PCOL
PCOL
SECTION DESCRIPTION
Overview of The lesson is intended to introduce the general principles of pharmacology,
Session enumerate end define the branches of pharmacology and review the different
pharmacokinetic principles.
• Pharmacology can be defined as the study of substances that interact with living
systems through chemical processes by binding to regulatory molecules and
activating or inhibiting normal body processes.
What is Pharmacodynamics?
What is Pharmacokinetics?
Define Drug.
• Drug is any substance, other than food, that is used in the prevention, diagnosis,
alleviation, treatment, or cure of disease.
3. Diagnostic agents
BaSO4 – for roentgenographic examination
4. Chemotherapeutic agent
• Selective toxicity
A. LADME
1. LIBERATION- release of the active ingredient (drug) from its dosage form
TRANSPORT MECHANISMS
- means of movement of drug molecule across cell membrane
3. CONVECTIVE TRANSPORT
- small drug molecules simply move along with fluid through the pores in cell walls
- MEDIATED through “water-filled pores/channels/aquaporins”
Eg. Grains of sand flowing down a drain along with running water
4. PINOCYTOSIS
- cell drinking
- ATP requiring transport
- Transport of large hydrophilic molecules
5. ACTIVE TRANSPORT
- from lower concentration to higher concentration
- “pushing a rock uphill”
- requires cellular energy
What are the different factors affecting the rate and extent of drug distribution?
Differentiate bound from unbound/free drug?
When is drug on its active form?
3. DISTRIBUTION
• Site of administration to the site of action (tissues or organs)
• The extent to which the drug passes into different tissues and fluid compartments
in the body.
4. METABOLISM/BIOTRANSFORMATION
• The chemical process by which the body converts an agent from its original form
to a more water-soluble form that can be excreted.
objective:
convert drug to forms which are less toxic, less active or inactive,
easily excreted
exception:
PRODRUG – inactive molecule converted to an active molecule
LIVER- most important site of drug metabolism, but it may also occur in renal
tissue, lungs, plasma or intestinal mucosa.
1. KIDNEY – most important/ primary organ in drug excretion for polar drugs.
a. Glomerular Filtration – a passive process by which small molecules and
drugs are filtered.
b. Tubular reabsorption – a passive process that follows Fick’s Law of
diffusion.
c. Active tubular secretion - a carrier-mediated active transport system that
requires energy.
2. SKIN- sweat
3. SALIVA
4. LUNGS – anesthetic
5. BILE – an active process that needs high drug concentrations for it to be
saturated.
- eg. Cefamandole, diazepam, digoxin, spironolactone, chloramphenicol
6. FECES- if the drug is given parenterally, an then observed in the feces, one can
assess that some of the drug was excreted in the bile.
- if given orally, some of the fecal drug excreted could represent
unabsorbed drug.
3. Gender-for IM injection
Male>Female in terms of vascular muscles
- Effects faster in male than female
4. PATHOLOGICAL FACTORS
- liver disease = biotransformation of drugs = toxic effects
5. GENETIC FACTORS
- ASIANS are fast acetylators
Acetylation (metabolizing INH) = antitubercular effects
Assessment A short examination regarding the topic discussed will be given the next meeting.
SUBJECT PHARMACOLOGY 1 TOPIC Pharmacodynamics/Kinetics
IG NUMBER IG-PHA-325LC WEEK NO 2
TERM PRELIM SESSION 3 of 30
DATE PREPARED May 2007 DURATION 1.5 HOURS
SECTION DESCRIPTION
Overview of The lesson is intended to discuss the different pharmacodynamic principles, and to
Session familiarize the students with the different drug-receptor theories.
Pharmacodynamics
- A branch of pharmacology that focuses on the study of the biochemical and
physiological effects of drugs and the mechanisms by which they produce such
effects.
1. Structural protein
Microtubule- important site of action
Drugs that inhibit microtubule synthesis/ spindle protein
A. Griseofulvin
B. Vinca alkaloids
C. Colchicine
2. Regulatory proteins
A. Channels/transport
Volgate-gated Na channel
Volgate-gated Ca channel
B. Carrier molecules
Na+-K+ ATPase pump- Digitalis glycosides
C. Enzymes
Xanthine oxidase - allopurinol
Cyclooxygenase - NSAID’s
ACE - ACE inhibitors
MAO - MAOI
Phenelzine
Isocarbozacid
Trancylpromine
Acetylcholinesterase- Tensilon
SECTION DESCRIPTION
Overview of This lesson is intended to discuss the different drug effects in the body. The lesson
Session covers concepts of agonism, antagonism and allosteric modulation.
Receptors
Type of Receptors
- location: nucleus
- onset: hours
-location: nucleus
-onset: hours
1. Affinity
• Ability to bind to receptor or target protein
• w/ or w/o effect
2. Intrinsic/biological property
• Ability to generate a series of biochemical effect leading to an effect.
FOUR VARIABLES:
1. maximal efficavy
2. slope
3. potency
4. ceiling dose
SLOPE
- reflects the ability of the drug to produce an effect. The STEEPER the slope, the
more readily the drug will bind with receptors.
POTENCY
- describes the relationship between the dose of the drug and the intensity of its
effect.
10 mg morphine = 1 mg hydromorphine
SECTION DESCRIPTION
Overview of This lesson is intended to discuss the different drug effects in the body. The lesson
Session covers concepts of agonism, antagonism and allosteric modulation.
Objectives 1. To identify the different classifications of drug according to thir interaction with
receptors
2. To classify drugs according their interaction with physiologic enzymes
3. To classify drugs according to their interaction with nucleic acids
1. AGONIST
• Able to bind
• With intrinsic activity
a. Full agonist
- Full response
- Stimulate all different variant of receptors even the new
receptors
b. Partial agonist
- Less than the expected response
2. ANTAGONIST
• With affinity but no intrinsic activity
• It prevents the binding if agonist
2. Inhibition of DNA or RNA biosynthesis occurs when drugs interfere with nucleic
acid synthesis. They are used primarily as antineoplastics in cancer
chemotherapy.
a. Drugs that interfere with DNA replication and function include intercalating
agents, alkylating agents, and antimetabolites.
b. Drugs that can damage and destroy DNA include compounds that can
produce free radicals and compounds that can inhibit topoisomerases.
c. Drugs that can interfere with microtubule assembly in the metaphase of cell
mitosis.
Define synergism.
What is the mathematical representation of an additive effect? Of synergistic effect?
Of Potentiating effect?
Give examples of drugs that exhibit additive effect.
Give examples of a synergism.
Give examples of potentiation.
DRUG EFFECT
The degree or extent of pharmacologic response or drug effect resulting from
drug action is determined in part by
SECTION DESCRIPTION
Overview of This lesson is intended to discuss the different pharmacotherapeutic principles. This
Session topic includes subtopics about drug development and clinical trials
Assessment A short quiz regarding the topic discussed will be given the following meeting
SUBJECT PHARMACOLOGY 1 TOPIC AUTACOID PHARMACOLOGY
IG NUMBER IG-PHA-325LC WEEK NO 4
TERM PRELIM SESSION 7 of 30
DATE PREPARED May 2007 DURATION 1.5 HOURS
SECTION DESCRIPTION
Overview of This lesson is intended to discuss Histamine pharmacology, its structure-activity
Session relationship, the mechanisms of its pharmacologic and physiologic actions, its
different physiologic effects and therapeutic uses, and the adverse effects associated
to it.
Lesson
Outline Histamine Pharmacology
1. Synthesis
a. Histamine is found in many tissues, including the brain; it is stored and found
in the highest amounts in mast cells and basophils. Mast cells, which are
especially abundant in the respiratory tract, skin (especially hands and feet),
gastrointestinal tract, and blood vessels, store histamine in a granule bound
in a complex with heparin, adenosine triphosphate (ATP), and an acidic
protein.
b. Release of histamine can occur by two processes:
(1) Energy- and Ca2+-dependent degranulation reaction. Immunoglobulin E
(IgE) fixation to mast cells (sensitization) and subsequent exposure to a
specific antigen induce the release of histamine from mast cells;
complement activation (Immunoglobulin G- or immunoglobulin M-
mediated) may also induce degranulation.
(2) Energy- and Ca2+ - independent release (displacement). Drugs such as
morphine, tubocurarine, guanethidine, and amine antibiotics induce
displacement. In addition, mast cell damage, which is caused by noxious
agents such as venom or by mechanical trauma, can release histamine.
2. Mechanism of action
B. Histamine agonists
Assignment
Read Histamine antagonists.
SUBJECT PHARMACOLOGY 1 TOPIC HISTAMINE ANTAGONISTS
IG NUMBER IG-PHA-325LC WEEK NO 4
TERM PRELIM SESSION 8 of 30
DATE PREPARED May 2007 DURATION 1.5 HOURS
SECTION DESCRIPTION
Overview of This lesson is intended to discuss Histamine pharmacology, its structure-activity
Session relationship, the mechanisms of its pharmacologic and physiologic actions, its
different physiologic effects and therapeutic uses, and the adverse effects associated
to it.
A. Classification
a. First-generation agents
(1) Alkylamines
(a) Alkylamines include chlorpheniramine and brompheniramine.
(b) These agents produce slight sedation.
(c) Alkylamines are the antihistamines used most frequently in the United
States.
(2) Ethanolamines
(a) Ethanolamines include diphenhydramine, doxylamine, and clemastine
(Tavist); dimenhydrinate is an equimolar combination of
diphenhydramine and 8-chlorotheophylline.
(b) Ethanolamines produce marked sedation; doxylamine is marketed
only as a sleeping aid.
(c) Ethanolamines also act as antiemetics.
(3) Ethylenediamines
(a) Ethylenediamines include pyrilamine, antazoline, and tripelennamine.
(b) Ethylenediamines produce moderate sedation and can cause
gastrointestinal upset.
(4) Piperazines
(a) Piperazines include meclizine and cyclizine.
(b) Piperazines produce marked adverse gastrointestinal effects and
moderate sedation.
(c) These agents are antiemetic.
(5) Phenothiazines
(a) Phenothiazines include promethazine and cyproheptadine.
(b) Phenothazines produce marked sedation.
(c) These agents have antiemetic activity.
(d) Phenothiazines are also weak alpha-adrenoceptor antagonists
(6) Methylpiperidines
(a) Methylpiperidines include cyproheptadine (periactin).
(b) Methylpiperidines have antihistamine, anticholinergic, and
antiserotonin activities.
b. Second-generation (nonsedating agents)
(1) Piperidines
(a) Terfenadine (Seldane)
a.1 Terfenadine is metabolized to the active metabolite
fexofenadine by a specific cytochrome P-450. This enzyme is inhibited
by certaim antibiotics (erythromycin) and antifungal agents
(ketoconazole).
a.2 The parent drug can interfere with cardiac potassium channels,
producing ventricular tachycardia; this can occur with large doses of
terfenadine or if other drugs block the P-450.
(b) Fexofenadine (Allegra). has been approved as a safer antihistamine of
this class.
(c) Astemizole (Hismanal). There are some reports of ventricular
tachycardia associated with astemizole; the mechanism of action is
likely the same as with terfenadine.
(d) Loratadine (Claritin). Poor CNS penetration.
(2) Alkylamines: acravistine (Semprex-D). Acravistine is not associated with
cardiac effects.
(3) Cetirizine (Zyrtec)
i. Cetirizine is not associated with cardiac abnormalities.
ii. Cetirizine has poor penetration into the CNS.
iii. Cetirizine is less sedating; it is ineffective for motion sickness or
antiemesis.
1. Treatment of allergic rhinitis and conjunctivitis. These agents are also used to
treat the common cold based on their anticholinergic properties, but they are only
marginally effective for this use. Diphenhydramine also has an antitussive effect not
mediated by H1-receptor antagonism.
2. Treatment of urticaria and atopic dermatitis, including hives
3. Sedatives. Several (doxylamine, diphenhydramine) are marketed as over-the-
counter (OTC) sleep aids.
a. Prevention of motion sickness
b. Appetite suppressants.
Assessment A chapter examination regarding histamine pharmacology will be given next meeting
SUBJECT PHARMACOLOGY 1 TOPIC SEROTONIN
AGONISTS/ANTAGONISTS
IG NUMBER IG-PHA-325LC WEEK NO 5
TERM PRELIM SESSION 9 of 30
DATE PREPARED May 2007 DURATION 1.5 HOURS
SECTION DESCRIPTION
Overview of This topic is intended to discuss Serotonin pharmacology which includes subtopics
Session regarding the synthesis, storage and release of serotonin, the physiologic effects and
pharmacologic uses, the different serotonin agonists and antagonists and the adverse
effects associated with the uuse of the agents.
Serotonin antagonists
1. Cycloheptadine (Periactin)
a. Cyproheptadine is a potent H1-receptor antagonist of the phenothiazine class;
it blocks both 5HT1- and 5HT2-receptors.
b. Cyproheptadine is used most frequently to limit diarrhea and intestinal spasms
produced by serotonin-secreting carcinoid tumors and postgastrectomy
dumping syndrome.
c. Cyproheptadine produces sedation and anticholinergic actions.
2. Ondansetron (Periactin)
a. Ondansetron is a 5HT3-receptor antagonist.
b. Ondansetron is highly effective in treating the nausea and vomiting associated
with chemotheraphy and radiation theraphy.
c. Administered intravenously or orally.
3. Granisetron (Kytril)
a. Similar in action and use to ondansetron.
b. Blocks 5HT3 receptors.
c. May be administered intravenously or orally.
4. Ketanserin (Sufrexal)
a. Ketanserin is a specific 5HT2-receptor antagonist; it also antagonizes alpha-
adrenergic, H1-, and dopamine receptors.
b. Ketanserin is an investigational drug that has been found to lower blood
pressure in experimentally induced hypertension.
5. Clozapine (Clozaril)
a. Clozapine is a specific 5HT2A- and 5HT2C – receptor antagonist.
b. Clozapine is an antipsychotic with reduced extrapyramidal effects.
6. Risperidone (Risperdal)
a. Risperidone is an antagonist at 5HT2A-, 5HT2C-, and dopamine (D2)-receptors.
b. Risperidone is an atypical antischizophrenic agent with reduced extrapyramidal
activity.
SECTION DESCRIPTION
Overview of This topic is intended to discuss the pharmacologic properties of eicosanoids. The
Session effects of eicosanoids in the body, its medical properties as well as the toxicity
associated with the use of eicosanoids would be also discussed.
Lesson Eicosanoids
Outline
What is the fatty acid precursor of eicosanoids?
What are the physiologic effects of eicosanoids?
How does eicosanoids contribute to inflammation?
What are the therapeutic uses of eicosanoids?
What is the difference between thromboxanes and prostaglandins?
What pathway leads to the formation of prostaglandins?
What pathway leads to the formation of leukotrienes?
A. Overview
1. Eicosanoids are a large group of autacoids with potent effects on virtually
every tissue in the body; these agents are derived from metabolism of 20-
carbon, unsaturated fatty acids (eicosanoic acids).
2. The eicosanoids include the prostaglandins, thromboxanes, leukotrienes,
hydroperoxyeicosatetraenoic acids (HPETEs), and hydroeicosatertraenoic acids
(HETEs).
B. Biosynthesis
1. Arachidonic acid, the most common precursor of the eicosanoids, is formed by
two pathways:
a. Phospholipase A2- mediated production from membrane phospholipids;
this pathway is inhibited by glucocorticoids.
b. Phopholipase C in concert with diglyceride lipase can also produce free
arachidonate.
2. Eicosanoids are synthesized by two pathways:
a. The prostaglandin H synthase (COX, cyclooxygenase) pathway produces
thromboxane, the primary prostaglandins (prostaglandin E, or PGE; the
prostaglandin F, or PGF; and prostaglandin D, or PGD), and
prostaglandin (PGI2).
(1) There are two forms of COX: Type 1 is located in endothelial
cells, kidney, gastrointestinal tract, and many other locations; type 2
is found in great abundance in connective tissues.
(2) The type 1 enzyme is expressed at fairly constant levels and
may provide protective action on gastric mucosa and in the kidney.
(3) The type 2 enzyme is highly inducible by numerous factors
associated with inflammation. Current theories suggest that the type
2 isoforms is predominantly associated with eicosanoids production
in inflammation.
b. The lipoxygenase pathway produces the HPETEs, HETEs, and the
leukotrienes.
(1) 5-Lipoxygenase produces 5_HPETEs, which are subsequently
converted to 5_HETEs and then to leukotienes.
(2) 12-Lipoxygenase produces 12-HPTES, which are converted to 12-
HETEs).
c. Additional metabolites of the HPETEs, hepoxillinsm and lipoxins, have
been identified, but their biological roles in unclear.
3. The eicosanoids all have short plasma half-lives (typically 0.5-5 minutes).
Most catabolism occurs in the lung.
a. Prostaglandins are metabolized by prostaglandin 15-OH dehydrogenase
(PDGH) to 15-keto metabolites, which are excreted in the urine.
b. Thromboxane A2 (TXA2) is rapidly hydrated to the less active TXB2.
c. PGI2 is hydrolyzed to 6-keto-PGF.
4. Various eicosanoids are synthesized throughout the body; synthesis can be
very tissue-specific:
a. PGI2 is synthesized in endothelial and vascular smooth musclecells.
b. Thromboxane syntghesis occurs primarily in platelets.
c. HPETEs, HETEs, and the leukotrienes are synthesized predominantly in
mast cells, white blood cells, airway epithelium, and platelets.
D. Therapeutic uses
1. Induction of labor at term. Induction of labor is produced by infusion of PGF2
(carboprost tromethamine) (Hemabate) or PGE2 (dinoprostone) (Prostin E).
2. Therapeutic abortion. Infusion of carboprost tromethamine or administration of
vaginal suppositories containing dinoprostone is effective in inducing abortion in the
second trimester. Currently, these prostaglandins are combined with mifepristone
(RU486) to induce first-trimester abortion.
3. Maintenance of ductus arteriosus. This action is produced by PGE1 (prostin VR)
infusion; PGE1 will maintain patency of the ductus arteriosus, which may be desitable
before syrgery.
4. Treatment of peptic ulcer. Misoprostol (cytotec), a methylated derivative of PGE1,
is approved for use in patients taking high doses of nonsteroidal anti-inflammatory
drugs (NSAIDs) to reduce gastric ulceration.
E. Adverse effects. Adverse effects of eicosanoids include local pain and irritation,
bronchospasm, and gastrointestinal disturbances, including nausea, vomiting,
cramping, and diarrhea.
Kinins
- One of a number of widely differing substances having pronounced and dramatic
physiological effects; some of the substances under this group are plant growth
regulators; others are polypeptides, formed in the blood by proteolysis secondary to
some pathologic process, that stimulate visceral smooth muscle but relax vascular
smooth muscle, thus, producing vasodilatation.
A. Biosynthesis
1. Kinins are found in the circulation and tissues as larger molecular weight
precursors, kininogens. Specific serine proteases called kallikreins convert
kininogens to the active kinins. Hagenman factor, trypsin, and kinins activate
Kallikreins.
2. The major kinins are a group of related peptides with potent actions as
vasodilators.
a. Bradykinin. Bradykinin has the following amino acid sequence:
ArgPro-Pro-Gly-Phe-Ser-Pro-Phe-Arg.
b. Lysyl-bradykinin (kallidin) has additional lysine residue on the N-
terminus of the bradykinin.
c. The des-Arg (removal of the C-terminal argiine) metabolites of both
bradykinin and kallidin are biologically active.
