Drug Absorption and Bioavailability
47 Questions
0 Views

Choose a study mode

Play Quiz
Study Flashcards
Spaced Repetition
Chat to Lesson

Podcast

Play an AI-generated podcast conversation about this lesson

Questions and Answers

Explain how the lipid solubility of a drug affects its ability to be absorbed in the body. Relate your answer to the permeability of cell membranes.

Lipid solubility enhances absorption because cell membranes are primarily lipid bilayers. Drugs with high lipid solubility can more easily diffuse across these membranes.

A drug has poor aqueous solubility. What challenges might this pose for its absorption, especially via the gastrointestinal tract (GIT)?

Poor aqueous solubility limits the drug's ability to dissolve in the fluids of the GIT, hindering its absorption across the epithelial lining.

Define bioavailability and explain why a drug administered intravenously (IV) has a bioavailability of 1 (or 100%).

Bioavailability is the fraction of a drug that reaches systemic circulation. IV administration bypasses absorption processes, delivering the entire dose directly into the bloodstream.

Explain why the oral route of drug administration may lead to variable drug absorption rates.

<p>Variations in gastric emptying, intestinal motility, interactions with food, and individual differences in GI flora can impact drug absorption.</p> Signup and view all the answers

How does the sublingual route of administration bypass first-pass metabolism, and why is this clinically significant?

<p>Venous drainage from the sublingual area enters systemic circulation directly, avoiding the liver. This results in higher bioavailability and quicker onset of action.</p> Signup and view all the answers

A patient with liver cirrhosis is prescribed an oral medication that undergoes significant first-pass metabolism. How might the patient's condition affect the drug's bioavailability, and what adjustments might be necessary?

<p>Liver cirrhosis impairs first-pass metabolism, potentially increasing the drug's bioavailability. Dosage adjustments may be needed to prevent toxicity.</p> Signup and view all the answers

Explain the relationship between a drug's permeability and its ability to cross biological membranes, and provide examples of molecular properties that influence permeability.

<p>Permeability determines how easily a drug can cross biological membranes. Lipid solubility, molecular size, and the presence of specific transporters affect permeability.</p> Signup and view all the answers

A new drug is developed that is highly charged at physiological pH. Predict how this property will affect its absorption and distribution in the body, and suggest potential routes of administration that might be more effective.

<p>A highly charged drug will have difficulty crossing lipid membranes, limiting oral absorption and distribution. Intravenous or subcutaneous routes might be more effective.</p> Signup and view all the answers

Explain how competition for plasma protein binding can lead to drug-drug interactions, providing a specific example.

<p>When two drugs compete for the same plasma protein binding sites, one drug can displace the other, leading to a higher concentration of free drug. For example, fonamide can displace warfarin, leading to increased bleeding.</p> Signup and view all the answers

A patient has a low albumin level due to liver disease. How might this affect the distribution and effect of a highly protein-bound drug?

<p>Lower albumin means less protein binding, resulting in a higher free drug concentration in the plasma. This can lead to an increased drug effect and potential toxicity.</p> Signup and view all the answers

Explain why lipid-soluble drugs tend to have a larger volume of distribution ($V_d$) compared to water-soluble drugs.

<p>Lipid-soluble drugs can easily cross cell membranes and distribute into tissues, including fat, leading to a larger volume of distribution. Water-soluble drugs are largely confined to the plasma and extracellular fluid.</p> Signup and view all the answers

How does blood flow to a tissue affect the rate at which a drug reaches equilibrium in that tissue?

<p>Higher blood flow allows drugs to reach equilibrium faster because more drug molecules are delivered to the tissue per unit of time.</p> Signup and view all the answers

Why is the blood-brain barrier significant for drug distribution, and what characteristics of a drug allow it to cross this barrier more readily?

<p>The blood-brain barrier is highly selective, protecting the brain from many substances. Lipid-soluble, non-ionized drugs cross it more easily.</p> Signup and view all the answers

A drug is primarily eliminated through hepatic metabolism. How might liver dysfunction affect the drug's half-life and potential for toxicity?

<p>Liver dysfunction impairs drug metabolism, increasing the drug's half-life. This can lead to accumulation and increased risk of toxicity.</p> Signup and view all the answers

Explain why a drug with a larger volume of distribution ($V_d$) typically has a slower excretion rate?

