Podcast
Questions and Answers
Given the principles of ion trapping and aspirin's pharmacokinetics ($pK_a = 3.0$), which of the following scenarios would least favor the systemic reabsorption of aspirin following oral administration?
Given the principles of ion trapping and aspirin's pharmacokinetics ($pK_a = 3.0$), which of the following scenarios would least favor the systemic reabsorption of aspirin following oral administration?
- Entry into renal tubular cells, followed by a shift into more alkaline (pH 7.4) environment.
- Passage through the gastric mucosa into the bloodstream where the pH is maintained at 7.4. (correct)
- Co-administration with an antacid that elevates gastric pH to 4.5, followed by passage into the jejunum.
- Excretion into acidic urine (pH 5.0) within the proximal tubules of the kidney.
A patient with chronic salicylate toxicity presents with metabolic acidosis. Considering the pH-dependent distribution of aspirin, what compensatory mechanism would paradoxically exacerbate aspirin's intracellular accumulation within renal tubular cells?
A patient with chronic salicylate toxicity presents with metabolic acidosis. Considering the pH-dependent distribution of aspirin, what compensatory mechanism would paradoxically exacerbate aspirin's intracellular accumulation within renal tubular cells?
- Increased renal clearance of bicarbonate ions, leading to further acidification of the urine.
- Administration of carbonic anhydrase inhibitors to promote bicarbonate excretion.
- Intravenous administration of sodium bicarbonate to raise systemic pH. (correct)
- Hyperventilation, causing a respiratory alkalosis and a transient increase in blood pH.
Following an aspirin overdose, forced alkaline diuresis is initiated to enhance drug elimination. Which statement elucidates the principal mechanism by which alkalinization of the urine accelerates aspirin excretion?
Following an aspirin overdose, forced alkaline diuresis is initiated to enhance drug elimination. Which statement elucidates the principal mechanism by which alkalinization of the urine accelerates aspirin excretion?
- Alkalinization of the urine increases the concentration of $H^+$ ions, which bind to aspirin, making it more water-soluble and easily excreted.
- Elevated urinary pH enhances the ionization of aspirin, trapping it within the renal tubules and preventing reabsorption. (correct)
- Alkaline urine promotes the active transport of aspirin out of the renal tubular cells into the urine.
- Increased urinary pH reduces the ionization of aspirin, facilitating its filtration at the glomerulus.
A research study is evaluating novel methods to mitigate aspirin-induced nephrotoxicity. Which of the following strategies, based on the principles of drug ionization and distribution, would most likely offer the greatest protective effect?
A research study is evaluating novel methods to mitigate aspirin-induced nephrotoxicity. Which of the following strategies, based on the principles of drug ionization and distribution, would most likely offer the greatest protective effect?
Considering the complexities of aspirin metabolism and pH-dependent distribution, in a scenario involving both gastric and renal handling of aspirin, which statement accurately correlates altered physiological conditions with aspirin's pharmacokinetic behavior?
Considering the complexities of aspirin metabolism and pH-dependent distribution, in a scenario involving both gastric and renal handling of aspirin, which statement accurately correlates altered physiological conditions with aspirin's pharmacokinetic behavior?
Given a compound with a volume of distribution ($V_d$) of 20 L and an elimination rate constant ($K_{el}$) of 0.15 $h^{-1}$, what adjustment to the infusion rate is required to maintain the same average steady-state plasma concentration if the bioavailability is reduced by 50%?
Given a compound with a volume of distribution ($V_d$) of 20 L and an elimination rate constant ($K_{el}$) of 0.15 $h^{-1}$, what adjustment to the infusion rate is required to maintain the same average steady-state plasma concentration if the bioavailability is reduced by 50%?
A novel drug exhibits non-linear pharmacokinetics, adequately described by the Michaelis-Menten equation. If the $V_{max}$ is 100 mg/hr and $K_m$ is 25 mg/L, what is the approximate rate of elimination when the drug's plasma concentration is 50 mg/L, assuming first-order kinetics do not apply?
A novel drug exhibits non-linear pharmacokinetics, adequately described by the Michaelis-Menten equation. If the $V_{max}$ is 100 mg/hr and $K_m$ is 25 mg/L, what is the approximate rate of elimination when the drug's plasma concentration is 50 mg/L, assuming first-order kinetics do not apply?
