FDN2_SM_WK2_DrugMetabolism Flashcards

Drug Metabolism

1
Q

What is the purpose of Phase I reactions?

A

Enzyme ~seduction~

These reactions make the drug more ~alluring~ to phase II enzymes by creating an active site for them

Keep in mind that some drugs are ~pre-lubed~ and do not need to undergo phase I reactions in order to be eliminated

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

What is the purpose of Phase II reactions?

A

Phase II reactions make the drug less lipophilic and more polar so it can be excreted

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

What is the most common pathway for drug elimination?

A

Phase I: CYP-dependent oxidation via CYP3A4

Phase II: Glucuronidation via UDP Glucuronyl Transfersase (UGT)

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

Name 3 relevant pro-drugs and describe what happens to them in phase I reactions

A

Prodrugs undergo bioactivation in phase I rections

Codeine -> Morphine via CYP2D6

Clopidogrel ->>> R-130964 (active form)
via CYP2C19 (rate-limiting enzyme)

Azathioprine ->>> glutathione nucleoties
TPMT facilitates excretion of an intermediate; this is accounted for in dosing.

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

Name 3 relevant drugs that do not need to undergo Phase I reactions, and explain why

A

Acetaminophen, Albuteral, Lorazapam

These drugs are ~pre-lubed~ with a group that facilitates Phase II reactions. Interaction with CYPs can create toxic intermediates

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

What is the only drug (that Silinsky knows of) that is not inactivated by glucuronidation?

A

Morphine

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

What drug-metabolizing enzymes are most likely to undergo polymorphisms?

A

CYP2D6, CYP2C9, CYP2C19

*CYP2D6 is the most common

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

What drugs are metabolized by CYP2D6?

A

Drugs for treating CNS disorders (antidepressants, antipsychotics, amphetamines, codeine, opioids)

Beta-Blockers (metroprolol, ___-olol drugs)

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

What drugs are metabolized by CYP2C9?

A

Warfarin

NSAIDS (Ibuprofen, Celecoxib)

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

What drugs are metabolized by CYP2C19?

A

Diazepam (and other ___-azapam benzos)

Proton-Pump Inhibitors (___-prazoles)

Clopidogrel

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

Describe the normal metabolism of acetaminophen

A

Normally, acetaminophen bypasses Phase I metabolism (it is pre-lubed)

It undergoes sulfation or glucuronidation and is safely excreted

If acetaminophen undergoes Phase I reactions via CYP2E1 or CYP3A4, toxic intermediates are created

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

Why is it a bad idea to take acetaminophen and drink alcohol?

A

High levels of aclohol induce CYP2E1

When CYP2E1 acts on acetaminophen, the toxic intermediate NAPQI (an evil electrophile) is created.

Under normal circumstances, the hepatocyte can be rescued if GST can conjugate glutathione to NAPQI. However, alcohol also inhibits GST.

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

What is NAPQI and how is it created?

A

NAPQI is an evil electrophile, created by Phase I metabolism of acetaminophin by CYP3A4 or CYP2E1

When it interacts with a nucleophilic macromolecule, it causes toxicity and liver cell death.

Conjugating glutathione to NAPQI via GST can create a nontoxic mercapturic acid conjugate that can be excreted

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

Which CYP causes benzo[a]pyrene to form epoxides? Why do we care about this?

A

CYP1A causes benzo[a]pyrene to form epoxides

We care becasue epoxides are higly reactive (aka evil). Cigarette smoke and other environmental pollutants contain benzo[a]pyrenes, and they basically cause themselves to become evil by inducing CYP1A

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

How does Ginkgo influence drug metabolism?

A

Ginkgo induces CYP2C9, CYP2C19, and CYP3A4

This increases the metabolism of warfarin, celecoxib, and omeprazole.

Incresed warfarin metabolism -> less effective, higher clotting risk

Increased celcoxib metabolism -> less effective pain management

Increased omeprazole metabolism -> less effective PPI activity

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

How does echinacea influence drug metabolism?