B. Actions
1. The effects of kinins are mediated by specific bradykinin receptors (B1 and
B2- receptors).
2. Kinins produce vasodilation by relaxation of precapillary arteriolar smooth
muscle (10 times more potent than histamine), as well as vasoconstriction of
capacitance vessels; these combined effects cause a rapid but transient fall in
bloof pressure.
3. Kinins stimulate gastrointestinal smooth muscle.
4. These agents increase renal blood flow and enhance chloride transport.
5. Kinins may play a role in the inflammatory process.
6. Kinins activate peripheral nerve endings that sense pain.
C. Therapeutic uses
1. Aprotinin (Trasylol) inhibits kallikreins and, thus, the formation of kinins; it
also inhibits other proteases, including plasmin.
2. Aprotinin is approved for use during cardiac bypass surgery based on its
anticoagulation properties that reduce the amount of blood needed for
transfusion during extracorporeal procedures.
3. Agents that alter kinin release are under investigation for the treatment of
inflammation.
SECTION DESCRIPTION
Overview of This lesson is intended to dicuss the Nervous system and its divisions.
Session
I. DIVISIONS
a. CENTRAL NERVOUS SYSTEM (CNS)
a.1 brain
a.2 spinal cord
b. PERIPHERAL NERVOUS SYSTEM (PNS)
Collection of nerves and ganglia scattered throughout the brain
Consists of 12 pairs of cranial nerves, 31 pairs of spinal nerves and
their branches to the entire body.
Transmits information to and from the CNS
A. Divisions of ANS
NEUROTRANSMITTERS
1 Acetylcholine- neurotransmitter released by pre-ganglionic neurons in both the
sympathetic and the parasympathetic divisions
• RELEASED by the POST-GANGLIONIC NEURONS in the
PARASYMPATHETIC DIVISION
Synthesis of NE
Tyrosine
Tyrosine hydroxylase
Dopa
Dopa decarboxylase
Dopamine
Dopamine b-hydroxylase
Norepinephrine
Assessment A short examination about the topic discussed will be given the following meeting
SUBJECT PHARMACOLOGY 1 TOPIC ANS-1
IG NUMBER IG-PHA-325LC WEEK NO 7
TERM MIDTERM SESSION 12 of 30
DATE PREPARED May 2007 DURATION 1.5 HOURS
SECTION DESCRIPTION
Overview of This lesson is inetended to dicuss the Functions of Autonomic Nervous System and its
Session effect in different organs.
Autonomic pathway
THUS, A NERVE IMPULSE ORIGINATING IN THE CNS FIRST SYNAPSES AT THE END
OF A PREGANGLIONIC FIBER IN A GANGLION, AND THEN SYNAPSES AT A POST
GANGLIONIC FIBER IN A MUSCLE FIBER OR GLAND.
ADRENERGIC RECEPTORS
Subtypes of adrenergic receptors
1. Alpha receptor
Location
1.1 alpha 1 smooth muscle tissue of peripheral vessels,
trigone and sphincter of the bladder, male
sex organ, and other tissues
1.2 alpha 2 located in pre-synaptic neurons
2. Beta receptor
2.1 Beta 1
2.2 Beta 2 bronchial smooth muscle
CHOLINERGIC RECEPTORS
Subtypes
1. Muscarinic receptor Located only in postganglionic effector cells
2. Nicotinic receptor NMJ, ganglia of both ANS divisions
Assessment
A short examination about the topic discussed will be given the following meeting
SUBJECT PHARMACOLOGY 1 TOPIC PARASYMPATHETIC DRUGS
IG NUMBER IG-PHA-325LC WEEK NO 8
TERM MIDTERM SESSION 13 of 30
DATE PREPARED May 2007 DURATION 1.5 HOURS
SECTION DESCRIPTION
Overview of This lesson is intended to discuss the effects of the Paraympathetic Nervous System
Session in the body, and the different agents that mimic the effects of the Parasympathetic
Nervous System.
Objectives
1. To discuss the receptors of the Parasympathetic nervous system
2. To enumerate the effects of parasympathomimetic agents in the receptors
located at different organs
3. To discuss the mechanisms of action of different cholinergic agonists
4. To classify the cholinergic agonists according to their chemical structure and
duration of actions
CHOLINERGIC AGONIST
PARASYMPATHOMIMETIC
B. Short-acting cholinomimetic
• Echothiophate
• Isofluorophate
• Malathion
• Parathion
Pharmacology
1. Cholinergic responses are mediated by both muscarinic and nicotinic receptors.
2. Cholinergic agonists act by mimicking the activity of endogenous acetylcholine at
muscarinic and nicotinic receptor sites.
a. Direct acting agonists interact directly with nicotinic and muscarinic
receptors.
b. Indirect acting agonists inhibit or block the activity of cholinesterase
enzymes, which metabolize endogenous acetylcholine to inactive
metabolites.
Organ Response
Heart
AV Node Decreased conduction velocity(negative dromotropy)
Atria,ventricles Decreased contraction force(negative inotropy)
SA node Decreased contraction rate(negative chronotropy)
Eye
Sphicter muscle Contraction, miosis
Ciliary muscle Contraction, accommodates near vision
Lung
Bronchial
Treatment of Organophosphate poisoning
1. Regenerator compounds
Used early in the course of poisoning
Facilitate removal of binding of Organophosphate to ACHE
Oxime derivatives (pralidoxime)
2. Physiologic antidote
Atropine –long term administration of acetylcholine is needed
Remove the enzyme to which organoP04 is bound
Assessment A short examination about the topic discussed will be given the following meeting
SUBJECT PHARMACOLOGY 1 TOPIC Parasympatholytic agents
IG NUMBER IG-PHA-325LC WEEK NO 8
TERM MIDTERM SESSION 14 of 30
DATE PREPARED May 2007 DURATION 1.5 HOURS
SECTION DESCRIPTION
Overview of This lesson is intended to discuss the pharmacology of the agents that reverse the
Session effect of parasympathetic nervous system.
PARASYMPATHOLYTIC DRUGS
Lesson
Outline What is the mechanism of action of parasympatholytics?
What is the prototype of anti-cholinergic drugs?
What are the effects of parasympatholytics?
What are the components of Twilight sleep?
Give examples of ganglionic blockers.
Parasympatholytics/Anti-cholinergic
1. Antimuscarinic
2. Antinicotinic
Anti-muscarinic
Atropine- Prototype drug
Effects
Bronchi bronchodilation
GIT Relaxation of intestinal walls and closure of sphincters
(constipation)
GUT Relaxation of bladder walls and closure of sphincters
(urinary retention)
Exocrine Absence of sweat (anhidrosis)
body temperature (hyperthermia)
Heart tachycardia,
Atropine
A tertiary amine
CNS effects seizures, psychosis, anxiety, restlessness
Effects according to target organ/tissue
Electrochemical dissociation
with observable electrical activity but
w/o cardiac contraction
Anti-nicotinic
Ganglionic blockers
Block both nicotinic receptors in sympathetic and parasympathetic ganglia
Non selective response
1. Hexamethonium
2. Mecamylamine
3. Trimethaphan
Effects
Pupils Para (miosis) mydriasis
Heart Para (bradycardia) tachycardia
GIT Para (diarrhea) constipation
GUT Para (urination) Urinary retention
Blood vessels S (vasoconstriction) vasodilation(hypotension)
Sweat glands S( sweating) absence of sweating
Assessment A short examination about the topic discussed will be given the following meeting
SUBJECT PHARMACOLOGY 1 TOPIC Sympathomimetic Drugs.
IG NUMBER IG-PHA-325LC WEEK NO 9
TERM MIDTERM SESSION 15 of 30
DATE PREPARED May 2007 DURATION 1.5 HOURS
SECTION DESCRIPTION
Overview of This topic is intended to dicuss the pharmacology of Sympathomimetic Drugs.
Session
1. To discuss the physiologic effects of the Sympathetic Nervous System
Objectives 2. To discuss the therapeutic indications of adrenergic agonists
3. To discuss the mechanisms of action of sympathomimetics
4. To classify the sympathomimetic drugs according to their mechanisms of action
5. To enumerate the adverse effects associated with the use of sympathomimetics
A. Chemistry
B. Pharmacology
1. Adrenergic peripheral responses are mediated both alpha and beta-
adrenoceptors.
A. Alpha Receptors:
B. Beta Receptors:
Assessment A short examination about the topic discussed will be given the following meeting
SUBJECT PHARMACOLOGY 1 TOPIC ADRENERGIC ANTAGONISTS
IG NUMBER IG-PHA-325LC WEEK NO 9
TERM MIDTERM SESSION 16 of 30
DATE PREPARED May 2007 DURATION 1.5 HOURS
SECTION DESCRIPTION
Overview of This lesson is intended to discuss the pharmacology of the different adrenergic
Session antagonists.
Sympatholytic or adrenergic blocking agents – a drug that lyses or blocks the effects
of the sympathetic nervous system
suffix
-zosin postsynaptic alpha-receptor blockers
terazosin, prazosin
2. beta-blockers
NON-SELECTIVE- Propranolol (prototype agent)
A. CARDIOSelective (B1 blocker)
Bisoprolol
Esmolol
Atenolol
Acebutolol
Metoprolol
B. with ISA (Intrinsic sympathomimetic activity)
Pindolol
Acebutolol
Labetalol
C. WITH MSA (MEMBRANE SATBILIZING ACTIVITY)
Should not be given as ophthalmic drops
Pindolol
Acebultol
Labetalol
Propranolol
Metoprolol
3. Mixed alpha-beta blocking effect
Carvedilol
Labetalol
C. Therapeutic Indications
1. Alpha-1 antagonists may be selective or nonselective. Phenoxybenzamine is
an irreversible antagonist because it forms covalent bonds with alpha-
receptors.
2. Phenoxybenzamine and phentolamine are used to relieve vasospasm in
Raynaud’s syndrome and for acute hypertensive emergencies resulting from
pheochromocytoma or from intake of MAO inhibitors or sympathomimetics.
3. Tolazoline, a similar agent, is used to treat neonatal pulmonary
hypertension.
4. Labetalol, an agent that possess both selective alpha-1 blocking activity and
nonselective beta-blocking activity, is used in the treatment of
hypertension.
5. Propranolol, a nonselective beta-antagonist, is used for prophylaxis of
angina pectoris, supraventricular and ventricular dysrhythmias and migraine
headache. It is also used as an anti-hypertensive, a negative inotropic
agent in hypertrophic obstructive cardiomyopathy, and a negative
chronotropic agent in anxiety and hyperthyroidism.
6. Beta-1 selective antagonists are used in the treatment of
hypertension,tachyarrhythmias and angina.
a. Metoprolol
b. Betaxolol
c. Atenolol
d. Acebutolol
7. Both beta-1 selective (betaxolol) and nonselective (timolol) blockers
decrease ciliary body production of aqueous humor and may be used in the
topical treatment of glaucoma.
D. Adverse Effects
1. Prazosin can cause sudden syncope with the first dose, orthostatic
hypotension, dizziness, headache, drowsiness, palpitations, fluid retention
and priapism.
2. Phenoxybenzamine can cause orthostatic hypotension, tachycardia,
inhibition of ejaculation, misosis and nasal congestion.
3. Propranolol can cause bradycardia and congestive heart failure, increased
airway resistance, hypertriglyceridemia, blood dyscrasias, psoriais,
depression, hallucinations, and transient hearing loss. Withdrawal from this
drug should be tapered since sudden withdrawal is cardiotoxic.
4. Metoprolol has adverse effects similar to those of propranolol, except that it
is less likely to increase airway resistance.
Assignment Tabulate the effects of adrenergic antagonists and their therapeutic indications.
Assessment A short examination about the topic discussed will be given the following meeting
SUBJECT PHARMACOLOGY 1 TOPIC skeletal muscle relaxants
IG NUMBER IG-PHA-325LC WEEK NO 10
TERM MIDTERM SESSION 17 of 30
DATE PREPARED May 2007 DURATION 1.5 HOURS
SECTION DESCRIPTION
Overview of The students are introduced to the pharmacology of skeletal muscle relaxants
Session
A. Pharmacology
1. The competitive nondepolarizing agents compete with acetylcholine for nicotinic
receptors at the neuromuscular junction. The agents decrease the end plate
potential so that the depolarization threshold is not reached. Competitive
nondepolarizing agents produce a surmountable blockade of neuromuscular
transmission in that adminstration of cholinesterase inhibitors or prejunctional
release of a large quantity of acetylcholine can relieve the blockade. In small
clinical doses and at low frequencies of stimulation, nondepolarizing muscle
relaxants act predominantly at the nicotinic receptor sites to compete with
acetylcholine. In larger doses, these drugs also enter the pore of an ion channel
to cause blockade. Nondepolarizing relaxants may also block prejunctional sodium
channel but not calcium channels. These agents produce a surmountable
blockade.
a. The surmountable block is a consequence of the postsynaptic blockade
produced by these agents as a result of tetanic stimulation, by releasing a
large quantity of acetylcholine, followed by a transient posttetanic
breakthrough or relief of the block.
b. The prinicipal active alkaloid in curare is tubocurarine. A closely related
trimethylate derivative is metocurine. Their most important structural feature
is the presence of tertiary-quaternary amine in which the distance between
the two cations is rigidly fixed at about 14 angstrom, twice the length of the
crirical receptor-binding moiety of acetylcholine.
c. A number of potent analogues have been developed. These include the
structurally similar isoquinolines atracurium, doxacurium, and mivacurium, as
well as the steroid derivative pancuronium, vecuroniumand pipecuronium.
d. The route of elimination is strongly correlated with its duration.
e. All steroidal muscle relaxants are metabolized to their 3-hydroxy, 17-hydroxy
or 3,17-dihydroxy products, mainly in the liver.
f. The 3-hydroxy metabolite may cause prolonged paralysis because it is more
slowly cleared than the parent compound.
g. The intemediate-duration muscle relaxants include vecuronium and
rocuronium and are more dependent on biliary excretion or hepatic
metabolism for their elimination. They are more commonly used than the
long-acting ones.
h. Vecuronium has a steroid nucleus like that of pancuronium differing only in
one nitrogen that is tertiary rather than quaternary.
i. Rocuronium has a pharmacokinetic profile similar to vecuronium.
j. Rapacuronium is the newest neuromuscular blocker and has the most rapid
onset. It is the preferred agent for rapid-sequence induction of anesthesia
and endotracheal intubation.
k. Atracurium is an isoquinoline derivative with many same characteristics as
vecuronium. The main product of metabolism is laudanosine and a related
quaternary acid, neither possess neuromuscular blocking properties.
l. Cisatracurium, one of the stereoisomers of atracurium but is less dependent
on hepatic inactivation, forms less laudanosine, and releases less histamine
from tissue stores.
m. Mivacurium and rapacuronium belong to the short-duration muscle relaxants.
n. The duration of action of mivacurium is prolonged among patients with
impaired renal function.
2. The noncompetitive depolarizing agents desensitize the nicotinic receptors at the
neuromuscular junction. These agents react with nicotinic receptors, decreasing
receptor sensitivity in a manner similar to that of excess released acetylcholine.
They depolarize the excitable membrane for a prolonged period; the membrane
then becomes unresponsive.
a. Noncompetitive agents are slender alipathic molecules.
b. Succinylcholine has a short duration(5-10 minutes) of action compared with
the other neuromuscular blocking agents. This results from its simple ester
functional group, which is rapidly hydrolyzed by plasma and liver
pseudocholinesterases. Its action may be prolonged, however, in patients
with an abnormal genetic variant of pseudocholinesterase, which has only
20% of the activity of the normal pseudocholinesterase.
- Phase I block(depolarizing). Succinylcholine reacts with the nicotinic
receptor to open the channel and cause depolarization at the motor end
plate , and this would spread and depolarize adjacent membranes causing
generalized disorganized contraction of muscle motor units. Succinylcholine
is not metabolized at the synapse resulting to suatined depolarization of
membranes making them unresponsive to additional impulses.
Furthermore, because excitation-concentration coupling requires end plate
repolarization and repetitive firing to maintain muscle tension, a flaccid
paralysis results. Phase I block is augmented, not reversed by
cholinesterase inhibitors.
- Phase II block(desensitizing)- the continued exposure to succinylcholine
results in the decrease of of initial end plate depolarization and the
mambrane becomes repolarized. Despite this repolarization, the membraine
is not easily depolarized because it is desensitized. During the latter part of
phase 2, the characteristics of the blockade are nearly identical to those of
a nondepolarizing block and are reversed by acetylcholinesterase inhibitors.
c. The initial metabolite of succinylcholine, succinylmonocholine is a weaker
neuromuscular blocker, which is further metabolized to succinic acid and
choline.
d. Neuromuscular blockade by both succinylcholine and mivacurium is prolonged
among patients with an abnormal variant of plasma cholinesterase of genetic
origin.
e. Dibucaine number- a test for the ability to metabolize succinylcholine. Under
normal conditions, dibucaine inhibits the normal enzyme about 80% and
abnormal enzyme only 20%.
SECTION DESCRIPTION
Overview of The students are introduced to the pharmacology of Local and General anesthetics
Session
Lesson
Outline GENERAL AND LOCAL ANESTHETICS
General anesthetics
• Induce combined state of analgesia, amnesia, loss of consciousness, inhibition of
sensory and autonomic reflexes, and skeletal muscle relaxation.
• The first scientific demonstration of drug-induced anesthesia during surgery was
in 1846, when William Morton used diethyl ether.
• James Simpson-discovered chloroform in Scotland
• 1790’s- Sir Humphry Davy discovered nitrous oxide
• 1930’s- Intravenous thiopental was introduced
• 1940’s- curare was used in anesthesia to achieve skeletal muscle relaxation
• 1956- the first modern halogenated hydrocarbon, halothane was introduced.
Characteristics
➢ Induce anesthesia rapidly and smoothly
➢ Permit rapid recovery of the patient once administration of the agent ceases
A. Chemistry
STAGES OF ANESTHESIA
• Guedel’s signs- a traditional description of the signs and stages of anesthesia
derives mainly from observation of the effects of diethyl ether, which has a slow
onset of central action owing to its high solubility in blood.
I. STAGE OF ANALGESIA: Analgesia without amnesia. In the later part of stage 1,
both analgesia and amnesia ensue.
II. STAGE OF EXCITEMENT: Delirious and excited but definitely amnesic. Irregular
respiration both in volume and rate, and retching and vomiting can occur.
III. STAGE OF SURGICAL ANESTHESIA: This stage begins with recurrence of
regular respiration and extends to complete cessation of spontaneous respiration.
Four planes of stage III have been described in terms of changes in ocular
movements, eye reflexes, and pupil size, which under specified conditions may
represent signs of increasing depth of anesthesia.
IV. STAGE OF MEDULLARY DEPRESSION: When spontaneous respiration ceases,
stage IV is present. The stage of anesthesia includes severe depression of the
vasomotor center in the medulla as well as the respiratory center. Without full
circulatory and respiratory support, death rapidly ensues.
• Atropine is used to decrease secretions, also dilates pupils.
• Tubocurarine and Succinylcholine are administered to affect muscle tone.
• Opioid analgesics exert depressant effects on respiration.
• The most reliable indications that stage III has been achieved are the loss of
eyelash reflexand establishment of a respiratory pattern that is regular in rate and
depth.
INHALED ANESTHETICS
• Depth of anesthesia is determined by the concentrations of anesthetics in the
central nervous system.
• The concentration of an individual gas in a mixture of gases is proportionate to its
partial pressure or tension.