A patient with end-stage renal disease requires a loading dose of Drug X to achieve a target plasma concentration of 2 mg/L. Given that the $V_d$ of Drug X is 0.7 L/kg and the patient weighs 70 kg, and considering that renal impairment reduces drug clearance by 60%, yet does not affect the initial volume of distribution, what loading dose should be administered?
A patient with end-stage renal disease requires a loading dose of Drug X to achieve a target plasma concentration of 2 mg/L. Given that the $V_d$ of Drug X is 0.7 L/kg and the patient weighs 70 kg, and considering that renal impairment reduces drug clearance by 60%, yet does not affect the initial volume of distribution, what loading dose should be administered?
A drug is administered intravenously, and its plasma concentration is measured at two time points: 2 hours (C1 = 15 mg/L) and 6 hours (C2 = 7.5 mg/L) post-administration. Assuming first-order elimination, what is the estimated time it will take for the plasma concentration to reach 1.875 mg/L?
A drug is administered intravenously, and its plasma concentration is measured at two time points: 2 hours (C1 = 15 mg/L) and 6 hours (C2 = 7.5 mg/L) post-administration. Assuming first-order elimination, what is the estimated time it will take for the plasma concentration to reach 1.875 mg/L?
A drug undergoes both renal elimination and hepatic metabolism. The renal clearance is 3 L/hr, and the hepatic clearance is 5 L/hr. If the drug's volume of distribution is 80 L, what is the drug's elimination half-life?
A drug undergoes both renal elimination and hepatic metabolism. The renal clearance is 3 L/hr, and the hepatic clearance is 5 L/hr. If the drug's volume of distribution is 80 L, what is the drug's elimination half-life?
Given the established mechanisms of action for lincosamides, which of the following scenarios would most likely attenuate their antibacterial efficacy in a clinically relevant context?
Given the established mechanisms of action for lincosamides, which of the following scenarios would most likely attenuate their antibacterial efficacy in a clinically relevant context?
In the context of chronic polymicrobial infections involving both aerobic and anaerobic bacteria, what is the most critical consideration regarding lincosamide usage, assuming resistance profiles are unknown?
In the context of chronic polymicrobial infections involving both aerobic and anaerobic bacteria, what is the most critical consideration regarding lincosamide usage, assuming resistance profiles are unknown?
Suppose a patient presents with a severe skin and soft tissue infection (SSTI) caused by Staphylococcus aureus. Prior to initiating lincosamide therapy, which preemptive pharmacogenomic test would offer the most direct insight into potential therapeutic failure?
Suppose a patient presents with a severe skin and soft tissue infection (SSTI) caused by Staphylococcus aureus. Prior to initiating lincosamide therapy, which preemptive pharmacogenomic test would offer the most direct insight into potential therapeutic failure?
A research team is investigating novel lincosamide derivatives with enhanced antibacterial activity. Which molecular modification strategy would MOST likely overcome existing resistance mechanisms and broaden the spectrum of activity?
A research team is investigating novel lincosamide derivatives with enhanced antibacterial activity. Which molecular modification strategy would MOST likely overcome existing resistance mechanisms and broaden the spectrum of activity?
Considering the pharmacokinetic and pharmacodynamic properties of lincosamides, what is the MOST probable explanation for their limited efficacy in treating central nervous system infections, despite demonstrating in vitro activity against causative pathogens?
Considering the pharmacokinetic and pharmacodynamic properties of lincosamides, what is the MOST probable explanation for their limited efficacy in treating central nervous system infections, despite demonstrating in vitro activity against causative pathogens?
A novel drug demonstrates high lipophilicity and undergoes Phase I metabolism, resulting in a slightly more polar metabolite that still exhibits poor water solubility. Which of the following strategies would MOST effectively facilitate renal elimination of this metabolite, considering both metabolic and physicochemical properties?
A novel drug demonstrates high lipophilicity and undergoes Phase I metabolism, resulting in a slightly more polar metabolite that still exhibits poor water solubility. Which of the following strategies would MOST effectively facilitate renal elimination of this metabolite, considering both metabolic and physicochemical properties?
A drug, primarily eliminated through hepatic metabolism, exhibits significant inter-individual variability in its clearance. Genetic analysis reveals polymorphisms in both CYP450 enzymes (affecting Phase I metabolism) and UDP-glucuronosyltransferases (UGTs) (affecting Phase II metabolism). If a patient is found to be a poor metabolizer for a specific CYP450 enzyme but an ultra-rapid metabolizer for a UGT enzyme relevant to the drug's metabolism, how would this most likely affect the drug's overall elimination rate and half-life?