A

Echinacea inhibits CYP1A2 and induces CYP3A4

This results in:

  • Reduced CYP1A2 activity, resulting in less epoxide formation and less toxicity due to environmental pollutants
  • Increased metabolism of the many CYP3A4 drugs (this can have mixed effects)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

How does St. John’s Wort influence drug metabolism?

A

Induces CYP3A4 -> mixed results

Induces CYP2C9 -> increased metabolism of warfarin and NSAIDS

Induces CYP2C19 -> increased metabolism of omeprazole, diazepam, clopidogrel

Induces CYP2E1 -> increased toxic metabolites of acetaminophen

Induces CYP3A -> increased metabolism of estrogen (decreased efficacy of oral contraceptives)

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

You prescribe acetaminophen with codeine to a patient post-op. They call two days later because their pain is poorly controlled. Which cytochrome P450 polymorphism might cause this?

A

CYP2D6 poor metabolizer

These individuals have imparied conversion of codeine to morphine, with reduced analgesic effects.

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

Which cytochrome P450 is involved in creating epoxides on benzo[a]pyrene?

A

CYP1A2

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

Which cytochrome P450 is involved in the metabolism of 50-60% of clinically prescribed drugs?

A

CYP3A4

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

Which cytochrome P450 polymorphism might be associated with needing a lower warfarin dose?

A

CYP2C9 slow metabolizer

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

What are the 5 different CYP-dependent Phase I oxidation reactions?

A
Hydroxylation
S-oxidation
N-oxidation
N-dealkylation
O-dealkylation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What are the 2 CYP-independent Phase I oxidations?

A

Dehydrogenation (pertains to ethanol)

Amine Oxidation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What are the 2 CYP-independent hydroletic Phase I reactions?

A

Ester Hydrolysis

Epoxide Hydrolases (protects against evil epoxides)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Describe the metabolism of Midazolam

A

Phase I: oxidation (N-dealkylation) mediated by CYP3A4

Phase II: Glucuronidation mediated by UGT

(They typical drug metabolism pathway; this applies to all _____-azolam benzodiazepine drugs)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What is something you need to keep in mind when prescribing the H2 histamine receptor blocker cimetidine?

A

Cimetidine inhibits all CYPs except CYP2E1;

This creates the potential for adverse drug interatctions

  • Inability to activate a pro-drug, resulting in a reduced effect
  • Inability to excrete a standard drug, resulting in an increased effect or toxicity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What are two examples of Methylxanthines?

What is their mechanism of action?

A

Caffeine and theophylline (treats respiratory diseases) are both methylxanthines

They are competitive inhibitors of adenosine (adenosine ususally inhibits cell function)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What are the 5 Phase II metabolism reactions?

A

SAGGMeth

Sulfation

Acetylation

Glucuronidation

Glutathione conjugation

Methylation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What is the role of glutathione in acetaminophen metabolism?

A

Glutathione conjugation (mediated by GST) can “rescue” hepatocytes from the toxic intermediate NAPQI.

NAPQI is created by CYP3A4 and CYP2E1.

If it is conjugated to glutathione, it can be safely excreted.

If instead it interacts with a nucleophilic macromolecule, it can lead to toxicity and hepatocyte death

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

Which cytochrome P450 polymorphism is associated with impaired metabolism of Proton-Pump Inhibitors?

A

CYP2C19 slow metabolizer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

Which cytochrome P450 is associated with the creation of the two active metabolizes of Diazepam?

A

CYP2C19

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

Which cytochrome P450 polymorphism is associated with a necessary increased dose of Metroprolol (A Beta1 blocker)

A

CYP2D6 ultrarapid metabolizer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

Why does CYP3A4 have such a broad substrate specificity?

A

CYP3A4 has a large active site cavity volume. It can bind to substrates of a wide variety of shapes and sizes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

Which cytochrome P450 is associated with caffeine and theophyline metabolism?

