Podcast
Questions and Answers
Concerning receptors with prolonged effects post-agonist removal, which mechanism most accurately delineates the sustained signaling observed in the absence of continued ligand binding?
Concerning receptors with prolonged effects post-agonist removal, which mechanism most accurately delineates the sustained signaling observed in the absence of continued ligand binding?
- Constitutive receptor activity independent of ligand presence drives continuous downstream effector activation.
- Stable, covalent modification of intracellular signaling proteins maintains signal transduction. (correct)
- Agonist-induced receptor phosphorylation prevents subsequent interactions with arrestins, prolonging signaling.
- Receptor internalization and subsequent lysosomal degradation result in irreversible signal termination.
Given the extended duration of action observed with certain receptor types, how do alterations in receptor turnover rate and trafficking dynamics modulate the overall cellular responsiveness to hormonal stimuli?
Given the extended duration of action observed with certain receptor types, how do alterations in receptor turnover rate and trafficking dynamics modulate the overall cellular responsiveness to hormonal stimuli?
- Impaired receptor recycling leads to receptor accumulation on the cell surface, desensitizing the cell to the agonist.
- Ubiquitination of receptors promotes enhanced association with chaperones, preventing premature degradation and maintaining responsiveness.
- Reduced receptor internalization impedes signal termination, resulting in prolonged activation even after agonist removal, potentially leading to tolerance. (correct)
- Increased receptor endocytosis diminishes sustained signaling by accelerating receptor degradation, enhancing overall sensitivity.
Considering receptors for steroid hormones and thyroxine, which statement accurately characterises their mechanism relative to classical plasma membrane receptors?
Considering receptors for steroid hormones and thyroxine, which statement accurately characterises their mechanism relative to classical plasma membrane receptors?
- These receptors are exclusively located on the plasma membrane to rapidly regulate ion channel conductance.
- They activate G proteins on the cell surface, leading to a rapid cascade of second messenger production.
- They are internalized upon ligand binding and initiate signaling through endosomal pathways exclusively.
- They directly modulate gene transcription by binding to intracellular receptors, influencing protein synthesis. (correct)
In the context of insulin receptor signaling, what mechanism primarily accounts for the prolonged downstream effects observed following the initial binding of insulin, beyond the immediate phosphorylation events?
In the context of insulin receptor signaling, what mechanism primarily accounts for the prolonged downstream effects observed following the initial binding of insulin, beyond the immediate phosphorylation events?
How does the dimerization status of the receptor tyrosine kinases following ligand binding influence the duration and intensity of downstream signaling cascades, and what post-translational modifications contribute to this regulation?
How does the dimerization status of the receptor tyrosine kinases following ligand binding influence the duration and intensity of downstream signaling cascades, and what post-translational modifications contribute to this regulation?
Given the heteropentameric stoichiometry characteristic of certain ligand-gated ion channels, what is the most precise biophysical consequence of disrupting the highly conserved intersubunit interactions at the α1-γ interface, considering cooperative binding kinetics?
Given the heteropentameric stoichiometry characteristic of certain ligand-gated ion channels, what is the most precise biophysical consequence of disrupting the highly conserved intersubunit interactions at the α1-γ interface, considering cooperative binding kinetics?
In the context of a pentameric ligand-gated ion channel with α1, α2, β, γ, and δ subunits, if a mutation within the γ subunit selectively impairs its physical interaction with the intracellular loop of the β subunit, what downstream effect on G-protein signaling is most probable?
In the context of a pentameric ligand-gated ion channel with α1, α2, β, γ, and δ subunits, if a mutation within the γ subunit selectively impairs its physical interaction with the intracellular loop of the β subunit, what downstream effect on G-protein signaling is most probable?
Considering a ligand-gated ion channel composed of α1, α2, β, γ, and δ subunits, what is the predicted impact on channel kinetics if the binding domain on the α1 subunit is mutated such that it exhibits a 100-fold increased affinity for an antagonist, but no change in agonist affinity?
