow to tissue capillaries


 Capillary permeability
 Degree of binding to plasma & tissue
proteins
Drug distribution depends on:

 Blood flow to tissue capillaries:


o Blood flow to tissue capillaries vary
o blood flow to brain, liver & kidney is higher
than that to skeletal muscles & adipose tissue
 Capillary permeability is determined by:
 Drug structure: Hydrophobic drugs, which

have no net charge, readily move across


biologic membranes. They dissolve in lipid
membranes and, therefore
 hydrophilic drugs, which have a positive or

negative charge, do not readily penetrate cell


membranes, and therefore, must go through
slit junctions
 Capillary structure: (brain continuous, liver
discontinuous)
Cross-section of liver and brain capillaries
Blood-Brain Barrier (BBB)

- To enter brain, drug must pass through


endothelial cells of capillaries of CNS or activelly
transported (specific transporters)
- Lipid-soluble drugs readily penetrate into CNS
because they can dissolve in membrane of
endothelial cells
- Ionised or polar drugs fail to enter CNS
because they are unable to pass through
endothelial cells of CNS
8 8
The BBB is created by the tight apposition of endothelial cells lining
blood vessels in brain, forming a barrier between circulation and
brain parenchyma (e.g. astrocytes, microglia)
Binding of drugs to plasma protein

 Plasma albumin is major drug-binding protein


- Bound drugs are pharmacologically inactive
- Only free drug (unbound drug) can act on
target sites in tissues (active)
- As concentration of free drug decreases due to
elimination (metabolism, excretion), bond drug
dissociate from protein
- Hypoalbumunemia may alter level of free
drug
10 10
Drug Metabolism

 Drugs are eliminated by biotransformation


and/or excretion into urine or bile
 Metabolism transform lipophilic drugs
into more polar products
 Liver is major site of drug metabolism
 Other sites kidney, intestines

11 11
 Some drugs are administered as inactive
compound (pro-drug) & must be
metabolised to their active forms
Kinetics of metabolism

 First-order kinetics:
- Metabolism is catalyzed by enzymes
- At low doses, drug metabolism is first
order – rate of metabolism is directly
proportional to drug dose

13 13
Kinetics of metabolism

 Zero-order kinetics:
- At high doses, drug metabolism is zero
order (no-Linear) that is constant &
independent of drug dose (because the
enzyme is saturated by high free-drug
concentration)
Effect of drug dose on rate of
metabolism
Reactions of metabolism

 Kidney cannot eliminate lipophilic drugs


that readily cross cell membranes and
reabsorbed in distal tubules
 Lipid-soluble drugs must be metabolised
in liver using two reactions called:
 Phase I

 Phase II

16 16
Phase I

 Converts lipophilic molecules into more


polar molecule by introducing or
unmasking polar functional group such
as –OH or –NH2
 Phase I reactions are catalsyed by
cytochrome P450 system (also called
microsomal mixed function oxidase)
Cytochrome P450 system

 Designated as CYP
 Is composed of many families of heme-
containing isozymes that are located in
most cells primarily in liver & GI tract
 There are many different genes & many
different enzymes known as P450
isoforms

18 18
 Most isoforms involved in metabolism
of drugs are:
 CYP3A4= (60% of drugs)

 CYP2D6= (25% of drugs)

 CYP2C9= (15% of drugs)

 CYP2C19= (15% of drugs)


Genetic Variability

 These enzymes exhibit genetic


variability, which has implication for
individual dosing regimens, determining
responsiveness & risk of adverse drug
reactions

20 20
CYP 450 inducers

 Certain drugs phenobarbital, rifampin,


carbamazepine increase synthesis of CYP
enzymes
 Thus increase metabolism of drugs
metabolised by these CYP enzymes,
decrease plasma concentration & decrease
therapeutic effect

21 21
CYP 450 inhibitors

 Inhibition of CYP isozymes can lead to serious


adverse events
 Important CYP inhibitors are erythromycin,
ketoconalzole, omeprazole
 Omeprazole is a potent inhibitor of three of CYP
isozymes responsible for warfarin metabolism
 If two drugs are taken together, plasma
concentrations of warfarin increase, which leads
to increase risk of hemorrhage and other serious
bleeding reactions 22 22
 Natural substances such as grapefruit juice
may inhibit drug metabolism
 Grapefruit juice inhibits CYP3A4 and, thus,

drugs such as amlodipine & clarithromycin,


which are metabolized by this system, have
greater amounts in systemic circulation—
leading to higher blood levels & potential to
increase therapeutic and/or toxic effects of
drugs
Phase II

 This phase consists of conjugation reactions


 If the metabolite from Phase I metabolism is
sufficiently polar, it can be excreted by
kidneys
 However, many Phase I metabolites are too
lipophilic to be retained in kidney tubules
 A subsequent conjugation reaction with
an endogenous substrate, such as
glucuronic acid, sulfuric acid, acetic acid, or
an amino acid
 Results in polar, usually more water-
soluble compounds that are most often
therapeutically inactive
Phase II

 Glucuronidation is the most common and


important conjugation reaction
 Drugs possessing –OH, HN2, COOH group
may enter phase II directly & become
conjugated without phase I

26 26
Biotransformation of drugs
Pharmacokinetics (III)

Dr mohammed Alsbou
Professor of Clinical Pharmacology
Department of Pharmacology
Faculty of Medicine, Mutah University

1
Drug elimination

 Removal of drug from body occurs via a


number of routes
 The most important being through kidney
into the urine
 Other routes include the bile, intestine,
lung, or milk in nursing mothers

2
A. Renal elimination of a drug

1. Glomerular filtration
2. Proximal tubular secretion (active
secretion)
3. Distal tubular reabsorption (passive
reabsorption)
4. Effect of drug metabolism on
reabsorption in distal tubule
3
A. Renal elimination of a drug

1. Glomerular filtration:
 Drugs enter kidney through renal arteries

 Free drug (not bound to albumin) flows

into Bowman’s space as part of the


glomerular filtrate

4
2. Proximal tubular secretion
(active secretion):

 Drugs that were not transferred into


glomerular filtrate
 Secretion occurs in proximal tubules by
active transport systems
 Competition between drugs for these
carriers can occur within each transport
system

5
3. Distal tubular reabsorption
(passive reabsorption):

 As drug moves toward distal tubule, its


concentration increases & exceeds that of
perivascular space
 Lipid-soluble drug, uncharged drug, may
diffuse out of kindney’s lumen, back into
systemic circulation (back-diffusion)

6
Drug elimination by kidney

7
Effect of drug metabolism on reabsorption
in distal tubule

 Most drugs are lipid soluble & would diffuse


out of kidney’s lumen when drug
concentration in filtrate becomes greater
than that in perivascular space
 To minimize this reabsorption, drugs are
modified primarily in liver into more ionized
or polar substances by phase I & II
reactions
8
Effect of drug metabolism on
reabsorption in distal tubule

9
Manipulating pH of urine

 Manipulating pH of urine to increase


ionized form of drug in lumen may be used
to minimize amount of back-diffusion
 Hence, increase clearance of an
undesirable drug

10
 As a general rule, weak acids can be
eliminated by alkalinization of urine
 Whereas elimination of weak bases may
be increased by acidification of urine

11
Examples

 A patient presenting with phenobarbital


(weak acid) overdose can be given
bicarbonate, which alkalinizes urine and
keeps drug ionized,
 Thereby decreasing its reabsorption

12
Examples

 If overdose is with a weak base, such as


cocaine,
 Acidification of urine with NH4Cl leads
to increase in its clearance

13
 Plasma clearance is expressed as
volume of plasma from which a drug is
removed in a given time (mL/min)

14
 Extraction ratio:
 The drugs enter kidneys at concentration
C1 and exit kidneys at concentration C2
The extraction ratio = C2/C1