A

CYP1A2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

Which cytochrome P450 is involved in creating toxic intermediates in acetaminophin metabolism?

A

CYP3A4 and CYP2E1

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

List some relevant actions of the CYP1As

A

CYP1A2: Phase I metabolism of methylxanthines (caffeine and theophyline)

CYP1A?: Causes environmental pollutants containing benzo[a]pyrene to become toxic

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

What are the three major processes in renal exretion of drugs?

A

Glomerular filtration, tubular secretion, passive tubular reabsorption

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

Describe glomerular filtration

A

As blood rushes through the glomerulus, filtrate enters the kidney through the glomerulus

The three layers that make up the glomerular filtration barrier determine what enters the filtrate, and what stays in the blood.

Small particles that are not bound to blasma proteins can easily enter the firltrate.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

Describe the Glomerular Filtration Barrier

A

The three membrane layers that achieve a size- and charge-sensitive filtration of drugs and solutes in the glomerulus.

  • Glomerular capilary endothelum
  • Porous glomerular basement membrane
  • Podocytes

These membranes contain fenestrae and negatively charged glycoproteins. The result is that negatively charged plasma proteins cannot enter (among other things)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

Describe tubular secretion

A

In tubular secretion, ionized drugs are secreted into the filtrate from the bloodstream.

This process is driven by transporters

  • SLC22 transporters uptake molecules from the capilaries into the tubular endothelium
  • SLC and ABC transporters promote the efflux of molecules from the endothelium to the limunal spacs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What transporters are involved in tubular secretion?

A

SLC: Organic Anion Transporters and Organic Cation Transporters

ABC: P-glycoprotein and MRP1

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What transporters are located on the basolateral side of the renal proximal tubule?

A

SLC transporters. This is the side that faces the capilaries

  • OATs and OCTs uptake anions and cations into the tubular endothelium
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What transporters are located on the aplical side of the renal proximal tubule?

A

SLC: OATs and OCTs

ABC: P-glycoprotein, MRP1

Together, these transporters move substances from the tubular endothelium into the lumen of the proximal tubule

44
Q

How would you promote the excretion of a weakly basic drug?

A

Basic drugs are charged in their protonated form.

If the pH of urine

To promote the excretion of a basic drug, make the urine more acidic

45
Q

Describe passive tubular reabsorption

A

Drugs and solutes are reabsorbed into the systemic circulation from the renal tubule.

Nonionized, lipid soluble forms of the drug are able to pass back into the interstitium

Changing the pH of the urine can promote the retention or excrection of acidic or basic drugs

46
Q

How would you promote the excretion of a weakly acidic drug?

A

Acidic drugs are charged in their deprotonated form

If pH of urine > pKa of the drug, the ionized form will dominate, and the drug will not be reabsorbed

To promote the excretion of an acidic drug, make the urine more basic

47
Q

In what scenarios would you want to promote excretion of a drug?

A

Overdose or toxicity due to high drug concentration

48
Q

Drug excretion via bile is analogous to what step in renal excretion?

A

Proximal tubular secretion

Both involve ABC and SLC transporters

49
Q

What transporters are invovled in dug excretion through the bile?

A

SLC: OAT and OCT transporters

ABCs

These transporters exist in canalicular membranes of hepatocyctes to secrete drugs into the bile

50
Q

A defect in which hepatocyte transporter is implicated in elevated cholesterol and phytoserol?

A

ABCG5/8

51
Q

A defect in which hepatocyte transporters is implicated in risk for myotoxicity with the use of statins?

A

SLCO1B1; Defects in this transporter result in imparied secretion of the drug into the bile

This results decreased excretion and increased plasma concentrations of the drug, which can lead to myotoxicity

52
Q

What kinds of drugs are most likely to be excreted in the bile?

A

Heavy drugs; molecular weight >300

53
Q

Describe enterohepatic circulation

A

A recycling system!