<p>A larger $V_d$ means more drug is in the tissues versus the plasma. Because excretion primarily occurs from what is in the plasma, it will take longer to eliminate.</p> Signup and view all the answers

How does tissue affinity affect the availability of a drug?

<p>If a drug binds strongly to certain tissues (e.g., mucopolysaccharides), its availability is reduced, as it is sequestered in those tissues and less free drug is available to exert its effects elsewhere in the body.</p> Signup and view all the answers

Differentiate between pharmacodynamics and pharmacokinetics, highlighting their respective focuses in the study of drugs.

<p>Pharmacodynamics focuses on the effects of drugs on the body, including their mechanisms of action. Pharmacokinetics focuses on how the body processes drugs, including absorption, distribution, metabolism, and excretion.</p> Signup and view all the answers

Explain how volatile general anesthetics induce a reversible change in the synaptic junction, acting independently of receptors.

<p>Volatile general anesthetics, acting independently of receptors, cause a reversible change in the synaptic junction by altering the lipid environment of nerve cell membranes, thus affecting ion channel function and neurotransmitter release.</p> Signup and view all the answers

Describe the process by which drugs that act as chelating agents work, and provide a specific example of their application.

<p>Chelating drugs work by binding to metallic ions, forming a complex that can be excreted from the body. This is used in treating heavy metal poisoning, such as lead or iron overload.</p> Signup and view all the answers

How do competitive antagonists affect the dose-response curve of an agonist, and why can their effect be overcome by increasing the concentration of the agonist?

<p>Competitive antagonists cause a parallel rightward shift in the dose-response curve of an agonist. Their effect can be overcome by increasing the concentration of the agonist because both compete for the same binding site on the receptor; a higher concentration of agonist displaces the antagonist.</p> Signup and view all the answers

Explain the mechanism by which non-competitive antagonists reduce the effect of an agonist, outlining why their effect cannot be reversed by increasing the concentration of the agonist.

<p>Non-competitive antagonists bind to the receptor in an irreversible or pseudo-irreversible manner, effectively reducing the number of receptors available to the agonist. Increasing the agonist concentration cannot reverse this effect because the antagonist does not compete for the same binding site or alters the receptor in a way that the agonist can no longer bind effectively.</p> Signup and view all the answers

Describe efficacy in the context of graded dose-response curves, differentiating it from potency.

<p>Efficacy refers to the maximum response an agonist can produce, representing the drug's ability to activate the receptor and elicit a maximal biological effect. Unlike potency, which is a measure of the drug concentration required to produce a given effect, efficacy reflects the magnitude of the response once bound.</p> Signup and view all the answers

Illustrate the relevance of understanding drug-receptor interactions in pharmacology, providing an brief example of how manipulating these interactions can lead to therapeutic benefits.

<p>Understanding drug-receptor interactions is crucial for designing drugs that selectively target specific receptors to produce desired therapeutic effects while minimizing unwanted side effects. For example, selective serotonin reuptake inhibitors (SSRIs) target serotonin transporters to increase serotonin levels in the brain and treat depression.</p> Signup and view all the answers

Describe the effect of a noncompetitive antagonist on the Emax (maximum effect) of an agonist and explain why this change occurs.

<p>If a noncompetitive antagonist is present, the Emax of the agonist will decrease. This is due to the fact that noncompetitive antagonists bind irreversibly (or nearly irreversibly) to the receptor, reducing the total number of receptors available for the agonist to bind to, irrespective of the agonist's concentration.</p> Signup and view all the answers

How does the EC50 value relate to a drug's potency, and what does a lower EC50 indicate about the drug?

<p>The EC50 value is the concentration of a drug that produces 50% of the maximal possible effect. A lower EC50 indicates that a smaller amount of the drug is needed to produce half of the maximal effect, meaning the drug has higher potency.</p> Signup and view all the answers

Explain why drugs that act independently of receptors are still important in pharmacology, giving examples of therapeutic effects they can produce.

<p>Drugs that act independently of receptors are important because they can produce therapeutic effects through various mechanisms, such as neutralizing stomach acid (antacids), binding metallic ions (chelating drugs), or altering osmotic pressure (osmotic diuretics and cathartics).</p> Signup and view all the answers

A patient is taking phenytoin for epilepsy. Knowing that phenytoin is metabolized by the liver, what other medication on the list could potentially alter phenytoin's blood concentration and how?