15
 Half-life (t1⁄2) of drug: is the time
required for drug concentration to change
by fifty percent

16
 Total body clearance:
 CL total or CLt, is the sum of clearances
from various organs
CL total = CL hepatic + CL renal + CL pulmonary + CL other

17
 When a patient has an abnormality that
alters half-life of a drug, adjustment in
dosage is required

18
Half-life of drug is increased by:

- Diminished renal plasma flow or hepatic


blood (cardiogenic shock, heart failure,
hemorrhage)
- Decreased extraction ratio—in renal disease
- Decreased metabolism— when another drug
inhibits its biotransformation or in hepatic
insufficiency, as with cirrhosis

19
Half-life of a drug may decrease by:

- Increased hepatic blood flow


- Increased metabolism

20
KINETICS OF CONTINUOUS
ADMINISTRATION

 PK describes time-dependent changes of


plasma drug concentration and total amount
of drug in body, following drug’s
administration by various routes:
A. IV infusion
B. Oral fixed-dose/fixed-time interval
regimens (e.g one tablet every 4 hours)

21
A. Kinetics of IV infusion
 Rate of drug exit from body increases
proportionately as plasma
concentration increases, and at every
point in time, it is proportional to plasma
concentration of drug

22
1. Steady-state drug levels in
blood:

 Following initiation of IV infusion, plasma


concentration of drug rises until rate of
drug eliminated precisely balances rate of
administration
 A steady-state is achieved in which
plasma concentration of drug remains
constant

23
 Rate of drug elimination from body = (CLt)(C)
 CLt = total body clearance

 C = plasma concentration of drug

24
2. Influence of rate of drug
infusion on steady state:

 Steady-state plasma concentration occurs


when rate of drug elimination is equal to
rate of administration

25
 At steady state,
input (rate of infusion) equals
output (rate of elimination)

26
Css = Ro/keVd = Ro/CLt
 Css = steady-state concentration
 Ro = infusion rate (mg/min)
 Ke =first-order elimination rate
 Vd = volume of distribution
 Because ke, CLt & Vd are constant for most
drugs showing first-order kinetics, Css is
directly proportional to Ro
27
 If infusion rate is doubled, plasma
concentration achieved at the steady
state is doubled

28
Effect of infusion rate on
steady-state concentration
of drug in plasma

29
3. Time required to reach
steady-state drug concentration:

 Concentration of drug rises from zero at


start of infusion to its ultimate steady-state
level (Css)

30
a. Exponential approach to
steady state:

 50% of steady state concentration of drug


is achieved in the (First t1/2)
 Waiting another half-life (Second t1/2)
allows drug concentration to approach 75%
of Css
 90% of steady state concentration of drug
is achieved in the Third t1/2
 A drug will reach steady-state in about
Four half-lives 31
Rate of attainment of steady-state
concentration of drug in plasma

32
b. Rate of drug decline when
infusion is stopped:

 When infusion is stopped, plasma


concentration of a drug declines (washes
out) to zero with same time course
observed in approaching steady state

33
c. Loading dose:

 A delay in achieving desired plasma levels


of drug may be clinically unacceptable
 Therefore, a “loading dose” of drug can
be injected as a single dose to achieve
desired plasma level rapidly
 Followed by an infusion to maintain steady
state (maintenance dose)

34
B. Kinetics of fixed-dose/fixed-
time-interval regimens

 Administration of a drug by fixed doses


(e.g. one tablet every 4 hrs) rather than by
continuous infusion is more convenient
 However, fixed doses, given at fixed-time
intervals, result in time-dependent
fluctuations in circulating level of drug

35
1. Single IV injection:
 Circulating level of drug

decreases exponentially
with time

36
2. Multiple IV injections:
 When a drug is given repeatedly at regular

intervals, the plasma concentration increases


until a steady state is reached

37
3. Orally administered drugs:

 Plasma concentration
of orally administered
drugs is influenced by
both the rate of
absorption and the
rate of drug
elimination

38
Plasma Site of
Dosage Effects
Concen. Action

Pharmacokinetics Pharmacodynamics
12
Plasma Concentration

10 TOXIC RANGE

6 THERAPEUTIC RANGE

2 SUB-THERAPEUTIC

0
0 1 2 3 4 5 6 7 8 9
Dose
DISPOSITION OF DRUGS

The disposition of chemicals entering the body (from C.D. Klaassen, Casarett and Doull’s Toxicology, 5th ed., New York:
McGraw-Hill, 1996).
LOCUS OF ACTION TISSUE
“RECEPTORS” RESERVOIRS
Bound Free Free Bound

ABSORPTION Free Drug EXCRETION

SYSTEMIC
Bound Drug CIRCULATION

BIOTRANSFORMATION
Plasma concentration vs. time profile of a
single dose of a drug ingested orally

14
Plasma Concentration

12
10
8
6
4
2
0
0 5 10 15 20
TIME (hours)
12
Plasma Concentration

10 TOXIC RANGE

6 THERAPEUTIC RANGE

2 SUB-THERAPEUTIC

0
0 1 2 3 4 5 6 7 8 9
Dose
LOCUS OF ACTION TISSUE
“RECEPTORS” RESERVOIRS
Bound Free Free Bound

ABSORPTION Free Drug EXCRETION

SYSTEMIC
Bound Drug CIRCULATION

BIOTRANSFORMATION
Bioavailability

Definition: the fraction of the administered


dose reaching the systemic circulation
for i.v.: 100%
for non i.v.: ranges from 0 to 100%

e.g. lidocaine bioavailability 35% due to


destruction in gastric acid and liver metabolism

First Pass Effect


Bioavailability
Destroyed Not Destroyed Destroyed
in gut absorbed by gut wall by liver

to
Dose systemic
circulation
PRINCIPLE

For drugs taken by routes other than the


i.v. route, the extent of absorption and the
bioavailability must be understood in
order to determine what dose will induce
the desired therapeutic effect. It will also
explain why the same dose may cause a
therapeutic effect by one route but a toxic
or no effect by another.
PRINCIPLE

Drugs appear to distribute in the body as if it


were a single compartment. The magnitude of
the drug’s distribution is given by the apparent
volume of distribution (Vd).

Vd = Amount of drug in body ÷ Concentration in Plasma

(Apparent) Volume of Distribution:


Volume into which a drug appears to distribute with
a concentration equal to its plasma concentration
Examples of apparent Vd’s for
some drugs
Drug L/Kg L/70 kg
Sulfisoxazole 0.16 11.2
Phenytoin 0.63 44.1
Phenobarbital 0.55 38.5
Diazepam 2.4 168
Digoxin 7 490
Elimination
of drugs from the body

M KIDNEY LIVER
A
J filtration metabolism
O secretion secretion
R (reabsorption)

M LUNGS OTHERS
I
N exhalation mother's milk
O sweat, saliva etc.
R
Elimination by the Kidney
• Excretion - major
1) glomerular filtration
glomerular structure, size constraints,
protein binding

2) tubular reabsorption/secretion
- acidification/alkalinization,
- active transport, competitive/saturable,
organic acids/bases
- protein binding

• Metabolism - minor
Elimination by the Liver
• Metabolism - major
1) Phase I and II reactions

2) Function: change a lipid soluble to more


water soluble molecule to excrete in kidney

3) Possibility of active metabolites with


same or different properties as parent
molecule

• Biliary Secretion – active transport, 4 categories


The enterohepatic shunt
Drug Liver

Bile Bile formation


duct
Biotransformation;
Hydrolysis by glucuronide
beta glucuronidase
gall bladder produced

Portal circulation

Gut
12
Plasma Concentration

10 TOXIC RANGE

6 THERAPEUTIC RANGE

2 SUB-THERAPEUTIC

0
0 1 2 3 4 5 6 7 8 9
Dose
Influence of Variations in Relative Rates of
Absorption and Elimination on Plasma
Concentration of an Orally Administered Drug

14 Ka/Ke=10
Plasma concentration

12 Ka/Ke=1

10
Ka/Ke=0.1
8 Ka/Ke=0.01
6
4
2
0
0 5 10 15 20

TIME (hours)
Elimination

• Zero order: constant rate of elimination


irrespective of plasma concentration.