Drugs that are glucuronidated in the liver are secreted into the bile and then dumped into the gut

Bacteria in the colon may cleave the glucuronide off of the drug, increasing its lipophilicity

The drug is now more likely to be reabsorbed into the hepatic circulation

54
Q

Why do some antibiotics (espcially rifampin) interfere with drug efficacy?

A

Antibiotics may disrupt the bacteria in the colon

The bacteria normally cleave glucuronide off of the drug, allowing the drug to be reabsorbed into the circulation.

If the bacteria are dead, more of the drug will be excreted, lowering the expected plasma concentration of the drug and reducing its efficacy

55
Q

For a drug that is excreted via glomerular filtration, how would you adjust dosing for a patient with renal failure?

A

You would want to lower the dosage of the drug; less will be filtered into the kidney, which means that more will remain in the plasma.

Lowering the dosage can reduce the risk of toxicity

56
Q

What factors impact the clearance of “high extraction drugs?”

A

Clearance depends on blood flow to the liver

Metabolism is not rate-limiting, and plasma-protein binding doesn’t really matter

Note: These drugs tend to have high first-pass metabolism

57
Q

What is the equation for hepatic clearance?

A

CL = (Rate of elimination) /[plasma concentration]

58
Q

What factors impact the clearance of “low extraction drugs?”

A

Hepatic metabolism and plasma protein binding

Blood flow to the liver is not rate limiting, because metabolism in the liver is slower, and dependent on the fraction of free drug in the blood (which is impacted by binding to plasma proteins)

59
Q

What are some examples of high extraction drugs?

A

Morphine, propanolol, lidocaine

60
Q

What are some examples of low extraction drugs?

A

Digoxin, diazepam, phenytoin, valproic acid, cimetidine

(DDPVC)

61
Q

How does binding to plasma proteins affect renal excretion of drugs?

A

More binding to proteins = less excretion

Bound proteins cannot enter glomerular filtrate

62
Q

What is the differnece between pharmacogenetics and pharmacogenomcis?

A

Both are related to the study of genes

Pharmacogenetics looks at individuals or families, and how their SNPs may affect drug response

Pharmacogenomics looks at genes that are associated with differential drug responses among groups of unrelated indivdiuals, on a population-wide level

63
Q

List a few specific pharmacogenetic or pharmacogenomic syndromes

A

Malignant hyperthermia susceptibility

G-6-P dehydrogenase deficiency

Pseudocholinesterase deficiency

64
Q

Describe the relevant pharmacogenomics of codeine

A

Codeine is a prodrug that requires converstion to morphine by CYP2D6

CYP2D6 ultrarapid metabolizers end up making a lot of morphine. This can lead to a dangerous functional morphine oversdose (increased sedation, respiratory depression)

CYP2D6 poor metabolizers have lower than expected analgesic effects from codeine

65
Q

Describe the relevant pharmacogenomics of clopidogrel

A

Clopidogrel is a pro-drug that is prescribed as an anti-platelet agent. CYP2C19 is required to activate the drug.

CYP2C19 ultrarapid metabolizeers are at an increased risk for bleeding

CYP2C19 poor metabolizers do not have adequate antiplatelet effects; they are at an increased risk for blood clots

66
Q

Which cytochrome P450 is relevant to codeine metabolism?

A

CYP2D6 activates codine to morphine

67
Q

Which cytochrome P450 is relevant to colpidogrel metabolism?

A

CYP2C19 activates clopidogrel

68
Q

Which cytochrome P450 is relevant to Omeprazole (and other ___-prazole) metabolism?

A

CYP2C19 and CYP3A4 facilitate excretion of omeprazole

69
Q

Which enzyme is relevant to Thiopurine drug metabolism? (ex: Azathiopurine)

A

Thiopurine S-Methyltransferase; required for excretion of the drug

70
Q

Which cytochrome P450 is relevant to warfarin metabolism?

