ADME Flashcards

1
Q

Q: What are the four stages of the ADME pathway?

A

A: Absorption, Distribution, Metabolism, Excretion.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Q: What must a drug presented in the intestine do to act in the body?

A

A: It has to negotiate a number of processes to get into the body and then into systemic circulation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Q: What happens to a drug immediately upon absorption?

A

A: Drugs must pass through the intestine and then the portal circulation, which takes them to the liver, ensuring any foreign chemical is taken to the liver to minimize the amount of drug entering the body.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Q: What is the fate of a drug after oral absorption? Serum conc

A

Absorption: After a drug is ingested orally, it is absorbed through the gastrointestinal tract into the bloodstream. The drug must reach a certain concentration in the blood (serum) to be effective, known as the effective serum concentration.
Therapeutic Effect: Once the drug reaches this concentration, it can exert its therapeutic effects on the target tissues or organs.

the drug dissolves in the stomach and intestines.
It is then absorbed into the blood through the walls of the intestines and enters the portal circulation, which carries it to the liver.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Q: What is the role of the liver in drug absorption?

A

A: The liver acts to convert foreign chemicals, including drugs, into forms that are more readily eliminated.

In the liver, a portion of the drug may be metabolized before it reaches the systemic circulation. This is known as the first-pass effect.
The remaining drug that is not metabolized enters the systemic circulation, where it can reach its target sites.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Q: What does Fick’s Law say about drug absorption?

A

Fick’s Law states that the rate of diffusion of a drug across a membrane is proportional to the concentration gradient, the surface area of the membrane, and the permeability coefficient of the drug.
A higher concentration gradient, larger surface area, and higher permeability all increase the rate of absorption.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Q: What are SLC influx transporters and their role in drug absorption?
.

A

Solute Carrier (SLC) transporters facilitate the uptake of drugs into cells.
These transporters, such as OATs (Organic Anion Transporters) and OCTs (Organic Cation Transporters), help move drugs from the intestinal lumen into the portal circulation.
They are crucial for the absorption of many drugs, especially those that are not readily absorbed by passive diffusion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Q: How does P-glycoprotein affect drug levels in the blood?

A

A:

P-glycoprotein decreases drug absorption in the intestines by pumping drugs back into the intestinal lumen.
This action reduces the amount of drug that enters the portal circulation and, subsequently, the systemic circulation.
By limiting absorption, P-glycoprotein helps regulate drug bioavailability and can contribute to drug resistance.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Q: How does drug ionization affect its disposition in tissues?

A

A:

Ionized drugs are less likely to cross cell membranes because they are more water-soluble and less lipid-soluble.
Non-ionized (neutral) drugs can more easily penetrate cell membranes due to their higher lipid solubility.
The degree of ionization depends on the drug’s pKa and the pH of the surrounding environment.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Q: What is the significance of tissue pH in drug absorption and disposition?

A

A:

Tissue pH affects the ionization state of a drug.
For example, weak acids are more likely to be non-ionized in the acidic environment of the stomach, enhancing their absorption.
Conversely, weak bases are more likely to be non-ionized in the more basic environment of the intestines, enhancing their absorption there.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Q: What is presystemic drug metabolism?

A

A:

Presystemic metabolism, also known as first-pass metabolism, refers to the metabolism of a drug in the liver and intestines before it reaches the systemic circulation.
This process can significantly reduce the bioavailability of orally administered drugs, as a portion of the drug is metabolized and inactivated before it can exert its therapeutic effect.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Q: How does plasma protein binding affect drug distribution?

A

A:

Drugs often bind to plasma proteins such as albumin.
Only the unbound (free) drug can cross cell membranes and exert a therapeutic effect.
Extensive protein binding can limit the amount of free drug available for distribution to tissues and can also slow down drug elimination.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Q: What is the first-pass effect?

A

A:

The first-pass effect is the rapid uptake and metabolism of an orally administered drug by the liver before it reaches the systemic circulation.
This effect can significantly reduce the bioavailability of the drug, as a large portion may be metabolized and inactivated on the first pass through the liver.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Q: What is bioavailability and how does it differ between oral and intravenous administration?