<p>Cimetidine could increase phenytoin blood concentrations because it inhibits liver enzymes, while Rifampin or Carbamazepine could decrease blood concentrations because they induce liver enzymes.</p> Signup and view all the answers

A drug is primarily eliminated through glomerular filtration. How would significant protein binding of this drug affect its rate of elimination and why?

<p>Significant protein binding would decrease its rate of elimination. Glomerular filtration is a passive process, and drugs bound to protein are not readily filtered.</p> Signup and view all the answers

A patient with a history of TB is prescribed rifampin. What adjustment to anesthetic dosage might be necessary and why?

<p>The anesthetic dosage might need to be increased. Rifampin induces liver enzymes, potentially increasing the metabolism and reducing the effectiveness of some anesthetics.</p> Signup and view all the answers

A patient is taking a drug that follows zero-order kinetics. If the drug's elimination rate is 10mg/hour, how much of the drug will be eliminated in 3 hours if the concentration remains high enough to saturate the elimination process?

<p>30mg</p> Signup and view all the answers

Why should clinicians exercise caution when prescribing medications to lactating mothers?

<p>Many drugs can be excreted in breast milk, potentially leading to neonatal toxicity.</p> Signup and view all the answers

A medication is known to be actively secreted in the proximal convoluted tubules of the kidneys. How would the co-administration of another drug that competes for the same active transport system affect the excretion of the first medication?

<p>It would likely decrease the excretion of the first medication. Competition for the active transport system would reduce the amount of the first drug being secreted into the tubules.</p> Signup and view all the answers

How could you adjust urine pH to enhance excretion of a toxic charged particle from the body and why would this adjustment work?

<p>If it is an acid, alkalinizing the urine would enhance excretion. If it is a base, acidifying the urine would enhance its excretion. Manipulating pH can trap molecules in the urine.</p> Signup and view all the answers

A patient presents with impaired kidney function. How would you expect this to affect the half-life of a drug that is primarily excreted renally and why?

<p>The half-life would likely increase. Impaired kidney function reduces the rate of drug excretion, causing the drug to remain in the body longer.</p> Signup and view all the answers

Explain how the rate of drug elimination differs between zero-order and first-order kinetics.

<p>In zero-order kinetics, the drug decreases at a constant rate, regardless of plasma drug concentration. In first-order kinetics, the drug elimination rate is proportional to the plasma drug concentration.</p> Signup and view all the answers

Define physiologic half-life (t½) in the context of drug elimination.

<p>Physiologic half-life (t½) is the time required for 50% of a drug to be eliminated from the body.</p> Signup and view all the answers

Describe the relationship between the rate of drug administration and the rate of drug elimination in the context of drug accumulation, assuming first-order kinetics.

<p>If the rate of drug administration exceeds the rate of elimination, the drug will accumulate in the body. Accumulation stops when the rate of elimination equals the rate of administration, reaching a steady state.</p> Signup and view all the answers

Explain how a drug's half-life influences the determination of appropriate dosing intervals.

<p>Drugs with shorter half-lives require more frequent dosing to maintain therapeutic levels, while drugs with longer half-lives require less frequent dosing.</p> Signup and view all the answers

What is meant by 'drug clearance', and what units are typically associated with it?

<p>Drug clearance refers to the volume of blood that is completely cleared of a drug per unit of time. It's mathematically equivalent to the rate of elimination divided by the drug's plasma concentration.</p> Signup and view all the answers

Explain why giving twice the dose of a drug with a short half-life does not necessarily double its duration of action.

<p>Due to its rapid elimination, a higher initial concentration diminishes quickly. The duration of action is more influenced by the half-life and the rate of elimination than by a single, larger dose.</p> Signup and view all the answers

Describe the purpose of a loading dose followed by maintenance doses for drugs with long half-lives.

<p>A loading dose rapidly achieves the desired therapeutic concentration, while maintenance doses sustain this level by replacing the amount of drug eliminated over time.</p> Signup and view all the answers

Differentiate between the rate constant of elimination (ke) and the physiologic half-life (t½) for a drug eliminated via first-order kinetics.

