Sie sind auf Seite 1von 60

PHARMACOLOGY

Lyceum Northwestern University


COLLEGE OF MEDICINE
➢ THE HISTORY OF PHARMACOLOGY
INTRODUCTION TO PHARMACOLOGY:
Materia Medica—the science of drug preparation and
The Nature of Drugs the medical uses of drugs—began to develop as the
Dr. Perfecto Soriano precursor to pharmacology.

Pharmacology can be defined as the study of François Magendie and his student Claude Bernard
substances that interact with living systems through
- began to develop the methods of experimental
chemical processes, especially by binding to regulatory
physiology and pharmacology in the late 18th and early
molecules and activating or inhibiting normal body
19th centuries.
processes.
Advances in chemistry and the further development of
Medical Pharmacology is often defined as the science
physiology in the 18th, 19th, and early 20th centuries
of substances used to prevent, diagnose, and treat
laid the foundation needed for understanding how
disease.
drugs work at the organ and tissue levels.
Toxicology is the branch of pharmacology that deals
Controlled clinical trial, were reintroduced into
with the undesirable effects of chemicals on living
medicine—only about 60 years ago—did it become
systems, from individual cells to humans to complex
possible to accurately evaluate therapeutic claims.
ecosystems.
Around the same time information accumulated about
drug action and the biologic substrate of that action,
the drug receptor. Studies of the local molecular
environment of receptors have shown that receptors
and effectors do not function in isolation; they are
strongly influenced by other receptors and by
companion regulatory proteins.

Pharmacogenomics—the relation of the individual’s


genetic makeup to his or her response to specific
drugs—is close to becoming an important part of
therapeutics.

Discovery that small segments of RNA can interfere with


protein synthesis with extreme selectivity has led to
investigation of small interfering RNAs (siRNAs) and
micro-RNAs (miRNAs) as therapeutic agents.
FIGURE 1–1 Major areas of study in pharmacology. The
actions of chemicals can be divided into two large Similarly, short nucleotide chains called antisense
domains. The first (left side) is that of medical oligonucleotides (ANOs), synthesized to be
pharmacology and toxicology, which is aimed at complementary to natural RNA or DNA, can interfere
understanding the actions of drugs as chemicals on with the readout of genes and the transcription of RNA.
individual organisms, especially humans and domestic These intracellular targets may provide the next major
animals. Both beneficial and toxic effects are included. wave of advances in therapeutics.
Pharmacokinetics deals with the absorption,
distribution, and elimination of drugs. Two general principles that the student should
Pharmacodynamics concerns the actions of the remember:
chemical on the organism. The second domain (right
side) is that of environmental toxicology, which is (1) That all substances can under certain circumstances
concerned with the effects of chemicals on all be toxic, and the chemicals in botanicals (herbs and
organisms and their survival in groups and as species. plant extracts, “nutraceuticals”) are no different from
chemicals in manufactured

1
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
drugs except for the much greater proportion of
impurities in botanicals;
✓ The Physical Nature of Drugs
Drugs may be solid at room temperature (eg, aspirin,
(2) That all dietary supplements and all therapies atropine), liquid (eg, nicotine, ethanol), or gaseous (eg,
promoted as health enhancing should meet the same nitrous oxide). These factors often determine the best
standards of efficacy and safety as conventional drugs route of administration.
and medical therapies.

*There should be no artificial separation between ✓ Drug Size


scientific medicine and “alternative” or The molecular size of drugs varies from very small
“complementary” medicine. Ideally, all nutritional and (lithium ion, MW 7) to very large (eg, alteplase [t-PA],
botanical substances should be tested by the same a protein of MW 59,050). However, most drugs have
randomized controlled trials (RCTs) as synthetic molecular weights between 100 and 1000.
compounds.
To have a good “fit” to only one type of receptor, a drug
molecule must be sufficiently unique in shape, charge,
and other properties, to prevent its binding to other
➢ GENERAL PRINCIPLES OF PHARMACOLOGY receptors.

To achieve such selective binding, it appears that a


THE NATURE OF DRUGS molecule should in most cases be at least 100 MW
Drug may be defined as any substance that brings about units in size.
a change in biologic function through its chemical Drugs much larger than MW 1000 do not diffuse
actions. readily between compartments of the body.
In most cases, the drug molecule interacts as an agonist
(activator) or antagonist (inhibitor) with a specific
molecule in the biologic system that plays a regulatory ✓ Drug Reactivity & Drug-Receptor Bonds
role. This target molecule is called a receptor. Drugs interact with receptors by means of chemical
forces or bonds. These are of three major types:
In a very small number of cases, drugs known as covalent, electrostatic, and hydrophobic.
chemical antagonists may interact directly with other
drugs, whereas a few drugs (osmotic agents) interact
almost exclusively with water molecules.
Covalent bonds are very strong and in many cases
Drugs may be synthesized within the body (eg, not reversible under biologic conditions.
hormones) or may be chemicals not synthesized in the Ex: covalent bond formed between the acetyl
body (ie, xenobiotics, from the Greek xenos, meaning group of acetylsalicylic acid (aspirin) and
“stranger”). cyclooxygenase, its enzyme target in platelets, is not
readily broken
Poisons are drugs that have almost exclusively harmful
effects. Electrostatic bonding is much more common than
Paracelsus (1493–1541) famously stated that “the dose covalent bonding in drug-receptor interactions but are
makes the poison,” meaning that any substance can be much weaker than covalent bonds. They vary from
harmful if taken in the wrong dosage. relatively strong linkages between permanently charged
ionic molecules to weaker hydrogen bonds and very
Toxins are usually defined as poisons of biologic origin, weak induced dipole interactions such as van der Waals
ie, synthesized by plants or animals, in contrast to forces.
inorganic poisons such as lead and arsenic.

2
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
Hydrophobic bonds are usually quite weak and are
probably important in the interactions of highly lipid-
soluble drugs with the lipids of cell membranes and
perhaps in the interaction of drugs with the internal
walls of receptor “pockets.”

Note: Drugs that bind through weak bonds to their


receptors are generally more selective than drugs that
bind by means of very strong bonds. This is because
weak bonds require a very precise fit of the drug to its
receptor if an interaction is to occur.

✓ Drug Shape
The shape of a drug molecule must be such as to permit
binding to its receptor site via the bonds. Optimally, the
drug’s shape is complementary to that of the receptor
site in the same way that a key is complementary to a
lock.

The phenomenon of chirality (stereoisomerism) is so


common in biology that more than half of all useful
drugs are chiral molecules; that is, they can exist as
enantiomeric pairs.

Note: Enzymes are usually stereoselective, one drug


enantiomer is often more susceptible than the other to
drug metabolizing enzymes. As a result, the duration of
action of one enantiomer may be quite different from
that of the other. Similarly, drug transporters may be
stereoselective.

✓ Rational Drug Design

Rational design of drugs implies the ability to


predict the appropriate molecular structure of a
drug on the basis of information about its biologic
receptor.
A few drugs now in use were developed through
molecular design based on knowledge of the three
dimensional structure of the receptor site.

3
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE

PRINCIPLES OF PHARMACOKINETICS
AND DRUG METABOLISM
Dr. Perfecto Soriano

➢ PHARMACOKINETICS
– refers on how the BODY acts on the DRUGS.
- The qualitative study of drug movement in,
through and out of the body.
Explanation:
- Intensity of effect is related to concentration of
- PK involves Absorption, Distribution,
the drug at the site of action. (Depend on its
Metabolism and Elimination (ADME), while PD
pharmacokinetic properties).
involves Efficacy and Toxicity.
• Pharmacokinetic properties of particular
- Process starts with PK which includes: Drug
drug is important to determine: dose, Drug concentration in blood and Drug
1. Route of administration concentration at target site but half way
2. Dose through the 3rd process PD also starts and
3. Onset of action continues with the Effect and Body’s Clinical
4. Peak Response
5. Action time
6. Frequency of dosing ➢ PHARMACOKINETIC PROCESS

➢ CLINICAL PHARMACOKINETICS
- Study of time course of a drugs movement
through the body
- Understanding of what the body does to or
with the drug.
-Application of Therapeutic Drug Monitoring
(TDM) and individualization of drug therapy
• TDM – branch of clinical pharmacology
that specializes in the measurement of
medication concentrations in the blood. ➢ DRUG TRANSPORTATION: Biological Membrane

➢ PHARMACODYNAMICS
- What the DRUGS do to the body.
- Drug concentration at the site of action or in
plasma is related to the magnitude of effect.

➢ PHARMACOKINETICS (PK) AND


PHARMACODYNAMICS (PD) RELATIONSHIP

- Bilayer of phospholipid and cholesterol


molecule = 100 Armstrong thickness
- EXTRINSIC AND INTRINSIC PROTEINS are
embedded in the membrane.

4
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
- GLYCOPROTEIN – on the surface. The drug which are UNIONIZED, LOW
- This proteins varies from cell to cell POLARITY AND HIGHER SOLUBILITY are
- Paracellular spaces and channels are also EASY to permeate membrane
present The drug which are IONIZED, HIGH
- Drug molecules can cross cell membrane by: POLARITY AND LOWER SOLUBILITY are
• Passive Transport DIFFICULT to permeate membrane
• Carrier – mediated (protein-mediated)
o Facilitated transport ➢ pH EFFECT
o Active transport - Most drugs are weak acids or weak bases
-Primary -The ionization of drugs may markedly reduce
-Secondary their ability to permeate membrane
- The degree of ionization of drugs is
determined by surrounding pH and their pKa
* pKa- index to express acidity of weak acids;
the smaller pKa value the stronger the acid is.

➢ HENDERSON-HASSELBALCH EQUATION
[𝑨 −]
𝒑𝑯 = 𝒑𝑲𝒂 + 𝒍𝒐𝒈
[𝑯𝑨]

pKa = negative logarithm of acid dissociation


constant
➢ PASSIVE TRANSPORT (Downhill movement) [A-] = ionized drug
- Most important transport mechanism for [HA] = unionized drug
most drugs.
-Majority of drugs diffuses across the ➢ IMPLICATION (Important)
membrane in the direction of concentration • Acidic drugs reabsorbed are largerly
gradient unionized in the stomach and absorbed
- No Active role of the membrane faster; while basic drugs are absorbed
-Proportional to lipid: water partition coefficient faster in the small intestines
-Lipid soluble drugs diffuse by dissolving in the • Ion trappinig
lipoidal matrix of the membrane • ACIDIC DRUGS are excreted faster in
- Characteristic: ALKALINE URINE.
• Not requiring energy
• BASIC DRUGS are excreted faster in
• Having no saturation ACIDIC URINE.
• Having no carriers
• Not resting competitive inhibition
- Affecting factors: ➢ FILTRATION
• Size of the molecules
• Lipid solubility
• Polarity
• Degree of ionization
• pH of the environment (fluid of body,
cell, blood, urine)
-REMEMBER:

5
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
• Carrying the substrate to the other side
of the membrane
- Passage of drugs through aqueous pores in • Dissociates
membrane or through paracellular spaces • Return back to its original position
- Lipid insoluble drugs can cross if the molecular -Move substrate of SINGLE CLASS (unipoters)
size is small
down a concentration gradient
-Majority of intestinal mucosa and RBC have - No energy dependent transport in the
small pores and drugs cannot cross
direction of electrochemical gradient
-CAPILLARIES have large paracellular spaces and -Similar to entry of glucose into the muscle
most drugs can be filter through this (GLUT 4)

➢ CARRIER-MEDIATED TRANSPORT
- Involves specific membrane transport ➢ ACTIVE TRANSPORT
proteins (transmembrane protein) known as - Energy dependent
drug transporter or carriers which is specific for - Can move solutes against concentration
the substrate: Physiological important ions, gradient
nutrients, metabolites, transmitters • Primary active transporter – generate
- They also translocate xenobiotics including energy THEMSELVEs (ex: ATP
drug metabolites
hydrolysis)
-Drug molecules:
• Secondary active transporter – utilize
1. Bind to the transporter
energy stored in voltage and ion
2. Translocated across membrane
gradients generated by primary active
3. Released on the other side of the
transporter (ex: NA/K ATPase)
membrane
- Symporters (co-transporter)
-Specific, saturable and inhibitable
- Antiporters (exchangers)
- Depending on energy requirement can be
either facilitative (passive) or active transport

➢ FACILITATIVE TRANSPORTER
-Higher to Lower concentration
- Transmembrane protein bind with their
substrate transiently (temporary)
- Conformational changes:

6
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
4. ANTIPORT/EXCHANGE TRANSPORT
- moving in opposite direction
➢ MAJOR DRUG TRANSPORTERS -mediates efflux of drug and metabolite
1. ATP- BINDING CASSETTE (ABC) -ex: Na+Cl dependent neurotransmitter
TRANSPORTER SUPER FAMILY
- Primary active transport ➢ ENDOCYTOSIS
• P- glycoprotein - Very little importance to drug translocation
- Intestinal mucosa, renal - Large protein molecules and metabolic waste
tubules and blood brain barrier -Either phagocytosis, pinocytosis or receptor-
-Mediated only EFFLUX OF mediated endocytosis.
SOLUTE from cytoplasm
detoxification ➢ PINOCYTOSIS
- It involves the invagination of a part of the cell
membrane and trapping within the cell of a
small vesicle containing extracellular
constituents.
- The vesicle contents can be released within
the cell or extruded from the other side of the
cell.

2. SOLUTE-CARRIER (SLC) TRANSPORTER


- Secondary active transport
- Energy to pump one solute derived from
downhill movement of another solute
(mostly Na)
• Organic Anion Transporting - It is important for the transport of some
Polypeptides (OATPs) MACROMOLECULES (ex: INSULIN through BBB)
- Central transporters in
disposition of drugs and other THE PROCESS OF PHARMACOKENETICS
xenibiotics (ADME)
- mediate wide variety of
endogenous substrate
• Organic Cation Transporters (OCTs)
- have specificity for a large
number of cationic substrate
that are both endogenous and
exogenous
- Both transporters are expressed in
LIVER and RENAL TUBULES, for
metabolism and excretion of drugs
3. SYMPORT/CO-TRANSPORT
- Concentration gradient is such that
both solute move in the same direction

7
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
I. ABSORPTION OF DRUGS • For Abraded Skin- Tannic acid
(can cause: Hepatic necrosis)
➢ Absorption – is the transfer of a drug from its - Cornea is permeable to lipid soluble
site of administration to the blood stream drug
- Most of the drugs are absorbed by the way of - Mucus membranes of mouth, rectum,
passive transport and vagina are permeable to lipophilic
-Intravenous (IV) administration has NO drugs
ABSORPTION
- Fraction of administered dose and rate of b. SUBCUTANEOUS AND
absorption are important INTRAMUSCULAR ROUTE
- Drugs directly reach vicinity of
➢ Factors affecting absorption: capillaries (passes capillary endothelium
• Drug properties – aqueous solubility, and reach circulation)
concentration, lipid solubility, molecular -Passes through the large paracellular
weight amd polarity pores
• Blood flow (vascularity) to the - Faster and more predictable than oral
absorption site administration
- Exercise and heat – increase
• Total surface area available for
absorption
absorption surface
-Adrenaline – decrease absorption
• Affinity with special tissue
• ROUTE OF ADMINSTRATION IS c. ORAL ROUTE
IMPORTANT! - Physical properties: physical state
(solid or liquid), lipid or water solubility
- Dosage Forms:
• Particle size
• Disintegration time and
dissolution rate
• Formulation
-Physiological factors:
• Ionization, pH effect
• Presence of food
• Presence of other agents
➢ ROUTE OF ADMINISTRATION:
➢ ORAL ADMINISTRATION: 1ST PASS
a. TOPICAL ROUTE METABOLISM
- Depends on lipid solubility – only lipid • Before the drugs reaches systemic
soluble drugs will penetrate intact skin (only circulation, the drug can be
few drugs are used therapeutically) metabolized in the liver. As a result, the
-Examples: concentration of drug in the systemic
• GTN, Hyosine,Nicotine, circulation will be REDUCED.
Testosterone and Estradiol
• Organophosphorous d. BUCCAL AND RECTAL
compounds – can cause - bypasses the liver
systemic toxicity if ingested