A drug, primarily eliminated through hepatic metabolism, exhibits significant inter-individual variability in its clearance. Genetic analysis reveals polymorphisms in both CYP450 enzymes (affecting Phase I metabolism) and UDP-glucuronosyltransferases (UGTs) (affecting Phase II metabolism). If a patient is found to be a poor metabolizer for a specific CYP450 enzyme but an ultra-rapid metabolizer for a UGT enzyme relevant to the drug's metabolism, how would this most likely affect the drug's overall elimination rate and half-life?
A pharmaceutical company is developing a novel prodrug that requires activation via hydrolysis followed by oxidation to exert its therapeutic effect. During preclinical studies, it's observed that species A exhibits significantly higher in vivo efficacy compared to species B. Given the metabolic pathways involved, which of the following enzyme activity differences between the two species would MOST likely explain this discrepancy?
A pharmaceutical company is developing a novel prodrug that requires activation via hydrolysis followed by oxidation to exert its therapeutic effect. During preclinical studies, it's observed that species A exhibits significantly higher in vivo efficacy compared to species B. Given the metabolic pathways involved, which of the following enzyme activity differences between the two species would MOST likely explain this discrepancy?
A research team is investigating a new drug that undergoes both oxidation and glucuronidation. They discover that a specific genetic polymorphism results in significantly reduced activity of the enzyme responsible for the oxidation step. Simultaneously, this polymorphism leads to a compensatory upregulation of the glucuronidation pathway. Assuming both pathways contribute equally to drug clearance in individuals without the polymorphism, what is the MOST likely effect of this genetic variation on the drug's overall clearance?
A research team is investigating a new drug that undergoes both oxidation and glucuronidation. They discover that a specific genetic polymorphism results in significantly reduced activity of the enzyme responsible for the oxidation step. Simultaneously, this polymorphism leads to a compensatory upregulation of the glucuronidation pathway. Assuming both pathways contribute equally to drug clearance in individuals without the polymorphism, what is the MOST likely effect of this genetic variation on the drug's overall clearance?
A patient with impaired renal function is prescribed a medication that is primarily eliminated through hepatic metabolism involving both Phase I (oxidation) and Phase II (glucuronidation) reactions. However, the resulting glucuronide metabolite is typically excreted renally. How would the patient's impaired renal function MOST likely affect the overall drug clearance, and what compensatory mechanism might occur?
A patient with impaired renal function is prescribed a medication that is primarily eliminated through hepatic metabolism involving both Phase I (oxidation) and Phase II (glucuronidation) reactions. However, the resulting glucuronide metabolite is typically excreted renally. How would the patient's impaired renal function MOST likely affect the overall drug clearance, and what compensatory mechanism might occur?
A novel drug, 'Hepatoreductase Inhibitor X,' undergoes extensive first-pass metabolism, rendering oral administration therapeutically ineffective. Which alternative route of administration would MOST likely circumvent this limitation, while also ensuring rapid systemic availability and minimizing hepatic exposure?
A novel drug, 'Hepatoreductase Inhibitor X,' undergoes extensive first-pass metabolism, rendering oral administration therapeutically ineffective. Which alternative route of administration would MOST likely circumvent this limitation, while also ensuring rapid systemic availability and minimizing hepatic exposure?
A research team is developing a new therapeutic agent, 'CardioProtect,' specifically designed to target myocardial ischemia. Initial studies reveal that CardioProtect undergoes nearly complete first-pass metabolism when administered orally. To optimize its efficacy in acute settings, which alternative route should be prioritized, considering both the speed of onset and the avoidance of hepatic degradation?
A research team is developing a new therapeutic agent, 'CardioProtect,' specifically designed to target myocardial ischemia. Initial studies reveal that CardioProtect undergoes nearly complete first-pass metabolism when administered orally. To optimize its efficacy in acute settings, which alternative route should be prioritized, considering both the speed of onset and the avoidance of hepatic degradation?
A patient presents with acute angina pectoris. Considering the need for rapid relief and the potential for significant first-pass metabolism, which formulation and route of administration of nitroglycerin would be MOST appropriate to maximize bioavailability and minimize the delay in therapeutic effect?
A patient presents with acute angina pectoris. Considering the need for rapid relief and the potential for significant first-pass metabolism, which formulation and route of administration of nitroglycerin would be MOST appropriate to maximize bioavailability and minimize the delay in therapeutic effect?