• BLOOD:GAS PARTITION COEFFICIENT- a useful index of solubility and defines
the relative affinity of an anesthetic for the blood compared to air.
• Desflurane, nitrous oxide, sevoflurane- are not very soluble in blood; have low
blood:gas partition coefficients.
• Methoxyflurane- very soluble in blood.
• Inverse relationship exists between blood solubility of an anesthetic and the rate
of rise in arterial blood. The higher the blood:gas partition coefficient, the slower
the induction of anesthesia.
• AN increase in the inspired anesthetic concentration will increase the rate of
induction of anesthesia by increasing the rate of transfer into the blood.
• Enflurane, isoflurane, and halothane- have relatively slow onset of anesthetic
effect.
• AN increase in pulmonary ventilation is accompanied by only a slight increase in
arterial tension of an anesthetic with low blood solubility or low coefficient but
can significantly increase tension of agents with moderate or high blood solubility.
• Depression of respiration by other pharmacologic agents, including opioid
analgesics, may slow the onset of anesthesia of some inhaled anesthetics if
ventilation is not controlled.
• AN increase in pulmonary blood flow slows the rate of rise in arterial tension,
particularly for those anesthetics with moderate to high solubility. A decrease in
pulmonary blood flow has an opposite effect and increases the rate of rise of
arterial tension of inhaled anesthetics.
• Highly perfused tissues exert greatest influence on arterio-venous concentration
gradient.
• The time to recover from inhalation anesthetics depends on the rate of
elimination of anesthetics from the brain after the inspired concentration of
anesthetic has been decreased.
• Inhaled anesthetics that are relatively insoluble in blood and brain are eliminated
at faster rates than more soluble anesthetics.
• The “washout” of nitrous oxide, desflurane, and sevoflurane occurs at a rapid
rate, which leads to rapid recovery from anesthetic effects.
• Halothane is approximately twice as soluble in brain tissue and five times more
soluble in blood tha nitrous oxide. Its elimination therfore takes place more
slowly, and recovery from halothane anesthesia is less rapid.
• Clearance of inhaled anesthetics by the lungs into the expired air is the major
route of their elimination from the body
• In terms of the extent of metabolism of inhaled anesthetics the rank order is:
methoxyflurane>halothane>enflurane>sevoflurane>isoflurane>desflurane>nitro
us oxide
B. Pharmacology
1. General anesthetics depress the CNS, producing a reversible loss of consciousness
and loss of all forms of sensation
2. Inhalational anesthetics are absorbed and are primarily excreted through the
lungs. Frequently these drugs are supplemented with analgesics, a skeletal muscle
relaxant, and an antimuscarinic agent
C. Therapeutic indications
1. Inhalational anesthetics are indicated to provide general surgical anesthesia
2. Non volatile anesthetics- indicated to induce drowsiness and provide
relaxation
3. Use of some previously popular volatile anesthetics has been discontinued
because of serious toxicity (chloroform) or because of the flammable and
explosive properties of the compounds (cyclopropane, diethylether)
D. Adverse effects
Depress respiration, circulation, and the CNS
They can decrease hepatic and kidney function (methoxyflurane)
Cause cardiac dysrrhytmia as a result of increased myocardial sensitivity to
cathecolamine (halothane)
General Anesthetics
• Halothane- hepatotoxic
• Enflurane and sevoflurane- nephrotoxic
• Thiopental- commonly sued in the induction of anesthesia
• Propofol (2,6-diisopropylphenol)
• Ketamine- dissociative anesthetic, related to PCP
Local Anesthetics
MOA: BLOCKS Na channels
- Produce reversible loss of sensation
- They do not produce a loss of consciousness
Chemistry
- Hydrophylic aminogroup linked through an ester or amide connecting - group to
a lipophilic armatic moiety
Esters
-Procaine
-Chlorprocaine
-Tetracaine
-has shorter duration of action since they are easily hydrolyzed by plasma esterases
Amides
-Mepivacaine
-Lidocaine
-Edtidocaine
-Prilocaine
-metabolized in the liver and has longer duration of action
Adverse effects
- high plasma concentrations can cause excitation, dizziness, tinnitus, disorientation
and muscle twitching
- Cardiovascular effects include myocardial depression, hypotension, decreases
cardiac output, heart block, bradycardia, ventricular arrhythmia and cardiac
arrest.
SECTION DESCRIPTION
Overview of This topic is intended to discuss the pharmacology of drugs’ use in pain management.
Session
1. To present the pathophysiology of pain
Objectives 2. To identify the drugs used in pain management
3. To elaborate the mechanisms of action of the different classes of drugs used in
pain mangement
4. To discuss the adverse effects associated with the drugs
Pathophysiology
• AFFERENT PAIN TRANSMISSION
A. Peripheral Stimulation
- The first step leading to pain is the activation of nociceptors
- Bradykinins, hydrogen, potassium, prostaglandins, histamine, leukotrienes and
serotonin sensitisize nociceptors.
- Nociception leads to action potentials that are transmitted along nerve fibers to
the spinal cord.
- Somatostatin, CCK,and substance P have been identified as possible
neurotransmitters in afferent nociceptive neurons.
- Substance P plays a role in enhancing the effectiveness of nociceptive
neurotransmitters that promote pain.
- Substance P blockade by capsaicin significantly reduces pain transmission.
- Nociceptive transmission takes place in the A-delta or C-afferent nerve fibers.
- Stmulation of the A-delta fibers evokes sharp, acute, well-localized pain, while
stimulation of C-fibers produces dull, aching and poorly localized pain
Kappa Analgesia
Less respiratory depression than mu
Less intense miosis than mu
Sedation
Reduced gastric motility
Dysphoria
Psychotomimetic effects
Delta Analgesia
CLINICAL PRESENTATION
• Pain with a known source is often localized, well described, and relieved with proper
analgesic therapy. Whereas, pain with no obvious origin is odten nonlocalized, ill
defined and not easily treated with conventional analgesics.
PRINCIPLES OF MANAGEMENT
1. Comprehensive Pain Management should determine the characteristics of the
patient’s pain complaint, clinical status and pain management history
a. Assessment of pain complaint should include chronology and symptomatology
of the presenting complaint such as location, onset, duration, intensity, quality,
provocative factors, temporal qualities, severity and pain history.
b. Assessment of clinical status should include the extent of underlying trauma or
disease.
c. Assessment of pain includesdrug allergies, analgesic response, onset, duration
and side effects.
2. Appropriate Pain Management
a. The primary pain goal is to improve patient comfort
b. For acute pain management, improved comfort can aid the healing and
rehabilitation processes
c. For chronic pain, the objective is to break the pain cycle.
NARCOTIC ANALGESICS
- include the opioid drugs. Because of their abuse potential, opioids are classified as
controlled drugs.
Mechanism of action
- endogenous opiates afford the body self-pain relieving mechanisms. These
endogenous peptides include the endorphins, enkephalins and dynorphins.
- Exogenous opiates are classified as agonists, antagonists and mixed opiates.
- The specific mechanism of opiate agonist is alteration of the effects of nociceptive
neurotransmitters, possibly nE and 5-HT.
Clinical Uses
- management of moderate to severe painof somatic or visceral origin
- the appropriate route of administration is individualized.
- Oral adminstration is preferred particularly for patients with chronic, stable pain.
- Intramuscular and subcutaneous are very commonly used in the post operative
period
- Intravenous for stable, chronic pain
- Epidural and intrathecal administrations are used for acute postoperative pain and
early management of chronic cancer pain.
- Rectal route for patients who cannot take oral narcotics
- Transdermal route is an alternative for patients with chronic pain and cannot
afford to take oral narcotics
Adverse Effects
- constipation
- nausea and vomiting
- sedation
- respiratory depression
- anticholinergic effects
- hypersensitivity
- CNS excitation
Drug Interactions
a. additive effects with CNS depressants, alcohol, BZD
b. Meperidine can cause severe excitation, sweating, rigidity and hypertension in
patients taking MAOIs.
Indications
a. Meperidine is used in severe pain. May precipitate tremors, myoclonus and
seizures
b. Fentanyl is used in severe pain
c. Methadone is effective in severe chronic pain, sedation can be a major problem
d. Propoxyphene is effective in moderate pain, it has weak analgesic properties and
most effective when used with nonnarcotic analgesics
e. Pentazocine is the 3rd line agent for modaerate to severe pain. May precipitate
withdrawal in opiate-dependent patients
f. Butorphanol and nalbuphine is the 2nd line agent for moderate to severe pain
g. Tramadol-maximum dose is 400mg/day
h. Codeine is used in moderate pain, has weak analgesic properties.
Morphine is the drug of choice in acute severe pain.
Assessment
A short exam regarding pain mangement next meeting.
SUBJECT PHARMACOLOGY 1 TOPIC Analgesics-2
IG NUMBER IG-PHA-325LC WEEK NO 11
TERM MIDTERM SESSION 20 OF 30
DATE PREPARED May 2007 DURATION 1.5 HOURS
SECTION DESCRIPTION
Overview of This topic is intended to discuss the pharmacology of drugs’ use in pain management.
Session
Lesson ANALGESICS
Outline II- NONNARCOTIC ANALGESICS
Nonsteroidal Anti-inflammatory Drugs (NSAIDs)
A. Overview
1. The inflammatory response is complex, involving the immune system and the
influence of various endogenous agents, including prostaglandins, bradykinin,
histamine, chemotactic factors, and superoxide free radical formed by the
action of lysozomal enzymes.
2. Aspirin, other salicylates, and newer drugs with diverse structures are referred
to as NSAIDs to distinguish them from the anti-inflammatory glucocorticoids.
NSAID are used to suppress the symptoms of inflammation associated with
rheumatic disease. Some are also used to relieve pain and fever.
B. Mechanism of actions
1. Anti-inflammatory effect
a. The anti-inflammatory effect of Nsaids is due to the inhibition of the enzyme
prostaglandin H synthase (cyclooxygenase, or COX), which converts arachidonic
acid to prostaglandins, and to TXA2 and prostacyclin. Most of the current
NSAIDs inhibit both COX-1 and COX-2 to about the same extent; compounds
more specific for COX-2 are in clinical trials.
b. Aspirin irreversibly inactivates COX-1 and COX-2 by acetylation of a specific
serine residue. This is an important distinction compared to other NSAIDs,
which reversibly inhibit COX-1 and COX-2.
c. NSAIDs have no effect on lipoxygenase and therefore do not inhibit the
production of leukotrienes.
c. Additional anti-inflammatory mechanisms may include interference with the
potentiative action of other mediators of inflammation, modulation of T-cell
function, stabilization of lysosomal membranes, and inhibition of chemotaxis.
2. Analgesic effect
a. The analgesic effect of NSAIDs is thought to be related to the peripheral
inhibition of prostaglandin, but it may also be due to the inhibition of pain
stimuli at a subcortical site.
b. NSAIDs prevent the potentiating action of prostaglandins on endogenous
mediators of peripheral nerve stimulation.
3. Antipyretic effect. The antipyretic effect of NSAIDs is believed to be related to
inhibition of the interleukin-1- and interleukin-6-induced production of prostaglandins
in the hypothalamus and the resetting of the thermoregulatory system, leading to
vasodilation and increased heat loss.
Therapeutic uses
1. Inflammation
a. NSAIDs are first-line drugs used to arrest inflammation and the accompanying pain
of rheumatic and nonrheumatic diseases, including rheumatoid arthritis, juvenile
arthritis, osteoarthritis, psoriatic arthritis, ankylosing spondylitis, Reiter’s syndrome,
and dysmennorhea. Pain and inflammation of bursitis and tendonitis also response to
NSAIDs.
b. NSAIDs do not significantly reverse the progree of rheumatic disease, but rather,
they slow destruction of cartilage and bone and allow patients increased mobility and
use of their joints.
c. These agents are administered chronically in doses well above those used for
analgesia and antipyresis and, at those dosages, are more likely to cause adverse
druf effects. Drug selection is generally dictated by the patient’s ability to tolerate the
adverse effects, and the cost of the drugs.
2. Analgesia. NSAIDs alleviate mild-to-moderate pain. They are less effective than
opioids, and they are more effective against pain associated with integumental
structures (Pain of muscular and vascular origin, arthritis, and bursitis) than with that
associated with the viscera.
3. Antipyresis. NSAIDs reduce elevated body temperature with little effect on normal
body temperature.
Pharmacologic properties
1. Salicylates are weak organic acids; aspirin has a pK of 3.5
2. These agents are rapidly absorbed from the intestine as well as from the
stomach, where the low pH favors absorption. The rate of absorption is increased
with rapidly dissolving or predissolved dosage forms.
3. Salicylates are hydrolyzed rapidly by plasma and tissue esterases to acetic acid
and the active metabolite salicylic acid. Salicylic acid is more slowly oxidized to
gentisic acid and conjugated with glycine to salicyluric acid and to ether and an
ester glucuronides.
4. Salicylates have a t1/2 of 3-6 hours after acute administration. Chronic
administration of high doses or toxic overdose increases the t1/2 to 15-30 hours
because the enzymes for glycine and glucuronides conjugation became saturated.
5. Unmetabolized salicylates are excreted by the kidney. If the Urine pH is increased
above 8, clearance is increased approximately fourfold as a result of decreased
reabsorption of the ionized salicylate from the tubules.
Therapeutic uses
1. salicylates are used to threat rheumatoid arthritis, juvenile arthritis, and
osteoarthritis, as well as other inflammatory disorders. 5-amino salicylates can be
used to treat Crohn’s disease.
2. Salicylic acid is used topically to treat plantar warts, fungal infections, and corns;
use is based on the destruction of keratinocytes and dermal epithelia by the free
acid.
Adverse effects
1. Gastrointestinal effects
a. Gastrointestinal effects are the most common adverse effects of hig-dose
aspirin use; these effects may include nausea, vomiting, diarrhea or
constipation, dyspepsia, epigastric pain, bleeding and ulceration.
b. These gastrointestinal effects are thought to be due to a direct chemical effect
on gastric cells or a decrease in the production and cytoprotective activity of
prostaglandins, which leads to gastric tissue susceptibility to damage by
hydrochloric acid.
c. The gastrointestinal effects may contraindicate aspirin use in patients with an
active ulcer. Aspirin may be taken with prostaglandins to reduce gastric
damage.
d. Substitution of enteric coated or timed release preparations, or the use of
nonacetylated salicylates, may decrease gastric irritation. Gastric irritation is not
prevented by using buffered tablets.
2. Hypersensitivity (intolerance)
a. Hypersensitivity is relatively common with the use of aspirin; hypersensitivity
results in rash, bronchospasm, rhinitis, edema, or an anaphylactic reaction with
shock, which may be life-threatening. The incidence of intolerance is highest in
patients with asthma, nasal polyps, recurrent rhinitis, or urticaria. Aspirin should
be avoided in such patients.
b. Cross-hypersensitivity may exist to other NSAIDs and to the yellow dye
tartrazine, which is used in many pharmaceutical preparations.
c. Hypersensitivity is not associated with sodium salicylate or magnesium
salicylate.
d. The use of aspirin and other salicylates to control fever during viral infections in
children and adolescents is associated with an increased incidence of Reye’s
syndrome, an illness characterized by vomiting, hepatic disturbances, and
encephalopathy that has a 35& mortality rate. Acetaminophen is recommended
as a substitute for children with fever of unknown etiology.
3. Miscellaneous adverse effects and contraindications
a. salicylates occasionally decrease the glomerular filtration rate, particularly in
patients with renal insufficiency.
b. Salicylates occasionally produce mild hepatitis, usually asymptomatic,
particularly in patients with systemic lupus erythematosus, juvenile, or adult
rheumatoid arthritis, or rheumatic fever.
c. These agents prolong bleeding time. Aspirin irreversibly inhibits platelet type
1 and type 2 cyclooxygenase and TXA2 production, suppressing platelet
adhesion and aggregation. The use of salicylates is contraindicated in patients
with bleeding disorders, such as hypothrombinemia, hemophilia, hepatic
disease, and vitamin K deficiency and use should be avoided in patients
receiving anticoagulants such as coumarin and heparin.
d. Salicylates are not recommended during pregnancy; they may induce
postpartum hemorrhage and lead to premature closure of the fetal ductus
arteriosus.
Drug interactions
1. The action of anticoagulants may be enhanced by their displacement by aspirin
from binding sites on serum albumin. Aspirin also displaces tolbutamide,
phenytoin, and other drugs from their plasma protein binding sites.
2. The hypoglycemic action of sulfonylureas may be enhanced by displacement from
their binding sites on serum albumin or by inhibition of their renal tubular secretion
by aspirin.
3. Usual analgesic doses of aspirin decrease renal excretion of sodium urate and
antagonize the uricosuric effect of sulfinpyrazone and probenecid; aspirin is
contraindicated in patients with gout who are taking uricosuric agents.
4. Antacids may alter the absorption of aspirin.
5. Aspirin competes for tubular reabsorption with penicillin G and prolongs its half-
life.
6. Corticosteroids increase renal clearance of salicylates.
7. Alcohol may increase gastrointestinal bleeding when taken with aspirin.
Toxicity
1. In adults, salicylism occurs as initial sign of toxicity after aspirin or salicylate
oversdose or poisoning.
2. In children, the common signs of toxicity include hyperventilation and acidosis, with
accompanying lethargy and hyperpnea.
3. Disturbance of acid base balance results in metabolic acidosis in infants and young
children and in compensated respiratory alkalosis in older children and adults.
Salicylate toxicity initially increases the medullary response to carbon dioxide, with
resulting hyperventilation and respiratory alkalosis. In infants and young children,
increases in lactic acid and ketone body production result in a metabolic acidosis.
With increased severity of toxicity, respiratory depression occurs, with
accompanying respiratory acidosis.
4. The uncoupling of oxidative phosphorylation by aspirin results in hyperthermia and
hypoglycemia, particularly in infants and young children. Nausea, vomiting,
tachycardia, hyperpnea, dehydration and coma may develop.
5. Treatment includes correction of acid base disturbances, replacement of electrolytes
and fluids, cooling, alkalinization or urine with bicarbonate to reduce salicylate
reabsorption, forced diuresis and gastric lavage or emesis.
Therapeutic effects:
a. The peripherally acting, nonnarcotic analgesics have several effects in common:
1. they are antipyretic
2. they are anti-inflammatory(except acetaminophen)
3. there is a ceiling effect to analgesia
4. they do not cause tolerance
5. they do not cause physical or psychological dependence
b. The efficacy of nonnarcotics is compared to aspirin. Several NSAIDs have shown a
superior effect to 650 mg of aspirin:
1. Diflunisal
2. Ibuprofen
3. Naproxen
4. Ketoprofen
Clinical Uses:
a. Generally, non-narcotic analgesics are used orally to manage mild to moderate
pain.
b. The NSAID ketorolac is administered intramuscularly and is useful in moderate to
severe pain, particularly in cases of drug addicts, excessive narcotic sedation and
respiratory depression.
c. Patients may vary in their response and tolerance to non-narcotic analgesics.
d. Several drugs such as diflunisal, naproxen, celecoxib, valdecoxib, leflunomide and
etanercept have long half-lives and, therefore,may be administered less frequently.
Rofecoxib, with a 17-hour half-life may be administered once daily
Adverse Effects:
a. Gastrointestinal effects: Most nonnarcotic analgesics cause GI symptoms
secondary to prostaglandin inhibition. Etanercept has not been associated with GI
side effects, whereas leflunomide has been associated with nausea and diarrhea.