• First order: rate of elimination proportional to


plasma concentration. Constant Fraction of drug
eliminated per unit time.

Rate of elimination ∝ Amount


Rate of elimination = K x Amount
Zero Order Elimination
Pharmacokinetics of Ethanol
• Ethanol is distributed in total body water.
• Mild intoxication at 1 mg/ml in plasma.
• How much should be ingested to reach it?
Answer: 42 g or 56 ml of pure ethanol (VdxC)
Or 120 ml of a strong alcoholic drink like whiskey
• Ethanol has a constant elimination rate = 10 ml/h
• To maintain mild intoxication, at what rate must
ethanol be taken now?
at 10 ml/h of pure ethanol, or 20 ml/h of drink.
Rarely Done DRUNKENNES Coma Death
First Order Elimination
dA/dt ∝A DA/dt = – k•A

14 DC/dt = – k•C
Plasma concentration

12 Ct = C0 . e – Kel •t
10 lnCt = lnC0 – Kel • t
8 logCt = logC0 – Kel •t
6 2.3
4 y = b – a.x
2
0
0 5 10 15 20

TIME (hours)
Plasma Concentration 10000

First Order Elimination


1000

100

10

1
0 1 2 3 4 5 6
Time
logCt = logC0 - Kel . t
2.303
Plasma Concentration Profile
after a Single I.V. Injection

10000
Distribution and Elimination
Plasma Concentration

Elimination only
1000
C0
100

10

Distribution equilibrium
1
0 1 2 3 4 5 6

Time
lnCt = lnCo – Kel.t

Vd = Dose/C0

When t = 0, C = C0, i.e., the concentration at


time zero when distribution is complete and
elimination has not started yet. Use this value
and the dose to calculate Vd.
lnCt = lnC0 – Kel.t

t1/2 = 0.693/Kel

When Ct = ½ C0, then Kel.t = 0.693. This is the


time for the plasma concentration to reach half
the original, i.e., the half-life of elimination.
PRINCIPLE

Elimination of drugs from the


body usually follows first order
kinetics with a characteristic
half-life (t1/2) and fractional rate
constant (Kel).
First Order Elimination
• Clearance: volume of plasma cleared of drug
per unit time.
Clearance = Rate of elimination ÷ plasma conc.
• Half-life of elimination: time for plasma conc.
to decrease by half.
Useful in estimating:
- time to reach steady state concentration.
- time for plasma concentration to fall after
dosing is stopped.
BLOOD OUT
CA CV

I
N

Blood Flow = Q

ELIMINATED
Rate of Elimination = QCA – QCV = Q(CA-CV)
SIMILARLY FOR
Liver Clearance = Q(CA-CV)/CA = Q x EF OTHER ORGANS

Renal Clearance = Ux•V/Px

Total Body Clearance = CLliver + CLkidney + CLlungs + CLx


Rate of elimination = Kel x Amount in body
Rate of elimination = CL x Plasma Concentration

Therefore,
Kel x Amount = CL x Concentration

Kel = CL/Vd

0.693/t1/2 = CL/Vd

t1/2 = 0.693 x Vd/CL


PRINCIPLE
The half-life of elimination of a drug (and
its residence in the body) depends on its
clearance and its volume of distribution

t1/2 is proportional to Vd
t1/2 is inversely proportional to CL

t1/2 = 0.693 x Vd/CL


Multiple dosing
• On continuous steady administration of a drug,
plasma concentration will rise fast at first then more
slowly and reach a plateau, where:
rate of administration = rate of elimination
ie. steady state is reached.
• Therefore, at steady state:
Dose (Rate of Administration) = clearance x plasma conc.
Or
If you aim at a target plasma level and you know the
clearance, you can calculate the dose required.
Constant Rate of Administration (i.v.)
Single dose –
Loading dose
7

6
Plasma Concentration

Therapeutic
5 level

3
Repeated doses –
2
Maintenance dose
1

0
0 5 10 15 20 25 30

Time
The time to reach steady
state is ~4 t1/2’s

Concentration due to
repeated doses

Concentration due to a single dose


Pharmacokinetic parameters
Get equation of regression line; from it get Kel, C0 , and AUC

• Volume of distribution Vd = DOSE / C0

• Plasma clearance Cl = Kel .Vd

• plasma half-life t1/2 = 0.693 / Kel

• Bioavailability (AUC)x / (AUC)iv


dC/dt = CL x C

dC = CL x C x dt

But C x dt = small area under the curve. For total


amount eliminated (which is the total given, or the
dose, if i.v.), add all the small areas = AUC.
Dose = CL x AUC and Dose x F = CL x AUC
Bioavailability = (AUC)o
70 (AUC)iv

60
Plasma concentration

50 i.v. route

40
30 oral route

20

10

0
0 2 4 6 8 10
Time (hours)
Variability in Pharmacokinetics

60
Concentration (mg/L)

50
Plasma Drug

40

30

20

10

0
0 5 10 15
Daily Dose (mg/kg)
PRINCIPLE

The absorption, distribution and


elimination of a drug are qualitatively
similar in all individuals. However, for
several reasons, the quantitative aspects
may differ considerably. Each person
must be considered individually and
doses adjusted accordingly.
RATIONAL USE OF DRUGS
AND MEDICATION ERRORS

Dr. Yousef Al-saraireh


Assistant Professor
Chairman of pharmacology department
1
Faculty of Medicine
CONTENTS

 RATIONAL USE OF DRUGS

 MEDICATION ERRORS

2
RATIONAL USE OF DRUGS
Rational use of drugs: "patients receive medications
appropriate to their clinical needs, in doses that meet
their own individual requirements, for an adequate
period of time, and at the lowest cost to them and their
community".

3
MEASURES TO ENSURE RATIONAL USE
OF DRUGS
The WHO advice several measures to ensure
rational use of drugs that include development of:
 National committee on drug use

 National list of essential drugs

 Use of clinical guidelines: by physicians as in


treating hypertension & asthma
 Some regulations and measures

4
IRRATIONAL USE OF DRUGS
Include:
 Poly-pharmacy (use of too many drugs)

 Poor compliance (non-adherence to instructions of


therapy)
 Misuse or inappropriate use of antimicrobials

 Over-use of injections

 Failure to prescribe in accordance with clinical


guidelines
 Inappropriate self-medication

5
SELECTION OF DRUGS
Choice of effective drugs should be based on:
1.Efficacy

2. Cost: affordable by patient and community

3. Chosen from Essential Drugs: These are


effective drugs that are commonly used in
community, and must always be available

6
MEDICATION ERRORS

Definition:
any preventable event that may cause or lead to
inappropriate medication use or patient harm while the
medication is in the control of the health care professional,
patient, or consumer ...
 Such events are related to professional practice, health
care products, procedures, and systems,
 including prescribing; order communication; product
labeling, packaging, and nomenclature; compounding;
dispensing; distribution; administration; education;
monitoring; and use.
7
What kinds of errors are most common?

the most common error involving medications was related to:


1. Administration of an improper dose of medicine,
accounting for 41 % of fatal medication errors.

2. Giving the wrong drug and using the wrong route of


administration each accounted for 16% of the errors.

Almost half of the fatal medication errors occurred in people


over the age of 60.

Older people may be at greatest risk for medication errors


because they often take multiple prescription medications.