A pharmaceutical scientist is tasked with reformulating an existing oral drug, 'Metabolase-X,' which exhibits poor bioavailability due to extensive first-pass metabolism. To enhance its therapeutic efficacy without altering the drug's chemical structure, which formulation strategy and route of administration would be MOST effective?
A pharmaceutical scientist is tasked with reformulating an existing oral drug, 'Metabolase-X,' which exhibits poor bioavailability due to extensive first-pass metabolism. To enhance its therapeutic efficacy without altering the drug's chemical structure, which formulation strategy and route of administration would be MOST effective?
A novel peptide-based drug, 'PeptiMax,' is being developed for the treatment of a central nervous system disorder. In vitro studies indicate that PeptiMax is highly susceptible to enzymatic degradation in the gastrointestinal tract and undergoes significant first-pass metabolism in the liver. Which innovative delivery strategy would be MOST appropriate to maximize its bioavailability and target the CNS effectively?
A novel peptide-based drug, 'PeptiMax,' is being developed for the treatment of a central nervous system disorder. In vitro studies indicate that PeptiMax is highly susceptible to enzymatic degradation in the gastrointestinal tract and undergoes significant first-pass metabolism in the liver. Which innovative delivery strategy would be MOST appropriate to maximize its bioavailability and target the CNS effectively?
Which of the following scenarios most accurately exemplifies the mechanism of pinocytosis in the context of cellular drug delivery?
Which of the following scenarios most accurately exemplifies the mechanism of pinocytosis in the context of cellular drug delivery?
In the context of targeted drug delivery exploiting pinocytosis, what is the most critical biophysical parameter to optimize for effective drug internalization?
In the context of targeted drug delivery exploiting pinocytosis, what is the most critical biophysical parameter to optimize for effective drug internalization?
A researcher is investigating the efficacy of a novel anticancer drug that utilizes pinocytosis for cellular entry. After conducting in vitro studies, it is observed that the drug is readily taken up by cancer cells but also exhibits significant uptake by healthy macrophages. Which of the following strategies would be most effective in mitigating off-target drug accumulation in macrophages while preserving its efficacy in cancer cells?
A researcher is investigating the efficacy of a novel anticancer drug that utilizes pinocytosis for cellular entry. After conducting in vitro studies, it is observed that the drug is readily taken up by cancer cells but also exhibits significant uptake by healthy macrophages. Which of the following strategies would be most effective in mitigating off-target drug accumulation in macrophages while preserving its efficacy in cancer cells?
Consider a scenario where a new therapeutic protein is designed to be delivered intracellularly via pinocytosis. However, upon internalization, the protein is rapidly degraded within lysosomes. Which modification strategy would most likely enhance the protein's intracellular stability and functional efficacy?
Consider a scenario where a new therapeutic protein is designed to be delivered intracellularly via pinocytosis. However, upon internalization, the protein is rapidly degraded within lysosomes. Which modification strategy would most likely enhance the protein's intracellular stability and functional efficacy?
In the effort to enhance drug delivery via pinocytosis, a research team explores employing a cell-penetrating peptide (CPP) to facilitate membrane interaction. Which parameter regarding the CPPs is most critical to optimize for maximized pinocytotic uptake without inducing extensive cellular toxicity?
In the effort to enhance drug delivery via pinocytosis, a research team explores employing a cell-penetrating peptide (CPP) to facilitate membrane interaction. Which parameter regarding the CPPs is most critical to optimize for maximized pinocytotic uptake without inducing extensive cellular toxicity?
Flashcards
Pinocytosis
Pinocytosis
Cell membrane folds inward, capturing a drug molecule within a vesicle and releasing it inside the cell.
Invagination
Invagination
A method where the cell membrane surrounds and engulfs a substance, bringing it inside without needing a receptor.
Trapping Drug Molecules
Trapping Drug Molecules
The act of enclosing or trapping a molecule within a vesicle.