1. Dyspepsia, ulceration, bleeding and perforation may occur
2. Patients most predisposed to severe GI effects include the elderly, those with
history of ulcers or chronic disease, and those who smoke or use alcohol
3. To minimize GI effects, the lowest possible analgesic dose should be used.
Aspirin, available as enteric-coated may minimize GI upset. Combination therapy
with GI protectant may be needed.
b. Hematological effects. Most nonnarcotic analgesics inhibit platelet aggregation.
The effect is produced by the reversible inhibition of prostaglandin synthase.
Aspirin is an irreversible inhibitor. Acetaminophen lack anti-platelet activity. Use of
anticoagulants is relatively containdicated in combination with aspirin or NSAIDs.
c. Renal effects. NSAIDs produce renal dysfunction. Selective COX-2 inhibitors are
not devoid of this effect.
- the mechanism of NSAID-induced renal dysfunction includes prostaglandin
inhibition, interstitial nephritis, impaired renin secretion, and enhanced tubular
water/sodium reabsorption.
- It is manifested by oliguria with sodium and water retention. This effect is
reversed upon discontinuation of NSAID therapy.
d. Miscellaneous effects
- Acetaminophen- hepatotoxicity at normal doses in patients with liver
disease and are chronic alcoholics
- Leflunomide- transient elevations in liver function tests, weight loss,
alopecia, rash an anemia. It is a category X drug.
- Aspirin- hypersensitivity reactions
- Etanercept- sepsis and reactions at injection sites
- Celecoxib- the only selective COX-2 inhibitor contraindicated with
sulfonamide allergy
Drug Interactions
a. Oral anticoagulants. Aspirin should be avoided in anticoagulated patients. Aspirin
inhibits platelet function and can cause gastric mucosal damage. Choline
magnesium trisalicylate or acetaminophen should be used if a nonnarcotic is needed
in an anticoagulated patient.
b. Methotrexate. Salicylates may enhace toxicity of methotrexate. The primary
mechanism is blockade of methotrexate renal tubular secretion by salicylates
resulting to pancytopenia or hepatotoxicity.
SECTION DESCRIPTION
Overview of This lesson and the succeeding topics are intended to introduce the pharmacology of
Session drugs that act on the central nervous system. In this lesson, the pharmacology of the
drugs used in the treatment of schizoprenia would be introduced.
SCHIZOPRENIA
• A disorder represented by a heterogenous syndrome of disorganized and bizarre
thoughts, delusions, hallucinations, inappropriate affect, and impaired social
functioning.
PATHOPHYSIOLOGY
Multifactorial, multiple pathophysiologic abnormalities may play a role in producing
the similar but varying clinical phenotypes.
A. NEUROTRANSMITTER CHANGES
• The most common pathophysiologic theories associated with the etiology of
schizoprenia have involved the dopaminergic system.
• Antagonism of DA receptors lead to schizoprenia
• Homovanillic acid (HVA)- concentrations in the CSF are correlated with
dopaminergic turnover in the synapse, and therefore HVA formation. A
hyperactive DA system should result in increased DA release from the presynaptic
termianl and increased metabolite formation.
• Da hypothesis may be more applicable to “neuroleptic-responsive psychosis.”
• Glutamatergic dysfuction has been suggested as the etiology in schizoprenia.
• The glutamatergic system is one of the most widespread excitatory
neurotransmitter systems in the brain.
• Dopaminergic innervation from the ventral striatum decreases the limbic system’s
inhibitory activity; thus, dopaminergic stimulation increases arousal.
• Glutamatergic deficiency produces symptoms similar to those seen in
schizoprenia.
• Serotonergic receptors are present on Daergic axons, and it is known that
stimulation of these receptors will decrease DA release, at least in the striatum.
• Atypical antipsychotics with potent 5-HT2 receptor antagonist effects have been
shown to reverse worsening of symptomatology induced by 5-HT agonist in
schizoprenic patients.
Increased concentrations of NE have been observed in limbic structures of patients
with chronic paranoid schizoprenia, but not with other subtypes.
• The dysregulation hypothesis maintains that aberrant homeostatic control
mechanisms cause erratic neurotransmission; that is, the homeostatic
mechanisms that control the relationships among neurotransmitter syntheis,
release, reuptake, metabolism, activity at receptors, and second messenger
systems are defective. This lack of hemeostais is manifested in dysfunction of
several processes including basal neurotransmission, biological rhythm, and
return to basal rate after pertubations on the system.
• The primary pathophysiologic abnormality in schizoprenia may occur in one of the
neurotransmitters(dopamine, glutamate, serotonin), with permissive properties of
other neurotransmitters.
Dopaminergic Tracts and Effects of Dopamine Antagonists
1. Nigrostriatal Tract
a. Origin. Substancia nigra (A9 area)
b. Innervation. Caudate nucleus, putamen
c. Function. Extrapyramidal system, movement
d. Dopamine Antagonist effect. Movement disorders
2. Mesolimbic Tract
a. Origin. Midbrain ventral tegmentum (A10 area)
b. Innervation. Limbic areas (amygdala, olfactory tubercle, septal nuclei), cingulate
gyrus
c. Function. Arousal, memory, stimulus processing, motivational behavior
d. Dopamine Antagonistic effect. Relief of psychosis
3. Mesocortical Tract
a. Origin. Midbrain ventral tegmentum (A10 area)
b. Innervation. Frontal and prefrontal lobe cortex
c. Function. Cognition, communication, social function, response to stress
d. Dopamine Antagonistic effect. Relief of psychosis
4. Tuberoinfundibular
a. Origin. Hypothalamus
b. Innervation. Pituitary gland
c. Function. Regulates prolactin release
d. Dopamine Antagonistic effect. Increased prolactin secretion
B. STRUCTURAL CHANGES
• Increased ventricular size, particularly in the third and lateral ventricles.
• Decrease in brain size
• Decreased cortical thickness
• Increased neuronal density
C. GENETICS
• If both parents have schizoprenia, the risk of producing a schizoprenic offspring
increases to approximately 40%
D. NEURODEVELOPMENTAL CHANGES
• This model proposes that genetic predisposition exists for schizoprenia and that
an unknown in utero disturbance occurs, probably in the second trimester of
pregnancy.
• Schizoprenic lesions are characterized by abnormalities in cell shape, position,
symmetry, connectivity and dysfunctional brain circuits.
• Gliosis, or proliferation of glial cells, is thought to occur as compensatory change
in degenerative brain disorders.
CLINICAL PRESENTATION
Schizoprenia is the most common form of functional psychosis. It is a chronic disorder
of thought and affect with the individual having a significant disturbance in
interpersonal relationships and ability to function in society on a daily basis.
Characteristic symptoms: Two or more of the following, each persisting for a
significant portion of at least a 1-month period:
1.Delusions
2.Hallucinations
3.Disorganized speech
4.Grossly disoganized or catatonic behavior
5.Negative symptoms
Note: Only one criterion is required if delusions are bizarre, if hallucinations consist
of a voice keeping a running commentary on the person’s behavior, or if there are
two or more voices conversing with each other.
Duration: Continuous signs of the disorder for at least 6 months. This must include at
least 1 month of symptoms fulfilling criterion. This 6 months may include prodromal
or residual symptoms.
DESIRED OUTCOME
• Pharmacotherapy is the mainstay in the treatment of schizoprenia, and it is
essentially impossible in most patients to implement effective psychosocial
rehabilitation programs in the absence of antipsychotic treatment.
ANTI-Psychotic Agents
HISTORY
Reseprine and Chlorpromazine- first drugs found to be useful in schizoprenia.
PHARMACOLOGY
Mechanisms of Action of Antipsychotics
• Strongly block postsynaptic D2 receptors in the central nervous system, especially
in the mesolimbic frontal system
• Change the amount of homovanillic acid(HVA), a metabolite of dopamine in the
cerebrospinal fluid, plasma and urine
• Atypical antipsychotics do not only block dopamine receptors but show strong
antagonistic effect at serotonin receptors.
• Typical or Tradition AP medications are putative dopaminergic antagonists, with
an affinity for D2 receptors that is greater than fo D1.
• Typical APs affect other neurotransmitter receptor systems, including blockade of
muscarinic, alpha1 adrenergic and histaminic receptors.
• As a rule, the lower potency APs are less specific for DA receptors and block other
receptors as well.
ATYPICAL ANTIPSYCHOTICS
MOA: Exact mechanisms are unknown. The mechanisms may be related to one or
more of the following pharmacodynamic effects: relative D1, D4 and D5 specificity,
relative selectivity for limbic dopaminergic receptors; 5-HT2, 5-HT6 and 5-HT7
antagonism; or alpha-1 adrenergic antagonism.
• Blockade of 5-HT2 receptors on presynaptic neurons on the striatum may lead to
increased Dopamine release, and therefore, less EPS.
• Atypical antipsychotics, with the exception of clozapine(CLZ) and sertindole
because of the associated side effects, have become the agents of choice in the
pharmacologic treatment of schizoprenia.
• Olanzapine(OLZ), Risperidone(RSP), quetiapine, sertindole and ziprasidone- have
superior efficay in the treatment of negative symptoms.
• Produce fewer or no acutely occurring extrapyramidal effects.
• They are described to be more effective, produce no tardive dyskinesia and does
not have much effect on serum prolactin.
• Clozapine- the prototypical agent
• Risperdone- low incidence of EPS at low to moderate doses. Effectivity is uperior
compared to haloperidol in the treatment of the negative symptoms.
• Olanzapine- has uperior efficacy in the treatment of the negative symptoms and
has very low incidence of extrapyramidal side effects.
• Quetiapine- minimal effect on negative symptoms
• Sertindole/Ziprasidone- has superior efficacy compared to typical antipsychotics,
with less Eps when used in doses 12 to 20 mg daily.
Typical/Traditional Antipsychotics
MOA: Putative Dopamiergic antagonists, with greater affinity to D2 receptors than D1.
• The primary therapeutic effects are thought to occur in the limbic system,
including ventral striatum, whereas, EPS are thought to be related to DA blockade
in the dorsal striatum.
• Tolerance develops but are less common.
• In the tuberoinfundibular system, APs block prolactin inhibitory factor, which is
DA acting at D2 sites.
• Side effects include dry mouth, constipation, sinus tachycardia, and orthostatic
hypotension, seen more commonly on low-potency typical APs.
A. Phenothiazines
- must have a nitrogen-containing substituent on the nitrogen-containing side-
chain substituent on the ring nitrogen which is responsible for its antipsychotic
property
- Phenothiazines in which the ring and side-chain nitrogen are separated by a
two-carbon chain have only antihistaminic or sedative property.
a. Alipathic (Chlorpromazine, CPZ) and piperidine (Thioridazine) derivatives are
the least potent
b. Piperazine derivatives are more effective even at lower doses. This group
includes trifluoperazine, perphenazine and fluphenazine.
B. Thioxanthenes
- exemplified by thiothixene
- slightly less potent than phenothizine analogues
- lack the ring nitrogen of phenothiazines and have a side chain attached to the
double bonds.
C. Butyrophenones
- chemically unrelated to phenothiazines but have similar activity
- haloperidol is the most widely used
- diphenylbutylpiperidines are closely related compounds
• they are more potent and have fewer autonomic effects.
Effects of Receptor Blockade
SECTION DESCRIPTION
Overview of This subtopic of antipsychotics is intended to discuss the pharmacokinetics and
Session physiologic effects of agents that are used in the treatment of schizoprenia.
Lesson Pharmacokinetics
Outline A. Absorption and Distribution
• Readily but incompletely absorbed and undergoes significant first-pass
metabolism
• Oral doses of chlorpromazine and thioridazine- 25 to 35% bioavailable
• Oral dose of haloperidol- 65% bioavailable
• Most antipsychotics are highly lipid-soluble and protein-bound
• They have large volumes of distribution since thay are sequestered in lipid
compartments of the body and have high affinity for selected neurotransmitter
receptors in the CNS, and have generally much longer clinical duration of action
than would be estimated fromtheir plasma half-lives.
B. Metabolism
• Most agents are completely metabolized by a variety of processes.
• Mesoridazine- major metabolite of thioridazine, which is more potent than the
parent compound and accounts for most of the effect.
C. Excretion
• They are excreted unchanged, because they are almost completely metabolized
to polar substances.
Psychological effects
• Most agents cause unpleasant subjective effects in nonpsychotic individuals; the
combination of sleepiness, restlessness, and autonomic effects create experiences
unlike those associated with more familiar sedatives or hypnotics.
Neurophysiologic effects
• Hypersynchrony may be focal or unilateral
Endocrine effects
• Amenorrhea-galactorrhea, false positive pregnancy tests, and increased libido
have been reported in women, whereas, men have experienced decreased libido
and gynecomastia.
Cardiovasular effects
• Orthostatic hypotension and high resting pulse rate frequently result from use of
the high-dose phenothiazines.
• Mean arterial pressure, peripheral resistance and stroke volume are decreased,
and heart rate is increased.
• Abnormal ECG with thioridazine. Changes include prolongation of the QT interval,
and abnormal St segment and T waves, the latter being rounded, flattened or
notched.
• Prolonged Qt interval with increased risk of arrhythmias- Sertindole
• Ziprasidone- risk of QT prolongation
Ophthalmologic Effects
• Impairment in visual accommodation results from paresis of ciliary muscles, an
Ach effect
• Photophobia
• May be managed using pilocarpine
• CPZ-opaque deposits in cornea and lens
• TRD- retinitis pigmentosis
Hematologic effects
• Transient leukopenia
• CLZ-agranulocytosis
Genitourinary system
• TRD-erectile dysfunction and impotence
• OLZ/quetiapine- can be used to prevent sexual dysfunction
• Sertindole- reduced ejaculatory volume
Dermatologic system
• Allergic reaction are rare
• Phenothiazine- absorbs UV light and energy, resulting in the formation of free
radicals, which can have damaging effects on skin.
• Blue-gray or purplish skin coloration with lens/corneal pigmentation-CPZ
INDICATIONS
• Schizoprenia is the primary indicationfor these drugs
• They are also indicated for schizoaffective disorders
• The manic episode in the bipolar disorder often requires treatment with
antipsychotic drugs, though lithium or VPA, supplemented with high-potency
BZD may suffice in milder cases.
• Olanzapine- acute phase of mania; shows antidepressant effect in schizoprenia
• Tourette’s syndrome
• Senile dementia of the Alzheimer type
• With antidepressants- to control agitation or psychosis in depressed patients
• OLZ, RSP, Quetiapine, Aripiprazole- better tolerated compared to other agents
• Ziprasidone- may be used in some cases of anxiety.
Non-psychiatric indications
• Antipsychotics, except thioridazine- have strong antiemetic property due to their
ability to block the dopamin receptor both centrally(CRTZ) and
peripherally(stomach).
• Prochlorphenazine and benzquinamide- marketed solely as antiemetics
• Phenothiazines with shorter side chains- have H1 receptor blocking abilities
preventing pruritus. In the case of promethazine, as preoperative sedatives.
• Droperidol(butyrophenone) + fentanyl= neurolepthanesthesia
DRUG INTERACTIONS
• With Lithium- safe, but irreversible encepalopathies were reported.
• With cigarette smoking- increased AP clearance
• With ACE inhibitors- additive hypotensive effects
Assessment Short Quiz regarding the topic discussed will be given the following meeting
SUBJECT PHARMACOLOGY 1 TOPIC MOOD DISORDERS-1
IG NUMBER IG-PHA-325LC WEEK NO 14
TERM FINALS SESSION 23 of 30
DATE PREPARED May 2007 DURATION 1.5 HOURS
SECTION DESCRIPTION
Overview of This lesson is intended to discuss the pharmacology of drugs used in the treatment of
Session mood disorders.
• Affective disorders
• Characterized into two major disturbance in mood:
- Bipolar
- Depressive
• Mood- defined as pervasive and sustained emotion that in extreme, markedly
affects the person’s perception of the world and the ability to adequately function
in the society.
• Bipolar disorders refer to patients who have episodes of mania and/or hypomania
usually alternating with depression episodes.
• Patients with depressive disorders do not have episodes of mani or hypomania.
EPIDEMIOLOGY
• Depression is two or three times as frequent in females than in males.
• It can occur at any age, but major incidence occurs at ages between 25-44 years
old.
• Depressive disorders and suicide tend to cluster in families, and first-degree
relatives of patients with depression are one and a half to three times more likely
to develop depression than normal subjects.
ETIOLOGY
• Alterations in brain monoamine neurotransmitters: Norepinephrine, serotonin and
dopamine play a major role in depressive disorders.
• Other factors include stressful events, medical illness an monoamine-depleting
drugs(reserpine).
PATHOPHYSIOLOGY
CLINICAL PRESENTATION
• Major depressive disorder is characterized by one or more episodes of major
depression.
• A major depression is characterized by five or more of the following symptoms for
2 weeks and represent a change in functioning, characterized by either depressed
mood or loss of interest/pleasure:
1. Depressed mood, most of the day, nearly everyday
2. Markedly diminished interest or pleasure in all, or almost all of activities
3. Significant weight loss or weight gain, or decrease or increase in appetite
4. Insomnia or hypersomnia nearly everyday
5. Observable psycomotor agitation or retardation everyday
6. Fatigue or loss of energy nearly everyday
7. Feelings of worthlessness or inappropriate guilt
8. Alogia
9. Recurrent thoughts of death and suicidal ideation
TREATMENT
Goals of Treatment:
a. to reduce symptoms of depression
b. to facilitate the person’s return to a premorbid level
General Approach
• Melancholic depression is characterized by a nearly complete absence of capicity
for pleasure, diurnal mood swings, early morning awakening, psychomotor
disturbances, excessive guilt and weight loss.
• A preferential response to Monoamine Oxidase Inibitors has been reported in
patients with atypical depression.
• Atypical depression is manifestion by any two or more of the following symptoms:
4. Weight gain or increase in appetite
5. Hypersomnia
6. Heavy feelings in arms and legs
7. Interpersonal rejection sensitivity
• Psychotically depressed individuals generally require either ECT or combination
therapy with an antidepressant plus an antipsychotic agent.
NON-Pharmacologic Therapy
• Psychotherapy is employed whenever the patient is able and willing.
• Psychotherapy alone is not recommended for the acute treatment of patients with
severe and/or psychotic major depressive disorder.
• Electroconvulsive Therapy (ECT) is a safe and very effective treatment for certain
severe mental illnesses. It is apllied when rapid response is needed.
• ECT is effective for all subtypes of major depressive disorder as well as other
selected psychiatric illnesses.
• Adverse effects of ECT include cognitive dysfunction, cardiovascular dysfunction,
prolonged apnea, treatment-emergent mania, headache, nausea and muscle
aches. Cognitive changes include confusion immediately after the seizure, and
retrograde and anterograde memory disturbance. Most cognitive disturbances are
transient.
• The light therapy is generally well-tolerated with minor visual complaints. Patients
undergoing light therapy should undergo periodic eye examinations.
Pharmacologic Therapy
Antidepressants can be classified in several ways. One approach is by hemical
structure, and another is by presumed mechanism of antidepressant activity.
ADVERSE EFFECTS
VENLAFAXINE
• The most commonly reported unwanted effects include nausea, constipation,
somnolence, dry mouth, dizziness, nervousness, sweating, asthenia, abnormal
ejaculation/orgasm, and anorexia.
• The effects are dose-related.
• It may also cause diastolic hypertension. Blood pressure should be monitored.
TRIAZOLOPYRIDINES
• Trazodone and nefazodone- produce minimal anticholinergic effects or 5-HT
agonist side effects, but can cause orthostatic hypotension.