8
Causes of medication errors:
1. Poor communication between health care providers
2. Poor communication between providers and their patients
3. Sound-alike medication names and medical abbreviations
4. Errors when prescribing, transcribing, dispensing, and
administering medications
5. Errors related to patient monitoring of the effects of
medications
6. Potential or actual confusion regarding look-alike drug or
vaccine names, and packaging similarities
7. Misuse or malfunction of medication-related tools (e.g.,
syringes, needles), equipment (e.g., tubing, infusion pumps),
and technology (e.g.,
barcode scanning).
9
Examples

1. A physician ordered a 260-milligram preparation of


Taxol for a patient, but the pharmacist prepared 260
milligrams of Taxotere instead

2. One patient died because 20 units of insulin was


abbreviated as "20 U" but the"U" was mistaken for a
"zero." As a result, a dose of 200 units of insulin was
accidentally injected.

3. A patient developed a fatal hemorrhage when given


another patient's prescription for the blood thinner
warfarin.

10
Medication errors may stem from:
1. poor communication,

2. misinterpreted handwriting,

3. drug name confusion,

4.lack of employee knowledge, and

5.lack of patient understanding about a


drug's directions.
11
Poor handwriting

12
Poor handwriting

13
DRUG NAME CONFUSION

14
DRUG NAME CONFUSION

15
MEDICATION ERROR PREVENTION

1. Patient communication
2. Intraprofessional communication
3. Education and training
4. Reporting
5. Electronic prescribing

16
THANKS

17
DRUGS USED DURING PREGNANCY
& LACTATION
PREGNANCY PHYSIOLOGY AND ITS EFFECTS ON PHARMACOKINETICS

Absorption
1. Gastrointestinal motility is decreased but there appears to be no major
affect in drug absorption except that reduced gastric emptying delays the
appearance in the plasma of orally administered drugs, especially during
labor.
2. Absorption from an intramuscular site is likely to be efficient because tissue
perfusion is increased due to vasodilatation.
DISTRIBUTION:
1. Total body water increases by up to 8 Litres, creating a larger space
within which water soluble drugs may distribute.
2. As a result of haemodilution, plasma albumin (normal 33-55 g/1)
declines by some 10 g/1. Thus there is scope for increased free
concentration of drugs that bind to albumin.
3. Unbound drug, is free to distribute, metabolized and excreted; e.g. the
free (and pharmacologically active) concentration of phenytoin is
unaltered, although the total plasma concentration is reduced.
METABOLISM
• Hepatic metabolism increases, but not the blood flow to liver.

• So, increased clearance of drugs such as phenytoin and theophylline

(elimination rate depends on liver enzyme activity)

• Drugs that are so rapidly metabolized that their elimination rate

depends on their delivery to the liver, i.e. on hepatic blood flow, have

unaltered clearance, e.g. pethidine.


ELIMINATION:
• Renal plasma flow almost doubles

• So there is rapid loss of drugs that are excreted by kidney

• e.g. amoxycillin, dose of which should be doubled for systemic infections

(but not for urinary tract infections as penicillins are highly concentrated

in urine).
PLACENTAL TRANSFER OF DRUGS
1. The placenta is not a perfect barrier to drugs and chemicals
administered to mother.
2. Thalidomide tragedy, showed that placenta was capable of
transferring drugs ingested by mother to fetus, with potential for
great harm.
3. On other hand, placental transfer of drugs administered to mother
has been used to treat fetal arrhythmias, congestive heart failure, &
other conditions.
FACTORS AFFECTING PLACENTAL DRUG TRANSFER &
FETAL TISSUE
(1) Physicochemical properties of drug

(2) Rate at which drug crosses placenta & amount of drug reaching the fetus

(3) Duration of exposure to drug

(4) Distribution characteristics in different fetal tissues

(5) Stage of placental & fetal development at time of exposure to the drug

(6) Effects of drugs used in combination


A teratogen is a chemical substance that can
induce a malformation during development.
TERATOGENESIS
PRINCIPLES OF TERATOLOGY
 Teratogens act with specificity. A teratogen produces a specific abnormality or
constellation of abnormalities. Eg. thalidomide produces phocomelia, and valproic
acid produces neural tube defects.

 Teratogens demonstrate a dose-effect relationship.

 Teratogens must reach the developing conceptus in sufficient amounts to cause


their effects.

 The effect that a teratogenic agent has on a developing fetus depends upon the
stage during development when the fetus is exposed.
MECHANISMS OF TERATOGENESIS

 Genetic interference, gene mutation, chromosomal breakage,


interference with cellular function, enzyme inhibition, and altered
membrane characteristics.

 The response of the developing embryo to these insults is failure of


cell–cell interaction crucial for development, interference with cell
migration, or mechanical cellular disruption.
EXAMPLES
PRESCRIBING IN PREGNANCY
•minimize prescribing;
•use ‘tried and tested’ drugs whenever possible in preference to new agents;
• use the smallest effective dose;
• remember that the fetus is most sensitive in the first trimester;
• consider pregnancy in all women of childbearing potential;
• discuss the potential risks of taking or withholding therapy with the patient;
• seek guidance on the use of drugs in pregnancy in the British National
Formulary, Drug Information Services, National Teratology Information
Service (NTIS);
• warn the patient about the risks of smoking, alcohol, over-the-counter drugs
and drugs of abuse.
DRUG USE DURING LACTATION

 Most drugs administered to lactating women are detectable in breast milk.


Fortunately, the concentration of drugs achieved in breast milk is usually low.
 Infant would receive in a day is substantially less than what would be considered a
“therapeutic dose.”
 If the nursing mother must take medications and the drug is a relatively safe one,
she should optimally take it 30–60 minutes after nursing and 3–4 hours before the
next feeding.
 Caution: Sedative-Hypnotics, Lithium Tetracyclines
THANK YOU
Pharmacovigilance & Adverse
Drug Reactions

Dr Mohammed Al-Sbou (MD, MSc, PhD)


Professor of Clinical Pharmacology
Faculty of Medicine, Mutah University

1
‘First of all be sure you
do no harm’

Hippocrates (460-370 BC)

2
Pharmacovigilance (PV)
 The root of
pharmacovigilance:
Pharmaco (Greek)= Drug
Vigilance (Latin)= to keep
awake or alert

3
Pharmacovigilance (PV)
 PV is concerned
with detection,
assessment &
prevention of
adverse reactions
to drugs (ADRs) or
any drug-related
problems

4
Drug-Related Problems
 Lack of efficacy
 Medication errors
 Drug misuse and abuse
 Overdose
 Quality issues:
 Manufacturing defects

 Contamination

 Counterfeit products

5
Why Pharmacovigilance?

 Because information collected during


pre-marketing phase are incomplete
with regard to possible ADR
 Tests in animals are insufficiently
predictive of human safety
Why Pharmacovigilance?
 In clinical trials:
- Patients are limited in number
- Conditions of use differ from those in
clinical practice
- Duration of trials is limited

7
Why Pharmacovigilance?
 Information about rare adverse reactions,
chronic toxicity, use in special groups
(children, elderly or pregnant women) or
drug interactions is often incomplete or not
available

Post-marketing surveillance by
companies is therefore essential
9
Definition of ADR
 An ADR is defined according to definition
of WHO “any response to a drug which is
noxious, unintended & that occurs at
doses used in man for prophylaxis,
diagnosis, or therapy of diseases’’

10
Epidemiology of ADRs
 ADRs represent a significant cause of
morbidity & mortality
 Many ADRs are mild, sometimes serious
& can cause death
 U.S, ADRs caused 100 000 deaths per
year, 4th & 6th leading cause of death
 About 50% of ADRs are preventable