Considering a ligand-gated ion channel composed of α1, α2, β, γ, and δ subunits, what is the predicted impact on channel kinetics if the binding domain on the α1 subunit is mutated such that it exhibits a 100-fold increased affinity for an antagonist, but no change in agonist affinity?
Given a heteromeric receptor composed of α1, α2, β, γ, and δ subunits, how would allosteric modulation at a site distal from the orthosteric binding pocket on the alpha subunits most likely impact agonist efficacy and potency?
Given a heteromeric receptor composed of α1, α2, β, γ, and δ subunits, how would allosteric modulation at a site distal from the orthosteric binding pocket on the alpha subunits most likely impact agonist efficacy and potency?
In a ligand-gated ion channel composed of α1, α2, β, γ, and δ subunits, how does phosphorylation of a tyrosine residue located within the intracellular loop connecting transmembrane domains TM3 and TM4 of the β subunit most directly influence receptor function?
In a ligand-gated ion channel composed of α1, α2, β, γ, and δ subunits, how does phosphorylation of a tyrosine residue located within the intracellular loop connecting transmembrane domains TM3 and TM4 of the β subunit most directly influence receptor function?
In patients with concomitant hepatic and renal dysfunction, which pharmacokinetic parameter is LEAST predictably altered, thereby necessitating individualized dosage regimen adjustments for drugs primarily cleared via these routes?
In patients with concomitant hepatic and renal dysfunction, which pharmacokinetic parameter is LEAST predictably altered, thereby necessitating individualized dosage regimen adjustments for drugs primarily cleared via these routes?
A patient with severe heart failure exhibits heightened sensitivity to digoxin, necessitating a dosage adjustment compared to individuals with normal cardiac function. Which underlying pathophysiological mechanism MOST directly contributes to this increased sensitivity?
A patient with severe heart failure exhibits heightened sensitivity to digoxin, necessitating a dosage adjustment compared to individuals with normal cardiac function. Which underlying pathophysiological mechanism MOST directly contributes to this increased sensitivity?
A patient with advanced cirrhosis exhibits ascites, jaundice, and hepatic encephalopathy. Which alteration in drug disposition is MOST likely to contribute to increased drug sensitivity and potential toxicity?
A patient with advanced cirrhosis exhibits ascites, jaundice, and hepatic encephalopathy. Which alteration in drug disposition is MOST likely to contribute to increased drug sensitivity and potential toxicity?
A patient with chronic kidney disease (CKD) and an estimated glomerular filtration rate (eGFR) of 25 mL/min presents with signs of digoxin toxicity despite being within the established therapeutic range for serum digoxin concentrations. What is the MOST probable explanation for this observation?
A patient with chronic kidney disease (CKD) and an estimated glomerular filtration rate (eGFR) of 25 mL/min presents with signs of digoxin toxicity despite being within the established therapeutic range for serum digoxin concentrations. What is the MOST probable explanation for this observation?
A patient with decompensated heart failure is prescribed intravenous furosemide. Despite adequate dosing, the patient exhibits minimal diuretic response and persistent fluid overload. Which of the following mechanisms MOST likely contributes to this resistance to furosemide's effects?
A patient with decompensated heart failure is prescribed intravenous furosemide. Despite adequate dosing, the patient exhibits minimal diuretic response and persistent fluid overload. Which of the following mechanisms MOST likely contributes to this resistance to furosemide's effects?
Given the pharmacological implications of isomeric variation in drug composition, which of the following best elucidates the potential clinical ramifications arising from the administration of a racemic mixture of a chiral drug?
Given the pharmacological implications of isomeric variation in drug composition, which of the following best elucidates the potential clinical ramifications arising from the administration of a racemic mixture of a chiral drug?
In the context of stereoisomerism and drug action, what is the most critical consideration when evaluating the therapeutic index of a drug formulated as a racemic mixture, especially when one isomer exhibits negligible therapeutic activity and the other possesses potential for severe adverse effects?
In the context of stereoisomerism and drug action, what is the most critical consideration when evaluating the therapeutic index of a drug formulated as a racemic mixture, especially when one isomer exhibits negligible therapeutic activity and the other possesses potential for severe adverse effects?
