Cology Lecture 1
Cology Lecture 1
Introduction to Pharmacology
Pharmacology is the branch of medicine and biology that focuses on the study of drug
action. It involves the study of how drugs interact with biological systems to affect
physiological functions. Pharmacology seeks to understand the origins, chemical properties,
biological effects, therapeutic uses, and potential toxicities of drugs.
1. Pharmacodynamics: The study of the effects of drugs on the body and the
mechanisms by which these effects occur.
2. Pharmacokinetics: The study of how the body absorbs, distributes, metabolizes, and
excretes drugs (often abbreviated as ADME).
3. Pharmacogenomics: The study of how an individual's genetic makeup affects their
response to drugs.
4. Toxicology: The study of harmful effects of chemicals, including drugs, on living
organisms.
The history of pharmacology is rich and spans centuries. Some key milestones include:
1. Ancient Times:
o Early humans used plants, herbs, and minerals for their medicinal properties.
Ancient civilizations like the Egyptians, Greeks, Chinese, and Indians
developed early pharmacological practices. For instance, Hippocrates (circa
460-370 BCE), the "Father of Medicine," is often credited with starting the
scientific approach to medicine and suggesting that disease had natural causes,
not divine ones.
o Galens' Works (circa 130-200 CE) on drugs, especially plant-based,
contributed to the development of the first pharmacological knowledge
systems.
2. 17th and 18th Centuries:
o The development of scientific method and the scientific revolution in Europe
advanced the field significantly.
o Paracelsus (1493–1541), a Swiss physician, revolutionized medicine by
promoting the use of chemicals and minerals in treatment, coining the term
"dose-response" and emphasizing the importance of dose in determining the
therapeutic effect.
3. 19th Century:
o Isolation of Active Compounds: In this era, scientists started to isolate pure
compounds from plants. Morphine (from opium) was isolated in 1804, and
quinine (from the bark of the cinchona tree) was recognized for its
antimalarial properties.
o Wilhelm Wundt and other early experimental psychologists laid the
groundwork for the study of the nervous system's interaction with drugs.
o Rudolf Buchheim (1820–1879) is often credited with founding modern
pharmacology. He established the first pharmacology department at the
University of Dorpat (now in Estonia) in 1847.
4. 20th Century:
o The Development of Modern Pharmacology: With the rise of molecular
biology, biochemistry, and biotechnology, pharmacology became increasingly
sophisticated. The development of synthetic drugs, antibiotics, and vaccines
revolutionized medical treatment.
o Penicillin, discovered by Alexander Fleming in 1928, marked a major
advancement in pharmacology, saving millions of lives from bacterial
infections.
o In the 1950s, the discovery of the first class of tranquilizers,
benzodiazepines, and the widespread use of antihistamines and oral
contraceptives marked significant achievements in therapeutic pharmacology.
5. Late 20th Century and 21st Century:
o The introduction of biologics, monoclonal antibodies, and the mapping of the
human genome has allowed pharmacologists to develop targeted therapies
based on genetic information.
o Personalized medicine is an emerging trend, with an emphasis on tailoring
drug treatments to individual genetic profiles.
o The development of immunotherapies and advances in nanomedicine have
expanded the scope of pharmacology in treating complex diseases, including
cancers and autoimmune disorders.
Scope of Pharmacology
Pharmacology is a vast and multidisciplinary field with numerous areas of study and
application. It plays a key role in:
Conclusion
Pharmacology is an essential field that not only drives the development of new and safer
medications but also ensures that current treatments are used safely and effectively in clinical
practice. Its scope is broad, from the basic molecular mechanisms of drug action to large-
scale public health implications. The field continues to evolve with advances in genetics,
biotechnology, and information science, promising more personalized and targeted therapies
for patients in the future.
Lecture 2
Nature and Source of Drugs
Drugs are chemical substances used to diagnose, treat, cure, or prevent diseases, alleviate
symptoms, or for other therapeutic purposes. The nature and source of drugs can be classified
based on their origin, chemical structure, and therapeutic use. The sources of drugs are
diverse and include:
1. Natural Sources
Drugs obtained from plants, animals, or microorganisms form the historical backbone of
pharmacology. Examples include:
• Plant-Derived Drugs:
o Many drugs are derived from plants, which produce bioactive compounds to
defend themselves against herbivores, diseases, or environmental stress.
o Example:
▪ Morphine (from the opium poppy) is used as a painkiller.
▪ Quinine (from the cinchona tree) is used to treat malaria.
▪ Aspirin (from willow bark) has anti-inflammatory, analgesic, and
antipyretic properties.
• Animal-Derived Drugs:
o These drugs are sourced from animal tissues or secretions.
o Example:
▪ Insulin (originally derived from bovine and porcine pancreases) is
used to treat diabetes.