11
Importance of ADRs
 Prolong length of stay in hospitals
 Increase costs of patient care
(£600 million NHS in UK)
 Commonest cause of drug withdrawal
from market (recall):
 ARBs (Valsartan, Losartan, Irbesartan) 2019
 Reductil (Sibutramine) 2010
 Valdecoxib (Bextra) 2005
 Rofecoxib (Vioxx) 2004
12
Classification of ADRs
 Classification of Rawlins & Thompson
Type A reactions
- Augmentation of known pharmacological effect of drug
- Predictable
- Dose related
- e.g. warfarin causing bleeding

Type B reactions
- Bizarre (idiosyncratic)
- Not dose dependent
- Unpredictable
- e.g. carbamazepine-induced skin rash
13
Warfarin-induced calf haematoma

14
Carbamazepine-induced Stevens
Johnson Syndrome (SJS)

15
 ADRs according frequency are divided into
very common, common, rare, very rare
 ADRs divided according to severity into
mild, moderate, severe

16
ADRs is considered serious if:
1. Causes death of patient
2. Life-threatening
3. Prolong inpatient hospitalisation
4. Causes significant or persist disability
5. Congenital abnormality

17
Risk Factors Predisposing to ADRs

 Age
 Long duration of treatment
 Polypharmacy
 Liver, kidney diseases

18
Causes of ADRs
1. Patient
2. Drug
3. Prescriber
4. Environmental factors

19
Causes of ADRs
1. The patient:
- Age (over 60)
- Genetic factors (e.g. polymorphisms
in CYP450)
- Previous history of ADR
- Hepatic or renal diseases

20
Causes of ADRs
2. The drug
- Narrow therapeutic index, e.g. warfarin,
digoxin
- Antimicrobials have a tendency to cause
allergy & may lead to type B reactions
- Ingredients of a formulation, e.g.
colouring, flavouring

21
Causes of ADRs
3. The prescriber:
- A drug is used for an inappropriately
long time
- At a critical phase in pregnancy
- Given with other drugs (drug-drug
interactions)

4. Environmental factors:
- Diet, smoking, alcohol

22
Drugs Most Commonly Causing ADRs

 Warfarin  Anticancer drugs


 Diuretics  Immunomodulators
 Digoxin  Analgesics
 Antibacterials  Biological &
 Steroids biosimilars
 Antihypertensives

23
Burden of Adverse Drug Reactions

Admission

In patients

A&E

Primary
care

24
Burden of Adverse Drug Reactions

Admission
(Inter J of Cl Phar & Ther,1998, 36(9):478-482)

In patients

(J Med J, 2010; 44(4);442-446)

25
26
 6.5% (n=1224) of admissions are due to ADRs
 Seven 800-bed hospitals are occupied by ADR
patients
 Death in 0.15% - equivalent to 5700 deaths per
year
 Cost NHS £600 million per annum

27
 16of 200 patients (8%) suffered from one
or more ADRs
 50% of ADRs were avoidable

 Onepatient died during admission and his


death was contributed to an ADR

28
29
Number of ADR Reports / Drug Class
Number of ADR Reports / System Organ Class
Why report suspected ADRs?
 Documentation of ADRs in patients’
records is often poor
 Physicians fear that reporting of ADR may
put them at risk
 Under-reporting is common phenomenon

32
Methods of Reporting ADRs
- Spontaneous reporting:
‘Yellow Card system’

33
Reporting Methods

1- Spontaneous reporting: (Voluntary)


- Doctors, nurses & pharmacists are supplied
with forms to record suspected ADRs
- Regional PV centers at hospitals
- Reporting ADRs to National
Pharmacovigilance Centre
- In UK & jordan this is called ‘Yellow Card
system’
34
Online ADRs reporting form

35
36
37
International Collaboration
 WHO International Drug Monitoring
programme, 86 member nations have
systems to record & report ADRs
 Member countries send their report to
Uppsala Monitoring Centre (Sweden)
where they are entered into WHO Database

38
- WHO database (vigibase) include 15
million case reports

39
 MedWatch is FDA
reporting system
in U.S. for adverse
effects of drugs

40
Jordan Food & Drug
Administration (JFDA)

Jordan Pharmacovigilance Centre

41
Pharmacovigilance Center for South
Jordan/ Alkarak Governmental Hospital

42
43
44
Pharmacogenetics
Dr. Mohammed Al-Sbou
Professor of Clinical Pharmacology
Faculty of Medicine-Mutah Uni

1
 The human genome project has led to an
explosion of genetic information that is freely
available to identify polymorphisms that may
determine drug response

2
 Advances in molecular genetics and
genotyping technologies during the last two
decades have led to identification of many
polymorphisms in phase I and phase II drug
metabolising enzymes, drug targets, and in
drug transporters

3
Individual Variation in Response to Drugs
 How individuals in a population are expected
to respond to a fixed dose of drug?
 Inter-individual variability:

- Some show less than usual response


- Most show usual response
- Others show more than usual response

4
Factors Determine Response to Drugs

Environmental
(Age, sex, race, concomitant diseases, diet,
smoking, alcohol)
 Genetic (polymorphisms drug metabolising
enzymes, receptors, drug targets)

6
Pharmacogenetics/Pharmacogenomics
 Pharmacogenetics: is study of variation in
drug response due to heredity & is used in
relation to genes determining drug
metabolism
 Pharmacogenomics is a more general term; it
refers to research area that comprises all genes
in the human genome that may determine
drug response

7
Benefits of
Pharmacogenetics/Pharmacogenomics
 The concept “The right medicine to the right
patient” is the basis of pharmacogenetics
(personalised or individualised medicine)
 Ultimate goals are to improve clinical
therapeutic outcome by:
- Increasing drug efficacy
- Increasing safety of drugs e.g. reducing
incidence of ADRs

8
Personalised or Individualised Medicine

9
Pharmacogenomic approach to personalized medicine.
Drug therapy is chosen for each patient based on their
particular genetic profile

10
Polymorphisms can occur in any gene that
encode:
- Drug metabolising enzymes
- Drug transporters
- Drug targets and receptors

11
Genetic polymorphisms of drug
metabolising enzyme genes
 The majority of phase I and phase II drug
metabolising enzymes are polymorphic
 The cytochrome P450 (CYP) enzymes are the
most important group of phase I enzymes
 Polymorphisms in cytochrome P450 genes can
cause enzyme products with abolished or
reduced or increased enzyme activity

12
Cytochrome P450 enzymes
 All genes that encode for families 1-3 are
polymorphic & their capacity to metabolise
drugs depends on the functional importance
and frequency of variant alleles

13
CYP450 Enzymes
 CYP2D6
 CYP2C9
 CYP2C19

14
CYP2D6
 CYP2D6 contributes to metabolism of large of
medications about 25% of all drugs, including:
 Antidepressants (TCAs, SSRIs)

 Antiarrythmics

 Analgesics

15
CYP2D6 Phenotypes
- Poor metabolisers( PM): lack functional enzyme
- Intermediate metabolisers (IM): carry two
alleles that cause reduce activity
- Extensive metabolisers ( EM): have two normal
alleles
- Ultra-rapid metabolisers (UM): multiple gene
copies

16
 Poor metabolisers can experience adverse
effects when treated with standard dose
 Ultra-rapid metabolisers require high doses
of drugs

17
Depression
- Tricyclic antidepressants are metabolised by
CYP2D6
 Disposition of nortriptyline is related to
number of active CYP2D6 alleles and
 Dose required to obtain same plasma drug
concentrations varies between subjects with
different CYP2D6 phenotypes

20
 Ultra-rapid metabolisers needed a 10-fold
larger dose of nortriptyline than poor
metabolisers to achieve the same plasma
concentration
 Ultra-rapid metabolisers require 500 mg of
doses compared to 50 mg in poor metabolisers

21
 Genetic polymorphisms of CYP2D6 gene may
be associated with ADRs and clinical response
to antidepressants
 30% of patients with ADRs to antidepressants
were PMs
 High incidence of UMs among non-responders
(20%)