Considering a scenario where a novel drug, synthesized as a racemic mixture, demonstrates in vitro activity as a potent agonist at a specific receptor, but exhibits markedly reduced in vivo efficacy. Which of the following mechanisms most plausibly accounts for this discrepancy?
Considering a scenario where a novel drug, synthesized as a racemic mixture, demonstrates in vitro activity as a potent agonist at a specific receptor, but exhibits markedly reduced in vivo efficacy. Which of the following mechanisms most plausibly accounts for this discrepancy?
A pharmaceutical company is developing a new drug currently formulated as a racemic mixture. During clinical trials, a subset of patients exhibits paradoxical responses, with some experiencing the expected therapeutic benefits and others exhibiting severe adverse reactions. What is the most appropriate next step to investigate the cause of these variable outcomes?
A pharmaceutical company is developing a new drug currently formulated as a racemic mixture. During clinical trials, a subset of patients exhibits paradoxical responses, with some experiencing the expected therapeutic benefits and others exhibiting severe adverse reactions. What is the most appropriate next step to investigate the cause of these variable outcomes?
In the development of a novel analgesic, a racemic mixture demonstrates potent pain-relieving effects in preclinical models. However, one enantiomer is found to possess a concerning off-target activity leading to significant cardiovascular risks. To optimize the drug's safety profile, which of the following strategies would be most effective?
In the development of a novel analgesic, a racemic mixture demonstrates potent pain-relieving effects in preclinical models. However, one enantiomer is found to possess a concerning off-target activity leading to significant cardiovascular risks. To optimize the drug's safety profile, which of the following strategies would be most effective?
Considering a scenario where two drugs, Drug A and Drug B, exhibit similar efficacy profiles but differ significantly in their receptor binding affinities, what is the most critical factor to consider when selecting between these drugs for chronic use in a patient with compromised renal function?
Considering a scenario where two drugs, Drug A and Drug B, exhibit similar efficacy profiles but differ significantly in their receptor binding affinities, what is the most critical factor to consider when selecting between these drugs for chronic use in a patient with compromised renal function?
In the context of drug development, if a novel compound demonstrates exceptional target receptor affinity in vitro but exhibits significantly reduced efficacy in vivo due to extensive first-pass metabolism and poor bioavailability, which optimization strategy would be most rational to pursue?
In the context of drug development, if a novel compound demonstrates exceptional target receptor affinity in vitro but exhibits significantly reduced efficacy in vivo due to extensive first-pass metabolism and poor bioavailability, which optimization strategy would be most rational to pursue?
In the context of receptor pharmacology, which of the following scenarios would MOST accurately describe a situation where competitive antagonism transitions into insurmountable antagonism?
In the context of receptor pharmacology, which of the following scenarios would MOST accurately describe a situation where competitive antagonism transitions into insurmountable antagonism?
Considering the intricacies of signal transduction pathways, which of the following mechanisms would BEST explain a scenario where prolonged exposure to an agonist leads to a paradoxical increase in receptor sensitivity, rather than the typical desensitization?
Considering the intricacies of signal transduction pathways, which of the following mechanisms would BEST explain a scenario where prolonged exposure to an agonist leads to a paradoxical increase in receptor sensitivity, rather than the typical desensitization?
Given two drugs, one highly potent with a narrow therapeutic window and the other less potent but with a wide therapeutic window, under what clinical scenario would the less potent drug be preferentially indicated despite requiring higher dosages?
Given two drugs, one highly potent with a narrow therapeutic window and the other less potent but with a wide therapeutic window, under what clinical scenario would the less potent drug be preferentially indicated despite requiring higher dosages?
Within the framework of pharmacodynamics, how does receptor cooperativity MOST profoundly influence the observed dose-response relationship, particularly in scenarios involving multimeric receptor complexes?
Within the framework of pharmacodynamics, how does receptor cooperativity MOST profoundly influence the observed dose-response relationship, particularly in scenarios involving multimeric receptor complexes?
If Drug X, a novel analgesic, exhibits comparable efficacy to morphine in clinical trials but requires a 100-fold higher dose to achieve the same level of pain relief, how would you comparatively assess the clinical utility of Drug X versus morphine, considering both acute and chronic use scenarios?