8
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
e. INTRAVENOUS ROUTE intestinal wall and/or liver or
- has no absorption phase excreted in bile

➢ ACCORDING TO THE RATE OF ABSORPTION: ➢ BIOAVAILABILTY AREA UNDER CURVE (AUC)


(Fastest to slowest) - is used to calculate the bioavailability of
1. Inhalation a drug
2. Sublingual - The area under the plasma drug
3. Rectal concentration-time curve (AUC) reflects
4. Intramuscular the actual body exposure to drug after
5. Subcutaneous administration of a dose of the drug and
6. Oral is expressed in mg*h/L
- This area under the curve is
7. Topical or Transdermal
dependent on the rate of elimination of
- Example: Nitroglycerine the drug from the body and the dose
administered.
• IV – immediate
• Sublingual – 1-3 minutes
• Per rectal – 40-60 minutes

➢ BIOAVAILABLITY

➢ ROUTE OF ADMISNISTRATION DETERMINES


BIOAVAILABILITY
- For drugs taken by routes other than
the IV route, the extent of absorption
and the bioavailability must be
- refers to the RATE AND EXTENT OF understood in order to determine what
ABSORPTION of a drug from dosage form as dose will induce the desired therapeutic
determined by its concentration-time curve in effect.
blood or by its excretion in urine - Its will also explain why the same dose
- It is the measure (fraction) of a drug that may cause a therapeutic effect by one
reaches the systemic circulation in route but toxic or no effect by another.
unchanged form
- Useful to compare two different drugs or ➢ BIOEQUIVALENCE
different dosage forms of same drugs - Therapeutic agent that has the same
- It is a concept for ORAL ADMISTRATION pharmacologic potency and
-BIOAVALABILITY of drug administration bioavailability as another drug at the
IV is 100% but unpredictable after oral same dose
ingestion because: - containing the same active ingredient
• The drug may be incompletely are bioequivalent if their rates and
absorbed extents of absorption (bioavailability) is
• The absorbed drug may undergo the same.
first pass metabolism in the - Drugs may
have the same

9
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
bioavailability but they may not be
bioequivalent = have the same
pharmacologic effect ➢ FACTORS INFLUENCING Vd
• Lipid solubility (Lipid: water partition
coefficient)
• pKa of the drug
• Affinity for different tissues
• Blood flow-brain vs fat
• Disease states
• Plasma protein binding

➢ REDISTRIBUTION
- Highly lipid soluble drugs get distributed to:
II. DISTRIBUTION OF DRUGS • High perfusion low capacity – brain, heart
and kidney
➢ Distribution – it is the passage of drug • Low perfusion high capacity – muscle and
from the circulation to the tissue and its fats
site of action - When plasma concentration of drugs falls,
-The extent of distribution of drug depends drug is withdrawn from this site prolonging the
on its: solubility, ionization at physiological action of this drug
pH (dependent on pKa), extent of binding -Greater the lipid solubility, the faster is its
to plasma and tissue proteins and redistribution (Thiopentene Na)
differences in regional blood flow, disease -Shorter acting drug can be prolonged by
like CHF, uremia, and cirrhosis. administering slowing and continuously –low
-Movement of Drug – until equilibrium perfusion high capacity tissues
between unbound drug in plasma and
tissue fluids. ➢ BRAIN AND CSF PENETRATION
• Blood-Brain-Barrier (BBB)
➢ VOLUME OF DISTRIBUTION (Vd) - includes capillary endothelial cells
• Apparent Volume of Distribution (which have tight junctions and large
- The volume that would accommodate intracellular pores) and an investment
all the drugs in the body, if the of glial tissue, over the capillaries
concentration was the same as in plasma - Similar barrier is located in the
-Expressed as in Liters choroid plexus
• Drug may distribute into any of all the • BBB is lipoidal and limits entry of non-
following compartments: lipid soluble drugs (Amikacin,
-Plasma Gentamicin, Neostigmine, etc.)
-Interstitial fluid - REMEMBER: Only lipid soluble
-Intracellular fluid unionized drug penetrate and have
• Total Body Fluid = approximately 42 liters action on the CNS
• Efflux carriers like P-glycoprotein
present in brain capillary endothelial
cell (also in intestinal mucosal, renal
tubular, hepatic canalicular, placental
and testicular

10
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
cells)extrude drugs that enter brain by • High degree of protein binding makes the
other processes drug long acting, because bound
- Inflammation of meninges of the fraction is not available for metabolism,
brain INCREASES PERMEABLITY of BBB unless it is actively excreted by liver or
• Dopamine does not enter brain, but its kidney tubules
precursor Levodopa does • Generally expressed plasma
concentrations of the drug refer to
➢ PLACENTAL BARRIER bound as well as free drugs
-Placental membrane are lipoidal and allows • In hypoalbuminemia, binding may be
free passage of lipophilic drugs, while reduced and high concentration of free
restricting lipophobic drugs
drugs may be attained (Phenytoin)
- Only lipid soluble drugs can penetrate and
limitation of hydrophobic drugs
➢ TISSUE STORAGE
- But higher concentration of lipophobic drugs
- Drugs may also accumulate in specific
in the maternal tissues gains access to the fetus.
organs or get bound to specific tissue
- Beside this:
constituent
-It’s an incomplete barrier – some influx
Organ Drugs
transporter operate (Thalidommide)
Heart and Digoxin
• Placental efflux – P-glycoprotein serves
skeletal muscle
a limiting factor Liver Digoxin, Chloroquine,
• Influx transporter Tetracyclin
Kidney Digoxin, Chloroquine
➢ PLASMA PROTEIN BINDING (PBB) Thyroid gland Iodine
- Most drugs possess physiocochemical affinity Brain Chlorpromazine, Isoniazid,
for plasma proteins. Acetazolamide
- ACIDIC DRUGS BIND TO PLASMA ALBUMIN Retina Chloroquine
and BASIC DRUGS BIND TO ALPHA 1- Iris Ephedrine, Atrophine
GLYCOPROTEIN Bones and teeth Tetracyclines, Heavy
-Extent of binding depends on the individual Metals
Adipose tissue Thiopental, Ether,
compound. Increasing concentration of drug
Minocycline, DDT
can progressively saturate the binding sites.
-The CLINICAL IMPLICATIONS of PBB are:
III. METABOLISM OF THE DRUG:
• Highly PPB drugs are largely restricted to
BIOTRANSFORMATION
the vascular compartment and tend to
have lower Vd
➢ Biotransformation –
• The PBB fraction is not available for action
chemical alteration of
• There is equilibrium between PBB fraction the drug in the body
of drug and free molecules of drug. -Aim: To convert non-
• The drugs with high physicochemical polar lipid soluble
affinity for plasma proteins (Aspirins, compounds to polar
sulfonamides, chloramphenicol) can lipid insoluble
replace the other drugs compounds to avoid
(Acenocoumarol, warfarin) or reabsorption in renal
endogenous compounds (bilirubin) with tubules.
lower affinity - Most
hydrophilic

11
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
drugs are less biotransformed and ❖ Phase II (Synthetic/Conjugation, anabolic) –
excreated unchanged (Streptomycin, result in inactive products/metabolites
Neostigmine and Pancuronium) -(Morphine to Morphine-6 –glucuronide and
- Biotransformation is required for active sulfate metabolite of Minoxil are
protection of body from toxi exceptions)
metabolites. - Both phases decrease lipid solubility, thus
➢ RESULT OF BIOTRANSFORMATION: increasing renal elimination
• Activation – few drugs are -Mostly occurs in liver, although some are
administered in inactive form metabolized in plasma, lung and gut.
(prodrug) and needs to be activated to
form active metabolite
- Stability, good bioavailability, less
side effect or toxicity, desired
pharmacokinetic properties
• Inactive drug (prodrug) to
active/enhanced activity
-Levodopa-dopamine
- Prednisome to Prednisolone
- Enapril to Enalprilat
• Inactivation – active drug and its PHASE I- OXIDATION
metabolite to inactive metabolites -Most important drug metabolizing reaction-
• Usually most drugs (Ibuprofen, addition of oxygen (negative) charge radical or
Paracetamol, Chloramphenicol, removal of hydrogen (positive) charged radical.
Propanolol, Lidocaine) -Insertion of Oxygen – short lived highly reactive
• Active drug to active product quinone/epoxide/superoxide
- Phenacetin to Acetaminophen or - Various oxidation reaction- are oxygenation or
Paracetamol hydroxylation of C, N or S atoms; N or O-
-Morphine to Morphine-6 –glucuronide, dealkylation, Oxidative deamination
digitoxin to digoxin -Examples: Barbiturates, Phenothiazines,
• No toxic or less toxic drug to toxic Paracetamol and Steroids
metabolites (Isoniazid to acetyl - Involve Cytochrome P-450 monooxygenases
(CYP), NADPH and Oxygen
isoniazid)
-More than 100 CYP P-450 isoenzymes are
- Mutagenecity
identified and grouped into more than 20 families
-Teratogenicity
(1, 2, 3…)
-Carcinogenicity
- Sub-families are identified as A, B, C…
-Hepatotoxicity
-In human, only 3 isoenzyme families are
important – CYP1, CYP2, CYP3
➢BIOTRANSFORMATION: CLASSIFICATION OR
-CYP 3A4/5 carry out biotransformation of
PHASES
largest number (30-50%) of drugs. In addition to
liver, this isoforms are expressed in intestine
❖ Phase I (Non-synthetic/functional/ catabolic) –
(responsible for 1st pass metabolism at this site),
metabolite maybe active or inactive and kidneys too.
- Functional group is generated, more - Inhibition of CYP3A4 by erythromycin,
chemically reactive clarithromycin, ketoconazole, itraconazole,
verapamil, ditiazem, and

12
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
constituents of grape fruit juice is responsible for • Conjugation of the drug or its phase I
untoward interaction with terfenadine and metabolite with an endogenous substrate-
astemizole polar highly ionized organic acid to be excreted
- Inducers of CYP3A4 are rifampicin, phenytoin, in urine or bile – high energy requirement
carbamazepine, Phenobarbital • Glucoronide conjugation - most synthetic
reaction
➢ NONMICROSOMAL ENZYME OXIDATION
• Compounds with hydroxyl or carboxylic acid
- Some drugs are oxidized by non-microsomal
group are easli conjugated with glucoronic acid
enzymes (mitochondrial and cytoplasmic:
(derived from glucose) by enzyme UDP-
• Alcohol – dehydrogenase
glucuronyl transferase
• Adrenaline –MAO and COMt
- Examples: chloramphenicol, aspirin,
• Mercaptopurine – Xantine Oxidase
morphine, metronidazole, bilirubin, thyroxine
• Drugs glucuromides, excreted in the bile can be
PHASE I – REDUCTION
hydrolyzed in the gut by bacteria, producing
-This reaction is opposite of oxidation and
beta-glucoronidase-liberated drug is
involves CYP 450 enzyme working in the
reabsorbed and undergoes the same fate-
opposite direction
-Examples: chloralhydrate, Enterohepatic recirculation (e.g.
chloramphenicol,levodopa,halothane and Chloramphenicol, Phenolpthlein, Oral
warfarin Contraceptives) and prolongs their action
Dopa decarboxylase • Acetylation: compounds having amino or
Levodopa Dopamine hydrazine residues are conjugated with the
help of acetyl CoA
PHASE I – HYDROLYSIS -e.g. sulfonamides, Isoniazid, Hydralazine,
-This cleavage of drug molecule by uptake of a Clonazepam, Procainamide.
molecule of water with the help of enzymes like • Genetic Polymorphism (slow and fast
esterases, amidases, peptidases acetylators
- Examples: Amides and polypeptides are
• Sulfate conjugation: the phenolic compounds
hydrolyzed by amidases and peptidases
and steroids are sulfated by sulfokinases
-Choline esters, procaine, lidocaine,
-e.g. Chloramphenicol, adrenal and sex steroids
procainamides, aspirin, carbamazepine-epoxide,
• Methylation: the amines and phenols can be
pethidine, oxytocin
-Hydrolysis occurs in liver, intestines, plasma and methylated. Methionine and cysteine acts as
other tissues methyl donor
-examples: adrenaline, histamine, nicotinic acid,
PHASE I – CYCLIZATION methyldopa, captopril, mercaptopurine
- Formation of ring structure from straight chain • Ribonucleoside/nucleotide synthesis:
compound activation of many purine and pyrimidine
- e.g. Proguanil antimetabolites used in cancer chemotherphy.
• Sulfate Conjugation: Phenolic and steroid
PHASE I – DECYCLIZATION compounds-sulfated by sulfottransferase
- Opening up a ring structure of the cyclic (SULTs)
molecule • Glycine Conjugation: Salicylates and other
-e.g. Phenytoin, Barbiturates drugs having carboxylic acid-conjugated with
glycine
PHASE II METABOLISM - Not a major pathway

13
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
• Glutathione Conjugation: Inactivates highly • A drug mau inhibit one isoenzyme while being
reactive quinines or epoxides intermediates – substrate of other isoenzyme – quinidine
Paracetamol -Quinidine is metabolized by CYP3A4 but
-When glutathione falls short (toxicity) toxic inhibits CYP2D6
products formed causing tissue damage • Some enzyme inhibitors- allopurinol,
-Minor Pathway erythromycin, verapamil, diltiazem,
omeprazole, metronidazole, ciprofloxacin and
➢ FACTORS AFFECTING BIOTRANSFORMATION sulfonamides
• Concurrent use of drug: induction and inhibition
• Genetic polymorphism
• Pollutant exposure from environment or ➢ HOFFMAN ELIMINATION
industry - Inactivation of drug in body fluids by spontaneous
• Pathological status molecular rearrangement without use if any
• Age enzyme

➢ METABOLIC ENZYMES
• Drug metabolizing enzyme is divided into 2 IV. EXCRETION OF DRUG
types:
-Microsomal Enzymes ➢Excretion is a transport procedure which the
-Non-microsomal Enzymes prototype drug (parent drug) or other
• Both of this is deficit in newborn-susceptible to metabolic products are excreted through
manydrugs (chloramphenicol, opiods) excretion organ or secretion organ
-Develop in 1st month partially and completely - Hydrophilic compounds can easily be
in 3 months excreted
• Amount and kind – genetically controlled,
altered by environmental factiors ➢ROUTES OF DRUG EXCRETION
-Drug response variation • Kidney
• Biliary excretion
➢ USE OF ENZYME INDUCTION • Sweat and saliva
• Congenital non-hemolytic jaundice (deficient • Milk
glucuronidation of bilirubin) • Pulmonary
- Phenobarbital hastens the clearance of
jaundice ➢ HEPATIC EXCRETION
• Cushing Syndrome – phenytoin enhances • Drugs can be excreted in bile especially when
degradation of adrenal steroids they are conjugated with glucuronic acid
• Chronic poisoning – faster metabolism of poison • Drug is absorbed, glucuronidated or sulfated in
• Liver disease the liver and excreted through the bile,
glucuronic acid/sulfate is cleaved off by bacteria
➢ ENZYME INHIBITION OF DRUG METABOLISM in the GIT, drug is reabsorbed (steroid
• One drug can inhibit metabolism of other if hormones, rifampicin,amoxicillin,
utilizes same enzyme or co-factors contraceptives
• However, not common because different drugs • Anthraquinone, heavy metal – directly excreted
are substrate of different CYPs (100 in the colon
isoenzymes of CYP – 450 alone)