• Trazodone- sedation, cognitive slowing, and dizziness are the most frequent dose-
limiting side effects. Priapism is observed in some patients.
• Nefazodone- light-headedness, dizziness, orthostatic hypotension, somnolence,
dry mouth, nausea and asthenia
AMINOKETONE
• Bupropion- nausea, dizziness, tremor, insomnia, vomiting, constipation, dry
mouth and skin reactions. Dose-dependent toxicity is manifested by seizures.
Pharmacokinetics
• Generally, TCAs are rapidly absorbed after oral administration. Bioavailability is
low as a result of the first-pass effect. They have a large volume of distribution
and concentration in the brain and cardiac tissues in laboratory animals. The
major metabolic pathways are demethylation, aromatic and alipathic
hydroxylation, and glucoronide conjugation.
• Fluoxetin has an elimination half-life of 2 to 3 days. The single-dose half-life of
norfluoxetine, the active metabolite is 7 to 9 days. Paroxetine and sertraline have
half-lives of 24 hours.
• Sertraline- has an active metabolite but it contributes minimally to pharmacologic
effects.
• Citalopram- half-life is 30 hours
• The SSRIs except fluvoxamine and citalopram, are extensively bound to plasma
proteins (94-95%).
• Mirtazapine-undergoes biotransformation via demethylation and hydroxylation
followed by glucoronide conjugation. CYPIA2 and CYPIID6 are responsible for the
formation of hydroxyl metabolite, while the CYPIIA4 may be responsible for the
formation of the N-desmethyl and N-oxide metabolite.
Altered Pharmacokinetics
• Factors that influence TCA plasma concentrations:
A. Disease states, genetics, age, cigarette smoking, and concurrent drug
administration.
B.Renal failure does not alter nortriptyline concentration but allows accumulation
of metabolite resulting to enhanced sensitivity to the drug
C. TCA plasma concentrations are increased in elderly patients
• Factors affecting half-lives of SSRI:
A. Liver cirrhosis increases half-lives of fluoxetine and norfluoxetine
B.Hepatic diseases provide a twofold increase in the plasma concentration of
paroxetine
C. Half-life of sertraline is 2.5 times greater in patients with mild stable cirrhosis.
D. With renal impairment, two to four-fold increase in paroxetine plasma
concentrations was observed.
E. Plasma concentrations increase with age.
• Factors that affect the AUC of Triazolopyridines:
A. Liver cirrhosis increases AUC of nefazodone and trazodone
• Factors affecting concentrations of Aminoketones:
A. Accumulation of metabolites of bupropion was observed among patients with
liver cirrhosis.
DRUG INTERACTIONS
A. Tricyclic Antidepressants
• TCAs are metabolized in the liver through the cytochrome P450 system, they may
interact with other drugs that modify hepatic enzyme activity or hepatic blood
flow.
• They can displace other drugs bound to proteins.
• They can reverse the hypotensive effects of certain sympatholytic
antihypertensives (guanethidine, methyldopa, clonidine) because of inhibition of
presynaptic uptake of antihypertensive or desensitization of the alpha2-adrenergic
receptor.
• TCAs may increase the vasopressor response to direct-acting sympathomimetics
such as NE, epinephrine and phenylephrine
• TCAs decrease the vasopressor response to indirect-acting sympathomimetics.
• Adverse effects would be additive with anticholinergic, sedative or hypotensive
drugs.
• MAOIs and TCAs may be coadministered safely in refractory patients with
apparent increased efficacy compared with monotherapy. However, severe
reactions and fatalities have occurred including hypertensive crises, hyperpyrexia,
excitation, and convulsion.
B. Selective Serotonin Reuptake Inhibitors
• The very slow elimination of fluoxetine makes it critical to ensure a 5-week
washout after fluoxetine discontinuation before starting a MAOI.
• Serotonin syndrome may occur upon administration with MAOI.
• Fluoxetine + Warfarin = increased risk of bleeding
• Fluoxetine + Carbamazepine/Phenytoin = increased anticonvulsant properties
• Fluoxetine + TCA = increased plasma concentration of TCA
• Paroxetine + Warfarin = increased bleeding time
• Sertralin + Warfarin = increased bleeding time
• Sertraline + Sec. Amine TCA /Carbamazepine = increased concentrations of TCA
and carbamazepine
• Cimetidine + Citalopram = reduced oral clearance of citalopram by 29%
• Citalopram + Fluvoxamine = increased plasma concentrations of citalopram
C. Other Agents
• Mirtazapine + MAOI = should not be permitted
Refractory Patients
• The majority of treatment-resistant depressed patients are likely the result of
inadequate therapy (relative resistance)
• Three primary pharmacologic approaches:
6. Current antidepressant may be stopped and a trial with an unrelated agent
may be initiated
7. The current antidepressant may be augmented by administration of lithium,
liothyronine, CBZ, VPA
8. Use concurrently two different classes of antidepressants
• Never coadminister MAOI with SSRI
Assessment A chapter examination regarding mood disorders will be given the following meeting.
SUBJECT PHARMACOLOGY 1 TOPIC MOOD DISORDERS-2
IG NUMBER IG-PHA-325LC WEEK NO 14
TERM FINALS SESSION 24 of 30
DATE PREPARED May 2007 DURATION 1.5 HOURS
SECTION DESCRIPTION
Overview of This lesson is intended to discuss the pharmacology of agents used in the management
Session of bipolar disorders.
EPIDEMIOLOGY
• Onset of mania after age 50 is rare, its prevalence increases during late
adolescence into early adulthood (15-30 y/o)
Etiology
A. GENETICS
- it has higher genetic risk than major depressive orders
- X chromosome may contribute susceptibilty to bipolar disorders
B. SECONDARY MANIA
- in this case, medications causing mania, agitation, or insomnia should be
discontinued
PATHOPHYSIOLOGY
A. NEUROTRANSMITTER THEORIES
- The monoamine hypothesis suggests a functional alterations of
neurotransmitters- NE, DA and 5-HT
- Excess catecholamines are associated with mania
- Deficiencies in neurotransmitters result to depression
- PERMISSIVE SEROTONIN HYPOTHESIS- low central 5-HT activity in both mania
and depression; 5-Ht plays a critical role in modulating CNS activity.
- Lithium-facilitates the release of 5-HT and increases postsynaptic 5-HT receptor
activity.
- L-tryptophan and fenfluramine(block 5-HT reuptake and enhances 5-HT
release) have been used with other antimanic agents to enhance efficacy.
- One hypothesis of the switch phenomenon from depression to mania involves
the balance of NE to DA. When NE activity is decreased, the DA activity
predominates, and this may switch hypomania to mania.
- Increased dopaminergic activity plays a role in causing hyperactivity and
psychosis associated with the severe stages of mania, and reduced DA activity
may cause depression. In this case, lithium decreases beta-adrenoceptor
number and blocks DA receptor sensitivity.
- Clonidine- an alpha2 agonist decreases NE release and has been used as an
adjuctive agent for agitation and insomnia.
- Antidopamine medications find their application in the acute manic phase of
illness.
- Antidepressants that augment NE, DA, 5-HT activity have been used to treat
bipolar disorders; however, these agents may cause switching to mania.
- GABA-the main inhibitory neurotransmitter in the brain, is involved in the
inhibition of NE and DA. It was hypothesized that the concentration of GABA is
deficient in cases of mood disorders. Several antimania drugs such as Lithium,
CBZ, VPA, clonazepam, and lorazepam enhance GABAergic activity.
- Glutamate- an excitatory neurotransmitter can be decreased by CBZ and
lamotrigine, and topiramate acts a glutamate-receptor antagonist.
- Acetylcholine deficiency or cholinergic-adrenergic imbalance- choline interacts
with catecholamines between IP and IP-choline secondary messenger systems.
In such cases, lithium increases RBC levels of choline, and choline
supplementation has been used in combination with lithium to treat rapid-
cycling bipolar patients.
C. NEUROENDOCRINE THEORIES
- Involves the hypothalamic-pituitary-thyroid axis
- Excessive thyroid activity may precipitate a manic episode by potentiating beta-
adrenergic activity
- Thyroid hormones are used to hasten the response of antidepressant activity
and to convert nonresponders to responders.
- Thyroid supplementations have been used to treat refractory rapid-cycling
bipolar disorders including episodes of mania and hypomania.
- Lithium blocks the release of thyroid hormones and CBZ decreases thyroid
indices.
CLINICAL PRESENTATION
2. MANIC EPISODE
- defined by a distinct period when the mood is abnormally and peristently
elecated, expansive or irritable and is accompanied by impairment in judgment,
and in social and occupational functioning. Acute mania begins abruptly, and
symptoms escalate over several days.
- Seasonal changes, stressors, sleep deprivation, antidepressants, bright light, or
Ect can precipitate a manic episode.
- It is characterized by euphoria, unrealistic grandiosity, flight of ideas, faster
speech, increased energy, psychomotor excitement, decreased need for sleep,
anger orirritability, heightened perceptual acuity and impulsivity.
- The first clue of a manic attack is a decrease in sleep; followed by short
attention span and flight of ideas.
- To consider a manic episode, there should be a persistent elevated mood
(greater than 1 week) with at least three of the following symptoms:
a. inflated self-esteem
b. decreased need for sleep
c. increased talking
d. racing thoughts
e. distractible
f. agitation
g. excessive involvement in pleasurable activities
3. HYPOMANIC EPISODE
- less severe form of mania in which the patient’s mood is elevated, expansive, or
irritable.
- During this episode, aptient may be more productive and creative
- Patients usually find this episodevery desirable because they have heightened
sense of well being, happiness, exhilaration, feel more powerful and productive,
and have increased energy.
- At least four days of abnormal and persistent mood elevation associated with
three of the following symptoms:
a. grandiosity
b. decreased need for sleep
c. increased talking
d. flight of ideas
e. poor attention
f. increased activity
4. MIXED
- bipolar disorder is a dynamic and constantly changing illness, so that
manifestations of either phase may occur simultaneously.
- Known as dysphoric mania
- Possess the criteria for both manic and depressive episode occurring nearly
evryday for at least 1-week.
2. RAPID CYCLING
- characterized by greater than four major depressive or manic episodes in 12
months.
Goals of Therapy
a. to resolve bipolar symptoms
b. to prevent future episodes
c. to minimize adverse drug effects
d. to comply with the treatment
e. to avoid stressors that may precipitate an acute phase
f. to decrease the risk of relapse after an episode has been controlled
• Acute manic episodes can be treated with lithium, CBZ, or VPA along with
adjunctive BZD for anxiety and insomnia.
• Severe manic episodes associated with psychosis and agitation require
antipsychotics alone or with BZDs for sedation along with lithium, CBZ, or VPA
until mania subsides.
• Monotherapy is preferred for long-term maintenance; however, combinations of
drugs may be necessary for patients with mixed episodes, rapid cycling, or those
with partial or nor response to monotherapy.
• Possible combinations include lithium and CBZ, lithium and VPA. The concomitant
use of multiple drugs may be needed to stabilize refractory patients or continuous
cyclers (lithium and CBZ with clonazepam, VPA and antipsychotic with BZD).
• Rapid discontinuation of effective prophylaxis with lithium therapy has been
associated with an increased risk of relapse and possibly a more severe and
nonresponsive type of disorder, gradual tapering should be done.
PHARMACOLOGIC THERAPY
Lithium
• 1970- Lihium carbonate was approved for the treatment of mania
• 1974- approved for bipolar disorder
• Long-term lithium therapy may be more effective in patients with fewer prior
episodes, with a history of euthymia or good functioning between episodes, and
with a family history of bipolar illness with a positive response to lithium.
• Less effective for severe mania with psychotic features, mixed episodes, rapid or
continuous cycling, alcohol and drug abuse, and in organic-induced mood states.
• Lithium
- a monovalent cation and competes with other monovalent and divalent cations
in body tissues and at receptor sites.
- Neuropharmacologic effects of lithium include blockade of the dopamine-
receptor suspersensitivity, decreases beta-adrenoceptor stimulation of
adenylate cyclase, and increases of 5-HT, acetylcholine and GABA function.
- It decreases neurotransmitter activity by acting at the postsynaptic secondary
messenger system
- It decreases receptor-G-protein coupling and decreases phosphoinositide
metabolism
- It is rapidly absorbed, no protein binding, is not metabolized and is excreted
unchaged in the urine and other body fluids.
- Side effects are innocuous and transient. Include SA block, T-wave flattening,
QRS widening, acne, exfoliative dermatitis,hair loss, rash and psoriasis. It is also
associated with hyperparathyroidism, hypothyroidism and weight gain.
- It is extremely toxic if taken in large doses.
- Lithium toxicity is manifested by: fine hand tremor, GI upset, fatigue, confusion,
lethargy, ataxia, dysarthria, nystagmus, emesis, increased deep-tendon
reflexes, muscle fasciculations.
• Valproic acid
- The exact mechanism is unknown but may be related to the inhibition of GABA
metabolism, stimulation of GABA synthesis and release, and augmentation of
postsynaptic inhibitory effect of GABA.
- It has lower incidence of adverse effects and generally well tolerated.
• Carbamazepine
- a dibenzazepine derivative, structurally related to TCAs.
- Blocks reuptake of NE, decreases NE release, increases Ach in striatum,
decreases DA and GABA turnover, bluck calcium influx through the NMDA-
glutamate receptor, and decreases the activity of adenylate cyclase.
- The most common adverse effects is associated with CNS toxicity.
- Neurologic side effects include drowsiness, vertigo, blurred vision, diplopia,
nystagmus, dysarthria, confusion and headache.
- Leukopenias were observed
Assessment
A chapter examination reagrding bipolar disorders next meeting.
SUBJECT PHARMACOLOGY 1 TOPIC ANXIOLYTICS
IG NUMBER IG-PHA-325LC WEEK NO 15
TERM FINALS SESSION 25 of 30
DATE PREPARED May 2007 DURATION 1.5 HOURS
SECTION DESCRIPTION
Overview of This lesson is intended to discuss the pharmacology of drugs used in the treatment of
Session Anxiety that includes anxiolytics and sedative-hypnotics.
Definition
• Anxiety is an emotional state commonly caused by the perception of real or
potential danger that threatens the security of an individual
Epidemiology
• Generally, anxiety disorders are group of heterogenous illnesses that develop
before age 30 and are more common in women and those with a family history of
anxiety and depression.
• Anxiety disorders are chronic in nature, patients are rarely completely symptom-
free.
ETIOLOGY
• Evaluation of anxious patients require a complete physical and mental status
examination, appropriate laboratory tests, toxicologic screen, and a thorough
knowledge of the patients’ medical, psychiatric and drug history.
-Anxiety symptoms are an inherent part of the initial clinical presentation in several
medical disorders, thus complicating the distinction between anxiety disorders and
medical disorders.
Drug-Induced Anxiety
• Two major drug classes that cause anxiety symptoms:
1. CNS stimulants
2. CNS depressants
PATHOPHYSIOLOGY
-Anxiety may result from alterations associated with multiple brain structures and
abnormal function in several neurotransmitter systems including norepinephrine(NE),
gamma-amino butyric acid(GABA), and serotonin(5-HT).
-The key brain structures that play an important role in anxiety states are:
- Amygdala, for assessment of fear and response to danger
- Locus ceruleus, located in the brainstem, is the primary site for NE-containing
site in the brain
- Hippocampus, integrates and consolidates traumatic memory and, along with
entorhinal cortex, contextual fear conditioning.
- Hypothalamus, the principal site for integrating neuroendocrine and autonomic
responses to threat.
NEUROCHEMICAL THEORIES
NORADRENERGIC MODEL
• The autonomic nervous system of anxious patients is hypersensitive and
overreacts to various stimuli with peripheral autonomic hyperactivity.
• Chronic, central noradrenergic overactivity down-regulates alpha2 receptors in
patients.
• Alpha2 receptors are hyperactive in some patients with panic disorder.
• Drugs with anxiogenic effects stimulate locus ceruleus firing and increase
noradrenergic activity.
• Drugs with anxiolytic or antipanic effects inhibit locus ceruleus firing, decrease
noradrenergic activity and block the effect of anxiogenic drugs.
SEROTONIN MODEL
• Serotonin is primarily an inhibitory neurotransmitter that is used by the neurons
originating in the raphe nuclei of the brainstem and projecting diffusely in the
brain.
• The diverse actions of serotonin were attributed to its 14 receptor subtypes.
PEPTIDE THEORY
• Cholecystokinin is an abundant peptide with receptors located through out the
CNS and high densities in the hypothalamus, limbic system, basal ganglia, cortex
and brainstem.
• CCk increases the activity of catecholamines in the locus ceruleus and co-exists
with GABA-producing neurons.
CLINICAL PRESENTATION
The DIAGNOSTIC and STATISTICAL MANUAL OF MENTAL DISORDERS, fourth edition
classifies anxiety disorders into several categories:
PANIC DISORDER
• It begins as a series of spontaneous, unexpected panic attacks, involving an
intense, terrifying fear, similar to that caused by life-threatening danger.
• The unexpected panic attacks are followed by at least one month of persistent
concern about having another panic attack, or a significant behavioral change
related to attacks.
• It usually lasts for no more than 20 to 30 minutes, with the peak intensity of
symptoms within the first 10 minutes.
SOCIAL PHOBIA
• The essential feature is a marked and persistent fear of social performance
situations in which embarrassment may occur
• Exists in two distinct forms:
1. Generalized- related to most social situations
2. Discrete-related to performance and is specific for one or two situations
• The fear and avoidance of the situation must interfere significantly with the
person’s daily functioning.
SPECIFIC PHOBIA
• A marked and persistent fear of a circumscribed object or situation.
TREATMENT
I. Generalized Anxiety Disorder
A. Goals of Therapy:
1. to reduce the severity, duration and frequency of the anxiety symptoms
2. to improve the patient’s overall functioning
3. to prevent anxiety symptoms
4. to improve quality of life
B. Nonpharmacologic Therapy
1. Short-term counseling
2. Stress management
3. Psychotherapy- for encouragement
4. Meditation
5. Exercise
6. Avoidance from caffeine, nonprescription stimulants and diet pills
C. Pharmacologic Therapy
BENZODIAZEPINES
• Drugs of Choice for treating GAD
• The BZ’s are the most effective and safe medication for the amelioration of
anxiety symptoms.
• All BZ’s are equally effective anxiolytics.
• Estazolam, flurazepam, temazepam, quazepam, and triazolam are used
therapeutically as sedative-hypnotics.
• Clonazepam is marketed as antipanic and anticonvulsant
• Midazolam is labeled for preoperative sedation
• Alprazolam is indicated for the traetment of panic disorder with or without
agoraphobia, as well as GAD.
Mechanism of Action:
• Potentiation in the inhibitory effect of GABA.
• Activation of the benzodiazepine receptor in the presence of GABA increases the
frequency of the chloride ion channel opening and increases in the influx of
chloride ions in the neuronal cell.
• The resultant negatively charged, hyperpolarized membrane prevents further
depolarization by excitatory neurotransmitters.
Pharmacokinetics
• Differences in lipid solubility influence pharmacokinetic properties.
• Diazepam and clorazepate are rapidly absorbed and quickly distributed in the CNS
because of their high lipophilicities.
• Onset of action of chlordiazepoxide is much slower because of decreased
lipophilicity, slower absorption and delayed passage into the CNS.
• In comparison with diazepam, lorazepam and oxazepam are relatively less
lipophilic and have a slower onset of effect. They have smaller volumes of
distribution and a resultant longer duration of action.