If Drug X, a novel analgesic, exhibits comparable efficacy to morphine in clinical trials but requires a 100-fold higher dose to achieve the same level of pain relief, how would you comparatively assess the clinical utility of Drug X versus morphine, considering both acute and chronic use scenarios?
Considering a scenario in which two drugs, Drug A and Drug B, both target the same receptor to manage hypertension, but Drug A has a significantly higher binding affinity and requires a much lower dose. However, Drug A also exhibits a markedly shorter half-life compared to Drug B. Which pharmacokinetic or pharmacodynamic factor would be most crucial in determining the preferred drug for a patient with a history of medication non-adherence?
Considering a scenario in which two drugs, Drug A and Drug B, both target the same receptor to manage hypertension, but Drug A has a significantly higher binding affinity and requires a much lower dose. However, Drug A also exhibits a markedly shorter half-life compared to Drug B. Which pharmacokinetic or pharmacodynamic factor would be most crucial in determining the preferred drug for a patient with a history of medication non-adherence?
In the realm of pharmacogenomics and individualized medicine, which of the following mechanisms BEST elucidates the phenomenon where a patient exhibits an unexpectedly exaggerated response to a standard drug dosage due to a rare genetic variant?
In the realm of pharmacogenomics and individualized medicine, which of the following mechanisms BEST elucidates the phenomenon where a patient exhibits an unexpectedly exaggerated response to a standard drug dosage due to a rare genetic variant?
Considering the complexities of biased agonism, which statement MOST accurately describes a scenario where a drug selectively activates one signaling pathway downstream of a G protein-coupled receptor (GPCR) while inhibiting another?
Considering the complexities of biased agonism, which statement MOST accurately describes a scenario where a drug selectively activates one signaling pathway downstream of a G protein-coupled receptor (GPCR) while inhibiting another?
Flashcards
What are pentameric receptors?
What are pentameric receptors?
Receptors with five transmembrane subunits (α1, α2, β, γ, δ).
Where does an agonist bind?
Where does an agonist bind?
The extracellular part of the receptor.
What triggers G-protein activation?
What triggers G-protein activation?
Binding of an agonist to the receptor.
What is a G-protein?
What is a G-protein?
Signup and view all the flashcards
α1, α2, β, γ, δ
α1, α2, β, γ, δ
Signup and view all the flashcards
Signaling Proteins
Signaling Proteins
Signup and view all the flashcards
Receptors with Persistent Effects
Receptors with Persistent Effects
Signup and view all the flashcards
Insulin
Insulin
Signup and view all the flashcards
Corticosteroids
Corticosteroids
Signup and view all the flashcards
Sex Hormones
Sex Hormones
Signup and view all the flashcards
What is potency?
What is potency?
Signup and view all the flashcards
What is efficacy?
What is efficacy?
Signup and view all the flashcards
Which is more important: efficacy or potency?
Which is more important: efficacy or potency?
Signup and view all the flashcards
Can potency overcome limited efficacy?
Can potency overcome limited efficacy?
Signup and view all the flashcards
What is a major limitation to increasing drug dosage?
What is a major limitation to increasing drug dosage?
Signup and view all the flashcards
Pharmacodynamics
Pharmacodynamics
Signup and view all the flashcards
What is a partial agonist?
What is a partial agonist?
Signup and view all the flashcards
What is a racemic mixture?
What is a racemic mixture?
Signup and view all the flashcards
What are isomers?
What are isomers?
Signup and view all the flashcards
Why do isomers matter?
Why do isomers matter?
Signup and view all the flashcards
How is a drug affected?
How is a drug affected?
Signup and view all the flashcards
Liver or kidney disease effect on drugs
Liver or kidney disease effect on drugs
Signup and view all the flashcards
Failing heart and drug sensitivity
Failing heart and drug sensitivity
Signup and view all the flashcards
Pathological status affect on drug response
Pathological status affect on drug response
Signup and view all the flashcards
Heart failure and drug effects
Heart failure and drug effects
Signup and view all the flashcards
Organ disease and drug response
Organ disease and drug response
Signup and view all the flashcards
Study Notes
- Pharmacology is a basic science focused on using small molecules to prevent, diagnose, or treat diseases.