▪ Heparin (an anticoagulant) is derived from the mucosa of pig
intestines.
• Microbial-Derived Drugs:
o Some drugs are produced by microorganisms (bacteria, fungi, etc.).
o Example:
▪ Penicillin, discovered by Alexander Fleming in 1928, is derived from
the fungus Penicillium and is one of the most important antibiotics in
modern medicine.
▪ Streptomycin (an antibiotic) is derived from Streptomyces bacteria.
2. Synthetic Sources
Many drugs are created artificially in laboratories using chemical processes. These synthetic
drugs are often developed to mimic the properties of naturally occurring substances or to
provide novel therapeutic effects.
• Example:
o Aspirin (acetylsalicylic acid) was originally derived from salicylic acid found
in willow bark but is now synthesized chemically.
o Barbiturates, a class of central nervous system depressants, were first
synthesized in the laboratory.
3. Semi-Synthetic Sources
These drugs are chemically modified versions of natural compounds. Semi-synthetic drugs
often have improved efficacy, safety profiles, or pharmacokinetic properties compared to the
original natural product.
• Example:
o Heroin is a semi-synthetic derivative of morphine.
o Amoxicillin is a modified version of penicillin.
4. Biotechnological Sources
The advent of biotechnology has enabled the production of drugs through recombinant DNA
technology, where genes coding for therapeutic proteins are inserted into microorganisms like
bacteria or yeast, which then produce the desired drug.
• Example:
o Monoclonal antibodies (like rituximab) are engineered using recombinant
DNA technology.
o Human growth hormone is produced using genetically modified bacteria.
1. Effectiveness: The drug must be proven to be effective for the conditions it is meant
to treat.
2. Safety: The drug must have a favorable safety profile, with minimal side effects when
used correctly.
3. Affordability: The drug should be affordable to the community and healthcare
system, ensuring broad access.
4. Availability: Essential drugs should be available at all times, in adequate quantities,
and in appropriate formulations.
5. Quality: These drugs must meet established standards of quality, potency, and purity.
The WHO Essential Medicines List (EML) is a list of medicines deemed essential for
meeting the basic health care needs of a population. It includes medications for a range of
common conditions, from infections and pain management to chronic diseases like diabetes
and hypertension.
• Example:
o Antibiotics like Amoxicillin.
o Antipyretics and analgesics like Paracetamol.
o Vaccines like DTP (Diphtheria, Tetanus, and Pertussis).
The goal is to promote the rational use of drugs, ensuring that patients receive the most
appropriate medication for their condition without unnecessary or expensive treatments.
The route of drug administration refers to the way a drug is taken into the body, which
determines its absorption, distribution, metabolism, and excretion (ADME). The choice of
route depends on factors like the drug's properties, the disease being treated, the desired
speed of action, and patient factors (e.g., age, ability to swallow, and condition of the
gastrointestinal tract).
• Administration: The drug is taken through the mouth in the form of tablets, capsules,
liquids, or powders.
• Advantages: Easy, convenient, and non-invasive.
• Disadvantages: Slow onset of action, drug may be degraded by stomach acid or
enzymes, some drugs may cause gastrointestinal irritation.
• Example: Paracetamol (acetaminophen), Aspirin.
• Administration: The drug is injected directly into the bloodstream through a vein,
providing immediate access to the circulatory system.
• Advantages: Fastest onset of action, complete bioavailability (100% of the drug
reaches the systemic circulation).
• Disadvantages: Requires medical supervision, higher risk of side effects or toxicity.
• Example: Morphine (for pain management), Saline solution.
• Administration: The drug is injected into a muscle, typically in the upper arm, thigh,
or buttock.
• Advantages: Faster than oral administration, but slower than intravenous; suitable for
larger volumes of medication.
• Disadvantages: Pain or discomfort at the injection site, potential for infection.
• Example: Vitamin B12, Vaccines.
5. Inhalation Route
• Administration: The drug is inhaled into the lungs, either as a gas, vapor, or aerosol.
• Advantages: Rapid absorption due to the large surface area of the lungs, direct action
on respiratory conditions.
• Disadvantages: Requires special equipment, not suitable for all types of drugs.
• Example: Albuterol (for asthma), Nitrous oxide (anesthesia).
6. Topical Route
7. Transdermal Route
• Administration: Drugs are absorbed through the skin, typically via patches.
• Advantages: Provides continuous, controlled release of the drug over time.
• Disadvantages: Limited to drugs that can penetrate the skin.
• Example: Nicotine patches (for smoking cessation), Fentanyl patches (for chronic
pain).
8. Rectal Route
• Administration: The drug is administered via the rectum in the form of suppositories
or enemas.
• Advantages: Can be used when oral administration is not possible (e.g., vomiting,
unconsciousness).
• Disadvantages: Uncomfortable, absorption may be unpredictable.
• Example: Paracetamol suppositories for children.