22
CYP2C9
 CYP2C9 metabolises a wide range of drugs
 Including drugs with narrow therapeutic indices
such as:
 Warfarin

 Phenytoin

 Non-steroidal anti-inflammatory drugs


(NSAIDs) including ibuprofen, diclofenac
and celecoxib

23
Warfarin and Bleeding
 Warfarin is one of the most widely prescribed
oral anticoagulant drugs
 It is used for:
 Prophylaxis and treatment of venous
thromboemolism
 Treatment of deep vein thrombosis (DVT)

 Atrial fibrillation (AF)

 In patients with prosthetic heart valves

24
Warfarin and Bleeding
 The main complication of warfarin therapy is
haemorrhage
 Genetic polymorphisms in CYP2C9 gives rise to
variants with altered enzymes activity
 Two allelic variants CYP2C9*2 (Arg144Cys) and
CYP2C9*3 (Ile359Leu) show 12% and 5% of
enzyme activity of the wild type CYP2C9*1 allele,
respectively, and are associated with decreased
warfarin dose requirements & increased risk
of bleeding
25
Peptic Ulcer
- Proton pump inhibitors (PPIs) are used for treatment
of gastric acid related diseases such as peptic ulcers,
gastro-esophageal reflux disease (GERD) & in
combination with antibiotics (amoxicillin &
clarithromycin) for eradication of Helicobacter
pylori (Hp)
- CYP2C19 metabolises several PPIs including
omeprazole and lanzoprazole
- Plasma concentrations of omeprazole, depend on
patient’s CYP2C19 phenotype

26
AmpliChip CYP450 Array
 The AmpliChip CYP450
Test provides comprehensive
detection of gene variations
including deletions and
duplications for the
CYP2D6 and CYP2C19
genes

27
Genetic Polymorphisms of Drug
Metabolising Enzyme Genes

 With respect to phase II enzymes, the most


important polymorphisms occur in N -
acetyltransferase-2 (NAT-2) and thiopurine
methyltransferase (TPMT)
 NAT-2 is involved in the metabolism of
isoniazid and sulphamethoxazole

28
Acetylation
- Most individuals are either rapid or slow
acetylators, but proportion varies between races
- The percentage of slow acetylators:
 90% in North African

 50% in Caucasian

 10% in Asian populations

29
Thiopurine S-methyltransferase (TPMT)

 TPMT catalyzes methylation of thiopurine


drugs such as 6-mercaptopurine & azathioprine
 These drugs are commonly used in treatment of
acute lymphoblastic leukaemia (ALL),
autoimmune diseases, inflammatory bowel
diseases, in organ & tissue transplantation
 Clinical testing for TPMT genetic polymorphisms
is available

30
 It has been shown that:
- 90% of population exhibit high TPMT activity
- 10% show intermediate activity
- 0.3% have low or absent enzyme activity

31
Genetic Polymorphisms in Drug Transporters

 Transporters are membrane proteins that play


crucial role in absorption, distribution &
elimination of drugs
 Genetic polymorphisms can occur in transport
proteins & may contribute to inter-individual
variation in drug response
 MDR1 (multi-drug resistant) P-glycoprotein-
Digoxin
 Serotonin transporter-antidepressant response

32
Genetic Polymorphisms in Drug targets
and Receptors
 Drug target genes including those coding for
receptors, ion channels and specific enzymes are
subject to genetic polymorphisms
 B2-adrenergic receptor: B2 agonist (salbutamol)
 Angiotensin converting enzyme (ACE):
ACE inhibitors (lisinopril)
 Vitamin K epoxide reductase complex
(VKORC): Warfarin

33
Practical Points
 Genetic is an important factor responsible for
failure to therapy & occurrence of adverse drug
reactions
 The goal of PGx is to maximize efficacy &
minimize toxicity, based on individual’s genetic
composition
 Individual variation in response to drug (some may
benefit, other fail to respond to treatment,
others may develop adverse effects)
34
Drug Therapy In Pediatric & Geriatric
Age Groups
Objectives

a. Discuss the principles of prescribing in pediatric and geriatric age groups.

b. Discuss the pharmacokinetic and pharmacodynamics differences in pediatric, geriatric and adult age

groups.

c. Describe how the efficacies of drugs vary according to age.

d. Describe different pediatric dosage forms & compliance in children.

e. Discuss important adverse drug reactions occurring in geriatric & pediatric age groups.
What is different from normal adult prescribing?

Children cannot be regarded as miniature adults in

terms of drug response, due to differences in body

constitution, drug absorption and elimination, and

sensitivity to adverse reactions.


Pediatric Group – Pharmacokinetics
Absorption:

o Gastro-intestinal absorption is slower in infancy, but absorption from intra-


muscular injection is faster.

o Infant skin is thin and percutaneous absorption can cause systemic toxicity

Distribution:

o Lower volume of distribution of fat-soluble drugs (e.g. diazepam) in infants.

o Plasma protein binding of drugs is reduced in neonates.

o Blood–brain barrier is more permeable in neonates and young children,


leading to an increased risk of CNS adverse effects.
Metabolism:

o At birth, the hepatic microsomal enzyme system is relatively immature.

o Drugs administered to the mother can induce neonatal enzyme activity


(e.g. barbiturates).

Excretion:

o All renal mechanisms (filtration, secretion and reabsorption) are reduced in


neonates.

o Subsequently, during toddlerhood, it exceeds adult values, often


necessitating larger doses per kilogram. E.g. the dose per kilogram of
digoxin is much higher in toddlers than in adults
PHARMACODYNAMICS

 Apparently paradoxical effects of some drugs (e.g. hyperkinesia with


phenobarbitone, sedation of hyperactive children with amphetamine)
are as yet unexplained.

 Augmented responses to warfarin in prepubertal patients occur at


similar plasma concentrations as in adults, implying a
pharmacodynamic mechanism.
PEDIATRIC DRUG DOSAGE

 Most drugs approved for use in children have recommended pediatric doses,
generally stated as milligrams per kilogram.

 Calculations of pediatric dosage:

o Surface area based (Young’s formula): Dose = 𝐴𝐴𝐴𝐴𝐴𝐴𝐴𝐴𝐴𝐴 𝐷𝐷𝐷𝐷𝐷𝐷𝐷𝐷 ×


𝐴𝐴𝐴𝐴𝐴𝐴(𝑦𝑦𝑦𝑦𝑦𝑦𝑦𝑦𝑦𝑦)
𝐴𝐴𝐴𝐴𝐴𝐴+12

o Body weight based (Clark’s rule): Dose = 𝐴𝐴𝐴𝐴𝐴𝐴𝐴𝐴𝐴𝐴 𝐷𝐷𝐷𝐷𝐷𝐷𝐷𝐷 ×


𝑊𝑊𝑊𝑊𝑊𝑊𝑊𝑊𝑊𝑊𝑊 (𝑘𝑘𝑘𝑘)
70
ADVERSE EFFECTS
• With a few notable exceptions, drugs in children generally have a similar adverse
effect profile to those in adults.

Some specific ADR examples are;

• chronic corticosteroid use, including high-dose inhaled corticosteroids, to inhibit


growth
• Tetracyclines are deposited in growing bone and teeth, causing staining and
occasionally dental hypoplasia
• Fluoroquinolone antibacterial drugs may damage growing cartilage
• Dystonias with metoclopramide occur more frequently in children and young
adults than in older adults
• Valproate hepatotoxicity is increased in young children
PEDIATRIC DOSAGE FORMS & COMPLIANCE
 Children under the age of five years may have difficulty in swallowing even small tablets, and
hence oral preparations which taste pleasant are often necessary to improve compliance. (Elixirs
& Suspensions)
 Pressurized aerosols (e.g. salbutamol inhaler) in children over the age of ten years, as
coordinated deep inspiration is required. Nebulizers may be used.
 Children find intravenous infusions uncomfortable and restrictive. Rectal administration is a
convenient alternative (e.g. metronidazole to treat anaerobic infections). Rectal diazepam is
particularly valuable in the treatment of status epilepticus. Rectal administration should also be
considered if the child is vomiting.
Rules of prescribing for Pediatric populations

 Calculate the doses for prescribed drugs based on weight of the patients.