14
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
➢ RENAL EXCRETION: URINE
• Most important channel for excretion of drugs ➢ TUBULAR SECRETION
• It eliminates water soluble substances • Certain cations and anions
• Amount of drug or its metabolite depends on: • Epithelium of the proximal tubules into
-Glomerular Filtration (GFR) tubulkar fluid via special consuming
-Tubular Reabsorption (TR) transport system
- Tubular Secretion (TS) • Energy dependent active transport- reduce
-Net Renal Excretion = (GFR + TS)-TR free concentration of drugs- further, more
secretion (protein binding is facilitated for
➢ GLOMERULAR FILTRATION (GFR) excretion of some drugs)
-Normal GFR – 120ml/min -OAT – organic acid transport
-Glomerular capillaries have pores larger than -OCT – organic base transport
usual -P-glycoprotein
-The kidney is responsible for excreting of all • Limited capacity
water soluble substances • Competitive inhibition can occur
-All nonprotein bound drugs (lipid soluble or • Bidirectional transport –blood vs tubular
insoluble) presented to the glomerulus are fluid
filtered • Utilized clinically – penicillin vs probenecid,
- Glomerular Filtration of drugs depends on probenecid vs uric acid (salicylate)
their plasma protein binding and renal blood • Quinidine decreases renal and biliary
flow-protein bound drugs are not filtered clearance of digoxin by inhibiting efflux
-Renal failure and elderly carrier P-gp
• If renal clearance is greater than
➢ TUBULAR REABSORPTION 120ml/min – TS is assumed to occur
• Passive diffusion depends on
• It reduces the amount of free form of drug
- Lipid solubility
• PPB drugs gets dissociates to get
– 99% of lipid soluble GF gets
eliminated via this route
reabsorbed
• OAT: penicillin, probenecid, urinc acid,
- Non-lipid soluble are unable to do so
- Ionization of drug at existing pH salicylates, indomethacin, methotrexate,
- Highly ionized drugs are not absorbed glucuronides, etc
• Depends on pH of urine, ionization, etc • OCT: thiazide, amiloride, triametene,
• Lipid insoluble ionized drug excreated as it furosemide, quinine, choline, cimetidine
is aminoglycosides (amikin, gentamicin, • Both transporters are bidirectional – blood
tobramycin) vs tubular fluid
• Changes in urinary ph can change the • Not well developed at birth-prolong action
excretion pattern of drugs of drugs (penicillin, cephalosporin)
-weak bases ionized more or less • Gets matured in infancy
reabsorbed in acidic urine • Progressively declines after the age of 50
-weak acids ionized more or less reabsorbed years and almost lowers of most drugs
in alkaline urine after 75 years old
• Utilized clinically in salicylate and barbiturate
poisoning – alkalinized urine (drugs with pKa: ➢ PULMONARY EXCRETION
5-8) - Gases and volatile drugs or particulate matter-
• Acidified urine- atrophine and morphine etc

15
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
irrespective of lipid solubility - CL= Rate of elimination/C
-Alveolar transfer of gas –partial pressure in blood -Example: if a drug has 20 mcg/min and RoE is
-example: General anesthesia, paraldehyde, 100mcg/min
alcohol -CL = 100/20 5ml/min

➢SALIVA AND SWEAT EXCRETION ➢ FIRST ORDER KINETICS (EXPONENTIAL)


-minor importance for drug excretion • Rate of elimination is directly
-example: lithium, potassium iodide, rifampicin proportional to drug concentration
and heavy metals • CL remaining constant
• Constant fraction of drug is eliminated
➢MILK EXCRETION
per unit time
- Most of drug enter – breast milk by passive
diffusion ➢ ZERO ORDER KINETICS (LINEAR); FEW DRUGS
-Lipid soluble and PBB drugs do it better
• The rate of elimination remains constant
-% of drug reaching infant is very less – majority
irrespective of drug concentration
of drugs can be given to lactating mothers
without ill effect • CL decreases with increase in
-But lactating mother should be prescribed with concentration
drugs with caution • Constant fraction of drug is eliminated
-Contraindicated drugs: amiodarone, per unit time
anthraquinone, chloramphenicol, ciprofloxacin, • Alcohol, theophyline, tolbutamide, etc.
cyclosporine, indomethacin, methotrexate,
tetracycline ➢PLASMA HALF LIFE
- Special precaution: ampicilin, aspirin, losartan, - Defined as time required for plasma
metoclopramide, sulfonamide concentration to be reduced to half (50%) of its
original value – 2 phase: rapid declining
➢ KINETICS OF ELIMINATION Mathematically: t 1/2= 0.693/k
• t1/2=In2/k
➢Pharmacokinetic Parameters • Ln2=natural logarithm of 2(0.693)
• K=elimination rate constant = CL/V
• Bioavailabilty (F): fraction of administered drug • t1/2= 0.693xV/CL
that reaches systemic circulation in unchanged • Complete drug elimination can occur in
form 4-5 half life
- F= amt. of drug that enters the systemic
circulation (AUC) dose administered

• Volume of Distribution (V): volume that


accommodate all the drugs in body; if
concentration throughout was the same as in
plasma
- V= dose administered/plasma drug
concentration
➢STEADY-STATE PLASMA CONCENTRATION
• Clearance (CL): volume of plasma containing
• Steady-state Plasma Concentration (Cpss)
the amount of drug that is removed from the
- Repeated drug
body in unit time
administration at

16
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
relatively short interval of time -Balances between doses administered and
-Input drug balance = clearance dose interval
- Cpss= dose rate/CL
- From this equation, it is clear that the dose rate ➢LOADING DOSE
can be calculated if CL and Target Cpss of drug is • Single or few quickly repeated dose to attain
unknown concentration rapidly
- Earlier equation is valid for drug route -Loading dose = (target Cp x V)/F
administration where the bioavailability (F) is • So loading dose is governed by V not CL or half-
100% but where if falls short the equation turns life of drug
to be
- Dose rate= (target Cpss xCL)/F
➢MAINTENANCE DOSE
- Both these equations stands valid for the drugs
• The amount of drug given to maintain the
following 1st order kinetics
steady state plasma concentration (Cpps) of
➢MICHELIS MENTEN KINETICS drug at regular interval so as to balance the
elimination
• Elimination changes from 1st order to zero
-Dose rate = (target Cpss x CL)/F
order kinetics over therapeutic range
• So it is dependent on CL or half-life of drug
• This signifies that till the saturation level of
drug, Cpss is related to dose rate
➢THERAPEUTIC DRUG MONITORING (TDM)
• But it turns out proportion beyond it
• Cpss of a drug depends on its F, V and
-Rate of drug elimination = (Vmax) (C)/Km+C
clearance – each of this parameters varies
-Km = plasma conc. At whihch elimination rate is
from patient to patient
half maximal
-C=plasma concentration of drug • Measurement of plasma concentration of drug
-Vmax=maximum rate of drug elimination after initial drug administration (based on
average patient) can give all this parameters of
➢PLATEAU PRINCIPLE OF DRUG ACCUMULATION an individual
• Repeated Dosing • This helps for the subsequent quantification of
- When constant dose of drug is repeated drug dose regimen
before the expiry of 4 half life- the higher peak
concentration is achieved after certain interval, ➢USE OF TDM
because some remnant drugs will be present in • Low safety margin – digoxin, anticonvulsants,
the body antiarrhythmics, theophylline,
-subsequent plasma concentration becomes aminoglycosides, lithium, TCA
constant and forms a plateau and fluctuates • Potentially toxic drugs
around desired therapeutic level of drug
-Desired therapeutic level reaches 4-5 half lives

17
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
Median lethal dose (LD50) - the dose that KILLS aprox
DRUG EVALUATION AND REGULATION 50% of the animals
Dr. Perfecto Soriano
These doses are used to calculate the initial dose to be
New drugs are discovered or developed through: tried in humans
-identification of new drug target (Usually taken as one hundredth to one tenth of the
-rational design of a new molecule NO-EFFECT DOSE in animals)
-screening for biological activity of large numbers of
natural products Limitations of preclinical testing
-chemical modification of a known active molecule -toxicity testing is time consuming and expensive
-large numbers of animals may be needed to obtain
Drug screening valid data
-done to define pharmacologic profile (activity and -toxic data from animals to humans are unpredicted
selectivity of the drug) -adverse effects are unlikely to be seen in preclinical
-desired result of screening is called a lead compound testing

Preclinical safety toxicity testing Evaluation in humans


“All drugs are toxic in some individuals at some dose”
Confounding factors in clinical trials
Preclinical toxicity testing includes most or all these A)the variable natural history of most diseases
procedures: -many diseases disappear spontaneously

Crossover design - alternating periods of administration


of test drug, placebo,standard treatment
- way to minimize errors

B) The presence of other diseases and risk factors


-Can be minimized by crossover technique and proper
selection and assignment of patient to a group

C) Subject and observer bias and other factors


Single blind design- minimize subjective bias, px doesn't
know if he/she is in taking placebo
Goals of preclinical toxicity is to:
Double blind design- minimize observer bias, px and
-identify potential human toxicities
personnel doesn't know the medication being used. A
-design tests to further define the toxic mechanisms
third party holds the code identifying the medication
- predicting the most relevant toxicities to be monitored
in clinical trials
Clinical trials

No-effect dose- MAXIMUM dose at which a specific


Phase 1
toxic effect is NOT seen
-The effects of the drug as a function of dosage are
Minimum lethal dose - the SMALLEST dose that is
established (20-100 healthy px) (if drug is expected to
observed to KILL any experimental animal
have toxic effect should be

18
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
done in px with the disease ex. Cancer aids px)
- nonblind or open, blinded and placebo controlled Adverse drug reactions
- harmful unintended response
-Done to determine the probable limits of the safe - Fourth leading cause of death (higher than
clinical dosage range pulmonary disease,AIDS,accidents and mobile
accidents)
Phase 2
-To determine the efficacy (proof of concept), and the Orphan drugs
doses to be used in any follow on trials (100-200 px with -drugs for rare diseases
the target disease)
-highest rate of failure
- single-blind design

Phase 3
-Certain toxic effects caused by immunologic processes
may first appear
- done to thousands of px
-phase 1 2 3 minimize errors caused by placebo effect
-double blind and cross over technique

Phase 4
- Monitoring the safety of the new drug under
actual conditions
- Has no fixed duration
- Low incidence drug effects are not generally
seen before phase 4

19
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
3. Nucleic acids
DRUG-RECEPTOR INTERACTION and
4. Polysaccharides
PHARMACODYNAMIC PRINCIPLES
Interaction
Dr. H.H. Maramba
DRUG MOLECULE (D)
OBJECTIVES
▪ Agonist (activator) (A)
▪ Drug-Receptor Interaction ▪ Antagonist (inhibitor)
▪ Agonists & Antagonists
▪ Graded Dose-Response TARGET MOLECULE
▪ Potency and Efficacy
▪ Quantal Dose-Effect ▪ Receptor (R)

Drug AFFINITY

Any substance that bring about a change in biologic


function through its chemical actions at the molecular
level
D + R = DR complex→ Effect
Pharmacodynamics
Receptor Principles
Actions of the drug on the body “what the drug does to
the body” 1. Receptor largely determine the quantitative
relations between dose or concentration of the
drug
✓ “the receptor’s affinity for binding a
drug determines the concentration of a
drug required to form a significant
amount of drug-receptor complexes”
2. Receptors are responsible for selectivity of drug
action
✓ “the molecular size, shape, and
electrical charge of a drug determine
whether it will bind to a particular
receptor among a wide array of
receptor/binding sites available at the
action site”
Drug Molecule (D) 3. Receptors mediate actions of pharmacologic
1. Appropriate size agonists and antagonists
2. Electric charge ✓ “the receptors mediate the action of
3. Shape the agonist (or antagonist) by producing
4. Anatomic composition an activated or inactivated response”
Michaelis - Menten Principle
Drug Receptor
➢ the amount of an enzyme is kept constant and
▪ A macromolecular component of the cell with the concentration of its substrate is gradually
which a drug (molecule ligand) interacts to increased, the reaction velocity will increase
produce a response until it reaches a maximum…
▪ Usually a protein molecule
Molecules capable of serving as drug receptors are: 1.
Enzymes
2. Membrane proteins

20
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
decreasing response
An activated receptor-effector
system is Ra
AGONISTS
1. Full agonists
2. Partial agonist
3. Inverse agonist

Full agonist
✓ Agonist drugs that bind to receptor sites and
activate the receptor effector system to the
maximum extent
Partial agonist
➢ Agonist drugs that bind to the same receptor
site, activates the receptor-effector system in
the same way but do not evoke maximum
response
Inverse agonist
➢ An agonist with a high affinity for the Ri form of
the receptor and stabilizes most of these
receptors preventing them from conversion to
their active form

DRUG-RECEPTOR INTERACTIONS
Agonistic (A)
➢ activate the agonist binding site
Antagonistic (B)
➢ Compete with the agonist
Allosteric Activator (C)
➢ Act at a separate site and increasing response
Allosteric Inhibitor (D)
➢ Act at a
separate site
a
n
d

21
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
ANTAGONIST (ED50) to produce an effect (50% of that drug’s
maximal effect)
1. Interacts with the receptor but does not change the
receptor
2. Blocks the action of other drug molecules
3. Effect is only observed in the presence of an agonist
4. Have Affinity but no Efficacy
Types of Antagonism
1. Competitive antagonism
2. Irreversible antagonism

Competitive antagonism
▪ These drugs bind to the receptor sites but
generally does not stimulate a response but
prevents the binding of the agonists to the ➢ Potency of a drug depends on the affinity of
same receptor sites. receptors for binding the drug and in part on
o High concentrations of the agonist drug the efficiency with which drug-receptor
can overcome the effect of a interaction is coupled to response.
competitive antagonist  Affnity is the propensity of the drug to
bind to a recptor.
Irreversible antagonism
▪ These drugs usually bind to the receptor sites in 2. Efficacy
an irreversible manner.
o A high concentration of the agonist ➢ Maximal effect of a drug which can be achieved
drug cannot completely reverse the with highest therapeutic doses
antagonistic drug’s effect ➢ Maximum effect (Emax)

RELATION BETWEEN DRUG DOSE AND CLINICAL


RESPONSE
Dose and Response in patients
▪ Graded dose-response relations
▪ Shape of dose-response curves
▪ Quantal dose-effect curves
GRADED DOSE-RESPONSE RELATIONSHIP
1. Potency
➢ Ability of the drug molecules to reach the
receptors and bind to these receptors
➢ Refers to the concentration (EC50) or dose

22
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
responses include: relief of headache for an
antimigraine drug, increase in heart rate of at
least 20 bpm for a cardiac stimulant, or 10
mmHg fall in diastolic blood pressure for an
antihypert
ensive.