<p>The rate constant of elimination (ke) represents the percent change in drug concentration per unit time, while the physiologic half-life (t½) is the time required for 50% of the drug to be eliminated. They are inversely related; a higher ke results in a shorter t½.</p> Signup and view all the answers

Explain how administering probenecid affects the duration of action of penicillin G and the pharmacokinetic principle behind this interaction.

<p>Probenecid prolongs the action of penicillin G by blocking its secretion, leading to slower excretion and increased duration of action. This relies on manipulating excretion to alter duration.</p> Signup and view all the answers

Describe the relationship between creatinine clearance and drug elimination in patients with renal insufficiency. How do adjustments in drug dosage or dosing intervals account for this relationship?

<p>Creatinine clearance directly correlates with drug elimination by the kidneys. In renal insufficiency, the initial loading dose remains the same, but maintenance doses are decreased, or dosing intervals are increased to prevent drug accumulation and toxicity.</p> Signup and view all the answers

Explain why a loading dose is sometimes necessary to achieve therapeutic drug levels rapidly. Give an example from the text.

<p>A loading dose rapidly achieves therapeutic levels by bypassing the 4-5 half-lives needed for standard dosing to reach effective concentrations. Amoxicillin, where a higher initial dose is used in significant infections, exemplifies this.</p> Signup and view all the answers

How does the concept of drug half-life relate to drug accumulation in the body, and what dosing adjustments are necessary to prevent accumulation?

<p>If the dosing interval is shorter than approximately four half-lives, the drug will accumulate in the body. To prevent this, the dosing interval must be extended, or the dosage reduced, to allow for complete elimination between doses.</p> Signup and view all the answers

Describe how inhibiting the metabolism of one drug with another can be clinically useful. Provide an example described in the text.

<p>Inhibiting the metabolism of a drug can prolong its action or increase its effectiveness. For example, allopurinol is used to block the metabolism of 6-mercaptopurine.</p> Signup and view all the answers

Flashcards

Pharmacology

The study of drugs, including their origins, composition, effects, and uses.

Pharmacodynamics

How drugs affect the body; identifies drug action sites and modes.

Pharmacokinetics

How the body affects the drug; includes absorption, distribution, metabolism, and excretion.

Antacids

Substances that neutralize stomach acids.

Signup and view all the flashcards

Chelating drugs

Drugs that bind to metallic ions.

Signup and view all the flashcards

Receptor

Drug target molecule on cell to initiate a biochemical cascade.

Signup and view all the flashcards

Agonist

Binds to receptors & stimulates them.

Signup and view all the flashcards

Antagonist

Binds to receptors & blocks the effect of an agonist.

Signup and view all the flashcards

Competitive antagonist

Binds reversibly, can be overcome by high agonist concentration.

Signup and view all the flashcards

Non-competitive antagonist

Binds irreversibly, cannot be overcome by high agonist concentration.

Signup and view all the flashcards

Dose & Route Effect

Dosage and administration route influence drug concentration.

Signup and view all the flashcards

Permeability

Drug's ability to cross cell membranes, crucial for absorption.

Signup and view all the flashcards

Lipid Solubility

Enhanced passage across cell walls for weak acids and bases.

Signup and view all the flashcards

Aqueous solubility

Charged, water-soluble molecules face difficulty crossing epithelial linings without being very small.

Signup and view all the flashcards

Bioavailability

Fraction of drug reaching systemic circulation (100% for IV).

Signup and view all the flashcards

First-pass metabolism

Drug passes from GIT -> Liver -> reduced bioavailability.

Signup and view all the flashcards

Oral drug administration

Common route, but drug needs lipid solubility and GI resistance.

Signup and view all the flashcards

Drug Interaction Source

The liver affects drug interactions through metabolism induction or inhibition.

Signup and view all the flashcards

Inducers

Increase the rate of drug metabolism.

Signup and view all the flashcards

Inhibitors

Decrease the rate of drug metabolism.

Signup and view all the flashcards

Kidney in Excretion

Primary site of drug removal from the body.

Signup and view all the flashcards

Glomerular Filtration

Passive, non-saturable kidney process.

Signup and view all the flashcards

Tubular Secretion

Active and saturable process in kidney tubules.

Signup and view all the flashcards

Drugs Excreted via Lungs

Gaseous anesthetics, paraldehyde, ETOH, and garlic.