• Administration: The drug is placed under the tongue (sublingual) or in the cheek
(buccal) for absorption through the mucous membranes.
• Advantages: Rapid absorption, bypasses the gastrointestinal tract and liver (first-pass
metabolism).
• Disadvantages: Limited to small doses and specific drugs.
• Example: Nitroglycerin (for chest pain), Lorazepam.
Conclusion
The nature and sources of drugs are diverse, ranging from natural substances to synthetically
engineered compounds, with modern biotechnology opening new frontiers in drug discovery.
The essential drugs concept ensures that life-saving medications are accessible to the
population, particularly in low-income regions. Additionally, understanding the various
routes of drug administration helps healthcare professionals choose the best method for
delivering drugs to achieve the desired therapeutic effect, taking into account patient needs
and the properties of the drug itself.
Lecture 3
In pharmacology, agonists and antagonists refer to the ways drugs interact with receptors in
the body to produce or block effects. These interactions are fundamental to understanding
how medications work and how they can be used therapeutically or controlled in cases of
addiction or overdose.
Agonists
1. Full Agonists: These agonists bind to a receptor and produce the maximum possible
response. Full agonists can produce the full biological effect associated with the
receptor activation.
o Example: Morphine is a full agonist at the opioid receptors, leading to strong
analgesic (pain-relieving) effects.
2. Partial Agonists: These agonists bind to the receptor but produce a less than
maximal response, even when all receptors are occupied.
o Example: Buprenorphine is a partial agonist at opioid receptors and is used
in opioid addiction treatment because it activates receptors but to a lesser
degree than full agonists like morphine or heroin, thus reducing the risk of
overdose.
Antagonists
An antagonist is a substance that binds to a receptor but does not activate it. Instead, it
blocks the receptor and prevents an agonist (or endogenous ligand) from binding and
exerting its effect. Antagonists are used therapeutically to block excessive receptor
activation or to treat conditions caused by overstimulation.
There are two types of antagonists based on how they interact with the receptor: competitive
antagonists and non-competitive antagonists.
Competitive Antagonists
A competitive antagonist competes with an agonist for binding at the same receptor site.
The effect of the antagonist can be overcome by increasing the concentration of the agonist,
because the agonist and antagonist are in a dynamic competition for the same receptor
binding site.
• Characteristics:
o They bind reversibly to the receptor (meaning the bond can be broken).
o The antagonistic effect can be overcome by increasing the concentration of
the agonist.
o They typically reduce the potency of the agonist but do not change the
maximum response that can be achieved.
• Example:
o Naloxone is a competitive antagonist at opioid receptors. It can reverse opioid
overdose by displacing the opioid (like morphine or heroin) from the receptor.
However, if the opioid concentration is high enough, the effects of the opioid
can overcome naloxone's antagonism.
• Clinical Relevance: Competitive antagonists are commonly used to treat overdose or
poisoning by competing with the substance of abuse (such as opioids) and blocking
their effects.
Non-Competitive Antagonists
A non-competitive antagonist binds to a different site (often called the allosteric site) on
the receptor, not the same binding site as the agonist. This binding causes a conformational
change in the receptor, making it less responsive to activation by the agonist. Because non-
competitive antagonists do not compete directly with the agonist, their effects cannot be
overcome by increasing the concentration of the agonist.
• Characteristics:
o They bind irreversibly (or very strongly) to the receptor or modify its
function in a way that cannot be reversed by increasing the agonist
concentration.
o The maximum response that can be achieved is decreased, as the receptor is
less responsive to activation.
o The drug’s effect is more long-lasting because of the receptor modification.
• Example:
o Ketamine is a non-competitive antagonist at NMDA (N-methyl-D-aspartate)
receptors, which play a role in pain sensation and memory. Ketamine blocks
these receptors, resulting in dissociative anesthesia and analgesia.
• Clinical Relevance: Non-competitive antagonists are often used to inhibit receptor
activity in chronic conditions or when it is important to block receptor activation for
prolonged periods.
Spare Receptors
The concept of spare receptors refers to the phenomenon where a full response can be
achieved without all the available receptors being occupied by an agonist. This can occur
when there are more receptors present than are needed to produce the maximum response.
The excess receptors are considered "spare".
Addiction
1. Tolerance: With repeated use, the body adapts to the drug, requiring larger doses to
achieve the same effect.
o Example: As a person continues to use alcohol, they may need increasing
amounts to achieve the same level of intoxication or euphoria.
2. Dependence: The body becomes reliant on the drug to function normally. When the
drug is not available, withdrawal symptoms occur.
o Example: Individuals who are physically dependent on opioids may
experience withdrawal symptoms like nausea, sweating, and anxiety when
they stop using the drug.