In Graded Dose-Response Curves (relationship), data is


obtained individually from a few study participants.
QUANTAL DOSE-EFFECT RELATIONSHIP
▪ Therapeutic index employs the use of the
quantal dose-effect curves
o Quantal Response = all or none
response
o The therapeutic index of a drug is the
ratio of the dose that produces toxicity
to the dose that produces a clinically
desired or effective response in a
population of individuals.
Therapeutic index

▪ TD50 is the dose of drug that causes a toxic


response in 50% of the population
▪ ED50 is the dose of drug that is therapeutically
effective in 50% of the population.
▪ Both ED50 and TD50 are calculated from
quantal dose response curves (QDRC), which
represent the frequency with which each dose
of drug elicits the desired response or toxic
effect in the population
Characteristics of QDRC
▪ Dose of drug in plasma is plotted in the
horizontal axis while the percentage of
individuals (animals or humans) that responds
or shows a toxic effect is represented in the
vertical axis.
▪ These curves measure all or none (positive or
negative) responses. Some examples of positive

23
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
6. Affinity Propensity of a drug to bind with a
receptor site
7. Michaelis-Menten The magnitude of a response
is attributed to the amount of available
receptors sites and the quantity of drug
molecules
8. Agonist Activates the receptor by binding at the
drug binding site
9. Antagonist Competes with the agonist for the
same binding site
10. Drug-Receptor Complex Product of a drug-
receptor coupling

Examples of drugs with Narrow TI


▪ Warfarin
▪ Lithium
▪ Digoxin
▪ Phenytoin
▪ Gentamycin
▪ Amphotericin B
▪ 5-fluorouracil
▪ AZT (zidovudine)

POP QUIZ
1. Receptor A macromolecule that binds with a
cell or ligand to produce a response
2. Allosteric Activator Binds to the receptor at a
separate site and increases the response
produced
3. Drug Substance that brings about a change in
biologic function through its chemical actions at
the molecular level
4. Quantal Relationship All-or-none response
5. Pharmacodynamics Actions of the drug on the
body

24
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
central nervous system through the cranial nerves
AUTONOMIC PHARMACOLOGY (especially the 3rd , 7th , 9th , and 10th ) and the third and
Dr. Perfecto Soriano fourth sacral spinal roots- “cranio-sacral”

Somatic Nervous System


➢ No ganglia present
➢ Largely non-autonomic
➢ Skeletal muscle is innervated by somatic
nerve nerves; controlling voluntary
actions
Autonomic Nervous System
➢ Has ganglia
➢ Largely autonomous (independent)
➢ Its activities are not under direct
conscious control

o Most of the sympathetic preganglionic fibers


terminate in ganglia located in the
paravertebral chains that lie on either side of
the spinal column.

o From the ganglia, postganglionic sympathetic


fibers run to the tissues innervated.

o The majority of parasympathetic preganglionic


fibers terminate on ganglion cells distributed
diffusely or in networks in the walls of the
innervated organs.
Enteric Nervous System

➢ a semiautonomous part of the ANS located in o Some preganglionic parasympathetic fibers


the GIT which specific function for the control terminate in parasympathetic ganglia located
of this organ system. outside the organs innervated.

• Consists of the myenteric o Because of the location of the ganglia, the


plexus (plexus of Auerbach) and preganglionic sympathetic fibers are short and
the submucous plexus (plexus the postganglionic fibers are long.
of Meissner)- they send sensory o Parasympathetic system: preganglionic fibers
input to the parasympathetic are longer and postganglionic fibers are short
and sympathetic nervous
systems and receive motor
output from them

The sympathetic preganglionic fibers leave the central


nervous system through the thoracic and lumbar spinal
nerves- "thoracolumbar system.”

The parasympathetic preganglionic fibers leave the

25
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
• Synthesized in the nerve terminal from
acetyl CoA (produced in mitochondria)
and choline (transported across the cell
membrane) by enzyme choline
acetyltransferase

• The rate-limiting step is probably the


transport of choline into the nerve
terminal - inhibited by hemicholinium

• Acetylcholine is actively transported into


its vesicles for storage by the vesicle-
associated transporter (VAT)- inhibited
by vesamicol

2. Release of acetylcholine

• Release of transmitter stores from


vesicles in the nerve endings requires the
Neurotransmitter Aspects of the ANS entry of calcium through calcium
Transmitter channels and triggering of an interaction
between SNARE(soluble N-
• Transmitter crosses the cleft by diffusion and
ethylmaleimide-sensitive-factor
activates or inhibits the postsynaptic cell by
attachment protein receptor) proteins.
binding to a specialized receptor molecule.
• SNARE proteins include v-SNARES
• Acetylcholine ( Ach)
associated with vesicles (VAMPs-
• Norepinephrine ( NE)
vesicle-associated membrane proteins:
• Dopamine
synaptobrevin, synaptotagmin) and t-
• The synthesis, storage, release, receptor
SNARE proteins associated with the
interactions, and termination of action of the
nerve terminal membrane (SNAPS,
neurotransmitters all contribute to the action of
synaptosome-associated proteins:
autonomic drugs
SNAP25, syntaxin and others)

Cholinergic Transmission • This interaction results in docking of the


vesicle to the terminal membrane and,
• Acetylcholine (Ach) is the primary transmitter in
with influx of calcium, fusion of the
all autonomic ganglia and at the synapses
membrane of the vesicles with the
between parasympathetic postganglionic
nerve ending membranes, the opening
neurons and their effector cells (cholinergic
of a pore to the extracellular space, and
fibers)
the release of stored transmitter.
• It is the primary transmitter at the somatic
• Botulinum toxins enzymatically alter
(voluntary) skeletal muscle neuromuscular
synaptobrevin or one of the other
junction.
docking or fusion protein to prevent the
1. Synthesis and storage

26
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
release process ADRENERGIC TRANSMISSION

3. Termination of action of acetylcholine Norepinephrine (NE) is the primary transmitter at the


sympathetic postganglionic neuron- effector cell
• The action of acetylcholine in the synapse is synapses in most tissues except sympathetic fibers to
normally terminated by metabolism to thermoregulatory (eccrine) sweat gland and vasodilator
acetate and choline by the enzyme sympathetic fibers in skeletal muscle (adrenergic fibers)
acetylcholinesterase in the synaptic cleft.
Dopamine may be a vasodilator transmitter in renal
• The product are not excreted but are blood vessels but NE is the vasoconstrictor of these
recycled in the body vessels

1. Synthesis and storage


• Inhibition ofacetylcholinesterase is an
important therapeutic (and potentially • The synthesis of dopamine and NE requires
toxic) effect of several drugs. several steps:
• After transport across the cell membrane,
4. Drug effects on synthesis, storage, release and
tyrosine is hydroxylated by tyrosine hydroxylase
termination of action of acetylcholine
to DOPA (dihydrophenylalanine),
• Synthesis- hemicholinium decarboxylated to dopamine; and (inside the
• Storage- vesamicol vesicle) hydroxylated to NE
• Release- botulinum toxin • Tyrosine hydroxylase is inhibited by metyrosine
• NE and dopamine are transported into the
vesicles and stored there
• Monoamine oxidase (MAO) is present on
mitochondria in the adrenergic nerve ending
and inactivates a portion of the dopamine and
NE in the cytoplasm
• MAO inhibitors may increase the stores of these
transmitters and other amines in the nerve
endings
• Vesicular transporter can be inhibited by
reserpine- resulting in depletion of transmitter
stores.

2. Release and termination of action

• Dopamine and NE are released from their nerve


endings by the same calcium-dependent
mechanism responsible for acetylcholine
release
• Metabolism is not responsible for termination
of action of catecholamine transmitters-
dopamine and NE
• Diffusion and reuptake (norepinephrine
transporter, NET or dopamine transporter, DAT)
reduce their

27
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
concentration in the synaptic cleft and stop
their action.
• Outside the cleft, these transmitters can be
metabolized- by MAO and catechol-O-
methyltransferase (COMT)- and the product of
these enzymatic reactions are excreted
• Determination of metanephrine,
normetanephrine,3-methoxy-4-
hydroxymandelic acid (VMA) and other
metabolites provides a measure of the total
body production of catecholamines
• Inhibition of MAO increases stores of
catecholamines and has both therapeutic and
toxic potentials

3. Drugs effects on adrenergic transmission

• Block NE synthesis- metyrosine

• Catecholamine storage- reserpine

• Catecholamine release- guanethidine

• Promote catecholamine release- amphetamine-


like agents

COTRANSMITTERS

• ATP
• Enkephalins
• Vasoactive intestinal peptide
• Neuropeptide Y
• Substance P
• Neurotensin
• Somatostatin
• The main role in autonomic function appears to
involve modulation of synaptic transmission
• The same substance function as primary
transmitters in other synapses

28
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE

CHOLINERGIC RECEPTORS

Acetylcholine receptor subtypes :


1. muscarinic
2. nicotinic

The term cholinoceptor denotes receptors (both


muscarinic and nicotinic) that respond to acetylcholine.

ADRENERGIC RECEPTORS

The term adrenoceptor is widely used to describe


receptors that respond to catecholamines such as
norepinephrine.

The adrenoceptors can be subdivided into (α-


adrenoceptor and β-adrenoceptor types on the basis of
both agonist and antagonist selectivity.

AUTONOMIC RECEPTORS

Cholinoceptors
Adrenoceptors
Dopamine receptors

DOPAMINE RECEPTORS

Describe receptors that respond to dopamine.

DA receptors: mesenterium, renal, cardiovascular


system, brain

Presynaptic regulation

29
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE

Functional Organization of Autonomic Activity


(Function of receptors)

A basic understanding of the interactions of autonomic


nerves with each other and with their effector organs is
essential for an appreciation of the actions of
autonomic drugs, especially because of the significant
"reflex" effects that may be evoked by these agents.

30
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE

31
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
For some organs, sensory fibers associated with the
parasympathetic system exert reflex control over motor
Central Integration
outflow in the sympathetic system.
• At the highest level--midbrain and medulla—
Thus, the sensory carotid sinus baroreceptor fibers in
the two divisions of the autonomic nervous the glossopharyngeal nerve have a major influence on
system and the endocrine system are integrated sympathetic outflow from the vasomotor center.
with each other, with sensory input, and with
information from higher central nervous system
centers. Similarly, parasympathetic sensory fibers in the wall of
• These interactions are such that the the urinary bladder significantly influence sympathetic
parasympathetic system a trophotropic one (ie, inhibitory outflow to that organ.
leading to growth) and the sympathetic system
Within the enteric nervous system, sensory fibers from
an ergotropic one (ie, leading to energy
the wall of the gut synapse on both preganglionic and
expenditure) that was activated for "fight or postganglionic motor cells that control intestinal
flight." smooth muscle and secretory cells.
• Sympathetic system is most active when the
body needs to react to changes in the internal
or external environment Integration of Cardiovascular Function
• During rage or fright the sympathetic system
• Autonomic reflexes are particularly important in
can discharge as a unit--affecting multiorgan
understanding cardiovascular responses to
systems.
autonomic drugs.
• Autonomic and hormonal control of
cardiovascular function.
At a more subtle level of interactions in the brain stem,
medulla, and spinal cord, there are important • The sympathetic nervous system directly
cooperative interactions between the parasympathetic influences four major variables: peripheral
and sympathetic systems. vascular resistance, heart rate, force, and
venous tone. It also directly modulates renin
production.
• The parasympathetic nervous system directly
influences heart rate.

32
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE

Pharmacologic Modification of Autonomic Function

• Because transmission involves different


mechanisms in different segments of the
autonomic nervous system, some drugs
produce highly specific effects while others are
much less selective in their actions.

The way on effects of autonomic drugs

1) Direct effect on receptor

• agonist:receptor →→ activation, similar to


transmitter
Presynaptic & Postsynaptic Regulation
• antagonist:receptor →→blockade
• Up- and down-regulation are known to occur in
2) To influence transmitter
response to decreased or increased activation,
respectively, of the receptors. • act on the transmitter biochemical synthesis
• Up- regulation : receptor number increase • act on the transmitter metabolism (enzymatic
inactivation of transmitter)
An extreme form of up-regulation occurs after
• act on the transmitterrelease
denervation of some tissues, resulting in
• act on the transmitter storage
"denervation supersensitivity" of the tissue to
activators of that receptor type

• down-regulation : receptor number decrease

33
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
NEUROTRANSMITTERS AND NEURORECEPTORS
CHOLINERGIC AGONISTS  Acetylcholine andnorepinephrine
Dr. Perfecto Soriano  All preganglionic neurons are cholinergic
 Parasympathetic postganglionic neurons are
CHOLINORECEPTOR – ACTIVATING (CHOLINERGIC cholinergic
AGONIST) AND CHOLINESTERASE – INHIBITING DRUGS
 Sympathetic postganglionic neurons are
adrenergic except sympathetic innervating
PARASYMPATHETIC DIVISION sweat glands, blood vessels in skeletal
muscle, and piloerection muscles, which are
 Preganglionic cell bodies in nuclei of brainstem or cholinergic
lateral parts of spinal cord gray matter from S2
– S4
o Preganglionic axons from brain pass to PARASYMPATHETIC NERVOUS SYSTEM
ganglia through cranial nerves RECEPTORS: CHOLINERGIC RECEPTORS
o Preganglionic axons from sacral region
pass through pelvic nerves to ganglia 1. MUSCARINIC RECEPTORS
 Preganglionic axons to terminal ganglia within - Associated with parasympathetic
wall of or near organ innervated functions and are located at the ends of
postganglionic neurons in peripheral
SITES OF CHOLINERGIC ACTIVTIY tissues (effectors, eg. Internal organs,
 Preganglionic synapses of both sympathetic and smooth muscle) innervated by the
parasympathetic ganglia parasympathetic system
 Parasympathetic postganglionic neuroeffector - Stimulation of the muscarinic receptors
junctions may result in either excitation or
 All somatic motor end plates on skeletalmuscles inhibition, depending on the organ
involved
A membrane protein: upon stimulation by
neurotransmitter, it causes the opening of ion channels
indirectly, through a second messenger. For this reason,
the action of a muscarinic synapse is relatively slow

34
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
MUSCARINIC RECEPTORS AND ITS LOCATION
 M1 receptor – nerve endings
 M2 receptor – heart and some nerve endings
 M3 receptor – effector cells, smooth muscle,
glands and endothelium
 M4 & M5 – CNS

MUSCARINIC ACTION
 Eye (miosis and set lens for near vision)
 Smooth muscle, exocrine glands
o Bronchoconstriction, ↑secretion, ↑
GI motility
o ↑ urination
 Heart & vascular beds (vagal stimulation
o ↓SA & AV conduction velocity
o ↓ force of atrial contraction DIRECT ACTING CHOLINERGIC AGONIST
o ↓ vascular resistance – due
to activation of receptors on CHOLINE ESTERS
endothelium
  ABSORPTION: polarity dependent (poor for
EDRF (endothelium Ach, quaternary ammonium), intravenous,
derived relaxation factor) subcutaneous and intramuscular for local
o Effects modified by reflexes effects (Ach)
 METABOLISM: highly dependent on the
2. NICOTINIC RECEPTORS susceptibility to acetylcholinesterase (AChE)
- Located in the autonomic ganglia of
both the sympathetic and
parasympathetic systems and the nerve
endings of the somatic motor system PROPERTIES OF CHOLINE ESTERS
(eg. motor nerves and skeletal muscle)
- Stimulation results in  Table 7 – 2 (page 100)
muscle contraction
- A channel protein that, upon binding by
acetylcholine, opens to allow diffusion
of cations
- NN – autonomic ganglia (NN)
- NMJ (NM) – muscle contraction 
paralysis