A

A:

Bioavailability is the fraction of an administered dose of a drug that reaches the systemic circulation in an active form.
Intravenous (IV) administration provides 100% bioavailability because the drug is delivered directly into the bloodstream.
Oral administration typically results in lower bioavailability due to first-pass metabolism and incomplete absorption.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Q: What are some methods to avoid the first-pass effect?

A

A:

Routes of administration that bypass the gastrointestinal tract and liver, such as intravenous (IV), sublingual (under the tongue), rectal, and transdermal (through the skin) routes, can avoid or reduce the first-pass effect.
These methods allow more of the drug to reach the systemic circulation directly.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Q: Why is intraocular and intranasal delivery advantageous?

A

A:

These routes target the local area directly, reducing the need for higher systemic doses.
They bypass the gastrointestinal tract and first-pass metabolism, leading to quicker onset and higher local drug concentrations with fewer systemic side effects.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Q: What is biotransformation and where does it mainly occur?

A

A:

Biotransformation is the process by which the body chemically alters drugs to make them more water-soluble and easier to excrete.
This process mainly occurs in the liver, but also in the kidneys, intestines, and other tissues.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Q: What is the role of CYP450 enzymes in drug metabolism?

A

A:

Cytochrome P450 (CYP450) enzymes are responsible for phase I reactions in drug metabolism, including oxidation, reduction, and hydrolysis.
These enzymes convert lipophilic drugs into more hydrophilic metabolites that can be more easily excreted.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Q: What are prodrugs and give an example?

A

A:

Prodrugs are inactive compounds that are metabolized in the body to produce an active drug.
An example is codeine, which is metabolized to morphine by the enzyme CYP2D6, providing its analgesic effect.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Q: How does genetic polymorphism affect drug metabolism?

A

A:

Genetic variations in metabolic enzymes, such as those in the CYP450 family, can lead to differences in how individuals metabolize drugs.
These variations can result in differences in drug efficacy and toxicity among individuals, requiring personalized dosing regimens.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Q: What are some important properties of CYP450 enzymes that can influence therapy?

A

A:

Readily Inhibited: CYP450 enzymes can be easily inhibited by drugs and chemicals because they have low substrate specificity, allowing for competition between different drugs.
Inducible: Exposure to certain drugs and chemicals can increase the amount of CYP450 enzymes present, enhancing the metabolism of drugs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Q: How does genetic variation in CYP450 enzymes impact pharmacokinetics?

A

A:

Pharmacogenetic Variation: Genetic differences in CYP450 enzymes can significantly impact drug metabolism and response, leading to variations in drug efficacy and toxicity among individuals.
Extensive Variation: CYP450 enzymes, like CYP3A4 and CYP2D6, exhibit extensive pharmacogenetic variation, affecting their ability to metabolize drugs.

23
Q

Q: What is CYP3A4 and where is it found?

A

A:

Member of CYP450 Family: CYP3A4 is a major enzyme of the CYP450 family.
Location: Predominantly found in the liver but also present in the intestines and other tissues.
Function: Metabolizes many drugs, including statins and HIV protease inhibitors.

24
Q

Q: How does grapefruit juice affect CYP3A4?

A

A:

Inhibition: Grapefruit juice contains compounds like bergamottin that inhibit CYP3A4, leading to increased drug levels in the blood.
Clinical Implication: This inhibition can cause higher concentrations of drugs metabolized by CYP3A4, increasing the risk of adverse effects.

25
Q

Q: What is the significance of inhibitory pharmacokinetic drug interactions?

A

A:

Transient vs. Long-Lived: Most inhibitory interactions are transient, but some, like those involving bergamottin, are long-lived and can significantly affect drug metabolism and safety.
Impact: These interactions are especially problematic for drugs with narrow therapeutic windows, where small changes in drug levels can lead to toxicity.

26
Q

Q: What are some inducers of CYP3A4 and their effects?

A

A:

Inducers: St. John’s Wort and other substances can induce CYP3A4.
Effect: Induction increases the enzyme’s activity, leading to lower drug levels and potentially reduced therapeutic efficacy.

27
Q

Q: How does induction of CYP genes occur?