 Ensure proper instructions to the care giver, including when the child vomits the given
medication after consumption.

 Ensure that all medicines are strictly out of reach of children at all times.

 Avoid prolonged treatment with drugs that have delayed complications (Steroids).

 Use antibiotics sparingly and only when required.

 Medications affecting the CNS need to be extensively reviewed and routinely monitored
to ensure minimal growth disturbances.
Older patients are not slowed
down adults!!!!
Geraitric Group - Pharmacokinetics

 Absorption: Little evidence of any major alteration in drug absorption with


age. However, conditions associated with age may alter rate at which some
drugs are absorbed. (Diabetic gastroparaesis, laxative abuse)

 Distribution: Elderly have reduced lean body mass, reduced body water.

 Metabolism: Capacity of liver to metabolize drugs does not appear to


decline consistently with age for all drugs.

 Elimination: Kidney is major organ for clearance of drugs from body, age-
related decline of renal functional capacity is important.
Pharmacodynamics
• Geriatric patients believed to be much more "sensitive" to action of many
drugs, implying a change in pharmacodynamic interaction of drugs with their
receptors. BUT, most of these are a result of changing Pharmacokinetics!
Rules of prescribing for the elderly

o Think about the necessity for o Take a careful drug history.


drugs. o Use fixed combinations of drugs
o Avoid drugs with negligible or rarely.
doubtful benefits. o Check Compliance.
o Think about the dose. o Think before adding a new drug to
o Think about drug formulation. the regimen.

o Assume any new symptoms may o Stopping is as important as Starting.


be due to drug side-effects.
Geriatric Prescribing - ADRs
Most frequent drug classes causing
ADRs and Age ADRs

o Incidence of ADR increases with age o Cardiovascular active agents


o Elderly receive more medicines
o Analgesics (opioid mainly)
o Incidence of ADR increases the more
o Antibiotics
prescribed medicines taken

o For patients aged>50 yrs o Hypoglycemic agents


o ADR rates – 5% for 1 or 2 medicines o Psychotropic agents
o Increased to 20% when >5 medicines
o Anticoagulants
THANK YOU
Management of Drug
Poisoning
/
Mutah Faculty of Medicine

14/11/2021 1
14/11/2021 2
Eric Doglas

14/11/2021 3
Drug Poisoning

14/11/2021 4
14/11/2021 5
14/11/2021 6
Causes of death in drug
poisoning
 CNS depression: Narcotics, sedative-hypnotics
 CVS toxicity: Digitalis, Cocaine

 Cellular hypoxia: Cyanide and CO

 Convulsions: Cocaine

 Organ system damage: Paracetamol

 Accidents

14/11/2021 7
ABCD of Poisoning treatment

 A: Airway
 B: Breathing

 C: Circulation

 D: Dextrose

14/11/2021 8
Prevention of further absorption of
the poison:
 Remove patient from the toxic environment
 Measures of decontamination:

 Removing toxins from:


Skin
GIT:
Emesis (not in petrolium nor in corrosive poisoning)
Gastric lavage
Activated charcoal

14/11/2021 9
Principles of treatment of poisoning
 ABCD of poisoning treatment
A: Airway, B: Breathing, C: Circulation, D:
Dextrose
 Diagnosis;history, exam, investigations
 Prevention of absorption of the poison:
Skin, GIT (Emesis, G lavage, Activated Charcoal)
 Specific
antidote
 Enhancing elimination of toxins by:
Haemodialysis or alteration of urinary pH
14/11/2021 10
Activated charcoal
 Reduces drug absorption
 Better than emesis or gastric lavage

 Safer, easier, adsorb toxic substances

 Binds to and inactivates many drugs

 Does not bind iron, lithium, corrosive acids


and alkali
 Given early within one hour of poisoning

14/11/2021 11
Specific antidote

 Paracetamol Acetylcysteine
 Iron Desferoxamine
 Digitalis Digoxin antibodies
 Benzodiazepines Flumazenil
 Opioids Naloxone
 OPI (CE inhibitors) Pralidoxime

14/11/2021 12
Enhancing Elimination of Toxins
 Haemodialysis:
Aspirin, Lithium, Carbamazepine

 Urinary pH alteration:
Urine alkalinazation: aspirin
Urine acidification: amphetamines

14/11/2021 13
Examples of Common Poisoning

14/11/2021 14
Paracetamol (Acetaminophen)
 Most common suicide drug
 Ingestion of 7 g total (adults) is toxic

 A highly toxic metabolite (NABQI) is


produced in the liver leading to depletion of
the protective hepatic glutathione
 Patient is asymptomatic initially

 After 24–36 hours, hepato-renal failure and


even death may occur
14/11/2021 15
Paracetamol poisoning

 Early treatment (within 8 hrs) is important


 N-acetylcysteine IV or methionine orally to
increase hepatic glutathione

14/11/2021 16
Pharmacokinetics of Paracetamol
 The highly toxic metabolite is N-acetyl-p-benzo
quinonimine (NABQI) conjugates with glutathione
 In overdose toxicity:
 Excess NABQI
 Glutathione depletion
 Then NABQI oxidizes thiol group of enzymes
 Leading to cell death
 Resulting in hepatic & renal tubular cell damage

14/11/2021 17
Paracetamol (Acetaminophen)
 Serum level > 200 mg/L after 4 hours of
ingestion suggests a risk for liver injury

 Acetylcysteine acts as a glutathione


substitute, binding the toxic metabolite

 Should be started within 8–10 hours if


possible
14/11/2021 18
Anti-muscarinic agents
(Atropine-like drugs)

 Hot, dry, flushed skin


 Blurred vision

 Delirium

 Tachycardia, mydriasis

 Treatment is supportive

14/11/2021 19
Aspirin (Salicylate)
 Ingestionof > 200 mg/kg
 Hyperventilation, respiratory alkalosis,
metabolic acidosis
 Hyperthermia

 Convulsions, coma

 CV collapse

14/11/2021 20
Aspirin (Salicylate)
 General supportive care
 Gastric lavage

 Activated charcoal

 IV fluid

 IV sod bicarbonate: renal elimination

 Severe poisoning: Haemodialysis

14/11/2021 21
Organophosphorous insecticide
poisoning
 Cholinergic crisis
Muscarinic & Nicotinic stimulation
 Pinpoint pupil, sweating, diarrhoea
 Urination, defecation

 Hypotension, bradycardia

 Treatment:
Atropine (anti-muscarinic)
Pralidoxime (enzyme reactivator)
14/11/2021 22
Other poisoning
 Iron:
Childhood poisoning; bleeding
Desferoxamine

 Opioids:
Drugs of abuse
CNS & respiratory depression
Naloxone IV

14/11/2021 23
Other poisoning
 Carbon monoxide (CO):
 Colorless, odorless gas
 Results from incomplete combustion
 Forming carboxyhaemoglobin
 Interfering with carrying of oxygen
 Leading to hypoxia

 Cyanide poisoning:
 Syncope, convulsions, coma
 Treatment: Cyanide antidote kit consists of:
 Nitrites: induce methemoglobinemia
 Thiosulfate: converts cyanide to thiocyanate

14/11/2021 24
New Drugs: Their
Development & Evaluation
Dr. Mohammed Al-sbou
Professor in Clinical Pharmacology
Faculty of Medicine, Mutah University