3. Reward System: Many drugs of abuse activate the dopaminergic reward system
(primarily the mesolimbic pathway), releasing dopamine, which contributes to
feelings of pleasure and reinforcement of drug-taking behavior.
o Example: Cocaine increases dopamine levels in the brain, leading to intense
euphoria. This reinforces the desire to continue using the drug.
4. Craving: The intense desire or compulsion to use the substance, often triggered by
environmental cues or emotional states.
o Example: A person addicted to alcohol might experience cravings when in
social situations where drinking is common.
Treatment of Addiction:
• Agonist substitution: Drugs like methadone (an opioid agonist) and buprenorphine
(a partial agonist) are used to replace more harmful substances like heroin and help
individuals reduce cravings and withdrawal symptoms.
• Antagonist therapy: Naltrexone, an opioid antagonist, blocks the effects of opioids
and alcohol, reducing cravings and preventing relapse.
Conclusion
Lecture 4
These terms refer to different phenomena associated with drug use and responses in the body.
Understanding these concepts is essential for clinicians and pharmacologists in prescribing
medications safely and effectively, as well as in managing potential adverse effects or
complications.
Tolerance
Tolerance is a physiological phenomenon where, with repeated use of a drug, the body
becomes less responsive to its effects. As a result, the same dose of the drug produces a
diminished effect, and higher doses may be required to achieve the same therapeutic effect or
euphoria.
• Mechanisms:
o Pharmacokinetic tolerance: The body becomes more efficient at
metabolizing or eliminating the drug, reducing the concentration of the drug
available at its site of action.
o Pharmacodynamic tolerance: The drug's target receptors become less
sensitive or the number of receptors decreases over time, making it harder for
the drug to exert its effect.
• Example: A person who regularly takes opioids may find that they need to increase
the dose to achieve the same level of pain relief or euphoria, because their body has
developed tolerance to the drug.
• Types:
o Cross-tolerance: Tolerance to one drug may result in tolerance to a
chemically similar drug. For example, tolerance to heroin may result in
tolerance to morphine.
o Acquired tolerance: Develops after prolonged exposure to a drug.
Dependence
Dependence refers to a state in which the body has adapted to the presence of a drug, and the
individual becomes reliant on the drug to function normally. Dependence can manifest as
physical dependence and psychological dependence.
1. Physical Dependence: The body has adapted to the drug, and abrupt cessation or a
significant reduction in the drug leads to withdrawal symptoms. These symptoms can
be physiological (e.g., sweating, tremors, nausea, increased heart rate).
o Example: Withdrawal from alcohol or opioids can lead to symptoms like
anxiety, tremors, sweating, and seizures (for alcohol) or nausea, vomiting, and
muscle pain (for opioids).
2. Psychological Dependence: This occurs when an individual feels a compulsive need
to take the drug, driven by the craving for its psychological effects, such as euphoria,
relaxation, or stress relief.
o Example: A person addicted to cocaine may feel a strong psychological urge
to use the drug even if they are physically not dependent on it.
Tachyphylaxis
Tachyphylaxis is a term used to describe the rapid development of tolerance after short-
term exposure to a drug. It refers to the phenomenon where, after only a few doses or a short
period of use, the drug’s effectiveness diminishes significantly. Unlike tolerance, which
typically develops gradually, tachyphylaxis occurs very quickly.
• Mechanisms:
o It can occur due to rapid depletion of neurotransmitters or receptors involved
in the drug's action.
o Another explanation is the desensitization or downregulation of receptors or
signaling pathways, causing the drug to lose its effect more rapidly.
• Example:
o Nitrates (e.g., nitroglycerin) used for angina often cause tachyphylaxis,
meaning that after repeated use over a short period, their effectiveness
diminishes, and higher doses may be required to achieve the same effect.
o Decongestants like pseudoephedrine can also lead to tachyphylaxis, where
the nasal passages become less responsive to the drug after repeated use.
• Clinical Relevance: Tachyphylaxis is important in the management of certain
medications to avoid rebound effects or loss of therapeutic efficacy. For example,
when using topical nasal decongestants, overuse can lead to rebound congestion due
to tachyphylaxis.
Idiosyncrasy
• Mechanisms:
o Genetic variations in drug-metabolizing enzymes can lead to idiosyncratic
reactions. For example, some individuals have genetic differences in enzymes
that metabolize certain drugs, causing unusual or extreme reactions.
o Immune system abnormalities may also play a role in idiosyncratic reactions.
• Example:
o Paracetamol (acetaminophen) toxicity: In some individuals with a specific
genetic variant, normal doses of paracetamol can lead to liver damage
because the drug is metabolized into a toxic compound.
o Sulfonamide drugs: Some people may have severe allergic reactions to
sulfonamide drugs (such as certain antibiotics) even though they are generally
safe for most people.