A

A:

Mechanism: Compounds like St. John’s Wort bind to receptors like PXR (Pregnane X Receptor), which then activate the transcription of CYP genes, increasing enzyme production.
Outcome: This leads to enhanced metabolism of drugs that are substrates for these enzymes.

28
Q

Q: What are CYP2D6 polymorphisms and their effects on drug metabolism?

A

A:

Genetic Variants: CYP2D6 exhibits multiple alleles, leading to different metabolic capacities among individuals.
Metabolizer Phenotypes: Includes poor, intermediate, extensive, and ultrarapid metabolizers, each with varying abilities to process drugs.

29
Q

Q: How do genetic polymorphisms impact CYP2D6 function?

A

A:

Single Nucleotide Polymorphisms (SNPs): SNPs can alter the amino acid sequence, affecting enzyme activity.
Consequences: Changes can result in enzymes that are less active, inactive, or truncated, impacting drug metabolism efficiency.

30
Q

Q: What is the clinical significance of CYP2D6 gene polymorphisms?

A

A:

Dosing and Efficacy: Variations can lead to differences in drug response, necessitating personalized dosing.
Adverse Effects: Poor metabolizers may experience toxicity with standard doses, while ultrarapid metabolizers may not achieve therapeutic levels.

31
Q

Q: How do poor metabolizers of CYP2D6 differ from extensive metabolizers?

A

A:

Poor Metabolizers: Carry two non-functional alleles, leading to significantly reduced enzyme activity and poor drug metabolism.
Extensive Metabolizers: Carry two active alleles, representing the normal metabolic capacity.

32
Q

Q: What are some common substrates for CYP3A4?

A

A:

Drugs Metabolized: Includes statins (for cholesterol), HIV protease inhibitors, and other widely prescribed medications.
Impact of Inhibition: Inhibition of CYP3A4 by substances like grapefruit juice can lead to increased levels of these drugs and potential toxicity.

33
Q

Q: What are SNPs and how do they affect CYP function?

A

A:

Single Nucleotide Polymorphisms (SNPs): Genetic variations involving single base pair changes in the DNA sequence.
Impact: SNPs can alter enzyme function by changing amino acid sequences, introducing stop codons, or affecting protein synthesis, leading to variability in drug metabolism.

34
Q

Q: How does St. John’s Wort affect drug metabolism?

A

A:

Induction of CYP3A4: St. John’s Wort induces CYP3A4, increasing the enzyme’s activity.
Effect on Drugs: This can lower the levels of drugs metabolized by CYP3A4, potentially reducing their effectiveness.

35
Q

Q: What is the role of PXR in CYP3A4 induction?

A

A:

Pregnane X Receptor (PXR): A nuclear receptor that, when activated by inducers like St. John’s Wort, binds to DNA and increases the transcription of CYP3A4 genes.
Result: Increased production of CYP3A4 enzymes, enhancing drug metabolism.

36
Q

Q: What are the implications of variable CYP2D6 activity in the population?

A

A:

Dosage Adjustments: Genetic testing can identify metabolizer status, guiding dosage adjustments to optimize therapeutic outcomes and minimize adverse effects.
Drug Selection: Alternative medications may be considered for poor or ultrarapid metabolizers to ensure efficacy and safety.

37
Q

Q: What are extensive metabolizers of CYP2D6?

A

A:

Normal Metabolism: Individuals with two functional alleles of CYP2D6, capable of metabolizing drugs at normal rates.
Clinical Relevance: These individuals typically respond well to standard drug doses without significant risk of adverse effects or therapeutic failure.

38
Q

Q: What are the effects of CYP450 enzyme inhibition on drug therapy?

A

A:

Increased Drug Levels: Inhibition can lead to higher plasma concentrations of drugs metabolized by these enzymes, increasing the risk of toxicity.
Drug Interactions: Co-administered drugs that inhibit CYP450 enzymes can cause significant pharmacokinetic interactions, necessitating dose adjustments or alternative therapies.

39
Q

Q: What is Phase II metabolism and its purpose?

A

A:

Definition: Phase II metabolism involves the conjugation of Phase I metabolites with highly polar endogenous molecules such as glucuronic acid or sulfate.
Purpose: To make drug molecules more water-soluble and less lipid-soluble, facilitating their excretion in urine or feces.