35
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
PHARMACOLOGIC EFFECTS MODES OF ACTION
 EYES  Direct acting cholinergics are lipid insoluble
o Contraction of ciliary muscle and  Do not readily enter the CNS so effects are
smooth muscle of the iris sphincter peripheral
(miosis) – aqueous humor outflow,  Resistant to metabolism by acetylcholinesterase
drainage of the anterior chamber (except for Acetylcholine chloride)
 Effects are longer acting than with acetylcholine
 CARDIOVASCULAR
(endogenous neurotransmitter)
o Bradycardia (possibly preceded by
 Widespread systemic effects when they
tachycardia)
combine with muscarinic receptors in cardiac
o Vasodilation (all vascular beds including
muscle, smooth muscle, exocrine glands and
pulmonary and coronary – M3) and
the eye
hypotension
o Reduction of the contraction strength
ADVERSE EFFECTS
(atrial and ventricular cells, IK+, Ica2+,
 Major contraindications:
diastolic depolarization, NO –
o Asthma  Bronchoconstrictor
inhibitable ATP?), negative inotropic &
actioncould precipitate an asthmatic attack
chronotropic effect (inhibition of
o Hyperthyroidism  Hyperthyroid
adrenergic activation), variable BP
patients may develop atrial fibrillation
effects
o Coronary insufficiency  Hypotension
induced by these agents can severely
 GI
reduce coronary blood flow, especially
o Increases in tone, amplitude of
if it is already compromised
contractions, and peristaltic activity of
o Acid – peptic disease  The gastric
the stomach and intestines
acid secretion produced by the choline
o Enhances secretory activity of the
esters can aggravate the symptoms of
salivary glands and GIT
acid – peptic disease
 URINARY BLADDER
POSSIBLE SIDE EFFECTS
o Increase ureteral peristalsis
 Sweating (very common)
o Contract the detrusor muscle of the
 Abdominal cramps
urinary bladder
 Sensation of tightness in the urinary bladder
o Increase the maximal voluntary voiding
pressure  Difficulty in visual accommodation for far vision
o Decrease the capacity of the bladder  Headache
o Relaxation of the sphincter  Salivation

 OTHER EFFECTS *Antidote – ATROPINE


o Increased secretion from all glands that
receive parasympathetic innervations *Epinephrine may be used to overcome severe
(salivary, lacrimal, tracheobronchial – cardiovascular or bronchoconstrictor responses.
increased respiratory secretion,
digestive and exocrine sweat glands)
o Increased tone and contractility of
bronchial smooth
muscle: BRONCHOCONSTRICTION

36
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
1. ACETYLCHOLINE  Pharmacologic effects
 Quaternary ammonium ester that is rapidly o Similar to acetylcholine
hydrolyzed by acetylcholinesterase and plasma
cholinesterase  Methacholine choride
 Therapeutic uses: o acetyl-b-methylcholine chloride
- Used as a MIOTIC in cataract surgery o PROVOCHOLINE
o May be administered for diagnosis of
bronchial hyperreactivity and asthmatic
FIGURE 7 -2 (pg. 99) conditions
 Molecular structures of four choline esters.
 Acetylcholine and methacholine are acetic acid 3. CARBACHOL
esters of choline and B-methylcholine,  Has a carbamic acid – ester link, which is not
respectively. readily susceptible to hydrolysis by
 Carbachol and bethanechol are carbamic acid cholinesterases
esters of the same alcohols.  Has all the pharmacologic properties of
acetylcholine. It exerts both nicotinic and
muscarinic effects.
FIGURE 7 – 3 (pg. 100)  Used ophthalmically as a miotic agent
 Structures of some cholinomimetic alkaloids

4. BETHANECHOL
 Structural features of both methacholine and
carbachol.
 Resistant to hydrolysis by cholinesterases
 Mainly muscarinic in action
 Used therapeutically for abdominal distention,
esophageal reflux, and urinary bladder
distention
 Alternative to pilocarpine to promote salivation
– Xerostomia (dryness of the mouth)
 Sjogren syndrome (immunologic disorder with
destruction of the exocrine glands) leading to
mucosal dryness
 Should be administerd ONLY by the oral or Q S
2. METHACHOLINE route for systemic effects
 Chemistry  Also used locally in the eye
o Differs chemically from acetylcholine by
the addition of a methyl group to the B
position of a choline. As a result:
• Hydrolyzed only by
acetylcholinesterase  has
a longer duration of action
than acetyl choline
• It becomes virtually a pure
muscarinic – acting agent

37
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
1. PILOCARPINE OTHER TERTIARY AMINES

 Isolated from Pilocarpus jaborandi, P.  NICOTINE


Microphyllus o A liquid, sufficiently lipid – soluble to be
 Plant family: Rutaceae (citrusfamily) absorbed across the skin
 Common name: Jaborandi (means slobber o Used as gum or transdermal patch
mouth plant in Tupi language) o Duration of action: 4 – 6 hours
 Origin: lowland wet forests of Tropical America, o Toxic effect associated with smoking
West Indies o Acute toxicity:
 Ethnomedical uses: members of the Tupiculture • Central stimulant actions
in Brazil chew leaves to induce salivation and 
sweating coma, respiratory arrest
 Chemical class: tertiary amine alkaloid • Skeletal muscle endplate
 MOA: cholinergic receptor agonist with strong depolarization blockade 
postganglionic (muscarinic receptor) depolarization blockade and
stimulation and mild ganglionic (nicotinic respiratory paralysis
receptor) stimulation • Hypertension and cardiac
 Muscarinic receptor stimulation effects: arrhythmias
o Salivation • Fatal dose: 40 mg or 1 drop of
o Intestinal motility pure liquid
o Pupil constriction o Treatment:
• Symptom directed
THERAPEUTIC USES
 Topical agent on eyes – constricts pupils  SUCCINYLCHOLINE
o Treats open – angle glaucoma by o N – receptor, moderately selective for
reducing intraocularpressure neuromuscular endplate (NM)
 Oral administration treats dry mouth o NM relaxation
(xerostomia) o Highly polar, IV administration
 Also used to reduce side effects of morphine o Duration of action: 5 to 10 minutes
treatment, including: o Treatment:
o Dry mouth • Symptom directed
o Constipation • Muscarinic excess resulting
o Urinary retention from parasympathetic
 Overdose may cause cardiovascular collapse ganglion stimulation can be
 Antedote is atropine (conversely, pilocarpine si controlled by atropine
sometimes used as an antedote in cases of • Central stimulation is usually
atropine poisoning) treated with anticonvulsants
o Chronic toxicity:
• Cardiovascular diseases

38
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
o Diarrhea
o Hypotension
2. MUSCARINE
o Bradycardia
 Compound isolated from various species of
o Bronchospasm
fungi:
o Amanita muscaria
o Amanita phalloides – Amanita phalloides: inhibits mRNAsynthesis – 24 h
deadly nightcap symptom free period followed by liver and kidney
o Amanita spp. malfunction, death within 4 to 7 days
o Inocybe spp.  Oxotremorine effects: neurobiological
o Clitocybe spp. research
 Antedote: 1 – 2 mg of atropine IM every 30
 Common name: amanita, fly agaric
mins
 Origin: Siberia, North America
 Habit: mushroom with red, orange, yellow 3. ARECOLINE
or cream colored cap with white spots.  From Areca catechu
Grows on forestfloor.  Plant family: Arecaceae (palmfamily)
 Common name: betelnut, betel, areca nut
 Ethnomedical/cultural uses:  Origin: tree in wet forests of Southeast Asia,
o Mushroom eaten by Indomalaysia, Oceania, probably originally from
Siberian indigenous people as
Sulawesi (Indonesia)
hallucinogen
 Ethnobotanical use:
o Dried mushrooms repel flies
o Used as a masticatory (chew): the
areca seed is rolled in leaves of Piper
 Compounds with hallucinogenic (CNS)
betel and mixed with gambir (a spice
activity:
made from the boiled leaves of Unicaria
o Ibotenic acid
gambir) and shell lime (which changes
o Muscimole
pH to release active compounds
o Muscazone
 Ethnomedical uses:
 Active compound: muscarine o Euphoretic
o Cardiac tonic
 Chemical class: quaternary ammonium
o Energizer
alkaloid
o Antihelminthic
o Less completely absorbed from the
 Chemical structure: arecoline is an alkaloid
GIT than tertiary amines
 Chemical derivative: arecoline 
aceclidine(glaucoma treatment in Europe)
 Chemical derivative: muscarine
  Arecoline stimulates both muscarinic
oxotremorin and nicotinic receptors (in ANS and
e CNS)

 Muscarinic effects:
o Diaphoretic
o Salivation
o Lacrimation
o Visual problems
o N/V

39
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
INDIRECTLY – ACTING CHOLINERGIC membrane
AGONIST ANTICHOLINESTERASE o Duration of action: 0.5 – 2 hours
(AChE)
PHARMACOLOGIC EFFECTS
2 MAJOR CHEMICAL CLASSES  Mimics the pharmacologic effects
 CARBAMIC ACID ESTERS (CARBAMATES): of acetylcholine
reversible  Enters and stimulates the CNS
o Physostigmine  Produces miosis – can antagonize the mydriasis
o Neostigmine induced by atropine – topical use
o Pyridostigmine  Duration of action: 2 – 4 hours
 PHOSPHORIC ACID  In large doses: causes fasciculation, then
ESTERS paralysis of skeletal muscle because of the
(ORGANOPHOSPHATES): irreversible accumulation of acetylcholine at the
o
Insecticide neuromuscular junction that results when
 Parathion acetylcholine is not broken down.
 Malathion
 Diisopropyl phosphofluoridate THERAPEUTIC EFFECTS
(DFP)  Treatment of atropine, phenothiazine, and
 Echothiophate tricyclic antidepressant intoxication
o Isofurophate  Treatment of glaucoma, especially simple and
 *EDROPHONIUM: is an alcohol (NOT an ester) secondary glaucoma
 Treatment of early stages of Alzheimer’s
MODE OF ACTION disease because of degeneration of cortical
 Both carbamates and organophosphate cholinergic axons
inhibitors bind to cholinesterase and udergo
prompt hydrolysis  the alcohol portion of ADVERSE EFFECTS
themolecule is then released.  Convulsion (high dose)
 The acidic portion (carbamate ion andphosphate  Bradycardia
ion) is released more slowly, and this retained  Paralysis of skeletal muscle (ACh accumulation
portion prevents the binding and hydrolysis of at NMJ)
endogenous acetylcholine, thus amplifying
acetylcholine effects whenever the transmitter
is released. 2. NEOSTIGMINE
 Synthetic reversible anticholinesterase that
A. CARBAMATES contains a quaternary nitrogen

1. PHYSOSTIGMINE MECHANISM OF ACTION


 Moderately polar but active orally
MECHANISM OF ACTION  Does not penetrate the blood-brain-barrier 
 Forms a reversible complex at the site of minimizes the toxicity due to inhibition of
acetylcholinesterase where acetylcholine is broken down acetylcholinesterase
 Both muscarinic and nicotinic receptors  It is destroyed by plasma esterases and is
 Pharmacokinetics: excreted in the urine
o Well – absorbed from the GIT, Duration of action: 2 – 4 hours
subcutaneous tissues and mucous

40
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
*EDROPHONIUM (SIMPLE ALCOHOL)
PHARMACOLOGIC EFFECTS  Reversible anticholinesterase
 Mimics the pharmacologic effects of  Pharmacokinetics:
acetylcholine o More rapidly absorbed and has a
 It has a direct action on nicotinic receptors n i shorter duration of action than
addition to blocking acetylcholinesterase neostigmine
 It reverses the neuromuscular blockade o Administered parenterally
produced by curare and its derivatives o Duration of action: 5 – 15 minutes
 The mechanism of action involves the release of  Pharmacologic effects:
increased amounts of acetylcholine from nerve o Similar to neostigmine except that with
endings, cholinesterase inhibition, and a direct doses it can stimulate the
action on skeletal muscle cholinergic receptors. neuromuscular junction without
affecting muscarinic effector organ
THERAPEUTIC USES  Therapeutic uses:
 Used to reverse the effects of competitive o Diagnosis of myasthenia gravis
blocking agents o Antagonism of curare – like agents
 Management of paralytic ileus and atony of the
urinary bladder
 Symptomatic treatment of myasthenia gravis B. ORGANOPHOSPHATE CHOLINESTERASE
INHIBITORS (IRREVERSIBLE
ANTICHOLINESTERASE
3. PYRIDOSTIGMINE
 Used in the chronic management of myasthenia 1. DIISOPROPLYFLUROPHOSPHATE (DFP)
gravis  Forms a covalent bond between its phosphorus
 Duration of action: 3 – 6 hours (longer than atom and esteratic site of cholinesterase
Neostigmine)  Limited to treatment of certain types of
 Adverse effects: generalized cholinergic glaucoma
stimulation
2. PARATHION
OTHER PHYSOSTIGMINE – LIKE  Used as insecticide
ANTICHOLINESTERASE  Active form is the metabolite – maraoxan
 Ambenonium – used in the symptomatic  Pharmacologic effects similar to DFP
treatment of myasthenia gravis
 Demecarium – used as an ophthalmic solution for 3. ECHOTHIOPHATE
the treatment of chronic glaucoma  Long – acting organophosphate cholinesterase
inhibitor with pharmacologic properties similar
to DFP
 Spontaneous regeneration of phosphorylated
enzyme can occur
Major use is in the treatment of glaucoma

41
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
MECHANISM OF ACTION of MUSCARINIC
CHOLINERGIC ANTAGONIST ANTAGONIST
Dr. Perfecto Soriano
• Act like competitive pharmacologic
antagonists: their blocking effect can be
ANTICHOLINERGIC DRUGS overcome by increase concentrations of
muscarinic agonist
1. ANTIMUSCARINIC
• M1 – Selective EFFECTS:
• Non – Selective
• Tissues sensitive to atropine: salivary
glands, bronchial and sweat glands:
2. ANTINICOTINIC
Intermediate – smooth muscles heart
• Ganglionic Blockers
• Atropine is highly selective to muscarinic
• Neuromuscular Blockers
receptor
MUSCARINIC ANTAGONIST • Systemic administration - short half life of –
2h
1. ATROPINE • Topical ocular administration – longer half
• Lipid from Atropa belladonna or deadly life
nightshade o Bind to iris pigments and are
• Naturally occurring atropine 1 (-) released over days, dark irises bind
hyoscyamine more
• Tertiary amine
• Clinical uses: Eye, CNS, PHARMACOLOGIC EFFECTS -Muscarinic
Antihistamines, Antipsychotic, receptor antagonists inhibit parasympathetic nerve
Antidepressant stimulation
• Relatively lipid soluble, well-absorbed • Relax Smooth Muscle
from the gut and across conjunctival • Increase Hear Rate
membrane • Increase conductivity in the heart
• Distribution is 30 minutes - 1 hour • Inhibit exocrine gland secretion

2. SCOPOLAMINE These effects are dose dependent – the higher the


• Derived from Hyoscyamusniger or dose the higher the effect
henbane
• Well distributed in CNS, rapidly
disappear in the blood, partially HEART
metabolized in the liver and partially • Standard dose
eliminated in the kidneys o Locks the effect of vagus nerve
• Elimination half-life - 2hours ▪ Increase heart rate
• Duration of action – 4-8 hours ▪ Increases AV conduction velocity
• Eyes – 72 hours or longer • Low dose
o When delivered by IV at low dose slows
3. IPATROPIUM heart rate by stimulating vagal motor
• Atropine Analog nucleus in the brain
• Used to treat sinus Bradycardia which can lead to
hypotension, ischemia if left untreated

42
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
CENTRAL NERVOUS SYSTEM GIT

• Atropine, scopolamine • Dramatic effects in motility and secretory


o Block muscarinic receptors functions of the gut
▪ Sedation, Excitement • Dry mouth is the frequent symptom
• Scopolamine • Less effective blockade of gastric secretion
o More sedating than atropine • Large dose may of atropine – decrease the acid
o Used as an adjunct to anesthesia pepsin and mucin secretion
o Rapidly and fully distributed in the CNS • Pancreatic and intestinal secretion less affected
than any other antimuscarinic drugs • Motility – stomach – colon diminished
o Slower long lasting sedative effects in the contraction
brain • Increased intestinal transit time
o Reduces tremors in Parkinson’s Disease • Temporary intestinal paralysis reestablish in 1-3
o Has marked central effects producing days
drowsiness and amnesia
o Most effective drug against motion sickness GUT
o Toxic dose: excitement, agitation,
• Ureter and bladder wall relaxed
hallucination
• Have no significant effects on the uterus
• Atropine
o Mild stimulation followed by a slower and SWEAT GLANDS
longer – lasting sedative effect
o Higher dose - > delirium, hallucination • Thermoregulatory sweating is suppressed by
• Ipatropium atropine
o Atropine analog • In adult – temperature is increased