• Oxazepam absorption is slow and peak levels are not obtained until 2 to 4 hours
after single dose.
• Benzodiazepines with slow absorption rates are not recommended for acute relief
of anxiety symptoms.
• Diazepam and Chlordiazepoxide- not administered through IM route.
• Lorazepam-provides rapid, reliable and complete absorption when administered
intramuscularly; preparation requires refrigeration.
• Many benzodiazepines are converted to N-desmethyldiazepam(N-DMDZ), an
active metabolite with a long elimination t1/2 of 36 to 200 hours.
• N-DMDZ is further oxidized to oxazepam, then conjugated, and excreted.
• Clorazepate is a prodrug and possesses no anxiolytic effects until metabolized to
N-DMDZ. Absorption is pH-dependent.
• Alprazolam, Lorazepam, Oxazepam- short t1/2
Adverse Effects
• CNS depression- most common AE.
- drowsiness
- sedation
- psychomotor impairment
- ataxia
• Transient mild drowsiness
• Disorientation, confusion, irritability, aggression and excitement
• Tolerance
• Anterograde amnesia
Benzodiazepine Discontinuation
• Withdrawal symptoms may persist for days to weeks.
• Common symptoms of withdrawal include:
1. anxiety
2. insomnia
3. restlessness
4. agitation
5. muscle tension
6. irritability
• Less frequently occurring symptoms:
1. nausea
2. malaise
3. coryza
4. blurred vision
5. diaphoresis
6. nightmares
7. hyperreflexia
8. ataxia
• Rarely occurring symptoms:
4. Tinnitus
5. Confusion
6. Paranoid delusions
7. Hallucinations
8. Seizures
9. Psychosis
• The onset of withdrawal symptoms in patients ingesting benzodiazepines with
short-elimination half-lives occurs much earlier than those with long-elimination
half-lives.
• If BZ therapy exceeds 6 weeks, a slow dosage taper over several weeks is
recommended.
• To treat withdrawal symptoms:
a. Diazepam-can be initiated as loading dose(40% of daily consumption),
followed by a daily tapering of 10%.
b. Clonazepam-an alternative agent
c. Phenobarbital-for mixed Bz and alcohol dependence.
Drug Interactions
• Alcohol + BZ – additive effect; lowers the therapeutic index of BZ
• CNS depressants + BZ – potentiation of adverse sedative effects
• Lorazepam + Clozapine – Respiratory suppression and death
• Cimetidine + BZ – inhibition of BZ metabolism
• Nefazodone/Fluvoxamine- increased alprazolam concentrations.
BUSPIRONE THERAPY
• An azapirone anxiolytic
• Possess no anti-convulsant , muscle relaxant, hypnotic, motor impairment and
dependence properties.
Mechanism of Action:
• Anxiolytic mechanism is unknown
• As a serotonin partial agonist, it binds presynaptically to the receptors in the
dorsal raphe and postsynaptically to receptors in hippocampus and cortical brain
areas.
• It also possesses both dopamine agonist and indirect dopamine antagonist
properties.
Pharmacokinetics
• After an oral dose, it is rapidly and completely absorbed, and undergoes
extensive first-pass metabolism.
• It is 95% protein bound.
• Mean elimination half-life is 2.1 to 2.7 hours
• It is eliminated primarily by oxidative metabolism and is converted into both
active and inactive metabolites.
Adverse Effects
• Lack of sedative properties
• Dizziness, nausea, headaches, nervousness and dysphoria
Drug Interactions
• Buspirone + Cyclosporine/haloperidol – increased levels of cyclosporine and
haloperidol
• Buspirone + MAOI – elevated blood pressure
• Buspirone + Disulfiram – mania
• Buspirone + Clomipramine – hypertension and anxiety
ANTI-Depressants
• Not the first line of agents for GAD because of their adverse effects.
• Imipramine and Trazodone- effective in GAD after 3 to 8 weeks of therapy.
• Low doses of SSRI’s are found to be effective to some patients.
• They are used as alternatives for individuals with contraindications to
benzodiazepine use or those with concomitant depressive symptoms.
• They are also used as adjuncts in the treatment of patients with a partial response
to BZ’s or buspirone.
SECTION DESCRIPTION
Overview of This lesson is intended to discuss the pharmacology of drugs used in Panic disorder- a
Session subtype of anxiety disorder.
PANIC DISORDER
Lesson
Outline What are the characteristics of a panic disorder?
What are the pharmacologic classes of drugs used in the threatment of panic
disorder?
Goals of Therapy
a. to completely resolved panic attacks
b. to markedly reduce anticipatory anxiety
c. to avoid phobic attacks
d. to maintain clinical response that allows patient to resume normal activities
Nonpharmacologic Therapy
• Patients should avoid substances that may precipitate panic attacks including
caffeine, drugs of abuse, and non-prescription stimulants
Pharmacologic Therapy
• Panic disorder is effectively treated with several drugs including the TCA
imipramine, the BZ alprazolam, the MAOI phenelzine, and SSRIs.
• Alprazolam, clonazepam, sertraline and paroxetine are approved for panic
disorders.
• SSRIs are emerging as first-line agents because of their improved tolerability.
• SSRIs and Clomipramine are found to be more effective than imipramine and
alprazolam.
• In patients whose illness is complicated by a history of alcohol or drug abuse,
benzodiazepines should be cautiously used, and a TCA, SSRI, or MAOI would be
more appropriate.
• Concomitant benzodiazepine and SSRI therapy for 1 to 3 weeks may be
indicated in persons with severe symptoms of anticipatory anxiety.
ANTI-DEPRESSANTS
A. TRICYCLIC ANTIDEPRESSANTS
• Imipramine effectively blocks panic attacks within 3 to 5 weeks; however,
maximal improvement does not occur until 6 to 10 weeks.
• Approximately 20-30% of patients experience stimulatory side-effects including
insomnia, jitterness, irritability and unusual energy.
• Imipramine and Clomipramine-most widely studied
• Desipramine and nortriptyline- possibly effective
• TCAs possess stimulatory effects, anticholinergic effects, orthostatic hypotension,
delayed onset of antipanic effects, and toxicity in an overdose.
• For patients who cannot tolerate the anticholinergic effects of TCAs, a switch to
an SSRI may be helpful.
• Weight gain is associated with long-term therapy
D. Other Agents
• Trazodone and maprotiline- effective for panic disorders
• Nefazodone- reduces panic symptoms in patients with comorbid depression
• Bupropion is ineffective in treating panic symptoms
• Venlafaxine- also helpful in preventing panic attacks
E. BENZODIAZEPINES
• Clonazepam and high-dose Alprazolam – effective in preventing panic attacks
• Diazepam and lorazepam – effective at high doses
• Therapeutic response to these agents occurs in 1 to 2 weeks, with further
improvement occuring at 4 to 6 weeks.
• Alprazolam- ideal for patients who need immediate relief.
Acute Phase
• The main goal of therapy in this phase is to reduce symptoms.
• The duration of this phase generally is 1 to 3 months depending on the choice of
medications.
• The guiding principle in the treatment of panic disorders is to start at adequately
low dose and to treat for an appropriate period of time.
• The duration of acute phase with antidepressants requires a minimum of 8 to 12
weeks.
• Low initial doses of SSRIs are recommended to avoid stimulatory side effects.
- The starting dose of paroxetine is 10 mg with dosage increase of 10mg
weekly; target dose is 40 mg
- Starting doses for fluoxetine are 2.5 to 5 mg/d to a dosage range of 10-20
mg/d by the end of 2 weeks.
- Fluvoxamine 25 to 50 mg/d was increased to 150mg/d in divided doses
- Sertraline initiated at 12.5 or 25 mg/d and titrated to 100 to 200 mg/d is
effective in panic disorder
• Phenelzine
- starting dose is 15 mg/d after evening meal every 3 to 4 days until 60 mg/d is
reached
- fluoxetine must be stopped 5 weeks before phenelzine can be started
- if taken after meals, orthostatic hypotension is less likely to occur.
- Anti-cholinergic effects are less severe than with TCAs.
- Unpleasant side effects subside after 3 weeks
- Hypertensive crisis following an ingestion of tyramine-containing foods or
sympathomimetic drugs is the most serious, potentially life-threatening
adverse effect manifested by severe headache with flushing accompanied by
heavy “thumping” of the myocardium.
• The duration of acute phase with BZs is approximately 1 month because response
is rapid and occurs within 1 to 3 weeks.
Continuation Phase
• The goals of therapy during this phase are to complete and extend the treatment
response obtained in the acute phase, especially with regards to phobic
avoidance.
• This phase may last for 2 to 4 months.
Assessment A short quiz regarding the topic discussed will be given next meeting
SUBJECT PHARMACOLOGY 1 TOPIC PARKINSON’S-1
IG NUMBER IG-PHA-325LC WEEK NO 16
TERM FINALS SESSION 27 of 30
DATE PREPARED May 2007 DURATION 1.5 HOURS
SECTION DESCRIPTION
Overview of This lesson intends to discuss the pathophysiology of Parkison’s disease and the
Session pharmacology of drugs used in the management of Parkinson’s disease
Incidence
1. It has a prevalence of 1 to 2 per 1000 of the general population and 2
per 100 among people older than 65 years.
2. Onset generally occurs between age 50 and 65; usually occurs in the 60s.
Diagnosis
1. Diagnosis depends on clinical findings.
2. Tests (including imaging) are most often used to rule out an etiology of
secondary Parkinson’s disease.
3. New technologies [e.g., positron emission tomography (PET) scan] are used to
visualize dopamine uptake in the substantia nigra and basal ganglia. The PET
scan measures the extent of neuronal loss in these areas, but it is not yet widely
available.
4. A specific form of single-photon-emission computed tomography (SPECT) could
be helpful for diagnosis of Parkinsonian syndromes and non-parkinsonisms,
particularly essential tremor.
Treatment
1. Nondrug treatment
a. Exercise is an important adjunctive therapy and is most beneficial. Although
exercise does not help with the symptoms of Parkinson’s disease, regular
focused exercise, stretching, and strengthening activities can have a positive
effect on mobility and mood.
b. Nutrition. Patients with Parkinson’s disease are at increased risk of poor
nutrition, weight loss, and reduced muscle mass. Examples of the beneficial
effects of proper nutrition in this group of patients include:
1. Sufficient fiber and fluid intake help prevent constipation associated with
Parkinson’s disease and the medications used to treat the disease.
2. Calcium supplementation helps to maintain the existing bone structure.
3. Excessive dietary protein in the late stages of the disease causes erratic
responses to levodopa therapy.
4. A large body of literature supports the pathophysiological role of antioxidants
in the relief of oxidative stress in Parkinson’s disease. High doses of
antioxidants and α-tocopherol or vitamin E are recommended in this group
of patients.
2. Drug therapy for symptomatic relief. Treatment is divided into two generalized
categories: symptomatic therapies and preventive or protective measures.
Neuroprotective strategies are used to slow the development and progression of
the disorder.
Neuroprotective Treatment
1. MAO B such as selegiline and tocopherol (vitamin E) acts as a scavenger of free
radicals.
2. Dopamine agonists serve as scavengers of free radicals and decrease dopamine
turnover, which reduces oxidative stress. During early development of the
disease, there are increases in oxidative stress. Four classes of drugs are
available:
a. Anticholinergics (for resting tremor)
b. Precursor of dopamine agonists (e.g., carbidopa/levodopa)
c. Direct-acting dopamine agonists (e.g., bromocriptine, pergolide)
d. Indirect-acting dopamine agonists
1. Decrease reuptake (e.g., amantadine)
2. Decrease metabolism (e.g., selegiline)
3. Drug therapy for treating associated symptoms
a. Tricyclic antidepressants are used to treat depression. They exhibit some
dopaminergic and anticholinergic effects.
b. β-Blockers, especially propanolol with its high lipophilicity, benzodiazepines,
and primidone, are medications used for action tremor. Usually patients
show a clinical response in low doses.
c. Antihistamines. Diphenhydramine hydrochloride has some mild
anticholinergic effects and is used for symptomatic release of mild tremor;
because of its adverse reaction in the CNS, it should be used with caution in
the elderly.
b. On-off effect describes oscillations in response (at the receptor site) and sudden
changes in mobility from no symptoms to full parkinsonian symptoms in a matter
of minutes. No direct relationship between the on-off effect and drug levels has
been found. Usually, a second drug is added to the therapy regimen to correct
the effect. Reducing the dose of one drug and adding a second drug may also
be helpful.
c. End-dose effect, known also as the wearing-off effect, occurs at a latter part of
the dosing interval; it happens after a few years of L-dopa therapy. Reduce the
single L-dopa dose and spread the total L-dopa dose over a larger number of
single doses. Change to a dopamine agonist and use a sustained-release
formulation of L-dopa.
7. Psychological rehabilitation provides support for patients and their families. Keep
in mind that patients with Parkinson’s disease have a high incidence of depression
and that, in later stages of the disease, they develop dementia.
8. Secondary effects of Parkinson’s disease include:
a. Cardiovascular effects, including orthostatic hypotension and arrhythmia
b. Gastrointestinal effects, including constipation and hypersalivation
c. Genitourinary effects, including increased urinary frequency and impotence
d. Central nervous system effects, including hallucinations, depression, and
psychosis
SECTION DESCRIPTION
Overview of This subtopic of Parkinson’s disease is intended to discuss the pharmacology
Session individual drugs used in the management of Parkinson’s disease.
Objectives 1. To discuss the classes of drugs used in the management of Parkinson’s disease
2. To discuss the mechanisms of action of the agents for Parkinson’s disease
3. To enumerate the adverse effects associated with the use of the drugs
4. Significant interactions
a. Side effects may be potentiated by other drugs with anticholinergic activity
such as antihistamines, antidepressants, and phenothiazines.
b. Anticholinergic agents increase digoxin levels.
c. When anticholinergic agents are taken with haloperidol, the following occurs:
1. Schizophrenic symptoms may increase.
2. Haloperidol levels may decrease.
3. The severity of (not the risk of) tardive dyskinesia may increase.
d. When phenothiazines are taken with anticholinergic drugs, the effects of the
phenothiazines decrease and the anticholinergic symptoms increase.
e. Patients in high doses of anticholinergics combined with levodopa should be
watched for decreased levodopa activity because of a delayed gastric
emptying time.
1. Mechanism of action
a. Levodopa is converted to dopamine by the enzyme dopa decarboxylase, which
elevates CNS levels of dopamine.
b. The sustained-release formulation is designed to release the drug over 4-6
hours, thereby inhibiting variation in plasma concentration and decreasing
motor fluctuation “off” time, or to improve overall dose response in patients
with advanced disease.
d.Significant interactions
1. A combination of antihypertensive drugs and bromocriptine could decrease
blood pressure.
2. Dopamine antagonists increase the effect of bromocriptine.
Pergolide
a. Mechanism of action. Pergolide is a semisynthetic ergosine derivative. In
Parkinson’s disease, it exerts its effect by directly stimulating postsynaptic dopamine
receptors in the nigrostriatal system.
1. It is 1000 times more potent than bromocriptine on a milligram basis.
2. It inhibits the secretion of prolactin, increases the serum concentration of
growth hormone, and decreases the serum concentration of luteinizing
hormone.
3. It is most commonly used as adjunctive treatment to levodopa/carbidopa
b. Administration and dosage
c. Precautions and monitoring effects
1. Hypersensitivity reactions to pergolide and other ergot derivatives can occur.
2. Caution must be used with patients who are at high risk for ventricular
arrhythmia, especially when doses higher than 3 mg/day are used.
3. Cardiac dysrhythmias. Adverse effects are similar to those experienced with
bromocriptine.
4. CNS effects include dyskinesia, hallucinations, somnolence, and insomnia.
5. GI effects include nausea, constipation, diarrhea, and dyspepsia.
6. Cardiovascular effects include premature atrial contractions and sinus
tachycardia (alone or in combination with levodopa).
7. Miscellaneous effects include transient elevations of aspartate
aminotransferase, alanine aminotransferase, and alkaline phosphatase, and
pleural thickening.
Significant interactions
1. Because pergolide is 90% protein bound, it must be used with caution with
other highly protein-bound drugs.
2.Antipsychotic agents (e.g., phenothiazines, haloperidol, metoclopramide)
combined with dopamine agonists decrease the dopamine action and decrease
the dopamine action and decrease the therapeutic action of the antipsychotic
agent.
2. Amantadine
a. Mechanism of action. Amantadine is an antiviral agent (used to prevent
influenza).
1. Amantadine increases dopamine levels at postsynaptic receptor sites by
decreasing presynaptic reuptake and enhancing dopamine synthesis and release.
2. It may also have some anticholinergic effects. It decreases tremor, rigidity,
and bradykinesia.
3. It can give in combination with levodopa as Parkinson’s disease progresses.
4. Clinical effects of amantadine can be seen within the first few weeks of
therapy, unlike the other antiparkinsonian medications (e.g., carbidopa/levodopa),
which need weeks to months to show their full clinical effects.
b. Administration and dosage
1. Amantadine should be started at 100 mg/day. This may be increased to 200-300
mg/day as a maintenance dose.
2. Patients experiencing a decline in response may benefit from the following:
a. Discontinuing the drug for w few weeks, then restarting it.
b. Using the drug episodically, only when the patients’ condition most needs a
therapeutic boost.
3. Amantadine is also available in liquid form for patients with dysphagia.
Significant interactions
1. Amantadine increases the anticholinergic effects of anticholinergic drugs,
requiring a decrease in the dosage of the anticholinergic drug.
2. Hydrochlorothiazide plus triamtrene decreases the urinary excretion of
amantadine and increases its plasma concentrations.
a. Pramipexole
1. Mechanism of action
a. D2 subfamily of dopamine receptors. Pramipexole fully stimulates the
dopamine receptors to which it binds. Its action may be related to its
capacity to function as an antioxidant and oxygen free-radical scavenger.
b. Pramipexole also has antidepressant activity in moderate depression, which
may be related to its preferential binding to the dopamine D 1-receptor
subtype.
c. Long-acting dopamine agonists appear to have a lower risk of inducing
abnormal movements. Their use as initial treatment in early Parkinson’s
disease seems warranted, particularly for those with disease onset at a
younger age.
4. Significant interactions
a. Cimetidine reduces renal clearance of pramipexole
b. No interaction with selegiline, probenicid, or domperidone
c. When combined with levodopa, the dosage of levodopa must be decreased
by 27%.
b.Ropinirole
1. Mechanism of action is similar to pramipexole.
b. Entacapone
1. Mechanism of action
a. Entacapone is a selective and reversible inhibitor of COMT and permits
additional levodopa to reach the brain. It does not have any anti-parkinson
effect of its own.
b. It acts only peripherally by inhibiting COMT.
c. It improves the duration of “on” time and decreases the duration of “off”
time.
d. It is indicated as an adjunct to those on levodopa/carbidopa therapy who
experience the signs and symptoms of end-of-dose “wearing-off.”
2. Administration and dosage
a. 200 mg with each dose of L-dopa up to 8 times daily with a maximum dose
of 1600 mg daily.
b. Rapid withdrawal could lead emergency signs and symptoms of Parkinson’s
disease such as hyperpyrexia and confusion (symptoms resembling
neuroleptic malignant syndrome).