40
Q

Q: What happens to Phase I metabolites in Phase II metabolism?

A

A:

Conjugation: Phase I metabolites are conjugated with polar molecules, making them more hydrophilic.
Excretion: These Phase II conjugates are then excreted rapidly in urine or feces.

41
Q

Q: Explain the role of UGT1A1 in drug metabolism and its genetic variability.

A

A:

Function: UGT1A1 is an enzyme involved in glucuronidation, a major Phase II metabolic pathway.
Polymorphisms: UGT1A1 has variants like *28, which affect enzyme activity. *28 involves an extra TA repeat in the promoter region, reducing enzyme efficiency.

42
Q

Q: What is the clinical significance of UGT1A1 polymorphisms?

A

A:

Drug Response: Individuals with certain polymorphisms (e.g., 28/28) may have reduced ability to metabolize drugs like irinotecan, leading to toxicity.
Diagnostic Tests: Genetic tests can identify these polymorphisms to guide personalized treatment.

43
Q

Q: How do Phase II enzymes protect against drug toxicity?

A

A:

Conjugation: They convert lipophilic drugs into hydrophilic metabolites, reducing their ability to cross cell membranes and interact with intracellular targets.
Excretion: Enhancing the elimination of these metabolites, thus reducing their potential toxicity.

44
Q

Q: What are the main routes of drug elimination from the body?

A

A:

Renal Excretion: Through glomerular filtration, active tubular secretion, and tubular reabsorption.
Biliary Excretion: Drug conjugates are excreted into bile and then into the intestines.
Other Routes: Pulmonary (exhalation), sweat, breast milk, hair, and saliva.

45
Q

Q: What factors influence renal excretion of drugs?

A

A:

Glomerular Filtration: Only free (unbound) drugs are filtered.
Tubular Secretion: Active transport of drugs from blood to urine.
Tubular Reabsorption: Passive diffusion of drugs back into the blood, influenced by urine pH.

46
Q

Q: How can urine pH be manipulated to enhance drug excretion?

A

A:

Alkalinization: Increasing urine pH with agents like sodium bicarbonate helps excrete weak acids.
Acidification: Decreasing urine pH with agents like ammonium chloride helps excrete weak bases.

47
Q

Q: Describe the role of ATP-binding cassette (ABC) transporters in drug excretion.

A

A:

Function: These transporters use ATP to actively efflux drug metabolites out of cells.
Importance: They help remove Phase II metabolites, preventing their accumulation and potential toxicity.

48
Q

Q: What is enterohepatic recycling and its impact on drug action?

A

A:

Process: Some drugs are excreted in bile, reabsorbed in the intestines, and returned to the liver.
Impact: This can prolong the drug’s presence in the body, extending its pharmacological effect.

49
Q

Q: What is the significance of pulmonary elimination of drugs?

A

A:

Volatile Substances: The lungs eliminate gases and volatile substances like anesthetics.
Drug Metabolites: Some drug metabolites, like those of erythromycin, can be exhaled, allowing measurement of metabolic activity.

50
Q

Q: How are drugs eliminated through sweat, breast milk, and other bodily fluids?

A

A:

Sweat: Elimination of drugs like THC, benzodiazepines.
Breast Milk: Excretion of cytotoxic agents, antihypertensives, and other medications.
Other Fluids: Drugs can also be excreted through saliva and hair.

51
Q

Q: How does irinotecan metabolism highlight the importance of Phase II glucuronidation?

A

A:

Active Metabolite: Irinotecan is converted to SN-38, which is toxic.
Glucuronidation: SN-38 is inactivated by UGT1A1 through glucuronidation.
Toxicity: Polymorphisms in UGT1A1 can impair this process, leading to severe side effects like myelosuppression and diarrhea.

52
Q

Q: What is the role of glucuronidation in drug metabolism?

A

A:

Process: Addition of glucuronic acid to drugs or their metabolites.
Result: Increases water solubility, facilitating renal or biliary excretion.

53
Q

Q: Explain the significance of pharmacogenetic testing in drug therapy.

A

A:

Personalized Medicine: Identifies genetic variations that affect drug metabolism and response.
Guidance: Helps tailor drug choice and dosing to individual genetic profiles, enhancing efficacy and safety.