- Clinical pharmacology involves the use of drugs in humans and includes drug-patient interaction.
- A drug is any chemical molecule that can interact with body systems and produce an effect.
Drug-Body Interactions
- Pharmacokinetics explains what the body does to the drug: "The effect of body on drug."
- Pharmacodynamics explains what the drug does to the body: "The effect of drug on body."
- ADME in pharmacokinetics stands for Absorption, Distribution, Metabolism, and Excretion.
Pharmacodynamics (Mechanism of Drug Action)
- Drug effects are produced through interaction with body control systems, notably regulatory proteins, or direct mechanisms.
- Direct mechanisms involve chemical or physical interactions.
Receptors
- Receptors are protein macromolecules, that when combined with a drug, may be activated or blocked.
- A ligand is any molecule that can combine with receptors.
- An agonist is a ligand that activates the receptor.
- An antagonist is a ligand that blocks the receptor.
- Affinity is the empathy of the receptor to the ligand.
- Affinity determines the number of receptors occupied by the drug.
Types of Receptors
- Types of receptors include Ion channel-linked receptors and G-protein-linked receptors.
Ion Channel-Linked Receptors
- Ion channel-linked receptors: These consist of an ion channel with 5 transmembrane subunits (α1, α2, β, γ, δ).
- The binding of an agonist to the extracellular part of the receptor causes the opening of the channel for a specific ion.
- Responses for the receptors are very fast, and the duration is very short.
- Nicotinic Ach receptors in the motor end-plate: The ion channel opens for Na ions in response to stimulation by Ach.
- Gama aminobuteric acid (GABA) receptors in brain: The ion channel opens for Cl ions in response to stimulation by GABA.
G-Protein-Linked Receptors
- G-protein-linked receptors: These consist of 7 membrane subunits.
- The binding of an agonist to the extracellular part of the receptor activates an intracellular G-protein.
- When a G-protein is activated, its a subunit binds to GTP to be phosphorylated, leading to stimulatory or inhibitory responses.
- The response here is slower than ion channel receptors, and the duration is longer.
- Stimulatory G-protein (Gs) increases the adenyl cyclase enzyme, which leads to increased cAMP and activation of protein kinases, like B1 and B2-adrenergic receptors.
- Inhibitory G-protein (Gi) decreases the adenyl cyclase enzyme, which leads to decreased cAMP inhibition of protein kinases, like a2- adrenergic receptors and M2 muscarinic receptors.
- Gq-coupled receptors increase inositol triphosphate (IP3) & diacylglycerol (DAG), where IP3 increases free intracellular Ca2+, like al-adrenergic receptors, M1 and M3 muscarinic receptors.
Tyrosine Kinase (TK)-Linked Receptors
- Tyrosine Kinase (TK)-Linked Receptors consist of 2 large domains: extracellular hormone-binding and intracellular TK-binding domains, connected by a transmembrane segment.
- The binding of agonists to the hormone-binding domain activates the intracellular domain, which then activates a TK enzyme, this starts activation of other proteins (known as signaling proteins).
- Insulin receptors are linked to TK.
Intracellular Receptors
- Intracellular Receptors: These are inside the cell either in the cytoplasm or directly on DNA.
- They regulate the transcription of genes in the nucleus or mitochondria.
- Agonists for these receptors need to enter inside the cell to reach them.
- Responses here are slow (time required for synthesis of new proteins), and effects persist for a long after the agonist is removed.
- Examples include receptors for corticosteroids, sex hormones, and thyroxin.
Types of Drug-Receptor Bonds
- Hydrogen bonds: Weak and reversible attraction.
- Ionic bonds (Electrostatic): Strong and reversible attraction between opposing charges.
- Covalent bonds: Very strong and irreversible bond; if it occurs, the receptor becomes permanently blocked.
Biological Response to Drug-Receptor Binding
- Agonist effect: the drug combines with the receptor and gives a response.