3. Precautions and monitoring effects
a. MAO. COMT and MAO are two major enzymes in the metabolism of the
catecholamines. Do not use together.
b. Drugs metabolized by COMT. Drugs that are metabolized by this pathway,
such as isoproterenol, epinephrine, norepinephrine, dopamine, and
dobutamine, as well as methyldopa may interact and may result in increased
heart rate, arrhythmias, and an excessive increase in blood pressure.
c. Hepatic function impairment. The majority of the drug is metabolized by the
liver; therefore, use caution in patients who have liver function abnormalities.
d. Fibrotic complication. Cases of retroperitoneal fibrosis, pulmonary infiltrates,
pleural effusion, and pleural thickening have been reported. These
complications may resolve when the drug is discontinued, but complete
resolution may not always occur.
e. Biliary excretion: Enatacapone is excreted by bile; therefore, use caution with
drugs known to interfere with biliary excretion, such as probenecid,
erythromycin, and ampicillin.
4. Other side effects. Dyskinesia/hyperkenesia, nausea, urine discoloration
(brownish-orange), diarrhea, and abdominal pain.
5. Drug interactions. May interact with drugs that are metabolized by the liver
cytochrome P450.
SURGICAL TREATMENT. All surgeries require needle insertion into the brain, which in
turn increases the risk of hemorrhage.
SECTION DESCRIPTION
Overview of This lesson is intended to present seizure disorders and the pharmacology of anti-seizure
Session and anti-convulsant agents.
MANIFESTATIONS
- Purposeless behavior is common.
- The affected person may have a glassy stare, may wander about aimlessly, and
may speak unintelligibly.
- Psychomotor (temporal lobe) epilepsy may lead to aggressive behavior (e.g.,
outbursts of rage or violence).
- Postictal confusion usually persists for 1-2 minutes after the seizure ends.
- Automatism (e.g., picking at clothes) is common and may follow visual, auditory
or olfactory hallucinations).
ETIOLOGY
Some seizures arise secondary to other conditions. However, in most cases,
the cause of the seizure is unknown.
PATHOPHYSIOLOGY
Seizures reflect a sudden, abnormal, excessive neuronal discharge in the
cerebral cortex. Any abnormal neuronal discharge could precipitate a seizure.
• NORMAL FIRING OF NEURONS – usually originate from the gray matter of one or
more cortical or subcortical areas, requires the following elements:
CLINICAL EVALUATION
• HISTORY – includes evaluation of the seizure, including interviews of the patient’s
family and eyewitness accounts to establish:
a. The frequency and duration of the episodes
b. Precipitating factors
c. The times at which episodes occur
d. The presence or absence of an aura
e. Ictal activity
f. Postictal state
THERAPY
• PRINCIPLES OF DRUG THERAPY
A. SEIZURE CONTROL – approximately 50% of epileptics achieve complete
seizure control through drug therapy.
- in another 25%, drugs reduce the frequency of seizures.
- epileptics generally require continuous drug therapy for at least 2 seizure-free
years before the drug discontinuation can be considered.
B.INITIAL TREATMENT
- Before anticonvulsive drug treatment is instituted, treatable underlying causes of
the seizure activity should be excluded.
- A single primary drug that is most appropriate for the seizure type must be
selected. If there is more than one appropriate primary drug, then age, sex, and
compliance of the patient must be considered.
- For patients with newly diagnosed epilepsy, administer low doses for a few days.
Patients may respond to a dosage that is lower that that traditionally prescribed
initially by their physicians, and this may have important implications in terms of
limiting adverse effects. The incidence of adverse effects increases with
increasing drug levels, even when the plasma concentrations are maintained
within the so-called therapeutic or optimal range.
- Approximately one-fourth or one-third of the maintenance dose of a single
medication is used to begin therapy; it is then increased over 3-4 weeks. The
exceptions are phenytoin or penobarbital, which can be started with the loading
or maintenance dose. The dose should be titrated until seizure control or
intolerable side effects occur.
- With the initiation of therapy, blood concentrations of medications should be
measured.
(1) To establish therapeutic ranges and dosage regimens based on symptomatic
toxicity or seizure frequency.
(2) To assess the patient’s compliance with therapy.
(3) To control the correlation among the dose, blood levels, and clinical
therapeutic levels or toxicity.
(a) Phenytoin follows nonlinear kinetics, as drug levels increase
dramatically (more than onefold) with only a small increase in the dose.
However, prior to this twofold increase in drug level with a small
increase in dose, there is a predictable linear increase with dose
increases; for this reason, it is recommended to increase the dose in
small increments to be able to predict when the drug follows nonlinear
kinetic. When this happens, that means the maximum rate of hepatic
enzyme clearance is reached and the body can no longer clear the drug
as it is introduced into the body.
(b) If physical examination reveals a new onset of nystagmus (except with
phenytoin, in which nystagmus develops before clinical intoxication),
ataxia, and unsteady, wide gait, the next dose increase should be
minimal.
(c) There is no justification for increasing drug dosage when a patient’s
seizures are fully controlled, even if the plasma concentration is below
the lower limit of the therapeutic range. If the patient continues to
have seizures without any evidence of adverse effects at a plasma
concentration near the toxic range, there are two approaches:
(i) Some increase the dosage according to clinical response up to
the highest tolerated limit.
(ii) Some do not increase the dosage because of the likelihood of
producing adverse effects.
(d) Carbamazepine has an autoinduction metabolism property, which
means that if the dose is increased twofold, blood levels increase less
than twofold because of increased metabolism.
(4) To determine the free drug level, which is helpful in patients who are in the
therapeutic range but have side effects or no response. The plasma protein
binding may be altered in these patients by some other disease state or
medication. Because of this alteration, there is more free drug available in
the system than the total level shows, especially with phenytoin, valproic
acid, and carbamazepine.
C. PARADOXICAL INTOXICATION – occurs when a high concentration of a single
drug causes increased frequency of seizures without classical adverse events.
This is common with hydantoins and carbamazepine. The proposed reason is
that their effect on the cerebellum is blocked at high concentrations.
Management usually requires no more than withholding enough doses of the
drug to allow the concentration to drift down.
Simple Partial
CHOICE 1: Carbamazepine (alone or combination)
CHOICE 2: Phenytoin
CHOICE 3: Primidone, Lamotrigine
CHOICE 4: Gabapentin, levetiracetam Zonisamide
Complex partial
CHOICE 1: Carbamazepine, Lamotrigine
CHOICE 2: Phenytoin
CHOICE 3: Phenobarbital, Zonisamide
CHOICE 4: Valproic acid, Primidone, Topiramate, Tiagabine
Primary generalized
CHOICE 1:Valproic acid
CHOICE 2: Carbamazepine
CHOICE 3: Phenytoin
CHOICE 4: Phenobarbital
TONIC-CLONIC
CHOICE 1: Lamotrigine
CHOICE 3: Valproic acid
CHOICE 4: Topiramate, Tiagabine
Absence
CHOICE 1: Lamotrigine*, Ethosuximide
CHOICE 2: Zonisamide, Valproic acid
Myoclonic
CHOICE 1: Valproic acid
CHOICE 2: Clonazepam
CHOICE 3: Zonisamide*
CHOICE 4: Felbamate*
Atonic
CHOICE 1: Valproic acid
CHOICE 2: Clonazepam
Status epilepticus
CHOICE 1: Diazepam
CHOICE 2: Phenytoin
CHOICE 3: Phenobarbital
Psychomotor
CHOICE 1: Phenytoin
CHOICE 2: Phenacemide
CARBAMAZEPINE
MECHANISM OF ACTION – chemically related to tricyclic antidepressants.
- mechanism of action is unknown in the treatment of seizure disorders.
- thought to act by reducing polysynaptic responses and blocking the posttetanic
potentiation.
SIGNIFICANT INTERACTIONS
- antiepileptic drugs (phenytoin, primidone, phenobarbital) – decrease the level
of carbamazepine (increase metabolism). VALPROIC ACID increases the level
of carbamezepine (decreases metabolism).
- OTHER MEDICATIONS (erythromycin, isoniazid, cimetidine, propoxyphene,
diltiazem, and verapamil increase the level of carbamazepine (decrease
metabolism).
PHENYTOIN
MECHANISM OF ACTION – inhibits the spread of seizures at the motor cortex and
blocks posttetanic potentiation by influencing synaptic transmission. There is an
alternation of ion fluxes in depolarization, repolarization, and membrane stability
phase and alternating calcium uptake in presynaptic terminals.
- Phenytoin is effective for the treatment of generalized tonic-clonic (grand mal)
seizures and for partial seizures, both simple and complex. It is not effective for
absence seizures.
ADVERSE EFFECTS
- The physician should be notified if any of the following adverse effects occur:
swollen or tender gums, skin rash, nausea and vomiting, swollen glands,
bleeding, jaundice, fever, or sore throat (i.e., signs of infection or bleeding).
SIGNIFICANT INTERACTIONS
➢ Antiepileptic drugs (carbamazepine, valproic acid, clonazepam, phenobarbital) –
decrease the level of phenytoin (increase metabolism.
➢ Phenytoin increases the conversion of primidone to Phenobarbital (increases
metabolism).
➢ Other medications such as disulfiram, isoniazid, chloramphenicol, and
propoxyphene increase the level of phenytoin (decrease metabolism). Drugs
whose efficacy is impaired by phenytoin include corticosteroids, digitoxin,
doxycycline, estrogens, furosemide, oral contraceptives, quinidine, rifampin,
theophylline, vitamin D and enteral nutritional therapy. Coumarin and warfarin
anticoagulants increase the serum phenytoin levels and prolong the serum half-
life of phenytoin by inhibiting the metabolism.
FOSPHENYTOIN
MECHANISM OF ACTION – water-soluble prodrug of phenytoin.
- it is converted to phenytoin by the bloodstream phosphatases, with a half-life of
about 8 minutes in both adults and children.
- It is indicated for patients who cannot take oral drugs, and in the acute
treatment for status epilepticus.
- administered via IV or intramuscular (IM) injection.
- characteristics similar to phenytoin.
ADVANTAGES
- an aqueous solution, unlike phenytoin, which is an alkaline solution; therefore,
there is no need to add propylene glycol and ethanol to the solution.
- Causes less soft-tissue injury at the site of injection. When administered by IM
injection, it is completely absorbed and has more predictable serum
concentration than IM-injected phenytoin.
VALPROIC ACID
MECHANISM OF ACTION – increases levels of GABA
- potentiates a postsynaptic GABA response by inhibiting the enzymatic response
for the catabolism of GABA.
- affects the potassium channel, creating a direct membrane-stabilizing effect.
ADVERSE EFFECTS
- Contact the physician if abdominal pain, nausea, vomiting, or anoxia occurs;
these could be symptoms of pancreatitis.
- CNS EFFECTS – tremor, ataxia, diplopia, lethargy, drowsiness, behavioral
changes, and depression.
- GI EFFECTS – nausea and increased appetite. Enteric-coated divalproex sodium
may reduce these side effects.
- DERMATOLOGICAL EFFECTS – alopecia and petechiae.
- HEMATOPOIETIC EFFECTS – thrombocytopenia, bruising, hematoma, and
bleeding.
- HEPATIC EFFECTS – minor elevations of aspartate aminotransferase (AST),
alanine aminotransferase (ALT), and lactate dehydrogenase (LDH).
- ENDOCRINE EFFECTS – decreased levels of prolactin, resulting in irregular
menses, and secondary amenorrhea.
- PANCREATIC EFFECTS – acute pancreatitis
- METABOLIC EFFECTS – hyperammonemia due to renal origin. Discontinuation
may be considered if lethargy develops.
SIGNIFICANT INTERACTIONS
- PRIMIDONE decreases valproic acid clearance (increases metabolism)
- PHENOBARBITAL and PHENYTOIN – displace protein binding, resulting in an
increased total phenytoin level and an increase or no change of free
phenytoin.
- CLONAZEPAM – increases CNS toxicity in patients on valproic acid.
- ASPIRIN – increases the level of valproic acid.
- WARFARIN – inhibits the secondary phase of platelet aggregation.
- ANTACIDS – increase the level of valproic acid.
- LABORATORY TESTS
- False-positive urine ketone tests may result among patients taking
valproic acid; thus, diabetic patients must use caution when using urine
tests.
- Antiepileptic drugs may alter thyroid function tests.
PHENOBARBITAL
MECHANISM OF ACTION – it increases the seizure threshold by decreasing synaptic
excitation by stimulating postsynaptic GABA-A receptor inhibitor responses as a CNS
depressant.
ADMINISTRATION AND DOSAGE
Adults = administered orally at 90-300 mg daily (in three divided doses or as a
single dose at bedtime).
Children = 3-6 mg/kg daily in two divided doses. Adjustment is made as needed.
PRECAUTIONS AND MONITORING EFFECTS
- It produces respiratory depression, especially with parenteral administration.
- It should be used with caution in patients with hepatic disease who may need dose
adjustments.
- It has sedative effects in adults and produces hyperactivity in children.
- Abrupt discontinuation produces withdrawal convulsions. If the drug must be
discontinued, another GABA-A agonist should be sunstituted.
ADVERSE EFFECTS
-Physician should be notified if any of the following adverse effects occur: sore
throat, mouth sores, easy bruising or bleeding, and any signs of infection.
CNS EFFECTS – agitation, confusion, lethargy, and drowsiness. Patients should avoid
alcohol and other CNS depressants.
SIGNIFICANT INTERACTIONS
-Antiepileptic drugs, such as valproic acid and phenytoin, increase the level of
Phenobarbital (decrease metabolism).
- Other drugs such as acetazolamide, chloramphenicol, cimetidine, and furosemide
increase the level of Phenobarbital (decrease metabolism). Rifampin, pyridoxine,
and ethanol decrease the level of Phenobarbital (increase metabolism).
PRIMIDONE
MECHANISM OF ACTION – it is a metabolite of Phenobarbital and
phenylethylmalonamide (PEMA), which has some anticonvulsive effects. It has
drug characteristics similar to Phenobarbital, with some differences in dose and
half-life.
ADMINISTRATION AND DOSAGE
- It has a short half-life of 7 hours, which may require three-times daily dosing.
- It is tolerated better if started at 50 mg at night for 3 days until the target daily
dose is reached.
ETHOSUXIMIDE
MECHANISM OF ACTION – it may inhibit the sodium-potassium adenosine
triphosphatase (Na+-K+ ATPase) system and the reduced form of nicotinamide-
adenine dinucleotide phosphate (NADPH)-linked aldehyde reductase (which is
necessary for the formation of γ-hydroxybutyrate, which is associated with the
induction of absence seizures).
- It reduces or eliminates the EEG abnormality; however, absence seizures are
the only seizures in which the normal EEG has clinical value (i.e., when the EEG
abnormality is corrected, the seizures are also controlled).
- It is relatively benign anticonvulsant with minimum protein binding.
ADMINISTRATION AND DOSAGE
It is usually given orally in an initial dose of 500 mg daily in adults and older
children and 250 mg daily in children ages 3-6 years. The dose may be raised by
250 mg every week to a maximum of 1.5 g daily in adults.
PRECAUTIONS AND MONITORING EFFECTS
- blood dyscrasias have been reported, making periodic blood counts necessary.
- there have been reports of hepatic and renal toxicity; thus, periodic renal and
liver function monitoring is necessary.
- cases of systemic lupus erythematosus have been reported.
ADVERSE EFFECTS
GI EFFECTS – nausea and vomiting. Small doses may lessen these effects. It
should be taken with food if GI upset occurs.
HEMATOPOEITIC EFFECTS – eosinophilia, granulocytopenia, leukopenia and
lupus.
CNS EFFECTS – drowsiness, blurred vision, fatigue, lethargy, hiccups and
headaches. Alcoholic beverages should be avoided with this medication.
PSYCHIATRIC-PSYCHOLOGICAL EFFECTS – confusion and emotional instability.
DERMATOLOGICAL EFFECTS – pruritus, photosensitivity, urticaria and Stevens-
Johnson syndrome.
GENITOURINARY EFFECTS – frequency of urination, vaginal bleeding, renal
damage, and hematuria.
MISCELLANEOUS EFFECTS – periorbital edema and muscle weakness. Patients
should also be advised of the risks of exposure to sunlight and ultraviolet
light.
CLONAZEPAM
MECHANISM OF ACTION – it is a potent GABA-A agonist, but its efficacy decreases
over several months of treatment.
ADMINISTRATION AND DOSAGE
Adults = oral agent given in an initial dose of 1.5 mg daily divided two or three
times. The dose may be increased to a maximum of 20 mg daily.
Children = 0.01-0.03 mg daily in two or three doses. The dosage may be
increased to a maximum of 0.2 mg/kg daily.
PRECAUTIONS AND MONITORING EFFECTS
- Patients with psychoses, acute narrow-angle glaucoma, and significant liver
disease should use this medicine cautiously.
ADVERSE EFFECTS
CNS EFFECTS – drowsiness, ataxia, and behavior disturbances in children;
these may be corrected by dose reduction.
RESPIRATORY EFFECTS – hypersalivation and bronchial hypersecretion.
MISCELLANEOUS EFFECTS – anemia, leukopenia, thrombocytopenia,
respiratory depression, anorexia, and weight loss.
SIGNIFICANT INTERACTIONS
- Antiepileptic drugs such as phenytoin, increase the level of clonazepam decrease
metabolism
- Other drugs. Clonazepam decreases the efficacy of levodopa and increases the
serum level of digoxin.
FELBAMATE
MECHANISM OF ACTION – the drug interacts with glycine modulatory site on N-
methyl-D-aspartate (NMDA) receptors. Blockade of NMDA may contribute to
neuroprotective effects of felbamate.
- it is used as a monotherapy or adjunctive therapy or without secondary
generalization in adults and generalized seizures associated with Lennox-Gestaut
syndrome in childen. The United States Food and Drug Administration (FDA)
recommended that use of felbamate be restricted to only those patients who are
refractory to other medications and in whom the risk-benefit relationship warrants
its use, because of severe hepatoxicity.
ADMINISTRATION AND DOSAGE
- Adults and children older than 14 years of age.
- Monotherapy, initially 1.2 g in three to four doses daily. The dosage may be
increased in 600-mg increments every 2 weeks to 2.4 g daily based on clinical
response and, thereafter, 3.6 g daily if necessary.
- Adjunctive therapy, 1.2 g in three to four divided doses daily, with reduction of
the dosage of other antiepileptic drugs by 20-33%. The dosage of felbamate
maybe increased in increments of 1.2 g at weekly intervals to a maximum of
3.6 g daily.
- Conversion to monotherapy initially 1.2 g daily in three to four doses, with
reduction of the dosage of other antiepileptic drugs by 33% at week 3. The
felbamate dosage may be increased to 3.6 g daily, and other antiepileptic drugs
discontinued or dosage further reduced in stepwise fashion.
- Children 2-14 years of age with Lennox-Gaustat syndrome, as adjunctive
therapy, initially 15 mg/kg daily in three to four doses. The dosage of other
antiepileptic drugs is reduced by 20%. The amount of felbamate may be
increased in increments of 15 mg/kg at weekly intervals to 45 mg/kg daily.
Further reduction in the dosage of other antiepileptic drugs may be necessary.
PRECAUTIONS AND MONITORING EFFECTS
- There are two very serious toxic effects, aplastic anemia and liver failure, which
lead to death for some patients.
- For aplastic anemia, the onset range from 5-30 weeks of initiation of therapy.
Weekly or biweekly CBCs are recommended initially.
- For liver, toxicity time between initiation of treatment and diagnosis of these
cases ranges from 14-257 days. It is recommended that liver function tests be
performed before initiation of therapy to identify patients who have evidence of
preexisting liver damage. Liver function tests should also be performed weekly
or biweekly. The FDA recommends that this drug be used only in patients who
are refractory to other medications and in whom the risk-benefit relationship
warrants its use.