Signup and view all the flashcards

Zero-Order Kinetics

Constant amount of drug eliminated per unit time.

Signup and view all the flashcards

Bronchodilators

Drugs that relax bronchial smooth muscle, opening airways.

Signup and view all the flashcards

Plasma Protein Binding

The process where drugs bind to proteins in the blood, mainly albumin.

Signup and view all the flashcards

Plasma Protein Binding determinants

Determined by the amount of tissue protein, the drug's binding constant, and competition from other drugs.

Signup and view all the flashcards

Volume of Distribution (Vd)

A theoretical volume representing how much space a drug occupies in the body.

Signup and view all the flashcards

Lipid-soluble drugs Vd

Drugs that easily cross cell membranes and distribute widely.

Signup and view all the flashcards

Unequal Drug Distribution

Drug molecules bind to mucopolysaccharides, nucleoproteins, phospholipids and accumulates in body fat, reducing drug concentration.

Signup and view all the flashcards

Blood-Brain Barrier (BBB)

A selective barrier protecting the brain, favoring lipid-soluble, non-ionized drugs..

Signup and view all the flashcards

Drug Elimination

Drugs are either converted to inactive metabolites or excreted unchanged, eliminating its intended use.

Signup and view all the flashcards

Drug Accumulation

Accumulation happens when drug dosage intervals are shorter than the time needed for complete elimination (around 4 half-lives).

Signup and view all the flashcards

Prolongation of Drug Action

Techniques or methods used to extend how long a drug remains active in the body.

Signup and view all the flashcards

Loading Dose

An initial, larger dose of a drug given to quickly achieve therapeutic levels in the body.

Signup and view all the flashcards

First-Order Kinetics

Drug elimination where a constant FRACTION is removed per unit time; rate depends on drug concentration.

Signup and view all the flashcards

Renal Insufficiency & Drug Dose

Reduced kidney function impacts drug elimination, requiring dose adjustments.

Signup and view all the flashcards

Hepatic Insufficiency & Drug Dose

Reduced liver functions requires monitoring serum levels

Signup and view all the flashcards

Physiologic Half-Life (t1/2)

Time required for 50% of a drug to be eliminated from the body.

Signup and view all the flashcards

Rate Constant of Elimination (ke)

Percent change in drug concentration eliminated per unit time.

Signup and view all the flashcards

Drug Clearance

Volume of blood completely cleared of a drug per unit time.

Signup and view all the flashcards

Steady State

State where the rate of drug elimination equals the rate of drug administration.

Signup and view all the flashcards

Study Notes

  • Pharmacology involves the study of drugs, including their origins, composition, pharmacokinetics, therapeutic uses, and toxicology.

General Principles of Pharmacology

  • Pharmacodynamics characterizes the action of drugs on the body's biochemical and physiologic systems, identifying sites and modes of action.
  • Pharmacokinetics describes the quantitative aspects of drug absorption, distribution, metabolism, and excretion, detailing the time course of drug and metabolite concentration at their site of action.

Drugs Acting Independently of Receptors

  • Antacids neutralize stomach acids.
  • Chelating drugs bind metallic ions.
  • Osmotically active drugs include diuretics like mannitol, and cathartics like methylcellulose.
  • Volatile general anesthetics cause reversible changes in the synaptic junction, with potency related to lipid solubility.

Drug Receptor Interactions

  • Drugs combine with specific target molecules on cells to initiate a biochemical cascade, leading to an effect.
  • Receptors may be proteins, carbohydrates, nucleic acids, or lipids.
  • Binding may involve ionic, covalent, hydrogen, or van der Waals bonds.

Drug Receptor Interactions (cont.)

  • Agonists bind to a receptor and stimulate it.
  • Antagonists bind to a receptor and decrease or block the effect of an agonist.
  • Competitive antagonists bind to the receptor and prevent agonist binding, can be overcome by high agonist concentrations, and produce a parallel right shift on the dose-response curve.
  • Non-competitive antagonists bind to the receptor in an irreversible way, prevent any agonist action, cannot be reversed high [agonist], and decrease height of dose-response curve.