• Clinical Relevance: Idiosyncratic reactions are unpredictable and may require
discontinuation of the drug and alternative therapy. Understanding an individual's
genetic predispositions can help minimize the risk of these reactions.
Allergy
Drug allergy is an immune-mediated reaction to a drug. It occurs when the immune system
mistakenly identifies a drug or one of its metabolites as a harmful substance and mounts an
immune response against it. Allergic reactions are typically predictable, and the severity can
range from mild skin reactions to severe, life-threatening conditions like anaphylaxis.
• Mechanisms:
o Type I Hypersensitivity: Immediate-type allergic reactions involving IgE
antibodies, such as hives, anaphylaxis, and asthma.
o Type II Hypersensitivity: Cytotoxic reactions, where antibodies bind to drug-
modified cells, causing cell destruction (e.g., hemolytic anemia).
o Type III Hypersensitivity: Immune complex-mediated reactions that can lead
to vasculitis and glomerulonephritis.
o Type IV Hypersensitivity: Delayed-type hypersensitivity mediated by T-
cells, causing rashes or contact dermatitis.
• Example:
o Penicillin: A common allergen that can cause rashes, angioedema, or severe
anaphylactic reactions in sensitive individuals.
o Sulfonamide antibiotics: Can cause allergic reactions such as skin rashes or
Steven-Johnson syndrome.
• Clinical Relevance: Allergies to medications should be documented in medical
records to avoid prescribing drugs that could trigger a serious immune response.
Immediate treatment with antihistamines, steroids, or epinephrine (in cases of
anaphylaxis) may be necessary. In some cases, allergy testing can help identify which
drugs should be avoided.
Summary Table
Conclusion
Lecture 5
Pharmacokinetics is the study of the movement of drugs within the body, which includes
the processes of absorption, distribution, metabolism, and excretion (often abbreviated as
ADME). These processes determine the drug’s onset of action, duration of effect, and
intensity of the response.
Here, we'll focus on absorption and distribution, the first two steps in pharmacokinetics.
1. Absorption
Absorption refers to the process by which a drug enters the bloodstream from its site of
administration. The rate and extent of absorption are key factors in determining the
bioavailability of a drug, i.e., the proportion of the drug that reaches the systemic
circulation in an active form.
1. Route of Administration: The method by which the drug is given greatly influences
absorption. For example:
o Oral route (PO): Drugs must pass through the gastrointestinal (GI) tract and
undergo absorption through the mucosal lining.
o Intravenous (IV): No absorption is needed, as the drug is directly introduced
into the bloodstream.
o Intramuscular (IM) and subcutaneous (SC) injections: Drugs are absorbed
into the bloodstream through the muscle or subcutaneous tissue.
o Inhalation and transdermal routes: These routes provide rapid absorption
via the lungs or skin.
2. Drug Formulation: The form of the drug (e.g., tablet, capsule, liquid, etc.) can
influence how quickly and efficiently it is absorbed.
o Example: Liquid drugs are usually absorbed more quickly than tablets
because they do not need to be dissolved first.
3. Solubility:
o Lipophilicity: Lipid-soluble (lipophilic) drugs tend to pass through cell
membranes more easily than water-soluble (hydrophilic) drugs.
o pH and Ionization: The drug’s ionization state can affect its absorption. Non-
ionized (neutral) drugs are generally more easily absorbed across cell
membranes.
4. Blood Flow: Absorption is faster in areas with a rich blood supply. For example,
drugs are absorbed more quickly in the small intestine (which has high blood flow)
than in the stomach (which has lower blood flow).
5. Gastric Emptying Time: The speed at which the stomach empties into the small
intestine can influence absorption. Some drugs are absorbed more effectively when
gastric emptying is rapid.
6. Presence of Food: The presence of food in the stomach can either enhance or inhibit
drug absorption. For example, fatty foods may increase the absorption of fat-soluble
drugs (e.g., vitamins A, D, E, K), while food might slow the absorption of other drugs
(e.g., antibiotics like penicillin).
7. Drug Interactions: Some drugs may affect the absorption of others. For example,
antacids may change stomach pH and interfere with the absorption of certain drugs.
8. Surface Area: Larger surface areas, such as the villi and microvilli in the small
intestine, increase the area for drug absorption.
Bioavailability (F)
Bioavailability is a measure of the extent and rate at which the active drug or its metabolites
enter systemic circulation and are made available to the site of action.
• Oral bioavailability is less than 100% due to the first-pass effect (explained below).
• IV drugs have 100% bioavailability because they are directly introduced into the
bloodstream.
First-Pass Effect:
• Drugs absorbed from the gastrointestinal tract are first carried to the liver via the
portal vein before entering the systemic circulation. During this passage through the
liver, some drugs undergo metabolism (biotransformation), reducing the amount of
active drug reaching the systemic circulation.
• Example: When morphine is taken orally, much of it is metabolized by the liver
before reaching the systemic circulation, reducing its bioavailability compared to IV
administration.