EYES THERAPEUTIC APPLICATIONS

• Mydriasis • CNS
• Cycloplegia o Parkinson’s Disease
• Reduction lacrimal secretion o Motion Sickness
• Antimuscarinic Drugs used in Pharmacology • Ophthalmologic Disorders
Duration of effect • Respiratory Disorder
Atropine 7 – 10 days o Ipatropium is used in asthma and CPD
Scopolamine 3 – 7 days
Homatropine 1 – 3 days
Cyclopentolate 1 day • Cardiovascular disorder
Tropicamide 0.25 day
o Mark vagal discharge – pain of MI –
Sufficient depression of SA and AV
RESPIRATORY SYSTEM o Node function
▪ Impair CO
• Bronchodilator
• Gastrointestinal Disorders
• Decreases secretion in the trachea and the
• Urinary disorders
possibility of laryngospasm
o Urinary urgency urolithiasis to relieve
• Ipatropium – inhalation in case of
spasms
bronchoconstriction
o Telterodine
o Imipramine – antidepressant agent used in
incontinence
o Propiverine

43
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
• Cholinergic Poisoning
• Hyperhidrosis
EFFECTS OF GANGLIONIC BLOCKING DRUGS
ADVERSE REACTION CNS
• Atropine fever • Reduction of nicotine craving and
• Acute angle – closure glaucoma ameboration(DI KO MAINTINDIHAN :D) of
• Bladder retention tourette’s syndrome(mecamylamine)
• Constipation
• Sedation, amnesia, hallucination, delirium and Eye
convulsions • Moderate mydriasis and cycloplegia
• Intraventricular conduction blockade, dilation of
cutaneous vessels “atropine flush” Bronchi

CONTRAINDICATION • Little effect

• Hyperthemia GI Tract
• Glaucoma • Markedly reduced Motility

GU Tract
NICOTINIC RECEPTOR ANTAGONIST • Reduced contractility of the bladder impairment
• Ganglionic blocking agents (e.g. Trimethaphan) of erection and ejaculation
o Block Nn Receptors at Sympathetic and
Heart
parasympathetic
o Effect on a tissue, depends on sympathetic • Slight tachycardia in young adults, reduction in
or parasympathetic system is dominant force of contraction and cardiac output
o No longer are used to treat chronic
hypertension Blood Vessels
o Trimethaphan is occasionally used in cases • Reduction in arteriolar tone
of hypertensive emergency, when extremely • Mark reduction in venous tone
high blood pressure must be lowered rapidly
• BP decrease
• Severe orthostatic hypotension
• Ganglionic Blocking Drugs
Glands
Chemistry and Pharmacokinetics
• Reductions in salivations, lacrimations, sweating
• Synthetic amine
and gastric secretion
• Tetraethylammonium (TEA) – short
duration of action Skeletal muscle
• Hexamethonium – first effective drug used
• No significant effect
to treate hypertension
• Mecamylamine – secondary ammonium
compound with improved GI absorption –
only ganglionic blocking agent available in
the US
• Trimethapan – inactive orally; IV infusion to
treat malignant hypertension – produces
controlled hypotension

44
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
Neuromuscular Blocking agents
- Inhibit neurotransmission at skeletal muscle
ADRENERGIC AGONISTS
- Important for producing complete skeletal Dr. Perfecto Soriano
relaxtion in surgery
- Causing muscle weakness and paralysis ADRENERGIC AGONIST SYMPATHOMIMETIC
1. Non-depolarizing blockers DRUGS
2. Depolarizing Blockers Includes:
• Sympathomimetic drugs
NONDEPOLARIZING NEUROMUSCULAR
BLOCKING AGENTS( CURARIFORM DRUGS) • Adrenomimetic drugs
• Adrenergic agonists
• positively charged e.g. Pancuronium, Tubocurarine
• Adrenoceptor agonists
• are not well absorbed from the GUT, ence they do
not cause poisoning when ingested with • ultimate effects of sympathetic stimulation are
mediated by release of norepinephrine from nerve
contaminated meat
terminals, which then activates adrenoceptors on
• do not cross blood-brain barrier postsynaptic sites
• Competitive antagonists of Ach at Nm receptors
• epinephrine acts as a hormone, whereas
• Produces a competitive block at the end plate norepinephrine acts as a neurotransmitter.
nicotinic receptor
• Drugs that mimic the actions of epinephrine or
• Causing flaccid paralysis that last 30- 60 mins norepineph- rine have traditionally been termed
• Paralyze: sympathomimetic drugs.
o small fast moving muscles of eyes and face
o larger muscles of the limbs and trunk Sympathomimetic drugs
o the intercostal muscles and the diaphragm • direct agonists; that is, they directly interact with and
• is used for relaxation of abdominal muscles for activate adrenoceptors.
surgical procedures without producing apnea • Others are indirect agonists because their actions are
dependent on their ability to enhance the actions of
SIDE EFFECTS endogenous catecholamines.
-These indirect agents may have either of two
• stimulate mast cells to release histamine different mechanisms:
• tachycardia (1) they may displace stored catecholamines from the
• hypotension adrenergic nerve ending (eg, the mechanism
• bronchospasm of action of tyramine), or they may decrease the
clearance of released
DEPOLARIZING NEUROMUSCULAR norepinephrine either by
BLOCKING AGENTS (2a) inhibiting reuptake of catecholamines already
released (eg, the mechanism of
Succinylcholine action of cocaine
(2b) preventing the enzymatic metabolism of
• binds to Nm receptors norepinephrine (monoamine
• 1st transient muscle contraction followed by oxidase and catechol-O-methyltransferase
sustained muscle paralysis inhibi- tors).
• Ultra short duration action (5-10mins)
Review of Sympathetic Activation
• Produce muscle relaxation before and during
surgery • ‘Fight’or‘Flight’onStress
• Heart
↑HR,contractility,conductionvelocity
• Vessels (arterioles)

45
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
-Skin, cutaneous, visceral : constrict
-Skeletal muscle, coronary: dilate
• Vessels (Vein): constrict Types and subtypes of adrenoceptors

• Eye • Adrenergic receptors located on smooth muscle,


-Radialmuscle,iris: contract cardiac muscle, exocrine glands, endocrine glands and
-Ciliarymuscle: relaxforfarvision on nerve terminals.

• Lung • the transmitter in all adrenergic neurons is NE


-Trachealandbronchialmuscle: relax • When NE and Epinephrine interacted with an
adrenoceptor, in some tissues the response is
Stomachandintestine excitatory while in other tissues it is inhibitory
• ↓Motility and tone • Two subtypes of adrenoceptors
• Sphincters : contraction • a - excitatory in most tissues(except - intestinal
smooth muscle)
• Secretion (intestine): inhibition
• b - inhibitory in most tissues (except - heart)
• Urinarybladder
Detrusororbladderwall: relax Rank Order of Potency
Trigone,sphincter: constrict
• Posteriorpituitary: ADHsecretion
• Liver: glycogenolysis,gluconeogenesis Pancreatic
B cell---stimulate insulin releaseSkeletalmuscle
• ↓contractility, glycogenolysis, K+ uptake
• Fatcells: lipolysis
• Uterus
-non-pregnant: relax
• Sweatgland: secretion
• Hair : piloerection

Adrenomimetic drugs
• The effects of adrenomimetic drugs are similar to
sympathetic activation. 1. α- receptorsEpi > NE >> Iso
2. β- receptorsIso > Epi > NE
• But why each adrenomimetic drug can produce
different responses? Type of adrenoceptor
• The differences in affinity to adrenoceptor subtypes • α1,α2
are responsible for different responses.
• β1,β2,β3n
• DA1, DA2

Mechanism of action of Adrenomimetic Drugs


• α 1 via coupling protein Gq
• α 2 via coupling protein Gi
• β1 ,β2,β3 viacouplingproteinGs
MECHANISM OF ACTION OF

46
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE

Classification of Sympathomimetics
• By chemistry
- Catecholamines
- Non-catecholamines
• By mode of action
-direct - acting
-indirect - acting
• By selectivity (to types of receptor)
Classification of Sympathomimetics
I. Catecholamines(CAs)
II. Non-catecholamines
A. Direct acting

ADRENOMIMETIC DRUGS • classifiedbyalpha,betareceptorsubtypes


• a1-selective,a2-selective,nonselective
• b1-selective,b2-selective,nonselective
Mechanism of action ofAdrenomimetic drug B. Indirect acting
-Releasers
Mechanism of action of Dopamine - Reuptake inhibitors
DA1 type
• cAMP Receptor types :
DA2 type 1. Alpha receptors
• cAMP - are coupled via G proteins in the Gq family to
Central Dopamine Receptor -different effect phospholipase C.
• D1-like: D1A, D1B, D5 - This enzyme hydrolyzes polyphosphoinositides,
leading to the formation of inositol 1,4,5-
• D2-like: D2, D3, D4 trisphosphate (IP3) and diacylglycerol (DAG). IP3
promotes the release of sequestered Ca2+ from
intracellular stores, which increases cytoplasmic free

47
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
Ca2+ concentrations that activate various calcium- CATECHOLAMINES: Norepinephrine and
dependent protein kinases. Epinephrine
- Alpha2 receptors are coupled to the inhibitory • Chemistry:
regulatory pro- tein Githat inhibits adenylyl cyclase a. Tyrosine is hydroxylated on the aromatic ring by
activity and cause intracellular cyclic adenosine tyrosine hydroxylase yielding dopa
monophosphate (cAMP) levels to decrease. (dihydroxyphenylalanine); this is the rate limiting step in
- It is likely that not only α, but also the β-γ subunits of biosynthesis of adrenergic transmitters.
Gi contribute to inhibition of adenylyl cyclase. b. Dopa is decarboxylated to give dopamine
B. Beta receptors c. Dopamine is hydroxylated on the β-carbon to give
- Activation of all three receptor subtypes (β1, β2, and norepinephrine
d. Epinephrineisformedintheadrenalmedullaby
β3) results in stimulation of adenylyl cyclase and
methylation of norepinephrine
increased cAMP
C. Dopamine receptor
Pharmacokinetics of Epinephrine and Norepinephrine
-D1 receptor is typically associated with the stimulation
a. Absorption is poor with oral administration because
of adenylyl cyclase
the drugs
-D2 receptors have been found to inhibit adenylyl
are rapidly conjugated and oxidized.
cyclase activity, open potassium channels, and decrease
b. Absorption is slow with subcutaneous administration
calcium influx.
because the drugs cause local vasoconstriction
c. Nebulized and inhaled solutions usually used for their
Receptor Regulation
actions on respiratory tract.
-desensitization may occur after exposure to
d. The drugs can be given IV, but route must be used
catecholamines and other sympathomimetic drugs.
with caution so that the heart does not fibrillate.
~One of the best-studied examples of receptor
e. The liver is an important in the degradation of
regulation
epinephrine and norepinephrine. The majority of the
~ Other terms such as tolerance, refractoriness, and
dose is metabolized by catechol O-methyltransferase
tachyphylaxishave also been used
(COMT) and monoamine oxidase (MAO), and
to denote desensitization
metabolites are excreted in the urine
~This process has potential clinical significance
because it may limit the therapeutic
response to sympathomimetic agents.
Pharmacologic Effects of Epinephrine
~major mechanism of desensitization that occurs
rapidly involves phosphorylation of • Interacts strongly with both β- and α-receptors.
receptors by members of the G protein- coupled • Its effects on some body systems depend on the
receptor kinase (GRK) family, of which concentration of epinephrine as well as type of
there are seven members. receptor.
~two major categories of desensitization of
responses mediated by G protein-coupled • At low concentration, β effects predominate, and, at
receptors: high concentration, α effects predominate.
1. Homologous desensitization
refers to loss of responsiveness exclusively of
the receptors that have been exposed to Effects onBloodPressure
repeated or sustained activation by an agonist. • A large dose of epinephrine administered IV causes
2. Heterologous desensitization an increase in blood pressure, with the systolic
refers to the process by which desensitization of pressure increasing more than the diastolic.
one receptor by its agonists also results
• The rise in pressure is due to:
in desensitization of another receptor that has not been
directly acti- vated by the agonist in - Vasoconstriction through activation of α- receptors.
question. - Increased ventricular contraction through activation
-Phosphorylation of these receptors enhances their of β- receptors
affinity for arrestins, a family of four proteins, of which - An initial increase in heart rate, which in the height
the two nonvisual arrestin subtypes are widely expressed of the vasopressor response, is slowed by a
compensatory vagal
DIRECT-ACTING SYMPATHOMIMETIC DRUGS discharge.

48
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
• Transport, or uptake into the cells, is an important
mechanisms for terminating the action of many
neurotransmitters. There are two active amine
Effects on the Heart transport systems:
• A direct effect on β1-receptors, producing a slight 1. A system in the plasma membrane transports amines
initial increase in heart rate, which is slowed by a from the extracellular fluids to the cytosol.
compensatory vagal discharge. 2. A system in the secretory granular membrane
• Increased in stroke volume transports amines into granules

• Increased in cardiac output • Deactivation by conversion to inactive metabolites


1. COMT adds a methyl group to the hnydroxyl of the
• A propensity toward arrhythmias catechol ring.
2. MAO removes the amine group. When metabolic
Effects on Smooth Muscle breakdown or transport is prevented (as in patients
• Depends on the predominant type of adrenergic taking MAO inhibitors or tricyclic antidepressants), then
receptor in the muscle the dosage of sympathomimetic agents must be reduced.
• Epinephrine relaxes GI smooth muscle (α2- and β-
receptor stimulation), whereas it usually increases Therapeutic Uses
sphincter contraction (α stimulation). 1. To treat bronchospasm
• Uterine contractions may be inhibited (β) or 2. For relief of hypersensitivity reactions; it is the
stimulated (α), depending on menstrual phase or state primary treatment for anaphylactic shock
of gestation. 3. To prolong the duration of infiltrative anesthesia
• In the bladder, the detrusor muscle relaxes (β), while 4. To restore cardiac activity in cardiac arrest
the trigone and sphincter contract (α) 5. To facilitate aqueous drainage in chronic open-angle
glaucoma.
• Bronchiolar smooth muscle relaxes (β2) 6. Norepinephrine is used for treating hypotension
during anesthesia when tissue perfusion is good.

Metabolic Effects
• An increase in glucose and lactate production via Adverse Effects of Epinephrine and NE
liver and 1. Anxiety
muscle glycogenolysis (β2). 2. Headache
3. Cerebral hemorrhage from the vasopressor effects.
• Inhibition of insulin secretion (α). 4. Cardiac arrhythmias, esp. in the presence of digitalis
• An increase in free fatty acids, mediated by cAMP and certain anesthetic agents
(β1) 5. Pulmonary edema from pulmonary hypertension.
• An increase in oxygen consumption
• Pharmacologic Effects of Norepinephrine Vascular Effects
• Equipotent to epinephrine in its reaction on β1- • Epinephrine exerts its action on small arterioles and
receptors, and slightly less potent on α- receptors. It precapillary sphincters. Its vascular effects include:
has very little effects on β2-receptors. - Decreased cutaneous blood flow
• An IV infusion raises both systolic and diastolic - Increased blood flow to skeletal muscle at low
pressure by constriction of vascular smooth muscle concentrations and decreased flow at higher
(α-receptors) concentrations.
• The increased peripheral vascular resistance - Increased hepatic blood flow with increased
produces a compensatory vagal reflex that slows the splanchnic vascular resistance.
heart rate. Cardiac output may actually decrease,
- Increased renal vascular resistance, producing
although coronary blood is increased.
decreased renal blood flow.
- Increased renal vascular resistance, producing,
Termination of Action decreased renal blood flow.