Vesicle
Vesicle
Signup and view all the flashcards
Release Inside the Cell
Release Inside the Cell
Signup and view all the flashcards
Total Drug Amount
Total Drug Amount
Signup and view all the flashcards
Drug Clearance Formula
Drug Clearance Formula
Signup and view all the flashcards
Loading Dose Calculation
Loading Dose Calculation
Signup and view all the flashcards
Volume of Distribution (Vd)
Volume of Distribution (Vd)
Signup and view all the flashcards
Elimination Rate Constant (Kel)
Elimination Rate Constant (Kel)
Signup and view all the flashcards
Aspirin Metabolism in Blood
Aspirin Metabolism in Blood
Signup and view all the flashcards
Aspirin Ionization in Blood
Aspirin Ionization in Blood
Signup and view all the flashcards
Aspirin in Acidic Urine
Aspirin in Acidic Urine
Signup and view all the flashcards
Aspirin Reabsorption
Aspirin Reabsorption
Signup and view all the flashcards
Aspirin and Renal Damage
Aspirin and Renal Damage
Signup and view all the flashcards
Phase I Reactions
Phase I Reactions
Signup and view all the flashcards
Phase II Reactions
Phase II Reactions
Signup and view all the flashcards
Water Solubility Effect
Water Solubility Effect
Signup and view all the flashcards
Renal Re-absorption
Renal Re-absorption
Signup and view all the flashcards
Conjugation
Conjugation
Signup and view all the flashcards
Lincosamides
Lincosamides
Signup and view all the flashcards
Smoking & Alcohol
Smoking & Alcohol
Signup and view all the flashcards
Isoniazid
Isoniazid
Signup and view all the flashcards
Azoles
Azoles
Signup and view all the flashcards
What is "other routes"?
What is "other routes"?
Signup and view all the flashcards
What is first-pass effect?
What is first-pass effect?
Signup and view all the flashcards
Give examples of "other routes"
Give examples of "other routes"
Signup and view all the flashcards
What is sublingual nitroglycerin?
What is sublingual nitroglycerin?
Signup and view all the flashcards
What is "drug effect"?
What is "drug effect"?
Signup and view all the flashcards
Study Notes
Pharmacokinetics
- It is the journey of a drug inside the body and its effect.
- Includes absorption, distribution, metabolism, and excretion.
- It discusses how the body affects the drug
Absorption
- It is the passage of a drug from the site of administration to the plasma
- Routes of administration include oral, rectal, sublingual, local, inhalation, and injections.
Mechanisms of drug absorption
Simple Diffusion
- Drugs move according to the concentration gradient from high to low concentration.
- It is a Passive process, as it requires no energy.
- The drug must be lipophilic to pass through the lipid membrane.
Carrier-Mediated Transport
- Achieved by specialized carrier molecules.
- Can be passive, moving according to the concentration gradient.
- Can be active, moving against the concentration gradient using energy.
Pinocytosis
- Part of the cell membrane invaginates and traps the drug molecule.
- The trapped drug molecule inside a vesicle is then released inside the cell.
- Large molecules such as hormones use this mechanism.
Factors Affecting Drug Absorption
Factor related to drugs
- Molecular size: Smaller drug molecules are absorbed better than larger ones.
- Dose: Absorption increases with increasing dose, up to a limit.
- Drug formulations: Absorption rates vary as sustained-release tablets < suspensions < powders
- Drug Combination: Certain drugs affect the absorption of others. Example: Vitamin C increases iron absorption.
- Lipid Solubility: Lipid solubility is most important factor as described by (drug ionization = pKa)
- Local effects of the drug: Some drugs affect their own absorption. Example: drugs producing vasoconstriction decrease their own absorption.
Factor related to absorbing surface
- Route of administration: IV route is the fastest, and rectal route is the slowest.
- Integrity of the absorbing surface: The absorbing surface or decrease in absorption
- Local blood flow: Ischemia decreases absorption.
- Specific factors that affect drug absorption include:
- Apoferritin enhances iron absorption
- The intrinsic factor is needed for Vitamin B12 absorption
- HCl is needed for aspirin absorption.
- First-pass metabolism: Metabolism at the absorption site decreases the concentration of the drug.
Relationship Between Lipid Solubility and Drug Ionization
- Theory of pKa = Lipid solubility = Drug ionization = Dissolution constant
- Definition: pKa is the pH at which 50% of the drug is ionized (non-absorbed), and 50% is non-ionized (absorbed).
- Principles:
- Ionized drugs, which are water-soluble, polar, and charged, are poorly absorbed.
- Unionized drugs, which are fat-soluble, non-polar, and non-charged, are more lipid-soluble and rapidly absorbed.
- For acidic drugs like aspirin
- They are more ionized in basic media and less ionized in acidic media.
- More absorbed in the stomach, less in the intestine.
- For basic drugs such as amphetamine and local anesthetics
- They are more ionized in acidic media and less ionized in basic media.
- More absorbed in the intestine, less in the stomach.
- Key principle: Drugs move better in a similar environment and worse in an opposite environment.
- When the pH of the media equals the pKa of the drug, the ratio of ionization or unionization is 1:1.