2. Distribution
Distribution refers to the process by which a drug is transported from the bloodstream to
various tissues and organs in the body. It is influenced by factors such as blood flow, plasma
protein binding, lipid solubility, and the permeability of cellular membranes.
1. Blood Flow:
o Organs and tissues with high blood flow, such as the heart, liver, kidneys,
and brain, typically receive drugs more quickly than tissues with lower blood
flow, such as muscle or fat.
o Example: Infections or inflammation can alter blood flow to tissues and affect
drug distribution.
2. Plasma Protein Binding:
o Many drugs bind to plasma proteins (like albumin) in the blood. Only the
free (unbound) drug is pharmacologically active and can cross cell
membranes to reach its site of action.
o Example: Warfarin, an anticoagulant, binds extensively to albumin. The
unbound fraction is the pharmacologically active form that can exert its
anticoagulant effects.
3. Volume of Distribution (Vd):
o The volume of distribution is a pharmacokinetic parameter that describes the
extent to which a drug is distributed in the body. It is calculated by comparing
the amount of drug in the body to the concentration of the drug in the blood or
plasma.
o Formula: Vd=Amount of drug in bodyConcentration of drug in plasmaVd =
\frac{{\text{Amount of drug in body}}}{{\text{Concentration of drug in
plasma}}}
o Drugs with a high volume of distribution tend to accumulate in tissues, while
drugs with a low volume of distribution remain primarily in the bloodstream.
4. Lipid Solubility:
o Lipophilic drugs (fat-soluble drugs) tend to distribute more widely throughout
the body, especially into fatty tissues, because they can easily cross lipid-rich
cell membranes.
o Example: Diazepam, a benzodiazepine, is highly lipophilic and distributes
widely throughout body tissues, including the brain and fat stores.
5. Blood-Brain Barrier (BBB):
o The blood-brain barrier is a selective permeability barrier that protects the
brain from potentially harmful substances in the bloodstream. It limits the
distribution of drugs to the brain.
o Lipophilic and small molecules tend to cross the BBB more easily.
o Example: Heroin (lipophilic) crosses the BBB easily and has rapid effects on
the brain, while penicillin (more hydrophilic) does not cross the BBB
effectively.
6. Tissue Affinity:
o Some drugs have a greater affinity for certain tissues and tend to concentrate
there.
o Example: Iodine in thyroid medications accumulates in the thyroid gland due
to its high affinity for iodine.
7. Barriers in the Body:
o Placenta: Some drugs cross the placenta and can affect fetal development,
which is important in pregnancy.
o Example: Alcohol and opioids can cross the placenta and affect the fetus.
8. Diseases: Certain diseases or conditions can affect drug distribution. For instance:
o Liver diseases can alter the distribution of drugs because the liver plays a
significant role in drug metabolism and protein synthesis.
o Renal disease can affect the volume of distribution and excretion of drugs.
The body can be considered as having different compartments in terms of drug distribution,
including:
1. Central Compartment: Primarily consists of the blood and organs with high
perfusion (e.g., heart, lungs, liver, kidneys). Drugs quickly distribute into this
compartment after administration.
2. Peripheral Compartment: Includes less vascularized tissues such as muscles and fat.
Drugs take longer to reach these tissues.
3. Tissues and Organs: Some drugs concentrate in specific tissues due to factors like
affinity, solubility, and protein binding.
Conclusion
In pharmacokinetics, absorption and distribution are key processes that determine how a
drug moves through the body and how it interacts with its target tissues. Absorption is
influenced by factors like the route of administration, drug formulation, and solubility, while
distribution is influenced by blood flow, plasma protein binding, lipid solubility, and other
physiological factors. These processes must be carefully considered in drug therapy to ensure
that drugs reach their targets
Following absorption and distribution, metabolism and excretion are the final two stages
of pharmacokinetics (the ADME process: Absorption, Distribution, Metabolism,
Excretion). These processes are critical for the elimination of drugs from the body, and they
determine the duration of action, toxicity, and potential drug interactions.
3. Metabolism (Biotransformation)
Metabolism is the process by which the body chemically alters drugs, usually to inactivate
them or make them more water-soluble, thus facilitating their excretion. The liver is the
primary organ responsible for drug metabolism, but other organs such as the kidneys,
lungs, and intestinal wall can also contribute.
1. Phase I (Functionalization):
o In Phase I, enzymes (mainly from the cytochrome P450 enzyme system, or
CYP450) introduce functional groups (such as hydroxyl, amino, or methyl
groups) to the drug molecule. This often makes the drug more hydrophilic
(water-soluble) or less active.
o Reactions: Oxidation, reduction, hydrolysis
o Example: CYP450 enzymes metabolize drugs like warfarin, diazepam, and
codeine.