A

CYP2C9 facilitates excretion of warfarin

71
Q

Describe the relevant pharmacogenomics of Omeprazole (and other ___ -azole drugs)

A

____-prazole drugs are proton pump inhibitors. CYP2C19 and CYP3A4 are required for metabilism and

CYP2C19 ultrarapid metabolizers clear the drug too quickly, resulting in poor treatment of gastric acidity

CYP2C19 poor metabolizers experience a higher efficacy of PPI drugs, but in children this may be associated with upper respiratory infections and strep

72
Q

Describe the relevant pharmacogenomics of Thiopurine drugs

A

Thiopurine drugs like Azathioprine are pro-drugs that are used to treat leukemia, rehumatic disease, IBD, and organ transplantation. They are activated by a metabolic pathway to glutathione nucleotides

Thiopurine S-Methyltransferase (TPMT) facilitates the excretion of an intermediate of the pathway, 6-mercaptopurine. If there is a loss of function/reduced function mutation in the TPMT gene, more glutathione nucleotides will be made.

This results in increased drug efficacy, but also risk for toxic effects

73
Q

What are Thiopurine drugs used for?

A

Thiopurine drugs are used to treat leukemia, IBD, rheumatic disease, and organ transplant

(Think Autoimmune disorders)

74
Q

Give some examples of Thiopurine drugs

A

Azathioprine, 6-mercaptopurine, and 6-thioguanin

75
Q

Describe the relevant pharmacogenomics of warfarin

A

Warfarin is an anticoagulent that is metabolized by CYP2C9.

CYP2C9 ultrarapid metabolizers will clear the drug quickly, putting the patient at risk for clotting

CYP2C9 poor metabolizers will clear the drug slowly, resulting in higher plasma concentration and risk for bleeding

Warfarin exerts its effects by inhibiting VKORC1, an endogenous enzyme required for sythesis of clotting factors

LOF mutations in VKORCI result in increased baseline INR (it takes longer for the patient to clot, putting them at increased risk for bleeding)

76
Q

What enzyme is relevant to warfarin pharmacokinetics?

A

CYP2C9

77
Q

What enzyeme is relevant to Warfarin pharmacodynamics?

A

VKORCI

78
Q

Describe a relevant exmample of somatic pharmacogenomics

A

Tumors may contain genetic variants that make them more or less responsive to anti-cancer drugs

Hyperactive EGF Receptors: This drug will respond really well to anti-cancer drugs that target the EGF receptor

Nonresponsive EGF Receptors: If anti-cancer drugs work to eradicate 99% of tumor cells, the ones that remain are likely resistant to the drugs due to a mutation that gives them resistance to the drug. Relapse due to growth of these resistant cells is hard to treat, because they are unresponsive to the anti-cancer drug.

79
Q

What are the “good and the bad” of gultathione?

A

Good: Glutathione conjugation via GST can protect against toxic metabolites and therefore prevent cancers from developing

Bad: If cancer does develop, high levels of GST in tumors can cause cancer cell proliferation and resistance to chemotherapy

80
Q

How would drinking grapefruit juice affect a patient taking statin drugs?

A

Grapefruit juice inhibits CYP3A4, resulting in slower metabolism of the drug. The effects of the statin will be potentiated.

81
Q

Suppose you get blackout drunk at a party and your “friend” gives you acetaminophen.

You are worried about the toxic effects that NAPQI may have on your system.

Luckily, you are a medical studnet and have free access to any chemical in the world. What antidote do you give yourself?

A

N-Acetylcysteine (NAC)

NAC conjugates directly with NAPQI, replaces glutathione stores, and can protect against extrahepatic injury

Giving extra glutathione may seem like it could work, but it would be ineffective because it can’t cross cell membranes.

82
Q

How would you interpret the genotype CYP2D6*1x3

A

The individual has an extra functional copy of the reference CYP2D6 gene; they may have increased CYP2D6 activity

83
Q

What is the equation for volume of distribution (Vd)?

A

Vd = (dose)/(plasma concentration)

Unit = L

84
Q

What does the volume of distribution (Vd) represent?