- Antagonist effect: the drug combines with the receptor but gives no response and prevents the receptor from binding to another drug.
Types of Responses to Drugs
- Graded Response: Increased proportionally to the dose of agonist; responses can be tested in one or more animals. It responds to most drugs.
- Quantal Response: Does not increase proportionally to the agonist; it is an all-or-none response, for a few drugs. It cannot be tested in one animal and must be tested in a group of animals.
- The heart's response to adrenaline is graded, and the prevention of convulsions by antiepileptic drugs is Quantal.
Effectiveness and Safety
- Efficacy refers to the ability of a drug to produce an effect, typically measured by Emax.
- Emax indicates the maximal response that a drug can elicit at full concentration.
- A full agonist produces a maximal response at full receptor occupancy, as opposed to a partial agonist, which can only produce a submaximal response, even at full occupancy.
- ED50 is the dose of a drug that gives 50% of the maximum effect.
- Potency refers to the dose that gives a desired effect in 50% of a test population.
- A "potent" drug gives its ED50 in smaller doses.
- Potency is generally less clinically important than efficacy
Safety
- TD50 (Toxic Dose) is the dose needed to cause a harmful effect in 50% of a test population.
- LD50 (Lethal Dose) is the dose needed to cause death in 50% of a test group of animals.
- The therapeutic index (TI) can be computed as LD50/ED50.
- It is a measure of safety, and drugs with a high TI are safer for clinical use.
- Warfarin has a narrow TI, and this require careful monitoring.
Antagonist Effect
- An antagonist combines with the receptor and does not activate it.
- It causes a pharmacological response by inhibiting the actions of endogenous substances or other drugs.
- Physiological Antagonism is an antagonism between two drugs producing opposite effects by the activation of different receptors.
Competitive Types
- If the antagonist binds to the same site as the agonist on the receptor, types include reversible and irreversible.
- Reversible: Weak bond with the block, reversed by high doses of the agonist, short duration of block with antagonists easily washed off receptors
- Irreversible: Covalent bond blocked, non-surmountable increase dose of agonist.
Non-Competitive
- Where the antagonist binds to another site on the receptor and prevents the action of the agonist.
Other Types of Drug Antagonism
- Chemical antagonism occurs when one acidic drug is added to a basic drug, causing precipitation.
- The addition of gentamycin (basic drug) to carpenicillin (acidic drug) causes a chemical complex.
- Adrenaline is the physiological antagonist of histamine.
- Administered protamine is used to counteract heparin overdose, because it carries a +ve charge, while heparin carries a -ve charge.
- One mg of protamine neutralizes 100 units of heparin.
- One drug prevents the absorption of another (antacids iron/asprin) or increases the metabolism of another (ex. rifampicin which decreases effect of oral contraceptive pills)
- One drug may increase the excretion of another, such as how NaHCO3 causes alkalinization of urine.
Ion Channels
- Drugs could modulate ion channels through four ways: physical block, acting as part of a receptor, modulation G-protein linked receptors, or modulation by intracellular ATP.
Enzymes
- Drugs could affect enzymes by acting as a competitive inhibitor, an irreversible inhibitor, a false substrate, and the induction or inhibition of hepatic microsomal enzymes activity
Carrier Molecules
- Carrier molecules are small protein molecules that carry organic molecules across the cell membrane when they are too large or too polar.
- Drugs can affect these molecules by blocking their recognition site.
Factors Affecting Dose-Response Relationship
- Factors include those related to the Drug or the Patient.
Factors Related to the Drug
- Drug shape (stereoisomerism)
- Molecular weight (MW)
- Time of drug administration, or chronopharmacology.
- Drug cumulation
- Drug combination
- Drug shape: Receptor sites are usually sensitive for one stereoisomer and not suitable for another.
- A drug must be administered according to the circadian rhythm of the body to get better responses and avoid side effects.
Drug Cumulation
- Drug cumulation: occurs when the rate of drug exceeds its rate of elimination, especially in those with liver or renal disease.