▪ Example: Codeine is converted into its active metabolite morphine
by the enzyme CYP2D6.
Outcome: Phase I metabolism can either activate or inactivate the drug. Some drugs,
like codeine, are prodrugs (inactive compounds) that are activated in Phase I.
2. Phase II (Conjugation):
o In Phase II, the drug or its Phase I metabolite undergoes conjugation with a
large, water-soluble molecule, such as glucuronic acid, sulfate, or
glutathione. This generally increases water solubility and facilitates
excretion in urine.
o Reactions: Conjugation with glucuronic acid, sulfate, or amino acids.
o Example: Acetaminophen (paracetamol) is conjugated with glucuronic acid
or sulfate in Phase II to facilitate excretion.
4. Excretion
Excretion is the process by which the body eliminates drugs and their metabolites, primarily
through the kidneys, but also through other routes like bile, lungs, and sweat.
Routes of Excretion:
1. Half-life (t₁/₂):
o The half-life of a drug is the time it takes for the concentration of the drug in
the plasma to reduce by half.
o Formula: t1/2=0.693×VdClt_{1/2} = \frac{0.693 \times Vd}{Cl}, where Vd
is the volume of distribution and Cl is the clearance.
o A drug's half-life is influenced by its metabolism and excretion rate. Drugs
with long half-lives stay in the body longer, while those with short half-lives
are eliminated more quickly.
2. Clearance (Cl):
o Clearance refers to the volume of plasma from which the drug is completely
removed per unit time.
o Formula: Cl=RateofeliminationPlasmadrugconcentrationCl = \frac{Rate of
elimination}{Plasma drug concentration}
o Clearance can be affected by renal function, liver function, and drug
interactions.
3. Steady State (Css):
o The steady-state concentration is reached when the rate of drug
administration equals the rate of drug elimination. This typically occurs
after 4-5 half-lives.
o The steady-state concentration is critical for drugs that need to maintain a
therapeutic level.
Clinical Relevance of Metabolism and Excretion:
• Example: In renal failure, drugs such as digoxin or lithium may accumulate and
cause serious toxicity.
Conclusion
Metabolism and excretion are critical processes for the elimination of drugs from the body.
Metabolism, primarily occurring in the liver, transforms drugs into more water-soluble
metabolites that can be excreted via the kidneys, bile, or other routes. Excretion is the final
step in drug elimination, ensuring that drugs do not accumulate in the body to toxic levels.
Understanding these processes is essential for determining appropriate drug dosages,
avoiding drug interactions, and minimizing the risk of toxicity in different patient
populations.
Lecture 7
In pharmacology, the concepts of enzyme induction and enzyme inhibition are crucial for
understanding how drugs can influence drug metabolism and affect the efficacy and toxicity
of other medications. Both processes involve changes in the activity of cytochrome P450
enzymes (CYP450), which are the primary enzymes responsible for the metabolism of
drugs in the liver. These processes can significantly alter the rate of drug metabolism,
influencing the half-life and steady-state concentration of drugs in the body.
1. Enzyme Induction
Enzyme induction occurs when a drug or other substance increases the activity of a specific
metabolic enzyme, leading to an increase in the rate of drug metabolism. As a result,
drugs that are metabolized by the induced enzyme may have reduced plasma
concentrations, leading to decreased therapeutic effects or, in some cases, requiring higher
doses to maintain efficacy.
Enzyme inhibition refers to the decrease in the activity of a specific enzyme, leading to
slower drug metabolism. This results in increased plasma concentrations of drugs that are
metabolized by the inhibited enzyme, potentially causing increased drug effects and an
increased risk of toxicity.
• Increased drug levels: Enzyme inhibition can lead to higher plasma drug
concentrations, which may increase the risk of toxicity.
• Prolonged effects: Drugs metabolized by inhibited enzymes may have prolonged
effects due to slower elimination.
• Altered drug interactions: Inhibitors can increase the plasma levels of other drugs
metabolized by the same enzyme, enhancing their effects and side effects.
1. Drug Interactions:
o Enzyme induction and inhibition can result in significant drug-drug
interactions. Clinicians must be aware of these interactions when prescribing
medications to ensure optimal therapeutic effects and avoid toxicity or loss
of efficacy.
2. Monitoring Drug Levels:
o For drugs that are substrates of inducible or inhibited enzymes, therapeutic
drug monitoring (TDM) may be required to adjust dosing and avoid adverse
effects.
o Example: Warfarin, a CYP2C9 substrate, requires close monitoring of INR
(International Normalized Ratio) because enzyme inducers (e.g., rifampin)
can decrease its effect, while enzyme inhibitors (e.g., fluconazole) can
increase its anticoagulant effect.
3. Impact on Drug Efficacy:
o Inducers may reduce the effectiveness of drugs, necessitating higher doses,
while inhibitors may cause toxicity, necessitating dose reductions or
discontinuation.