Summary of Antagonists

  • Competitive Antagonists bind reversibly to an agonist and it can be overcome by a large amount of agonist
  • Non-competitive Antagonists bind irreversibly and cannot be overcome by an agonist

Graded Dose-Response Curves

  • The response of a system to increasing doses of a drug (agonist) can be analyzed.
  • Agonists are drugs that bind to a receptor which results in stimulation.
  • The effect is analyzed by plotting the response versus the log of drug concentration (dose).
  • Efficacy is the maximum response by an agonist, which escalates as effect heightens (seen along the y-axis).

Graded Dose-Response Curves (cont.)

  • Potency measures how much drug is needed to produce a given effect.
  • Potency is expressed as the concentration that can give a 50% response (EC50).
  • Less drug needed to produce an effect indicates a more potent drug.
  • Potency increases as the curve shifts left on the x-axis.

Quantal Dose-Response Curves

  • Demonstrates the minimum drug dose needed to produce a pre-determined response in a polulation.
  • A percentage of the population is plotted against the Log (dose).
  • ED50 (median effective dose) is the dose that will produce an effect in 50% of the population.
  • TD50 is the minimum dose that produces a specific toxic effect in 50% of the population.
  • LD50 is the minimum dose that will kill 50% of the population.

Quantal Dose-Response Curves (cont.)

  • The therapeutic index (TI) is the ratio of the dose required to produce a toxic or lethal effect to the therapeutically effective dose.
  • A TI greater than 4 is considered good.
  • TI = TD50/ED50 or LD50/ED50.

Summary of Dose-Response Curves

  • Drugs with high efficacy but low potency reach a high level of response with a greater dose.
  • A low therapeutic index (TI) indicates a high incidence of side effects at usual doses.

Summary of High TI values

  • A higher the therapeutic index is favorable and indicates a low incidence of side effects at usual doses.
  • A high Therapeutic Index signifies a safer drug.
  • Drug companies aim for a ratio of at least 4.
  • Anything less than 2 requires close patient monitoring, such as with lithium for manic disorders.

Pharmacokinetics

  • Factors that effect drug in the site of action include routes of administration. circulatroy systems, other copartments, distribution, metabolism and excretion.

Drug Absorption

  • Permeability affects drug absorption because of a drugs ability to cross the cell wall, lipid solubility, aqueous solubility.
  • Correlates to ability to cross cell wall
  • Weak acids & bases more lipid soluble
  • Charged (ions), water soluble molecules are excluded from crossing epithelial lining of skin & GIT (unless very small)

Factors affecting absorption (cont.)

  • Bioavailability is the fraction of a drug that reaches systemic circulation.
  • A bioavailability of "1" means 100% and indicates the drug was given intraveneously.
  • <1 bioavailability, indicates permeability
  • Oral administration could result in reduced bioavailability due to incomplete absorpition or the first-pass metabolism in the liver.

Routes of Administration

  • An oral route of administration is most commun, safe, economical, and convenient
  • Drugs must be lipid soluble and resistant to GI acid, GI digestive enzymes, and GI bacterial flora.
  • Rate and degree of absorbtion can vary.

Routes of Administration (cont)

  • Sublingual (buccal) administration enters venous drainage, leading to systemic circulation and bypassing the liver.
  • It's good for self-administration and rapid onset, like nitroglycerin for angina pectoris.
  • Suited for metabolized drugs in the liver.

Routes of Administration (cont.)

  • The rectal route has less of a first-pass effect compared to oral routes.
  • Rectal administration is useful in cases of vomiting or unconsciousness.
  • Absorption is irregular.
  • The intravenous (IV) route allows for rapid and complete delivery to target tissues.
  • Useful in emergencies and for drugs highly metabolized by the liver or poorly absorbed from the GI tract.
  • allows precise titration of dose levels.

Routes of Administration (cont.)

  • Intramuscular (IM) administration is contraindicated for patients on anticoagulants.
  • Aqueous solutions are absorbed rapidly and oil solutions are absorbed slowly.
  • With Subcutaneous (SC) administration, small volumes are used and drugs are slowly absorbed

Routes of Administration (cont.)