A

The amount of the administered drug that is in tissues, organs, or otherwise sequestered out of the systemic circulation.

This measurement is only meaningful right after the drug has been distributed (before it is metabolized or excreted)

85
Q

What factors affect Volume of Distribution (Vd)?

A

Patient factors: age, gender, body muscle/fat proportion, level of hydration, water distribution (pregnancy, edema)

Drug Factors: High lipid solubility, low ionization = higher Vd

Vd is unaffected by half-life or clearance

86
Q

What is the equation for half-life of a drug?

A

For first order drugs t1/2= 0.693*Vd/Cl

For zero-order drugs, half life cannot be calculated by a simple equation; the time it takes for 50% of the drug to be eliminated will depend on how much drug is in the system

87
Q

What is the equation for Clearance (CL)?

A

CL = (rate of elimination)/(plasma concentration of the drug)

Unit: L/hr or mL/min

CL is constant for first order drugs; the rate of elimination increases or decreases with the [drug]

CL is not constant for zero order drugs

88
Q

What does the Clearance (CL) of a drug mean?

A

The volume of plasma cleared of the drug per unit time.

89
Q

What is the equation for rate of elimination?

A

Rate of Elimination = CL * [drug in plasma]

90
Q

How is Rate of Elimination different for zero order and first order drugs?

A

First order drugs: Rate of Elimination is a fixed fraction of the [drug]; this is not a saturated system, and can change based on the [drug]

Zero order drugs: Rate of Elimination is a fixed amount of drug. It doesn’t depend on the [drug]; think of it like a saturated system that is going as fast as it can

91
Q

What is the equation for steady state concentration (Css)?

A

For IV drugs: (infusion rate)/(CL)

For oral drugs: (Oral dose)/(dosing interval * CL)

92
Q

If you are administering a constand dose, about how long will it take a patient to reach Css?

A

~3.3 half-lives to reach 90% Css (which is pretty much the goal)

93
Q

What is the equation for loading dose (LD)?

A

LD = Css * Vd

94
Q

What does it mean if a drug has a high Vd?

A

A high Vd means that a large amount of the drug gets sequestered in tissues or other spaces outside of the systemic circulation

95
Q

If a drug has a low Vd, how is the Css affected?

A

It isn’t; Css doesn’t depend on Vd

Css = Infusion rate/CL

The goal for Css is to have a steady [drug] in the systemic circulation. After a drug has been distributed to tissues and other spaces outside of the systemic circulation, it kind of just stays there and doesn’t affect [drug}

96
Q

What is the equation for maintenence dose?

A

IV drug: I = Css * CL

97
Q

What kind of transporter is p-glycoprotein?

A

ATP Binding Cassette

(Directly energy dependent = Involved in active transport)

98
Q

Which genotypes of CYP2D6 would result in ultrarapid metabolism?

A

More than 2 copies of *1 or *2

99
Q

Which genotypes of CYP2D6 would result in poor metabolism?

A

Two copies of anything that isn’t *1 or *2

*1 or *2 plus any other allele = within normal range

(*4 and *5 are most common)

100
Q

Which genotypes of CYP2C19 would result in ultrarapid metabolism?

A

*1/*17 or *17/*17 (17 is ultrarapid)

101
Q

Which genotypes of CYP2C19 would result in poor metabolism?

A

Any genotype that doesn’t contain *1 or *17

(*2 and *3 are common)

102
Q

In which populations are CYP2D6 poor metabolizers most common?

A

Caucasians (8-10%)

African Americans (1-3%)

103
Q

In which populations are CYP2D6 ultrarapid metabolizers most common?

A

Ethiopians and Saudi Arabians (30%)

104
Q

In which populations are CYP2C19 poor metabolizers most common?

A

Asians (15-20%)

Caucasians (3-5%)

African Americans (3-5%)

105
Q

In which populations are CYP2C9 poor metabolizers most common?

A

Caucasians (10-35%)