4. Special Populations:
o Elderly, liver disease patients, or those with genetic polymorphisms in
metabolic
enzymes may experience altered responses to drugs due to enzyme inhibition or induction.
Personalized medicine, such as adjusting doses based on genetic testing or monitoring drug
levels, is increasingly being employed to tailor therapy.
Conclusion
Both enzyme induction and enzyme inhibition can significantly influence drug metabolism,
with inducers speeding up metabolism and potentially lowering the effectiveness of other
drugs, and inhibitors slowing down metabolism, potentially leading to drug toxicity.
Clinicians must understand these processes to manage drug interactions, adjust dosages
appropriately, and ensure patient safety.
Lecture 8
Kinetics of Elimination
The kinetics of elimination describe how drugs are removed from the body and involve two
primary processes: metabolism (biotransformation) and excretion. The rate of elimination
is a key factor in determining the half-life of a drug, which ultimately influences its duration
of action, frequency of dosing, and potential for accumulation in the body.
The kinetics of elimination can be understood using two main models: zero-order kinetics
and first-order kinetics. These models describe how the drug concentration in the plasma
decreases over time.
1. First-Order Kinetics
Mathematical Representation:
Where:
o kek_e is the elimination rate constant (fraction of the drug eliminated per
unit time)
o C(t)C(t) is the concentration of the drug at time tt
• The half-life in first-order kinetics is given by:
t1/2=0.693ket_{1/2} = \frac{0.693}{k_e}
This shows that the half-life is independent of the initial concentration and remains
constant over time.
Graphical Representation:
• The plasma concentration of the drug decreases in an exponential fashion over time.
A graph of concentration vs. time results in a logarithmic decay (a straight line on a
semi-logarithmic scale).
2. Zero-Order Kinetics
In zero-order kinetics, the rate of elimination is constant, meaning the drug is eliminated at
a fixed rate, regardless of its concentration in the bloodstream. This occurs when the
elimination mechanisms (such as liver enzymes or renal clearance) are saturated or when the
drug is present in high concentrations, overwhelming the metabolic or excretory capacity.
• Constant elimination rate: A fixed amount of the drug is eliminated per unit of time,
irrespective of the drug concentration. This can lead to drug accumulation if dosing
is not adjusted.
• Half-life (t₁/₂): In zero-order kinetics, the half-life is not constant because the
elimination is independent of the drug concentration. As the concentration decreases,
the half-life can change.
Mathematical Representation:
o k0k_0 is the elimination rate (fixed amount of drug eliminated per unit time).
• The concentration of the drug decreases linearly with time, leading to a non-
logarithmic decline in a concentration vs. time graph.
Graphical Representation:
• The concentration vs. time graph shows a linear decline in the drug concentration
over time, unlike the exponential decline seen in first-order kinetics.
The half-life is an important pharmacokinetic parameter that quantifies the time it takes for
the plasma concentration of a drug to decrease by half. The half-life depends on the
elimination rate and is used to determine how frequently a drug should be dosed to maintain
effective concentrations.
• First-Order Kinetics:
t1/2=0.693ket_{1/2} = \frac{0.693}{k_e}
• Zero-Order Kinetics:
t1/2=C0k0t_{1/2} = \frac{C_0}{k_0}
Where C0C_0 is the initial concentration of the drug, and k0k_0 is the constant
elimination rate.
4. Clearance (Cl)
Clearance (Cl) is a pharmacokinetic parameter that describes the volume of plasma from
which a drug is completely removed per unit of time. It is important because it helps define
the elimination rate of the drug. Clearance can occur through different routes (e.g., renal,
hepatic) and is a key factor in determining half-life.
Where:
The steady state is the point at which the rate of drug administration equals the rate of
elimination. After multiple doses of a drug, steady state is generally achieved after 4-5 half-
lives of the drug.
First-Order Kinetics:
• Predictable elimination: Drugs that follow first-order kinetics are eliminated at a rate
proportional to their concentration, making it easier to predict their behavior in the
body.
• Dosing frequency: Since the half-life is constant, it allows for consistent drug levels
over time if the drug is administered at regular intervals.
• Toxicity risk: Since the half-life is fixed, drug accumulation to toxic levels can be
managed with proper dosing and monitoring.
Zero-Order Kinetics:
Conclusion
The kinetics of elimination describe how drugs are removed from the body through
processes of metabolism and excretion. The two main types of elimination kinetics are first-
order and zero-order kinetics. Drugs following first-order kinetics are eliminated at a rate
proportional to their concentration, with a constant half-life, while drugs following zero-
order kinetics are eliminated at a constant rate, independent of concentration, leading to
potential saturation and toxicity risk. Understanding these kinetic models is essential for
dosing strategies, monitoring drug levels, and preventing drug toxicity.