- With every unit change in pH of the media ionized to unionized ratio changes 10 folds
- Acidic drug in pH medium with unionized ionized ratio
- ↑pH 2 unit shows a 1:100 ratio and is 100 % ionized.
- ↑pH 1 unit shows a 1:10 ratio with 10/90 ionized
- pH = pKa shows a1:1 ratio, with 50%/50% ionized
- ↓ pH 1 unit shows a 10:1 ratio with 90/10 ionized
- ↓ pH 2 unit shows a 100:1 ratio and is 100 % union
- Acidic drug in pH medium with unionized ionized ratio
- Basic drugs in pH medium with unionized ionized ratio
- ↑pH 2 unit shows a 100:1 unit and is 100 % union
- ↑pH 1 unit shows a 10:1 ration with 90/10 ionized
- pH = pKa shows a1:1 ratio, with 50%/50% ionized
- ↓ pH 1 unit shows a 1:10 ratio with 10/90 ionized
- ↓ pH 2 unit shows a 1:100 with 100% ionized
Clinical Significance of pKa
Understanding Absorption and Excretion of Weak Acids, e.g., Aspirin
- Aspirin is an acidic drug with a pKa of 3.5
- pH values in different body locations:
- Stomach: 1.5
- Upper Intestine: 6.5
- Lower Intestine: 7.5
- Blood and Body Organs (stomach cells & renal tubular cells): 7.4
- Urine: 4.6 to 8.2
- Tubular Cells: 7.4
Actions of apsirin
- Aspirin in the stomach
- Acid conditions, making it more unionized than ionized
- Resulting in better absorption in the stomach
- Aspirin in the intestine
- Basic conditions, making it more ionized than unionized
- Resulting in less absorption in the intestine.
- Aspirin inside the stomach wall (body pH) exhibits ion trapping
- pH inside the cells is 7.4, aspirin becomes more ionized and less able to diffuse out.
- This can lead to cell damage and gastric ulcers.
- Aspirin in the blood
- Aspirin is metabolized into salicylic acid with a pKa of 3.
- At a blood pH of 7.4, it is more ionized and remains in the blood.
- Aspirin in the urinary tract at acidic pH
- It becomes less ionized and may be reabsorbed.
- Aspirin inside tubular cells exhibits 2nd ion trapping → and can damage to renal tubules.
Understanding Absorption & Excretion of Weak Base
- Example: Amphetamine (pKa = 9.9)
- Opposite effects compared to acidic drugs like aspirin.
Treating Drug Toxicity Based on pKA
- Toxicity with Acidic Drugs (Aspirin):
- Treat by alkalinization of urine:
- Using sodium bicarbonate to increase drug ionization in the urine
- Prevents reabsorption and promote excretion.
- Treat by alkalinization of urine:
- Toxicity with Basic Drugs (Ephedrine):
- Treat with acidification of urine:
- Using ammonia to increase drug ionization in the urine
- Prevents reabsorption and promotes excretion.
- Treat with acidification of urine:
Drug Trapping Sites (Ion Trap)
Acidic Drugs:
- Trapped in stomach wall after absorption, which may cause ulcers when taken after eating.
- Trapped in the nephron wall after reabsorption from urine, leading to kidney injury. Alkalinization of urine helps excretion. Alkaline Drugs:
- Trapped in milk and tumors due to higher unionization in plasma at pH 7.4
- Can cross into milk & tumor due to pH between 6.5 and 7, become ionized, and get trapped.
- Taking the drug after lactation and alkalizing blood increases trapping, which is harmful to the baby but good for killing tumor cells.
Increasing or Decreasing Drug Action
- Local anesthetics are weak bases.
- Adding bicarbonate to the anesthetic maintains it in a non-ionized state, which increases penetration.
- Tissue infection and inflammation can make tissues acidic, increasing ionization of local anesthetics, and decreasing penetration.
Bioavailability of Drugs
- Fraction of unchanged drug reaching systemic circulation after administration by any route compared to the administered dose
- Intravenous bioavailability is 100% since the entire dose reaches the bloodstream
- Bioavailability measured by:
-
AUC: Area under the curve
-
AUC oral AUC IV
- Oral bioavailability =
- Importance: Determine effectiveness and duration of a drug at different routes especially for drugs like:
- Digoxin
- Phenytoin
- Warfarin
- Prednisolone
- Chloramphenicol
- Factors that affect bioavailability:
- Absorption
- First-pass for oral routes
Distribution
- Transport of drug from site of administration to all body tissues through circulation.