Drug Combination
- Drug combination: very common in practice used in the following effects:
- Summation of addition: where the combined effect of 2 drugs is equal to the sum of their individual effects
- Synergism: where the combined effect is greater than the sum of their individual effects
- Potentiation: where a similar effect is achieved as synergism, however one drug increases other drugs with little to no effect
- Antagonism: The drug abolishes the effects of the other drugs.
Factors Related to the Patient
- Age, sex, and weight.
- Pathological status
- Pharmacogenetic factors, or idiosyncrasy. This is genetic abnormality in drug.
- Hyporeactivity to drugs
- Hyperactivity to drugs
Pharmacogenetic Factors (Idiosyncrasy)
- Pharmacogenetic Factors is an abnormal drug response caused by genetic abnormality in drug metabolism.
Examples of Heritable Conditions Causing Exaggerated Drug Response
- A) Pseudo-cholinesterase deficiency.
- B) Glucose-6-phosphate dehydrogenase (G6PD) deficiency
- C) Thiopurine methyl-transferase (TPMT) deficiency
- D) Acetylator phenotypes
Examples of Heritable Conditions Causing Decreased Drug Response
- Resistance to coumarin (warfarin) anticoagulants
- Resistance to vit D (vit D-resistant rickets)
- Resistance to mydriatics
Hyporeactivity to Drugs
- Hyporeactivity to drugs, also known as tolerance or tachyphylaxis.
- Tolerance involves a progressive decrease in drug response with successive administration over a long period.
- Tachyphylaxis involves an acute type of tolerance that occurs very rapidly.
Mechanisms
- Receptor desensitization
- Receptor down-regulation
- Exhaustion of mediators
- Increased metabolic degradation of a drug by induction of hepatic enzymes
- Learning how to actively overcome a certain drug-induced effect through practice
Hyperreactivity to Drugs
- Rebound Effect: Recurring of symptoms in exaggerated form.
- Withdrawl Effect (Syndrome): addition of new symptoms is caused when a drug is stopped suddenly.
N.B.
- Beta-blockers, Clonidine, Cimetidine, Corticosteroids, Morphine, Warfarin should not be topped suddenly.
Factors Affecting Dose-Response Relationship
- Factors affecting the dose response relationship include pharmacogenetics.
Pharmacogenetics
- Study of variation in single drug response due to genetic variation of single genes.
- Variations in around 20 provide useful predictions of reactions to 80-100 drugs.
Examples of Pharmacogenetics
-
- Pseudo-cholinesterase (PCHE) deficiency includes performing biochemical test on those who show exaggerated sensitivity to succinylcholine.
-
- G6PD deficiency
-
- Thiopurine methyltransferase (TPMT)
Principles of Drug Interaction
- Drug interactions in vitro: anti-pseudomonal penicillins and aminoglycosides form complexes in infusion.
- Drug interactions in vivo:
A. Absorption
- Tetracycline + Ca2+, Mg2+ and Al3+ result in complex formation.
- Cholestyramine +digitalis and thyroxin result in inhibited absorption of digitalis/thyroxin.
- Anticholinergic drugs decrease intestinal motility
- Prokinetic drugs increase intestinal motility
- Antiacids decrease absorption of salicylates.
- Ketoconazole is poorly absorbed in absence of gastric acidity.
B. Distribution
- Sulfonamides displace bilirubin from plasma protein in premature infants which can lead to kernicterus.
- Phenylbutazone displaces warfarin which leads to excessive bleeding.
C. Metabolism
- Inhibition or induction of microsomal metabolism:
- MAO Inhibitors inhibits metabolism of benzodiazepines.
D. Excretion
- Reducation in urinary elimination
- Probenecid decrease Penicillin excretion & Quinidine decreases digoxin excretion Changes in urinary volume:
- Increase volume cause decrease reabsorption
B. Pharmaco-Dynamics Interaction
- Types
- SYNERGISM: increase of one drug toxicity
- POTENTIATION: CNS depression of opioids
- CHANGES: ↑ digitalis toxicity with potassium and decrease toxicity when K is high.
Studying That Suits You
Use AI to generate personalized quizzes and flashcards to suit your learning preferences.