49
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
- Increased arterial and venous pulmonary pressure. • These are similar to the adverse effects of
epinephrine.
• Overdosage by inhalation can produce fatal
- Increased coronary blood flow, caused indirectly by ventricular arrhythmias
an increase in the work of heart, and mediated by
local effectors. • Tolerance to the desired effects occurs with overuse
in the asthmatic.

Isoproterenol
• Pharmacokinetics Dopamine
• Absorptionof orally administered isoproterenol is • Chemistry
unreliable - Intermediate in the synthesis of norepinephrine
• It is readily absorbed when given parenterally or as • Pharmacokinetics
an inhaled aerosol.
- Resembles epinephrine and noepinephrine
• It is principally metabolized by COMT; MAO plays
a smaller role than in epinephrine or noepinephrine • Central Dopamine Receptors (D1, D2, D3)
metabolism. - The central D1- receptor site is excitatory and
directlyactivates the adenylate cyclase system.
- The D2 receptor site is inhibitory in some brain
Pharmacologic Effects tissues and uses cAMP as 2its intracellular messenger.
• Has an N-alkyl substitution, which makes it act Pituitary-related side effects of neuroleptics are
almost entirely on β-receptors and have a little effect thought to be mediated through D2 recptors in the
on α- receptors. pituitary.
• IV infusion produces a reduction in peripheral - The D3 receptor is localized in the limbic system
vascular resistance in skeletal muscles and in renal and is not found in the pituitary. It is principally
and mesenteric vascular beds. associated with emotional and cognitive behavior.
• Diastolic blood pressure falls, but because there are
Pharmacologic Effects
increased venous return positive inotropic and
chronotropic effects, cardiac output is increased. • Important neurotransmitter in the CNS
• Systolic blood pressure may increase, but mean • Direct-agonist, acting on β1-receptors and releasing
pressure decreases. norepinephrine from nerve terminals. The result is a
positive inotropic effect on the myocardium.
• Renal blood flow decreases in normotensive
individuals, but it increases in patients with • Low or intermediate doses of dopamine reduce
nonhemorrhagic shock. arterial resistance in the mesentery and kidney; this
raises the glomerular filtration rate. The effect is
• Relaxation of both bronchial and GI smooth muscle
mediated by a receptor for dopamine.
occurs.
• A release of free fatty acids occurs; hyperglycemia is • At higher dose, it acts on α-receptors and causes
vasoconstriction with a consequent reduction in renal
less than
function.
• Pancreatic islet cells are activated, stimulating
insulin secretion.
Therapeutic Uses:
• Cadiogenic and septic shock
Therapeutic Uses • Chronic refractory Congestive Heart Failure
• Bronchodilator
Adverse Effects
• Cardiac stimulant
• Excessive sympathomimetic activity
Adverse Effects

50
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
• Anginal pain, arrhythmias, nausea and hypertension • In patients taking β-blockers increases the risk of
can occur, but these effects are short-lived because cardiac irregularities, myocardial infarction, and
dopamine has rapid metabolism. intracranial hemorrhage
• Rebound nasal congestion can occur in chronic use
as nasal decongestant.
Dobutamine
• Pharmacokinetics
- Not absorbed when given orally Methoxamine
- Half-life- 2 minutes when given IV • Direct-acting stimulator of α-adrenergic receptors
with pharmacologic properties similar to those of
• Pharmacologic Effects phenylephrine.
- Resembles dopamine chemically, it is a direct β1-
• It produces little CNS stimulation.
receptoragonists. It has a greater inotropic than
chronotropic effect. • Its large pressor effects results from an increase in
- Does not act on dopaminergic receptors. total peripheral resistance.

• Therapeutic uses • It causes a reflex bradycardia.


- Used to improve myocardial function in CHF. • Used therapeutically in hypotensive state and to end
attacks of paroxysmal atrial tachycardia
- Oxygen demands are less than with other
sympathetic agonists because it causes minimal β2- Adrenergic Agents
changes in heart rate and systemic pressure.
• Pharmacologic Effects
Adverse Effects - Primarily β2- agonists has a relaxing effect on
bronchial smooth muscle and show little effect on
• Increases AV conduction- used with caution in atrial cardiac β1- receptors.
fibrillation.
• Therapeutic Uses:
• Similar with other catecholamines - Used therapeutically for the treatment of bronchial
asthma or bronchospasm, where their lack of cardiac
stimulation is a decided advantage
Phenylephrine
• Administration
• Pharmacologic Effects - Inhaled
- Direct-acting sympathomimetic agent - Oral
- Similar to norepinephrine- but less potent and has - Parenteral
longer duration of action.
- Vasoconstriction, increased atrial pressure, reflex Adverse Effects
bradycardia occurs with parenteral administration. • Similar to other sympathomimetic agents
• Therapeutic Uses • Regular used of short-acting β2 agonists can be
- Nasal decongestant associated with increased bronchial
- hyperresponsiveness to metacholine challenge.
Pressor agent
- Provide local vasoconstriction: • Overuse of β2-agonists by patients with asthma has
~10% ophthalmic solution been associated with increased mortality
~Adjunct for use with local anesthetics • Even though their effects are primarily bronchial,
- For relief of paroxysmal atrial tachycardia β2- agonists should be used in patients with
cardiovascular disease or hyperthyroidism because of
Adverse Effects its effects on β1- receptors of the heart.
Specific Agents:
• Large doses cause cardiac irregularities.
• Ophthalmic solutions, like intranasal solutions- • Metaproterenol has more rapid onset of
systemically absorbed bronchodilating

51
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
action in asthma than either albuterol or terbutalline • When administered IV, its action is similar to that of
• Albuterol- more bronchoselective than isoproterenol epinephrine. However,
when given as an inhalant 1. its pressor response occurs more slowly and lasts 10
~ Effects is apparent within 15 times longer.
min., maximal at about 60-90 min., last about 3-4 hour 2. Its potency is 1/120 that of epinephrine in producing
• Salmeterol is a long-acting β2 selective adrenergic and equivalent pressor response.
agonist inhalant used for maintenance treatment of • Ephedrine increases arterial pressure by causing
asthma,with or without inhaled corticosteroids. It also peripheral vasoconstriction and cardiac stimulation.
appears to inhibit the release of inflammatory • Its effects on the bronchi and other smooth muscles
mediators from lung tissue are qualitatively similar to those of epinephrine.
~ Not recommended for acute treatment of
bronchospasm, bronchodilation begins in 15 minutes but • It causes CNS stimulation, which can result in effect
can last up to 12 hours such as insomia, nervousness, nausea and agitation.
~ Because it is a long-acting, a short acting β2-agonist • Tachyphylaxis occurs with repeated administration
can be administered as needed to control acute
symptoms.
Methamphetamine & Hydroxyamphetamine
• Methamphetamine
• Terbutalline- when used in the treatment of bronchial - Related chemically to both amphetamine and
asthma, has longer duration of action than ephedrine.
metaproterenol. It may have more side effects than - Mixed-acting sympathomimetic agent, it is equal to
other sympathomimetic agents. amphetamine in central stimulant activity and is more
- Otheruses:Tocolyticagent potent than ephedrine in pressor activity.
- Administration:oral,inhalation,subcutaneous - Therapeutic indications are limited to those that make
• Bitolterol is a prodrug that is converted in the lung to use of CNS effects, such as narcolepsy
colterol, an active catecholamine. It may have a
longer duration of action than terbutalline,
metaproterenol, or albuterol. It may cause tremor due
• Hydroxyamphetamine
- Has an action resembling that of ephedrine but without
to stimulation of skeletal muscle, in addition to the
CNS
adverse effects seen with all sympathomimetic agents
effects.
• Other agents: Salbutamol, Procaterol, Bambuterol - It is used as a mydriatic, decongestant, and pressor
agent.

MIXED-ACTING SYMPATHOMIMETIC DRUGS


Mephentermine
Ephedrine
• Mixed-acting sympathomimetic
• Pharmacokinetics
- Absorbed when taken orally • Its peripheral vasopressor actions are similar
- Resistant to COMT and MAO, so that its action is tothoseof methamphetamine
prolonged. • It lacks significant central actions
• Pharmacologic Effects • Major therapeutic use in the treatment of
- Mixed-acting sympathomimetic agent- both direct and hypotension
indirect actions
~ Its primary action is indirect. It causes the release of
norepinephrine from the storage in nerve terminals, Metaraminol
apparently by competing with norepinephrine for
transport into the granules. • Mixed-acting sympathomimetic that is similar to
~ It also produces direct stimulation of adrenergic norepinephrine in its action- less potent than NE
receptors. • Increases diastolic and systolic pressure via
vasoconstriction, and

52
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
produces a martked reflex bradycardia • Alpha-receptor antagonists may be reversible or
• It has little stimulant effect on the CNS irreversible in their interaction with these receptors.
• Its major therapeutic use is in the treatment of
hypotensive state. • Reversible antagonists dis- sociate from receptors,
and the block can be surmounted with sufficiently
high concentrations of agonists;

• Irreversible drugs do not dissociate and cannot be


surmounted.

• Effects of an irreversible antagonist may persist long


after the drug has been cleared from the plasma

CLASSIFICATION OF ADRENERGIC RECEPTOR


ANTAGONISTS
• α-receptor antagonist drugs cause a lowering of
peripheral vascular resistance and blood pressure

• Alpha-receptor antagonists often cause orthostatic


hypotension and reflex tachycardia

• Nonselective (α1 = α2, Table 10–1) blockers usually


cause significant tachycardia if blood pressure is
lowered below normal

A. Alpha Adrenergic Blockers


1.) Non-selective-
a.) Irreversible
b.) Reversible

2.) Alpha 1-selective


3.) Alpha 2-selective

ADRENERGIC RECEPTOR ANTAGONISTS B. Beta Adrenergic Blockers


• In clinical therapeutics, nonselective α antagonists are 1.) Non Selective
used in the treatment of pheochromocytoma (tumors 2.) Beta 1-selective
that secrete catecholamines) 3.) Beta 2- selective

C. Alpha Adrenergic Receptor Antagonists


• α1-selective antagonistsare used in primary 1. ALPHA RECEPTORS mediate many important
hypertension and benign prostatic hyperplasia. actions of
endogenous catecholamines. These include:
• Beta-receptor antagonist drugs are useful in the
treatment of hypertension, ischemic heart disease, a. ALPHA 1 mediated VASOCONSTRICTION,
arrhythmias, endocrinologic and neurologic disorders, b. ALPHA 2 receptor mediated INHIBITION OF THE
glaucoma, and other conditions. RELEASEOF NE and ACh,
c. ALPHA 2 mediated INHIBITION OF INSULIN
SECRETIONAND INHIBITION OF LIPOLYSIS,
Mechanism of Action d. ALPHA 2 mediated CONTRACTION OF BLOOD
VESSELS INSKIN AND

53
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
MUCOSA (these receptors are preferentially activated ACTIVATION OF ALPHA 1 RECEPTORS IN
by circulating catecholamines, whereas alpha 1 receptors VASCULAR SMOOTH MUSCLE, and BETA 1
are activated by NE released at sympathetic nerve RECEPTORS IN THE HEART among others.
terminals). • the net result is an increase in MEAN BLOOD
e. alpha 2 mediated central INHIBITION OF PRESSURE and a REFLEX BRADYCARDIA
SYMPATHETIC TONE.
the most important effects of alpha 1 & 2 adrenergic Other Actions of Alpha Adrenergic Antagonists
antagonists are on the 1.ALPHA 1 ANTAGONISTS
CARDIOVASCULAR SYSTEM. • can BLOCK ALPHA 1 RECEPTORS THAT
MEDIATE CONTRACTION OF NONVASCULAR
• these cardiovascular effects are mediated in large part SMOOTH MUSCLE such as the trigone and
by the effects of the antagonists on sympathetic nerve sphincter muscles of the bladder, leading to decreased
endings, and by effects on the CNS resistance to urinary outflow
• the antagonism which is observed in most cases is
competitive 2. Activation of Alpha 2 receptors causes inhibition of
insulin secretion
CARDIOVASCULAR EFFECTS OF ALPHA 1 • BLOCKADE of these receptors FACILITATES
ANTAGONISTS INSULIN RELEASE
Blockade of α receptors in other tissues elicits miosis
(small pupils) and nasal stuffiness
ALPHA ADRENERGIC ANTAGONISTS (Specific
1.ALPHA 1 ANTAGONISTS Agents)
• BLOCK VASOCONSTRICTION induced by
endogenous catecholamines 1. PHENOXYBENZAMINE
• the resulting fall in peripheral resistance leads to a • is an irreversible alpha blocker that blocks both alpha
FALL IN MEAN BLOOD PRESSURE 1 and alpha 2 receptors with slight PREFERENCE
• the magnitude of this effect is dependent upon the FOR ALPHA 1 RECEPTORS
degree of sympathetic tone at the time the antagonist • irreversible; long acting
is administered. • it causes a DECREASE IN PERIPHERAL
• thus, there is less hypotension in the supine than in the RESISTANCE,
standing patient and a REFLEX TACHYCARDIA due to hypotension
• the decrease in blood pressure leads to a reflex mediated baroreceptor effects, and also because it blocks
alpha 2 receptors, resulting in increased release of NE
tachycardia the effects are exaggerated if
the drug also has alpha 2 antagonist effects, because
• HYPOTENSIONis particularly prevalent when
sympathetic reflexes are active, such as in the
antagonism of alpha 2 receptors facilities release of NE
standing patient, or during stress
presynaptically to cause a further tachycardic effect
• they block the vasoconstriction and hypertensive • a MAJOR SIDE EFFECT THEREFORE IS
POSTURAL HYPOTENSION
effects of exogenous sympathomimetics.
for example, pressor responses to phenylephrine (a pure • also seen are cardiac arrhythmias
alpha 1 agonist) are completely blocked. • others ejaculation is inhibited, miosis, nasal stuffiness,
nausea and vomiting
CARDIOVASCULAR EFFECTS OF ALPHA 2
ANTAGONISTS • Phenoxybenzamine blocks pressor responses to
2. ALPHA 2 ANTAGONISTS exogenous catecholamines, and in fact causes reversal of
the pressor response to EPI into a depressor response
• increase sympathetic outflow by an action on the CNS
this potentiates the RELEASE of • it also inhibits the uptake of catecholamines into nerve
NOREPINEPHRINE terminals and extraneural uptake sites
• from sympathetic nerve endings leading to • it irreversibly inhibits 5-HT and histamine receptors
in higher doses than those reqired
for alpha receptor blockade

54
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
• duration of the effect of the drug is 48 hours to many • it has 1000 times greater affinity for alpha 1 vs alpha
days 2 receptors
• has short elimination half-life • it is a short acting drug with a duration of about 7 to
• a major use of the drug is in the management 10 hours
ofHYPERTENSION inPHEOCHROMOCYTOMA • PRAZOSIN causes a decrease in TOTAL
prior to PERIPHERAL RESISTANCE but NOT an
surgery INCREASE in HEART RATE (since alpha 2
• to REVERSE VASOCONSTRICTION IN SHOCK receptors are not inhibited)