Sites Of drug distribution
- Plasma (3 liters)
- Extracellular water (9 liters)
- Intracellular water (29 liters).
- Key Point: a Volume of distribution (Vd) of more than 41 liters means the drug moves to tissues.
Volume of Distribution (Vd)
- Apparent volume of water into drug is distributed after distribution equilibrium = time Distribution all over the body.
- Total amount of the drug within the body divided by the drug concentration in the plasma
- Equation: (L)Total amount of drug in body/ Plasma conc. of drug after distribution equilibrium
- Calculated as: Total Vd for 1 KG is Vd x 70kg if Large Vd = more tissue distribution
- If Vd for Digoxin is 6 L/kg (total = 420 L).
- Each 1 kg of tissue takes concentration drugs in 6 L of plasma.
- Then Each 1 kg of tissue takes 6-fold the conc of digoxin 1 L of the plasma.
- Then each 1L plasma takes a 1/6 of digoxin conc in 1 kg of tissue.
- To distribute digoxin equally between all body tissues (70 kg) and plasma, there is a desire for imaginary volume of plasma = 6 * 70 = 420 L
Volume of distribution dependdant of the type
- small Vd occurs when:
- Lipid solubility is low
- Large drug size
- There is a high degree of plasma protein binding.
- There's a low level of tissue binding.
- Large Vd occurs when:
- Lipid solubility is high
- Small drug size
- There is a low degree of plasma protein binding.
- There is a high level of tissue binding
Significance of site distribution
-
Use determination of site of drug distribution to check if Drug is confined to Blood or can
be removed by dialysis. -
if a Total Vd < 5L then drug Confined to blood and be removed by dialysis
-
if Total Vd < 15 L then the drug restricted to ECF.
-
if Total Vd15-41 L the drug distributed Throughout all body water.
-
if Total Vd > 41L then drugs bind extensively To the tissue proteins and Cannot be removed by the dialysis
Calculation
- Calculation of total amount of the drug in the body by a single measurement of the - - - - plasma concentration.
- Calculation of drug Clearance = Vd x Kel. Calculation of the loading dose as (total): drug amount= Vd X plasma Concentration
Binding of Drugs to Plasma Proteins
- Most drugs bind to plasma proteins; the bound form is inactive, and the free unbound part is active.
- The bound form is gradually released, the plasma concentration of the drug decreases
- Albumin:
- Most important plasma protein
- Binds mainly to acidic drugs and some basic drugs.
- Other plasma proteins
- Globulin and Glycoproteins bind mainly basic drugs.
Advantages of Plasma Binding
- Slows the rate of drug action but not for stopping, the action is better for prophylaxis due to ↑duration after single in the blood.
Disadvantages of plasma binding:
- Is not effective, as a bound form.
- Drug interaction, if given to other drugs to plasma.
- Increases when drugs have High plasma protein bound.
- Change in the binding occurs
- Plasma protein binding gets altered and is so free that the leading causes a serious issue.
Binding of Drugs to Fats
-
Clinical Significance
- Cross the BBB And exerted CNS effects and
- Stored in the body fats
-
In the same way vitamin A stored in fat cells of river For six months until the Rapid elimination of drug to fat by redistribution
-
Strong lipophilic drug (thiopental) when injected intravenously goes into two phases
- The First Phase
-
The drug goes into the organs that are the organs and redistributes into the bloodstream -. The process is abrupt termination of the effect of the drug
-
-
Second phase of drug distribution to the less vascular tissue occurs after an initial distribution, a phase of a highly vascular organ. The effects of anesthetic and other drugs is ultra acting although one over is 3-6 hours
-
After five minutes the liver (terminating its effect) and redistributes in the body’s after and after that.
-
Can make thiopental a longer repeat to saturate fat distribution.
-
Selective Accumulation of Drugs
- Tissue half-life of certain organs occurs.
- So leave plasma rapidly:
- short plasma
- so that have short plasma and the time is still being taken multiple times per day.
- Or may be have to get into.
- They may be taken once daily or more due to selective accumulation in organs prolong duration hours Carbimazole used to treat hyperthyroidism is selective, like is only 3 hours to 6, is selective and used at the thyroid gland or tissue and used once daily.