- Photoallergy or phototoxicity may occur; patients should take protective
measures against exposure to ultraviolet light or sunlight.
- Instruct patients to store medication in its own tightly closed container at room
temperature away form excessive heat, direct sunlight and moisture.
ADVERSE EFFECTS
- If bleeding and blushing occurs, contact your physician.
- Potential to cause aplastic anemia (bone marrow).
- Patient should be monitored for these toxicities by CBCs and liver function tests
weekly or biweekly until discontinuation of any sign of these toxicities occurs.
CNS EFFECTS – insomnia, headache, anxiety, hyperactivity, and fatigue.
CARDIOVASCULAR EFFECTS – peripheral edema, vasoldilation, hypotension, and
hypertension.
OCULAR EFFECTS – diplopia and blurred vision.
GI EFFECTS – anorexia, weight decrease and nausea.
HEMATOLOGICAL EFFECTS – lymphadenopathy, leukopenia, and
thrombocytopenia.
METABOLIC/NUTRITION EFFECTS – hypokalemia and hyponatremia.
SIGNIFICANT INTERACTIONS
- Felbamate and phenytoin. Felbamate causes increase in phenytoin plasma
concentration. Phenytoin doubles felbamate clearance, resulting in 45%
decrease in steady-state trough levels.
- Felbamate and carbamazepine. Felbamate causes a decrease in
carbamazepine levels and an increase in carbamazepine metabolites. In
addition, carbamazepine causes a 50% increase in felbamate clearance,
resulting in a 40% decrease in steady-state through levels.
- Felbamate and valproic acid. Felbamate causes an increase in valproic acid
levels, but valproic acid does not affect felbamate levels.
- ADVERSE EFFECTS: signs and symptoms associated with increased plasma
level and toxicity are anorexia, nausea, vomiting, insomnia and headache.
GABAPENTIN
MECHANISM OF ACTION – an analogue of GABA
-it increases GABA turnover, but it does not bind to GABA or any other
established neurotransmitter receptor.
-its mechanism of action is currently unknown, although it binds to a specific
receptor in the brain and inhibits voltage-dependent sodium currents.
-it has been shown to be effective as an add-on drug in patients with partial
seizure with or without secondary generalization.
ADMINISTRATION AND DOSAGE
- Patients older than 12 years receive 900 mg to 1.8 g daily, administered as
adjunctive therapy in three divided doses. Titrating to an effective dose
normally can be achieved within 3 days by initiating therapy with 300 mg and
then increasing the dose in 300-mg increments over the next 2 days to
establish a dosage of 900 mg daily in three doses. If necessary, the dosage
may be increased to 1.8 g daily. To minimize potential side effects, especially
somnolence, dizziness, or fatigue, the first dose on day 1 may be administered
at bedtime.
- 3-12 years of age = 10-15 mg/kg/day in 3 divided doses to up to 25-50
mg/kg/day.
- The drug is primarily excreted renally; therefore, the dosage should be
adjusted for patients who have compromised renal function.
- The drug does not bind to plasma protein. There are no significant
pharmakokinetic interactions with other commonly used antiepileptic drugs.
- If gabapentin is discontinued or an alternate anticonvulsant medication is
added, it should be done gradually over a minimum of 1 week.
ADVERSE EFFECTS
- Common side effects are somnolence, ataxia, dizziness, fatigue and
nystagmus.
CNS EFFECTS – somnolence, dizziness, ataxia and fatigue.
GI EFFECTS – dyspepsia, dryness of mouth, constipation, and increased
appetite.
OCULAR EFFECTS – diplopia, blurred vision and nystagmus.
SIGNIFICANT INTERACTIONS
- Antacids and gabapentin. Antacids reduce the bioavailability of gabapentin by
20%; gabapentin could be taken 2 hours after antacid use.
- Cimetidine and gabapentin. Cimetidine decreases the renal excretion of
gabapentin by 14% and consequently increases gabapentin plasma levels
(however, this amount is not clinically significant).
- Oral contraceptives and gabapentin. Oral contraceptives increase the level of
norethindrone by 13%; this amount may not be clinically significant.
LAMOTRIGINE
MECHANISM OF ACTION – Its antipeileptic effect is similar to that of phenytoin. Its
effect may be due to inhibition of voltage-dependent sodium currents and
reduction of sustained repetitive neuronal activity.
- it is indicated for the treatment of partial seizures and secondary generalized
tonic-clonic seizures that are not controlled with other drugs.
- it is also used to treat Lennox-Gastaut syndrome.
- it is a broad spectrum, as well tolerated as monotherapy, and probably the least
teratogenic of the first-line agents. It may aggravate severe myoclonic epilepsy.
ADMINISTRATION AND DOSAGE
- Adults (older than 16 years) = 50 mg/day in two divided doses [patients taking
valproic acid (VPA) should be given 25 mg every other day], up to 100 mg/day
(up to 25 mg daily with VPA treatment)
- 2-12 years of age = 0.6 mg/kg/day in two divided dosed (0.15 mg/kg/day on
VPA treatment), up to 1.2 mg/kg/day (up to 0.3 mg/kg/day with VPA treatment)
- the smallest available chewable dispersible tablet is 5 mg. Them after 2 weeks,
increase by 0.3 mg/kg/day in one to two divided doses, up to 200 mg/day.
- swallow chewable dispersible tablet whole, chewed, or in dispersing water or
diluted fruit juice. If chewed, consume a small amount of water or dilute fruit
juice to aid in swallowing. To disperse, add the chewable dispersible tablet to
a small amount of liquid (1teaspoon or enough to cover the medication).
- Approximately 1 minute later, when the tablet is completely dispersed, swirl the
solution and consume the entire quantity immediately.
- for patients taking valproic acid, the initial dose is 50 mg daily for 2 weeks,
followed by maintenance doses of 100-200 mg daily in two divided doses.
- reduced clearance in the elderly necessitates dosage reduction.
- patients with hepatic impairment may require dosage reduction because of
reduction in metabolism.
PRECAUTION AND MONITORING EFFECTS
- the value of monitoring plasma concentration has not been established.
- caution should be used for patients taking this drug. It may adversely affect the
patient’s metabolism or complicate the elimination of the drug because of renal,
hepatic, or cardiac impairment.
- lamotrigine binds to melanin and can accumulate in melanin-rich tissue over
time. Periodic ophthalmological monitoring is recommended.
- photosensitization (photoallergy and phototoxicity) patients should take
protective measures against exposure to ultraviolet light and sunlight.
- serious rashes requiring hospitalization have been reported. The incidence of
rashes, including Stevens-Johnson syndrome, is approximately 1% in patients less
than 16 years old and 0.3% in adults. Are cases of toxic epidermal necrolysis or
rash-related death have occurred. Most rashes occur within 2-8 weeks of initial
treatment.
ADVERSE EFFECTS
- most common side effects are dizziness, diplopia, ataxia, blurred vision, nausea,
and vomiting.
CNS EFFECTS – headache, dizziness and ataxia
GI EFFECTS – nausea, vomiting, diarrhea and dyspepsia.
OCULAR EFFECTS – diplopia, blurred vision, and vision abnormality.
DERMATOLOGICAL EFFECTS – pruritus and rash may form similar to that found
when using phenytoin and carbamazepine. In many cases, the rash disappears
during continued therapy, but 1-2% of patients with rash represent a more
serious allergic reaction. Occassionally, patients have developed the Stevens-
Johnson syndrome. Concomitant use with valproic acid may increase the
likelihood of serious rash. The occurrences of life-threatening rash that were
reported developed within 2-8 weeks following therapy; other cases of rash
have been reported developing up to 6 months after therapy. The incidence of
rash is higher in children than in adults.
- monotherapy during pregnancy found no teratogenic effect.
SIGNIFICANT NTERACTIONS
- Carbamazepine decreases lamotrigine concentration by 70% and increases
carbamazepine levels.
- Phenobarbital or primidone decreases lamotrigine concentration by 40%.
- Valproic acid decreases the metabolism of lamotrigine and extends its half-life
to 60 hours, which necessitates a dose reduction.
TOPIRAMATE
MECHANISM OF ACTION – a derivative of fructose.
- it decreases rapid hippocampal neuronal firing, possibly because of sodium-
or calcium-channel inhibition.
- it is also a weak carbonic anhydrase inhibitor and a sodium-channel blocking
agent.
- it potentiates the the activity of GABA. It has been shown to be effective
adjunctive therapy for partial seizure treatment in adults, tonic-clonic seizures,
infantile spasms, and Lennox-Gastaut syndrome.
ADMINISTRATION AND DOSAGE
- 17 years and older = 25-50 mg/day, up to 400 mg/day in two divided doses.
- 2-16 years of age = 1-3 mg/kg/day, up to 5-9 mg/kg/day in two divided
doses.
- Topiramate is 80% bioavailable, and food does not affect its bioavailability.
- Dose adjustment is necessary for patients with renal and hepatic
impairments.
- Enzyme-inducing anticonvulsive drugs can decrease topiramate levels, but it
has significant effect on metabolism of other anticonvulsive drugs.
- initial treatment is 50 mg daily, followed by titration to an effective dosage.
More than 400 mg daily has not been shown to improve response.
PRECAUTION AND MONITORING EFFECTS
- Increased incidence of kidney stones (renal calculus) in older patients who
received this drug. Patients should be advised to increase intake of fluids.
- Paresthesia is a common side effect of anhydrase inhibitors.
ADVERSE EFFECTS
- Physician should be notified if any of the follwoign side effects occur:
▪ Breast pain in females
▪ Nausea and tremor, which are dose-related side effects
▪ Back pain, chest pain, dyspepsia or leg pain.
CNS EFFECTS – psychomotor slowing, difficulty with concentration and
speech, somnolence, fatigue, asthenia, weight loss, cognitive
disturbances and difficulties, tremors, dizziness, ataxia and headache.
GI EFFECTS – nausea, vomiting and gastroenteritis.
GENITOURINARY EFFECTS – renal calculi
CV EFFECTS – chest pain, palpitation and vasodilation
OCULAR EFFECTS – abnormal vision, eye pain and diplopia
HEMATOLOGICAL EFFECTS – anemia, epistaxis, leukopenia and aplastic
anemia.
SIGNIFICANT INTERACTIONS
- Phenytoin and carbamazepine will increase clearnace
- Topiramate increases the clearance of other drugs that are cleared by
cytochrome P450 (CYP450).
TIAGABINE
MECHANISM OF ACTION – designed to act on the inhibitory action of GABA by
blocking its uptake, thereby prolonging its action after synaptic release.
- it is indicated as adjunctive therapy for partial seizures and secondary
generalized tonic-clonic seizures.
ADMINISTRATION AND DOSAGE
- Starting dose of 4 mg daily for 2 weeks may be increased 4-8 mg weekly
thereafter, to a maintenance dose of 32-56 mg daily.
- Maximum recommended dosage for children is 32 mg daily; maximum
recommended dosage for adults is 56 mg daily.
- A high-fat meal decreases the rate of tiagabine absorption but does not
affect the extension of absorption. Tiagabine should be taken with food.
PRECAUTIONS AND MONITORING EFFECTS
- generalized weakness: moderately severe to severe weakness has been
reported. It resolves in all cases after reduction in dose or discontinuation of
therapy.
- ophthalmic effects, as indicated by animal studies, include the possibility for
residual binding to retina and melanin binding; this finding, however, has not
been confirmed in human studies. Periodic ophthalmological monitoring is
necessary.
DERMATOLOGICAL EFFECTS – possibility of severe rash to Stevens-Johnson
syndrome, as reported in clinical studies.
ADVERSE EFFECTS
CNS EFFECTS – confusion, dizziness and fatigue.
GI EFFECTS – upset stomach, nausea, mouth ulceration and anorexia.
SIGNIFICANT INTERACTIONS
Phenobarbital, phenytoin, and carbamazepine will increase tiagabine clearance
ZONISAMIDE
MECHANISM OF ACTION – not well known.
- it may block the sensitive sodium channels and T-type calcium channels.
- It is an effective agent for refractory partial seizures, generalized seizure
indicated for adjunct therapy for partial seizure for adults, infantile spasm,
mized seizure types of Lennox-Gestaut syndrome, myoclonic, and generalized
tonic-clonic seizures.
ADMINISTRATION AND DOSAGE
- adults and children older than 16 years of age = 100 mg daily; within 2
weeks, increase to 200 mg/daily in 2-week bases, up to 600 mg daily.
- can be taken with or without food.
PRECAUTIONS AND MONITORING
- may increase mean concentration of serum creatinine and BUN; renal function
should be monitored periodically.
- may increase serum alkaline phosphatase
- may produce drowsiness
SIDE EFFECTS
- Contact the physician if sudden pain and abdominal pain occurs or if blood in
urine is detected; these symptoms could indicate a kidney stone. Increase fluid
intake to decrease the risk of stone formation. Also, contact the physician if
fever, sore throat, oral ulcer, or easy bruising is seen; these symptoms could be
due to a hematological complication.
SIGNIFICANT INTERACTIONS
-It induces liver enzymes by increasing metabolism and through clearance of
zonisamide and decreases half-life.
- Food will delay absorption but will not affect the bioavailability of the drug.
LEVETIRACETAM
MECHANISM OF ACTION – a pyrrolidone derivative and is chemically unrelated to
other antiepileptic drugs.
- it displays inhibitory properties in the kindling model in rats.
- it is used as an adjunctive therapy in the treatment of partial seizure in
adult patients.
ADMINISTRATION AND DOSAGE
- staring dose of 1000 mg/day given in two divided doses may be increased
every 2 weeks, to a maximum of 3000 mg/day.
PRECAUTIONS AND MONITORING EFFECTS
- hematological effects are minor, but there is a statistically significant decrease
compared to placebo in total man RBC count, mean hemoglobin, and mean
hematocrit.
- decrease in WBC count and neutrophil count
- also causes drowsiness
SIDE EFFECTS
- MOST COMMON: somnolence, weakness (asthenia), infection and dizziness.
CNS – somnolence, dizziness, depression, nervousness
CARDIOVASCULAR – palpitation, tachycardia, vascular insufficiency
RESPIRATORY – pharyngitis, rhinitis, increase cough
MISCELLANEOUS – weakness, headache, infection
SIGNIFICANT INTERACTIONS
- it does not influence the plasma concentration of existing antiepileptic drugs,
and other antiepileptic drugs do not influence the pharmacokinetic effects of
levetiracetam.
• SURGERY
If seizures do not respond to therapy, surgery may be performed to remove the
epileptogenic brain region. The most common is cortical excision (lobectomy).
COMPLICATIONS
- The risk of mother and fetus should be discussed, including the risks of fetal
malformation associated with antiepileptic drugs and other genetic factors.
DRUG THERAPY
➢ If the patient is seizure free for at least 2 years, withdrawal of the drug should
be considered. If antiepileptic drug therapy is necessary, a switch to
monotherapy should be made if possible.
➢ Five antiepileptic medications have been used or studied in pregnant patients:
carbamazepine, Phenobarbital, phenytoin, primidone, and VPA. Monotherapy of
lamotrigine during pregnancy found no teratogenic effect. Congenital
malformations associated with these drugs include craniofacial abnormalities,
cardiac defects, and neuronal tube defects. Most of the studies consider paternal
genetic factors, environmental factors, drug dosing or combination therapy.
➢ Antiepileptic drugs interfere with folate metabolism. Administration of folic acid,
4 mg daily, and multivitamins decreases the risk of malformation, especially of the
neuronal tube.
➢ Vitamin K, 10 mg per day for the last 1 or 2 months of gestation, will help to
prevent neonatal hemorrhage, especially in cases of phenytoin or Phenobarbital
use.
➢ Seizure disorder and oral contraceptives. Gabapentin, lamotrigine,
levetiracetam, and tiagabine do not affect the efficacy of oral contraceptives.
➢ Seizure disorder and the elderly population. New generations of antiepileptic
drugs such as levetiracetam and gabapentin may be more useful due to low levels
of protein binding and safer side effects. Also, new generations have less
potential for drug interactions than agents eliminated from the liver.
MONITORING
➢ Free serum antiepileptic drug levels should be monitored monthly, immediately
before the next dose, and the dose should be adjusted to the lowest dose
providing adequate control.
➢ Serum alphafetoprotein levels should be checked and ultrasonography should be
preformed at 16 weeks of gestation to evaluate for fetal neuronal tube defects.
An alternative to these tests is amniocentesis, especially if the mother is taking
valproate or carbamazepine.
➢ Comprehensive ultrasonography should be performed at 18 and 22 weeks of
gestation for patients taking antiepileptic drugs that cause cardiac anomalies.
• Intrapartum plans should include IV administration of a short-acting
benzodiazepine. If there is concern about fetal or maternal respiratory
depression, administering intravenous phenytoin or intramuscular Phenobarbital
should be considered. Clotting studies should be performed, and 1 mg vitamin K
should be given to the infant. Nurses and physicians should be alerted for
possible hemorrhage of the infant and apprised that the infant may experience
antiepileptic drug withdrawal.
SECTION DESCRIPTION
Overview of This lesson is intended to discuss the different pharmacologic properties of ethanol,
Session alongside with the different substances of abuse.
DRUGS OF ABUSE
Tolerance- a decreased response to the effects of the drug. It may either be:
a. Metabolic- increased disposition of drug after chronic use
b. Behavioral – ability to compensate for drug affects
c. Functional- compensatory changes in receptors, effector
enzymes,or membrane actions
Opioids
- The nepenthe (free from sorrow) in odyssey- contains opium
Clinical aspects
- symptoms of opioid withrawal: lacrimation, rhinorrhea, yawning, and sweating.
- Followed by restless sleep, weakness, chills, gooseflesh, nausea, vomiting, muscle
aches and involuntary movements
• Ethanol- the sedative-hypnotic with the longest history of both use and abuse.
• Barbiturates- introduced in 1903
• Meprobamate- 1954, a nonbarbiturate sedative-hypnotic that lacks the
disadvantage of older agents
• BZD- 1960, now the drugs of choice for sedation-hypnosis.
Mickey Finn
• Chloral hydrate
Flunitrazepam
• Roofies
Rapid-onset BZD
• Associated with date rape
STIMULANTS
• Caffeine- Most widely used social drug worldwide
• Nicotine- one of the most widely used licit drugs
• Cocaine- used for at least 1200 years by natives of South American Andes. Koller
is credited with first using the drug as topical anesthetic for eye surgery
• Mathampethamine- Methedrine, “speed”
• MDMA- “ecstacy”, methylenedioxymethampethamine
• Cathinone- a plant from Catha edulis, possess amphetamine-like effects.
• Speedball- cocaine and heroin
HALLUCINOGENS
• LSD- lysergic acid diethylamide, most common hallucinogen
• Atropa belladonna and Datura stramonium- deliriants
• Mescaline, psylocibin
• Phencyclidine- PCP, angel dust
Marijuana
• Cannabis
• Have three major cannabinol compounds: cannabidiol, tetrahydrocannabinol and
cannabinol.
Inhalants
• Nitrous oxide- “head-shops”
• Ether and chloroform- nephrotoxic, hepatotoxic, and peripheral nerve damage
• Organic nitrites- sexual enhancer
• Amyl nitrite- poppers
• Isobutyl nitrite- locker room, rush
Steroids
• Heavy users may develop edema and jaundice
• Clinical findings may include hypertrophied muscles, acne, oily skin, hirsutism in
females, gynecomastia in males and needle puncture.
Final examination
Assessment