• to RELIEVE VASOSPASM in RAYNAUD’S • DECREASE ARETERIAL PRESSURE with little


phenomenon change in cardiac output

2.PHENTOLAMINE and TALOZOLINE 2. TERAZOSIN

• are competitive antagonists of both alpha 1 and alpha • is a close structural analogue of prazosin with similar
2 receptors pharmacological effects but an intermediate duration
of action, is approximately 18 hours
• as such their effects on the cardiovascular system are
similar to those of phenoxybenzamine 3. DOXAZOSIN
• reversible , short-acting Phentolamine (non-selective); • is another structural analogue with similar
Tolazoline (slightly selective) pharmacological effects to Prazosin, but with a long
• used to control ACUTE HYPERTENSIVE duration of action as 36 hours.
EPISODES treatment test for • amajoruse ofthesedrugs,PRAZOSINanditsanaloguesis
PHEOCHROMOCYTOMA in the treatment of HYPERTENSION
• treatment of PERSISTENT PULMONARY • Alpha 1 receptors in the trigone muscle of the bladder
HYPERTENSION in and in the urethra contribute to the resistance to
neonates outflow of urine.
• experimentally used to relieve vasospasm in • Alpha 1 receptor blockers reduce this resistance
Raynaud’s especially when impaired bladder emptying is the
phenomenon result of BENIGN PROSTATIC HYPERPLASIA
• duration of action: • Adverse effects of these drugs include the so called
• Oral: 2–4 days Parenteral: 20–40 minutes “FIRST DOSE PHENOMENON”that is marked
postural hypotension & fainting 30-90 minutes after
• these drugs also have a parasympathetic effect on GI the first dose or with the addition of a second
smooth muscle and gastric secretions antihypertensive drug to a patient already taking one
• toxicity is like that of Phenoxybenzamine of these agents
** PHentolamine
• Pheochromocytoma Hypertensive episodes 4. TAMSULOSIN
Pulmonary Hypertension • is another alpha 1 blocker with little effect on alpha 1-
• Adverse Effects of PHENTOLAMINE and B receptors
TOLAZOLIN: ARRHYTHMIA and ANGINAL • considerable efficacy at the alpha 1-A receptor which
PAIN is believed to be predominant in human bladder and
can produce PARADOXICAL HYPERTENSION prostate
induce GASTROINTESTINAL STIMULATION • it is used in the treatment of BENIGN PROSTATIC
HYPERPLASIAbecause it decreases the resistance to
ADRENERGIC RECEPTOR ANTAGONIST urinary flow with less hypotensive effects on blood
pressure
1. PRAZOSIN **although all the alpha blockers have been tested and
• is the prototype of a family of potent and VERY are relatively effective in BPH
SELECTIVE
ALPHA 1 RECEPTOR ANTAGONISTS

55
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
5. ALFUZOSIN 3. Sometimes used in treatment of peripheralvasospastic
Is another related compound used in the treatment of diseaseRAYNAUDS disease to
perfusion
benign prostatic hypertrophy
4. To enhance urinary flow in benign
PRazosinos
PROSTATICHYPERPLASIAmajor therapy is
Tamsolusin
SURGERY
Alfuzosin
5. Treatment of local EXCESS CONCENTRATION of
Terazosin
aVASOCONSTRICTOR in order to prevent necrosis
Doxazosiiiiiiiiiiiiiiiiiiiin – longest half-life
TOXICITY of ALPHA BLOCKERS
MISCELLANEOUSDRUGSwithAlpha
1. POSTURAL HYPOTENSION
Adrenergicblocking activity:
2. REFLEX TACHYCARDIA
• the ERGOT Alkaloids where the first adrenergic 3. Other ARRHYTHMIAS
blocking agents to be discovered Ergot is a FUNGUS 4. Parasympathomimetic effects of Phentolamine and
which grows on rye which contains many biologically Tolazoline
active components
• ERGOT ALKALOIDSare weak alpha-blocking agent
and are serotonin antagonists BETA ADRENERGIC ANATAGONISTS
CLASSIFICATION:
• PARTIAL AGONIST at alpha-adrenergic receptors
Pharmacologic Effects: 1. RECEPTOR SELECTIVITY
-CNS stimulats
-irregular respiration A.Non-selective
-confusion
-anxiety Propranolol

• Directly stimulate smooth muscle Sotalol

• Significant elevation of BP via peripheral Acetabulol (β1>β2)


vasoconstriction Timolol
Labetalol
• ERGOTAMINE tartrate treatment in MIGRAINE Nadolol
headache
Pindalol
• ERGONOVINE maleate powerful OXYTOXIC but
lacks the adrenergic blocking activity of ergotamine B. Beta 1- receptor selectivity
• causes direct contraction of uterine smooth muscles Acetabulol
and used to decrease postpartum bleeding Esmolol
Metoprolol
• METHYLERGONOVINE maleate used to decrease Nebivolol
postpartum bleeding Atenolol
• METHYLSERGIDEused as prophylactic agent for
migraine attack C. Beta 2-receptor selectivity Acetabulol
• ERGOTISMis a disease characterized by profound Butoxamine
VASOCONSTRICTION ISCHEMIA and
GANGRENE of the affected limb
D. Alpha-Beta Blockers Labetolol Carvedilol (β1
• in the Middle Ages it was known as ST. selective, α3 antagonist)
ANTHONY”S FIRE was relieved by visiting the
shrine of St. Anthony which was Cetiprolol
located in an area of France where bread was seldom
contaminated with the fungus 2. PARTIAL AGONIST ACTIVITY
Pindolol Acetabulol(causesomebronchodilation)
THERAPEUTIC USES OF ALPHA ADRENERGIC
RECEPTOR BLOCKERS: Celiprolol (β2 agonist)
1. HYPERTENSION
2. PHEOCHROMOCYTOMA

56
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
drugs should be used with great caution, if at all in
BETA ADRENERGIC RECEPTOR BLOCKERS patients with bronchospastic disease since even
• Beta- blocking drugs occupy β receptors and relatively selective blockers have some affinity for the
competitively reduce receptor occupancy by beta 2 receptor
catecholamines and other β agonists.
3. METABOLIC EFFECTS OF BETA BLOCKERS
• β-blocking drugs in clinical use are pure antagonists CATECHOLAMINES

A. INTRODUCTION • promote glycogenolysis & mobilize


1. Cardiovascular effects of Beta Blockers glucose in response to hypoglycemia.

• since catecholamines have positive inotropic & • BETA BLOCKERS (Beta 3 receptor) attenuate the
chronotropic effects on the heartBETA BLOCKERS lipolytic response to sympathetic nervous system
SLOW the HEART RATE and DECREASE activation which is an important energy source for
MYOCARDIAL CONTRACTILITY exercising muscle. Selective beta 1 antagonists may
cause these effects less frequently than non-selective
• when sympathetic activation is low, these drugs have antagonists.
modest effects
• Nonselective Beta blockers must therefore be used
• however, when sympathetic activation is high, such as with great caution in diabetics. A selective Beta 1
during exercise & stress, beta-blockers attenuate the blocker is usually preferable in this case.
expected increase in the heart rate and diminish
cardiac output
SPECIFIC AGENT OF BETA RECEPTOR
• Beta blockers tend to decrease the capacity to work BLOCKING DRUGS
• exercise performance is impaired less by selective 1. PROPRANOLOL
Beta 1 blockers • After oral administration, propranolol is almost
• Beta blockers have important effects on cardiac completely absorbed but undergoaes extensive first
rhythm and automaticity pass metabolism in the liver

• they reduce the sinus rate, decrease the rate of • the extent of clearance by the liver varies radically
depolarization of ectopic pacemakers, and slow across patients however and results ina s much as a 20
conduction in the atria & AV node fold difference in plasma concentration of the drug
after oral administration to different persons
• Beta blockers DO NOT LOWER BLOOD
PRESSURE in NORMAL MANhowever they do • this has obvious significance in patients with liver
reduce blood pressure in hypertensive man. The disease
mechanism of this effect has not been satisfactorily • completely absorbed in the GIT
explained
• 90% of the drug in the circulation is bound to plasma
• some beta blockers have peripheral vasodilating proteins
effects via different mechanisms which may • elimination half-life is 3 hours, prolonged half-life in
contribute to their capacity to lower blood pressure
larger dose
(These include celiprolol and nebivolol)
• WHEN USED AS AN ANTIHYPERTENSIVE
• Chronic prophylactic therapy with a beta blocker in AGENT THE FULL RESPONSE ON THE BLOOD
patients who have had a myocardial infart appears to
PRESSURE MAY NOT DEVELOP UNTIL AFTER
help prevent the recurrence of a second fatal
SEVERAL WEEKS OF ADMINISTRATION
myocardial infarct.
• Abrupt withdrawal of Propranolol after chronic
2. Pulmonary effects of Beta Blockers therapy can lead to the development of withdrawal
symptoms
• Beta blockade usually has little effect on pulmonary
function in normal man
PHARMACOLOGIC EFFECTS:
• however in ASTHMATICS – they can cause the • decrease heart rate & cardiac output
threatening bronchoconstriction.
• prolong systole
• Although beta 1 selective blockers maybe less likely
to cause respiratory problems in asthmatics, these

57
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
• decrease total coronary blood flow and oxygen lower incidence of CNS side effects seen with this
consumption drug.

• reduces blood flow in most tissues except the brain • Not metabolized and is excreted unchanged in urine.
lowers BP • Adverse effect-similar to Propranolol
• inhibit renal secretion of rennin
• decrease Na excretion increases airway resistance by
β2 blockade interferes carbohydrate and fat metabolism 3. TIMOLOL
• Therapeutic uses treatment of HYPERTENSION in • it is a nonselective blocker with a SHORT
combination with DIURETIC PROPHYLAXIS of DURATION of action
angina pectoris prophylaxis of SUPRAVENTRICULAR • it is well absorbed from the gut and is subject to
and VENTRICULAR ARRYTHMIAS moderatefirst pass metabolism
• long term prophylaxis in patients who have had MI • it is metabolized by the liver and only a small fraction
management of HYPERTROPIC OBSTRUCTIVE isexcreted by the kidney in unchanged form
CARDIOMYOPHATIES to reduce force myocardial • Timolol is an example of a beta blocker which is used
contraction management of HYPERTHYROIDISM in glaucoma to reduce the rate of synthesis of aqueous
humor
• decrease heart rate prophylaxis for migraine
headaches • the ocular form of timolol is well absorbed into the
circulation and adverse effects can occur in patients
SIDE EFFECTS: with asthma or congestive heart failure
• Other new drugs with similar mechanisms of action
• include GI distress (relatively unopposed include Penbutolol, and Pindolol
parasympathetic nervous system
• CNS effects such as nightmares, insomnia, fatigue, 4. LABETOLOL
sexual dysfunction and depression, • it is an example of a nonselective beta blocker which
• induce heart failure especially in patient with MI, also blocks alpha 1 receptors.
rapid withdrawal can lead to hypersensitivity of beta- • it also inhibits uptake of NE from the synaptic cleft (a
adrenergic receptors-anginal attacks cocaine-like effect).
• arrhythmias or MI • its pharmacokinetics are like those of Propranolol, is
• increases airway resistance well absorbed, highly metabolized and very variably
• hypoglycemic action of insulin can be augmented so on first pass through liver, mainly metabolized by
rash, fever, purpura, contraindicated in patients with liver.
Raynaud’s phenomenon • it is used in the treatment of
PHEOCHROMOCYTOMA and HYPERTENSIVE
2. NADOLOL EMERGENCY,where its alpha 1 effects promote
• is a non-selective beta blocker with a LONG vasodilation and hypotension, while its beta 1 effect
DURATIONOF ACTION helps reduce reflex tachycardia.

• is poorly lipid soluble and incompletely absorbed


from the GI tract • ADVERSE EFFECTS:

• interindividual variability in its bioavailability is less • postural hypotension,


than with Propranolol • jaundice,
• it is extensively metabolized & is EXCRETED • same to other β-blockers
INTACT IN THE URINE • can be administer IV in hypertensive emergencies
• as such KIDNEY DISEASE is a cause for concern, other new drugs with a similar mechanism of action
and may result in an accumulation of NADOLOL
because NADOLOL is poorly lipid soluble, • include Carvedilol, and Bucindolol
• it is thought that there will be lower concentrations of 5. SOTALOL
Nadolol in the brain,and this may contribute to a

58
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
• non-selective β-blocker without intrinsic • it has a somewhat longer duration of action
sympathomimetic or local anesthetic activity thanmetoprolol.

• 6. PINDOLOL 3. ESMOLOL

• is non-selective β-blocker with intrinsic • it is a selective Beta 1 antagonist with a VERY


sympathomimetic alpha-adrenergic activity, no VERY SHORTDURATION OF ACTION which is
rebound tachycardia upon withdrawal also devoid of intrinsic sympathomimetic activity and
membrane-stabilizing activity

• 7. ACETABULOL • it is given by IV infusion


• β-blocker with mild intrinsic sympathomimetic • it has a half life of about 8 minutes in plasma, and is
activity, β1>β2 effects metabolized by plasma esterases

• ADVERSE EFFECTS: same with other β-blocker • it is used ONLY for the treatment of ATRIAL
agents, arthritis, myalgia and arthralgia • TACHYCARDIA.
PHARMACOLOGY OF SELECTIVE BETA • 4. NEBIVOLOL
BLOCKERS
• it is the MOST SELECTIVE BETA 1
ANTAGONIST clinically
• 1. METOPROLOL
• available which is devoid of intrinsic
• it is a selective Beta 1 antagonist, whose sympathomimetic activity,but has the ability to reduce
pharmacokinetics systemic vascularresistance
• are like propranolol which is also devoid of intrinsic • is effective in treating hypertension and heart failure
sympathomimetic activity and membrane-stabilizing
activity • other new drugs with a similar mechanism of action
include Bisoprolol, Acebutolol
• inhibits the inotropic and chronotropic cardiac
responses to Isoproterenol •
• 1/50th as potent as Propranolol in inhibiting the • 5. BETAXOLOL
vasodilator response to Isoproterenol • selective β1 antagonist, once daily administration,
• long acting morelipophilic than Atenolol, topical administration
for the treatment of GLAUCOMA
• well-absorbed orally
• LESS SIDE EFFECTS IN ASTHMATIC PATIENTS • AGENTS THAT INHIBIT THE ACTION OF
side effects- similar to Propanolol ADRENERGIC NERVES
1. RESERPINE
• inhibits the uptake of norepinephrine
2. ATENOLOL • BP decreases
• it is also a selective Beta 1 antagonist which is also • triggers reflex tachycardia
devoid of intrinsic sympathomimetic activity and
membrane- stabilizing activity. • Sedation-depletion of catecholamine in the brain
treatment in HYPERTENSION
• it is a poorly lipid soluble drug
• ADVERSE EFFECTS: sedation,
• about half of an oral dose is absorbed, but most of
thisreaches the systemic circulation if there is no first • psychic depression→suicide abdominal cramps and
passmetabolism in the liver diarrhea GI ulceration
• as a result, peak concentrations of this drug in the • high incidence of breast carcinoma
plasmaare not so variable across patients •
• it is excreted unchanged in the urine • 2. GUANETHIDINE

59
PHARMACOLOGY
Lyceum Northwestern University
COLLEGE OF MEDICINE
• impairs the response to sympathetic stimulation by
inhibiting the release of neurotransmitter
• displaces norepinephrine from intraneuronal storage
granules
• slow onset of action after oral administration
• rapidly cleared by the kidney
• Therapeutic use: POTENT LONG ACTING
HYPERTENSIVEAGENT
• ADVERSE EFFECTS:
• postural hypotension, syncope-esp. in strenous
activities, diarrhea,
• edema
• Contraindicated in patient taking MAO inhibitors
Antihypertension effects can be reversed by
Ephedrine or
• Phenylpropanolamine

3. GUANADREL
• similar to Guanethidine
• less morning hypotension
• less diarrhea
• contraindicated in patient with MAO inhibitors
causes water retention-given with diuretics
4. BRETYLIUM
• produces block in the release of epinephrine
• inhibits re-uptake of norepinephrine into nerve
terminal antiarrhythmic agent
• poorly absorbed orally
• strong hypotension

60

Das könnte Ihnen auch gefallen