Metabolism
- Bio-transformation is the process of chemical alteration of drugs in the body
- Primary site of chemical alteration of drugs is the liver. Other organs include the kidneys, lungs, and GIT
- metabolism aims to convert lipid soluble drugs to water soluble:
- Facilitate drug Excretion (urine, bile, milk, sweet)
- decrease tissue penetration and renal reabsorption
Biochemical Reactions
Phase 1 reaction
- oxidation
- reduction
- hydrolysis
- If the drug is converted into the water, with one of the reaction does not mean to inter phase 2 metabolites
Phase 2 reaction
- Conjugation
- Coupling of water, with water and insoluble and forming water to form water sugar
Oxidation Reactions
- Most common reaction. Oxygen to drug or is removed from the drug
Principle of Oxidation REACTIONS
- By oxidizing enyzyms or more organs
Enzymes
- are more enzymes liver especially
- 2-microphones sub cellular membrane or vascular
- 3 cytochrome P450 enzymes Cp450) 4 Family of Enzyme.
microsomal liver enzyme inducers and Inhibitors include
Enhancers
- that the rates of the drugs leading to the therapeutic failure or Must of except of acetaminophen and a clinical examples of some include accelerating rate of A graft injection.
Inhibitors
- the rate of some leading to ↑ and clinical Ciprofloxacin or .
Non-Microsomal Oxidation
- Soluble enzymes in cytoplasm mitochondrial.
- Ex: xanthine converts xanthine to uric acid through oxidatives, oxidizes, oxidation by oxidase, oxidizes catecholamines, and serotonin's.
Reduction reactions
- less common than oxidation. types:
- Microsomal.
- Non-microsomal.
Hydrolysis Reactions
- TYPE
- mainly non-microbial bodies
Main Types
types
- non specifc esterss
- Peptidases
NOTE
- if drug is Water is available from the start then metabolism Excretion
Phase 2 Reactions (Conjugation)
Definition:
- Coupling of a drug metabolite transfer to make water and secreted through
- Types of endogenous reactions
- Glucuronide for a reaction which is one, where the glucuronic is conjucation and is conjured from it. Drugs will be Metabolized
- Glucuronide is conjugated secreted and rapidly in excreted. It may involve bacteria that has a circulation increased, E.g. birth controlling pills
- Glutathione-Using glutathione acid.
- non Microbial reactions Include Acetylation (isoniazide, salicylic acid). Sulffation
First-Pass Metabolism (Pre-Systemic Elimination)
- Metabolism of drugs at admin, site before reaching action.
oral route:
- Gut first-pass metabolism Gastric acidity destroys benzylpenicillin
- Digestive enzymes digest hormones as insulin
- COMT in intestinal mucosa metabolize L-dopa -Hepatic: the first pass metabolized to and lung through inhalation.
- if drug is partial the doses are High drug dose first pass effect if it is partial with a smaller dose . or complete if there were small inhalators and
- A low does to avoid
Bio Factors Affect Drug Metabolism
genetic factors the vitamins that decreases amino or increases with malnutrition. Other effects may increase or decrease based on extreme malnutrition
- Age- cause hepatic amino acid. -Hepatic- decreased -Hepatic enzymes- Increase and lead to toxicity.
Excretion and Elimination
Clearance as a Channel of Elimination
- The volume of formula for minute and its Rate or concentration in Vd as with T
- Time taken for one out of two of plasma concentrated to drop
- If one of and in one way to glomerus
- if limited. Not exceed than blood filtered rate
- Is greater, then also the rate of blood
Route of elimination
- Kidney, major Route filtered through Active systems tubular the and
- The Biles and then small bowl followed by
- Lungs then the mouth.
Clinical Importance
- Clinical in portance of the kidney route and the way can adjust for the to avoid coming to the right, with the right dose to the
- Elimination half-life
- Definition Time taken 50% to fall in place
- Calculation 0.69 and one
Clinical time
Clinical with a specific can lead to toxic levels depending on it, The drug if or not is to toxicity for long will improve better Determining the dose over the of all levels one can and the drug to make it effective depends on side effects which may vary if it is already that the side effects are caused or may be of . The use of that certain cases a clinical to better know exactly what one has the patient at
steady state conc
The Steady The administration barriers that the that those this equation If the or If is a one with If and occurs to and.
- Steady Conc with the
- Determine every order and give calculate drug effect
- Time to observe will the has will
Time needed to observe toxicity
- Zero order will till improvement
Kinetic Eliminatlion
- the alpha from the the followed B is and the of is to and
type kinetic order, elimination
clinical or
- The drug in what can be followed
- first elimination
- with toxic known
Studying That Suits You
Use AI to generate personalized quizzes and flashcards to suit your learning preferences.