L30 Pharmacogenetics Flashcards

1
Q

What is the range of variations in drug responses?

A
  • severe adverse effects

- lack to therapeutic activity

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

what are the factors that can influence the drug response phenotype?

A
  • age
  • gender
  • disease
  • genetic variation!
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

define pharmacogenomics?

A

study of the role of inheritance in variation in drug responses

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

why is pharmacogenomics important to study?

A

because adverse drug reactions are a major cause of morbidity and mortality

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

what are the 3 types of genetic variation that can influence pharmacotherapy?

A
  1. variation in proteins involved in drug metabolism/transport
  2. variation in drug targets (pharmacodynamic variation)
  3. variation in proteins associated with unique (idiosyncratic) adverse drug effects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

what does absorption, distribution and elimination (what the body does to the drug) define?

A

pharmacokinetics

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

what describes what the drug does to the body?

A

pharmcodynamics

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

what is the most common factor responsible for pharmacogenetic variation in drug responses?

A

genetic variation in enzymes that catalyze drug metabolism

*most enzymes in drug metabolism are POLYMORPHIC!

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

give 4 examples of the polymorphic drug metabolizing enzymes?

A
  • butyrylcholinesterase (pseudocholinesterase) (BChE)
  • N-acetyltransferase 2 (NAT 2)
  • cytochrome P450 2D6 (CYP2D6)
  • thiopurine S-methyltransferase (TPMT)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

what type of drug is succinycholine?

A

depolarizing neuromuscular blocker

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

what is the function of succinylcholine?

  • what receptor does it bind to?
  • what NT does it act like?
  • what does it cause to happen on the end plate?
A
  • binds to nicotinic receptors
  • acts like ACh
  • depolarizes the end plate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

what is the major difference between ACh and succinylcholine?

A

succinylcholine is not metabolized at the synapse effectively like ACh (using AChE)

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

what type of onset and duration does succinyl choline have?

A

fast onset

short duration

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

what breaks down and metabolizes succinylcholine?

A

plasma butyrylcholinesterase

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

what happens with genetic variation in butyrylcholinesterase?

  • main problem
  • result
  • cause (what is the defected gene)
  • how it is transmitted?
A
  • decreased rate of metabolism of succinyl choline
  • prolonged paralysis
  • defect in BCHE gene
  • autosomal recessive trait
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

what does N-acetyltransferase 2 (NAT 2) catalyze?

A

ACETYLATION of isoniazid (for TB) and other drugs

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

what are the 2 classes of patients tc with isoniazid?

A
  • slow acetylators
  • fast acetylators

*The rate is inherited!

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

will slow acetylators have high or love blood levels of drug?

A

high because they metabolize the drug SLOWER

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

will fast acetylators have high or low blood levels of drug?

A

low because they metabolism the drug FASTER

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

slow acetylators of isoniazid are ____ for an autosomal recessive allele of the enzyme with lower activity levels

A

homozygous

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

what acetylators are more prone to toxicity of drugs that are metabolized by acetylation? Why

A

slow acetylators are prone to toxicity of drugs because they take a long time to metabolize the drugs so the levels are going to rise int he blood and can create serious adverse effects on the pt.

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

____ may cause neuropathy and hepatotoxicity

A

isoniazid

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

______ and _____ may cause SLE

A

hydralazine and procainadmide

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

_____ may cause hypersensitivity reactions, hemolytic anemia, SLE

A

sulfonamides

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

what drugs were used when studying the CYP2D6 polymorphism?

A

antihypertensive - debrisoquine

oxytotic - sparteine

26
Q

if the ratio of decrisoquine/4-hydroxydebrisoquine metabolic ratio is high, what does that indicate?

A

poor metabolizer

27
Q

if the ratio of decrisoquine/4-hydroxydebrisoquine metabolic ratio is low, what does that indicate?

A

ultrarapid metabolizer

28
Q

poor metabolizers are _____ for the recessive alleles coding for enzymes with low activity

A

homozygous

29
Q

extensive metabolizers (normal) are ______ or _____ for the “wild type” allele

A

homozygous

heterogygous

30
Q

some ultra rapid metabolizers can have ______ copies of the CYP2D6 gene

A

multiple (up to 13)

31
Q

inter individual differences are important with CYP2D6 because they metabolize many commonly prescribes drugs like…

A
  • beta blockers
  • antipsychotic
  • opiods
  • antidepressants
32
Q

what drug is ineffective in poor metabolizers and why?