1
New Drug Development
 Idea or hypothesis
 Design & synthesis of substances
 Studies on tissues & animal (preclinical studies)
 Studies on man (clinical studies)
 Official license (registration & market
authorization)
 Post-marketing studies

2
Aims of Therapeutic Evaluation
 To assess efficacy, safety & quality of new
drugs
 To expand indications for the use of current
drugs
 To protect public health

3
Drug Development
 Drugs are chemical substances useful in
prevention & diagnosis & treatment of diseases
 The process of drug development may be
abandoned at any stage including after
marketing (safety, inadequate efficacy)

4
Drug Development
 New drug development is enormously expensive
 Cost of development of a new chemical entity
from synthesis to market US $ 500 million
 The process may take 10-15 years

5
Origin of Drugs
 Natural sources:
 Plant origin like morphine, digoxin, atropine

 Micoorganisms as fungi & bacteria synthesizing


antibiotics
 Animal origin like hormones (insulin), heparin

 Mineral origin like iron, calcium

6
Origin of Drugs
 Synthetic when synthesized chemically in
laboratories
 These represent majority of drugs, as they are
easily manufactured & cheaper like aspirin,
paracetamol & propranolol

7
Medicines
 Medicines are drugs formulated in a suitable
way for administration & use by patients
 Medicines consist of the active drug
combined with excipients that give it shape,
size, stability & other criteria as starch, Arabic
gum & many other substances

8
Therapeutic Investigation
 There are three questions to be answered during
drug development:
1. Does the drug work?
2. Is it safe?
3. What is the dose?

9
Phases of Drug Development
1. Pre-clinical studies in animals
2. Clinical studies in human

10
1. Pre-clinical studies in animals including:
A. General pharmacology studies:
 Pharmacokinetic studies
 Pharmacodynamic studies
 Dose, preparation & routes of
administration

11
1. Pre-clinical Studies in Animals including:
B. Toxicological studies
 Acute toxicity
 Special toxicity studies:
 Reproductive system

 Mutagenesis (mutation production)

 Oncogenesis (malignancy)

 Teratogenicity (harmful effects on


foetus)

12
2. Clinical Studies in Human
 These are carried out in
humans in clinical trials
centers & in hospitals
under supervision of qualified
investigators
 These include:

13
2. Clinical studies in human
 Phase 1 studies
 Phase 2 studies
 Phase 3 studies
 Phase 4 studies

14
Phase 1 Studies
(Human pharmacology)
 These are performed on a limited number of
healthy volunteers (20-50 subjects)
The aims of these trials are:
 Study of the general pharmacology of drug

 Pharmacokinetics (ADME)

 Pharmacodynamics (biological effect)

 Tolerability, efficacy & safety (associated


adverse effects)
15
Phase 2 Studies
(Therapeutic exploration)
 These are carried out on a limited number of
patients (50-300) to:
 General pharmacology of drug in patients
 Pharmacokinetics
 Pharmacodynamics
 Establish safety of drugs
 Assess potential therapeutic effects,
comparison with placebo
16
Phase 3 Studies
(Therapeutic confirmation)
 Randomized control trials
 These include multi-centre comparative
studies on a large number of patients (250-
1000) to establish therapeutic efficacy & safety,
comparison with existing drugs
 Short term efficacy & safety

17
Phase 4 studies
(Therapeutic use)
 These include post-marketing surveillance
(post-authorization studies) (2000- 10,000) to
look for possible long term effects of drugs
 Long term efficacy & safety

18
Phases of Drug Development

19
20 20
Clinical Trials

 Clinical trials are carefully and ethically


designed controlled experiments performed
on human beings to evaluate certain aspects of
drug studies

21
Aims of clinical trials
 Whether treatment is of value
 Magnitude of that value compared with other
remedies
 Type of patients in whom it is of value
 Best method of applying treatment (how often,
dosage of drug)
 Disadvantages & dangers of treatment

22
Fundamental to any clinical trial are:
 An hypothesis
 Definition of primary endpoints
 Method of analysis
 A protocol

23
Other factors when designing a trial:

 Characteristics of patients
 Size of trial
 Duration
 Method of monitoring
 Use of interim analyses

24
Subjects included in the studies are either:
 Healthy normal volunteers or
 Patients

25
Patients excluded from clinical trials include:

 Children
 Pregnant women
 Mentally ill patients

26
Techniques to avoid bias
 Randomization:
- Introducing element of chance into selection &
allocation of subjects to treatments
 Blinding

27
Criteria of clinical trials (CCT)

 Objective: should be clear & limited to one aim


 Careful design: A protocol should be prepared
that shows design of the CCT prepared by clinical
pharmacologist, physician & statistician
 Crossover design: when each subject is
randomized to a sequence of two or more
treatment, and he acts as his own control for
treatment comparisons

28
Criteria of clinical trials
 Clinical trials may be of non-crossover design
recruiting different subjects as a control group
 Balanced regarding sex, age, weight & disease state
 Double-blind technique when neither investigator
nor subject knows about treatments they are
receiving. This technique is important to:
 Eliminate investigator bias
 Eliminate patients or subject bias
 Allow the use of placebo

29
 Single-blind technique is described when
investigator knows but patient does not
know treatment given to him
 Control group is used who will receive either
placebo or a standard therapy
 Statistical analysis should be planned initially
including the proper tests used

30
The use of placebo
 It is a pharmacologically inert substance
identical in all aspects to the active treatment
indistinguishable from it
It is intended to:
 Eliminate observer or investigator bias
 Detect non-pharmacological effects of drugs
(placebo effects)
 A control for statistical comparison

31
Conditions that do not require use of
placebo
 Therapeutic studies as it is unethical to
deprive patients of treatments. A standard
therapy is chosen instead of placebo
 When the active compound can be identified
e.g. a vasodilator, alkaptonuria (nitisinone)
 Dose-finding studies
 Pharmacokinetic studies

32
Ethical Considerations in Clinical Trials
 Declaration of Helsinki
 The declaration of Helsinki (1964, 1975) sought to
clarify the ethical principles governing clinical
research involving human subjects emphasizing
informed consent & proper scientific research
design. It is the mission of doctor to safeguard
health of people. The doctor’s knowledge &
conscience are dedicated to the fulfillment of this
mission
33
Recommendations are essential as a
guide to doctors in clinical research:

 Risks & benefits must be carefully assessed


 Nature, purpose & possible hazards must be
explained to subjects by doctor

34
Recommendations are essential as a
guide to doctors in clinical research:

 Informed written consent must be obtained


 Subjects must be free to withdraw from clinical
trial anytime
 The investigators should discontinue research, if in
their judgment it may if continued be harmful to
subjects

35
DevelopAKUre

 4 years international multicenter


clinical trials, funded by European
commission (FP7) - € 6 million
 12 European partners & Jordan
(Faculty of medicine-mutah university)
 3 trial sites: UK (Liverpool), France
(Paris) & Slovakia (Piestany)
 Aims: to study the potential
effectiveness & safety of nitisinone in
treating alkaptonuria (AKU)
37
SONIA 2 (Suitability of Nitisinone in AKU )

o 140 patients
o Patients were from Spain,
France, Belgium, Italy,
Netherlands, Germany,
Slovakia & Jordan
o 19 Jordanian patients
National Institute for
Rheumatic diseases,
Slovakia
38
Procedures

• Bloods: • Urine: • Yearly:


• HGA, tyrosine, • HGA, tyrosine, • Abdominal ultrasound,
nitisinone nitisinone Audiometry
• Bone/muscle/cartilage • Bone & cartilage • ECG, Echocardiogram
markers markers
• Bone density scan
• Biochemistry profiles, • Metabolomics (Dexa)
metabolomics
• Isotope scintigraphic
• Genetics scan
• Acute phase reactants, • Medical Photographs
cytokines
 www.ClinicalTrials.gov

39

You might also like