  • Topical administration is utilized on the skin, vagina, eyes, ear, nose, and throat.
  • Transdermal administration is effective for the systemic delivery of drugs to the skin.
  • Absorption is slow, such as with nicotine or nitroglycerin patches. Greater absorption occurs with topical lidocaine

Routes of Administration (cont)

  • Intrathecal (IT) administration delivers the drug into subarachnoid space (lumbar puncture) or ventricular system (Ommaya reservoir).
  • This route bypasses the blood-brain barrier and blood-CSF barrier.
  • Useful for drugs with slow CNS penetration or rapid high CSF concentrations such as with meningitis and spinal anesthesia.

Routes of Administration (cont.)

• Intra-arterial (IA) administration allows for delivery of high concentrations to selective sites. • Used for X-ray contrast studies such as angiograms. • Inhalation can be used to administer gaseous and volatile drugs such as anesthetics and bronchodilators for asthma.

Drug Distribution

  • Plasma protein binding is a key factor in drug distribution.
  • It is determined by amount of tissue protein (albumin), the binding constant for the drug, and the fact that binding is non-specific, so several drugs may compete for the same binding site.

Drug Distribution (cont.)

  • Volume of Distribution (Va) = Total drug in body(g) / Plasma [drug].
  • Lipid-soluble drugs exhibit a Volume of Distribution greater than total body water.
  • Drugs that bind strongly to proteins have a Volume of Distribution approaching plasma volume.
  • Greater Volume of Distribution = slower excretion rate

Drug Distribution (cont)

  • Unequal distribution of drugs occurs
  • Tissue affinity involves binding to mucopolysaccharide, nucleoprotein & phospholipid reduces availability of drug
  • Body fat acts as a reservoir for lipid-soluble drugs.
  • Blood-Brain Barrier is highly selective for lipid-soluble, non-ionized drugs.
  • High blood flow allows drugs to reach equilibrium quickly (e.g. brain).

Drug Distribution (cont.)

  • Clinical Correlation drug competition can impact plasma protein binding and explain drug-drug interactions.
  • Both fonamide & coumarin bind to proteins.
  • Administration of fonamide to patient on chronic warfarin can displace it causing dangerously high levels of free warfarin in the blood, leading to severe bleeding

Drug Elimination

  • Pharmacologic effects are terminated by transformation of drug to an inactive metabolite prior to excretion, excretion of unchanged drug, or active metabolite

Drug Elimination (cont.)

  • Metabolism and transformation mainly occurs in the liver (most important site).
  • Metabolic enzymes and hepatic microsomal enzymes are found in smooth endoplasmic reticulum (e.g. cytochrome P-450 system).
  • Other enzymes are located in the mitochondria (e.g. monoamine oxidase), cytosol (e.g. alcohol dehydrogenase), or lysosomes.

Drug Elimination (cont.)

  • Metabolism & Transformation occurs in two phases, Phase 1 & Phase 11
  • Phase I reactions mostly encompass oxidations, reductions, or hydrolysis.
  • Phase II reactions conjugate the drug or metabolite.
  • Conjugations involve adding an endogenous substance (e.g. carbohydrate or sulfate).
  • Phase two Inactivates drug or metabolite to hydrophilic form to facilitate excretion

Drug Elimination - Phase 11 (cont.)

  • Conjugation occurs with glucuronic acid, sulfate, amino acid, or acetylation.
  • Enterohepatic circulation involves:
    • conjugated drug being actively secreted in bile,
    • drugs being hydrolyzed in the small intestine,
    • most bile salts being reabsorbed in the terminal ileum,
    • drugs potentially being excreted in feces, urine, or saliva, or being reabsorbed.

Drug Elimination (cont)

  • Factors that affect hepatic metabolism is age; very young or old are impaired.
  • Genetics regulate activity of N-acetyltransferase
  • Genetics influence metabolism of procainamide (Rx arrhythmia), dapsone (Rx autoimmune disease) and isoniazid (Rx TB)

Factors that affect Hepatic Metabolism (cont.)

  • Hepatic insufficiency impairs metabolism such as with cimetidine to Rx peptic ulcer.
  • Drug interactions may competitively inhibit the metabolism of microsomal enzymes leading to an increasing metabolism
  • Reduced Hepatic Blood Flow can be caused by Congested Heart Failure (CHF) & drugs that reduce cardiac output (e.g. propanolol)

Metabolic Summary

  • Drug interactions is an important interaction that is metabolized in the liver.

  • There are inducers and inhibitors that affect metabolism.