A

codeine

requires CYP2D6-catalyzed conversion from codeine to morphine

33
Q

what metabolizers can overdose with standard doses of codeine and suffer from respiratory depression or arrest?

A

ultrarapid metabolizers

34
Q

what does TPMT catalyze?

A

S-methylation of the anticancer thiopurines (6 mercaptopurine and azathioprine)

35
Q

____ of the anticancer thiopurine inactivates them!

A

methylation

36
Q

thiopurines have a ________therapeutic index and some pts may suffer from life-threatening _____

A

narrow

myelosuppression

37
Q

about 1/300 individuals are ____ for a polymorphism that leads to LOW TPMT activity

A

homozygous

38
Q

the individuals who are homozygous for low TPMT activity have an increased risk for ____ when treated with standard doses of thiopurine drugs , so these pts have to be tx with _____ of the standard dose

A

myelosuppression

1/10

39
Q

why does warfarin make dosing problematic?

  • therapeutic window
  • variability
A

because it has a narrow therapeutic winnow and

wide inter individual variability

40
Q

what does under-anticoagulation result in

A

thrombosis

41
Q

what does over-anticoagulation result in

A

bleeding episodes

42
Q

warfarin

  • what type of mixture?
  • what are the 2 types?
  • are they both metabolized by the same enzyme?`
A
  • racemic
  • S-warfarin***** (3-5x more potent) and R warfarin
  • stereoismoers are metabolized by DIFFERENT enzymes
43
Q

which gene is most involved in the metabolism of S-warfarin?

A

CYP2C9 - VERY polymorphic!

44
Q

pts who carry the variant alleles require ____ doses of warfarin to achieve an anticoagulant effect and they also have an increased risk for ____ during warfarin therapy

A

decreased

increased

45
Q

what is the molecular target for warfarin?

A

vitamin K epoxide reductase

46
Q

the gene that encodes the enzyme for vitamin K epoxide reductase complex 1 (VKORC1) how a number of _____

A

polymorphisms

47
Q

what do idiosyncratic drug reactions result from? what is a classic example?

A

interactions between the drug and a unique aspect of the physiology of the individual patient

deficiency of G6PD

48
Q

what is the normal function of G6PD

A

protects RBCs from oxidative injury

49
Q

what does diminished G6PD impair?

A
  • cell can’t form NADPH which is important for the maintenance of the reduced glutathione pool
  • this results in a decrease in the cellular detoxification of free radicals and peroxides formed within the cell
50
Q

what is the most common G6PD polymorphism? how much enzyme function is reduced with this polymorphism?

A

G6PD A

causes a 90-95% reduction of the enzyme function

51
Q

what are some other drugs that can cause oxidative stress on RBCs?

A
  • sulfonamides
  • antimalarials
  • chloramphenicol
52
Q

what happens if a pt with G6PD deficiency is exposed to

  • sulfonamides
  • antimalarials
  • chloramphenicol
  • FLAVA beans
A

may develop hemolytic anemia

53
Q

Describe malignant hyperthermia

A

-potentially fatal genetic disorder of skeletal muscle

54
Q

how is malignant hyperthermia triggered?

A

by volatile inhalation anesthetics (halothane) and depolarizing skeletal muscle relaxants (succinylcholine)

55
Q

how is malignant hyperthermia transmitted?

A

autosomal dominant - but its rare

56
Q

what is one of the main causes of death due to anesthesia?

A

malignant hyperthermia

57
Q

what are the sx/s of malignant hyperthermia syndrome?

A
  • tachycardia
  • hypertension
  • severe muscle rigidity
  • hyperthermia
  • hyperkalemia
  • acidosis
58
Q

what is the cellular mechanism behind malignant hyperthermia?

  • altered control of what?
  • defected gene?
A

altered control of Ca2+ release from the SR

defect in the ryanodine receptor gene (RYR1)

59
Q

what is the most reliable test to establish susceptibility to malignant hyperthermia?

A

the Caffeine-Halothane muscle contracture test

60
Q

how was drug genetic variation measured then and now?

A

then - urinary excretion levels
now - DNA -based tests
—Amplichip CYP450 tests

61
Q

what can the AmpliChip CYP450 test determine?

A

CYP2D6 and CYP2C19 genotypes