  • Inducers include Barbiturates, Phenyton (Rx epilepsy), Rifampin (Rx TB), and Carbamazepine

  • Inhibitors include Cimetidine, Ketoconazole and Isoniazid

Drug Excretion

  • The kidney is the primary site of the excretion process.
  • Glomerular filtration occurs with protein binding
  • Glomerular filtration is passive and non-saturable.
  • Tubular secretion is active and saturable.
  • Tubular secretion mainly happens in proximal convoluted tubules.
  • Passive: neutral molecules enhance the secretion of toxic charged particles

Drug Excretion (cont.)

  • Lungs excrete of gaseous anesthetics, paraldehyde (sedative & hypnotic), EtOH, and garlic

  • GI tract drugs that are secreted into the liver biliary tract and are eliminated in the feces

Drug Excretion (cont.)

  • Sweat, saliva, tears and breast milk are minimally involved in excretion.
  • Lactating mothers require medical supervision to ensure drugs that excreted in breast milk not cause neonatal toxicity

Drug Decay Curves

  • It's based on kinetic the model where the body is a single compartment
  • Describes the time course of drug in the body
  • Occurs when an elimination process is saturated
  • Zero-order kinetics eliminates a constant amaount, not a fraction of drugs over a a given time(e.g., ETOH)

Drug Decay Curves (cont.)

  • First-order kinetics are processes necessary for absorpiton or not saturable
  • First-order kinetics are concentration dependent
  • First-order kinetics shows a constant elimination per unit time
  • First-order kinetics shows that the rate drug removal is proportional to concentration in the plasma
  • First-order kinetics show a concentration of drug diminishes logarithmically with time

Drug Decay Curves (cont.)

  • The rate of elimination can be desccribed in 2 ways:
    • Physiologic half-life is the time (t1/2) required for 50% of a drug to be eliminated.
    • Rate constant of elimination (ke) = percent change per unit time

Pharmaokinetics:Drug Decay Curves

  • On a graph of plasma, a Plasma [drug] decreases to the with zero-order kinetics.
  • An initial Plasma [drug]decreases over time with first order kinetics

Summary of Drug Decay Curves

  • With Zero-order kinetics: drug decrease at a constant rate regardless of plasma drug concentration
  • With First-order kinetics: drug elimination rate is proportional to plasma drug concentration

Drug Acculmulation

  • Drug Clearance is the volume of blood that can completely cleared of a drug per unit time.

  • Repeated doses may cause drug acculmulation

  • Assuming First-Order kinetics is that if the administration is greater than the rate of elimination occurs

  • Acculmulation stops when rate of elimination equals rate of administration to reach a steady state

Clinical Implications of Dose

• Determine dose interval necessary to obtain desired level of drug •Drug with short half-life: •Giving twice the dose does not double the duration of action • Drug with long half-life: • Larger loading dose, followed by smaller maintenance doses (e.g. penicillin)

Clinical Implications : Half-Life (cont)

  • A drug accrues, therefore it takes approximately four half lives to reduce dosage.

Clinical Dose Implications (cont)

  • Prolongation of Drug Action can be achieved by
    • Frequent doses,
    • Coating tablets (time release), -Depots, -Slow excreting and inhibiting the drug metabolism.

Clinical Dose Implications

  • Loading dose is used to produce a therapeutic effect without delay of 4-5 half lives

Disease States Requiring Adjustment of Dose & Dosing Levels

  • Renal Insufficiency requires the creatinine clearance to be high to eliminate the drug by kidney. The initial dose needs to be high, then decrease the intervals of the increased dose.
  • With Hepatic Insufficiency the monitor of serum levels, or clinical signs of toxicity needs to be performed.

Studying That Suits You

Use AI to generate personalized quizzes and flashcards to suit your learning preferences.

Quiz Team

Related Documents

Pharmacology Lecture Notes PDF

Description

Explores drug absorption in the body, focusing on lipid solubility, aqueous solubility challenges, and bioavailability. Details intravenous administration benefits and oral route variability. Highlights first-pass metabolism effects and permeability factors.

More Like This

Pharmacokinetics: Oral Drug Absorption
109 questions
Pharmacology: Bioavailability and Drug Absorption
50 questions
Drug Absorption and Bioavailability
55 questions
Use Quizgecko on...
Browser
Browser