Lecture 5: Intro to Biotransformation, Pharmacogenomics, and Clinical Trials Flashcards

1
Q

What is Biotransformation?

A

chemical modification of lipophilic, unionized, or large compounds to terminate their actions and facilitate elimination

  • polar, small molecular volume xenobiotics eliminated through renal excretion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What is a Prodrug?

A

an inactive drug that undergoes biotransformation to become an active drug

  • may have a protective functional group that gets removed, allowing the drug to become active
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Besides the liver, what are 4 other locations in the body that have considerable biotransforming activity?

A

GI tract, lungs, skin, and kidneys

  • express smaller quantities of the the enzymes required than the liver
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What is the First-Pass Effect?

A
  • oral drugs are absorbed in the small intestine and transported to the liver via hepatic portal system for metabolism
  • parenteral routes do not undergo first-pass biotransformation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What happens to estrogen and morphine when undergoing First-Pass metabolism?

A
  • GI flora increases the bioavailability of estrogen by increasing enterohepatic cycling
  • only 25% of oral morphine doses are bioavailable, making parenteral administration much more desirable
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What happens during Phase I of biotransformation? (What does it do, what causes it, and where?)

A
  • catabolic rxn that unmasks a functional group on the drug (oxidation, reduction, hydrolysis)
  • rxns carried out by mixed function oxidases (MFOs)
    • CYP, FMO, mEH/sEH
  • rxns occur in lipophilic ER membranes in liver
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What happens during Phase II of biotransformation? (What does it do, what causes it, and where?)

A
  • anabolic rxns that form a conjugate of the phase I product (polar w/high molecular weight)
  • dependent on glucuronic, sulfuric, acetic, and amino acids
  • takes place in the liver and is a faster than Phase I rxns
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What is the most abundant cytochrome P450 enzyme?

A

CYP3A4

  • involved in metabolism of 50% of clinically used drugs
  • uses oxygen and hydrogen from NADPH to carry out oxidation of substrates
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What are the 4 other major cytochrome P450 enzymes?

A

CYP1A2, CYP2A6, CYP2D6, CYP2E1

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

What is Succinylcholine and what enzyme is used to metabolize it?

A
  • depolarizing neuromuscular blocking drug

- genetic defect in pseudocholinesterase = metabolize at 50% the normal rate

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

What is the Slow Acetylator phenotype and who does it most commonly affect?

A
  • autosomal recessive = dec. N-acetyltransferase lvls
  • isoniazid, hyralazine, caffeine, other amines metabolized at slower rates = hepatotoxicity
  • 50% of US and 83% of French populations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What are 5 common P450 inducers? (PEBRP)

A

phenytoin, ethanol (chronic), benzo[a]pyrene, rifampin, phenobarbitol

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

What is the effect of Grapefruit juice on P450?

A
  • irreversibly inhibits CYP3A4 and alters the bioavailability of drugs taken orally
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What is the effect of allopurinol and mercaptopurine on xanthine oxidase?

A
  • allopurinol treats excess uric acid and inhibits xanthine oxidase
  • xanthine oxidase metabolizes mercaptopurine (cancer treatment drug), so coadministration with allopurinol increases toxic effects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

How can acetaminophen become hepatotoxic?

A
  • normally 95% undergoes glucuronidation while 5% is biotransformed by P450s
  • when excess intake occurs, hepatic GSH is depleted faster than can be regenerated, so more toxic metabolites accumulate = hepatotoxicity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Genetic variation of CYP2D6 and Codeine/Morphine

A
  • codeine (prodrug) is converted to active morphine, which binds to mu-receptors in CNS and is used to manage mild-moderately severe pain
  • people with polymorphisms in CYP2D6 may experience insufficient pain relief OR side effects (drowsiness and respiratory depression)
17
Q

Genetic variation of CYP2C19 and Clopidogrel

A
  • clopidogrel is acidic antiplatelet prodrug that is converted to active thiol metabolite (only 15% of dose) that causes antiplatelet activity
  • people with polymorphisms in CYP2C19 (reduced function) are at an inc. risk for adverse cardiovascular effects (acute coronary syndrome)
18
Q

Genetic variation of UGT1A1 and Irinotecan

A
  • irinotecan is a topoisomerase I inhibitor prodrug (first line chemotherapy for colon/rectum carcioma)
  • metabolized to SN-38 metabolite (TOXIC)
  • UGT1A16 and UGT1A128 polymorphisms are at inc. risk of neutropenia and diarrhea due to SN-38 buildup
19
Q

Genetic variation of Thiopurine S-Methyltransferase (TPMT) and azathioprine, 6-mercaptopurine (6-MP), and 6-thioguanine (6-TG)

A

`- normal enzyme attaches methyl group to aromatic/heterocyclic sulfhydrl compounds (deactivates)

  • 6-MP/6-TG are activated by HGRPTase to form TGNs (thioguanine nucleotides) that are anticancer agents and have bone marrow toxicity
  • TPMT and xanthine oxidase normally inactivate 6-MP and 6-TG, but polymorphisms can lead to a buildup of toxic products
20
Q

Genetic variation of G6PD and Rasburicase

A
  • G6PD exclusive source of NADPH and glutathione in RBCs
  • Rasburicase manages high uric acid lvls in cancer pts on chemotherapy; converts uric acid to allantoin
  • hydrogen peroxide is a biproduct that can be reduced by glutathione, so use is NOT permitted in pts. with a G6PD deficiency (African/Mediterranean)
21
Q

Genetic variation of OATP1B1 and simvastatins

A
  • transporter responsible for uptake of weak acid drugs (statins) and bilirubin
  • polymorphism rs4149056 in SLCO1B1 genes can cause low lvls of OATP1B1
  • individuals with low lvls are at increased risk of toxicity to skeletal muscle if taking simvastatins to reduce serum lipids (prevent cardiac events)
22
Q

CYP2C9 alleles and S-warfarin

A
  • CYP2C92 and CYP2C93 are alleles that lead to reduced metabolism of S-warfarin and are more common in European populations
  • CYP2C95/6/8/11 alleles are more prevalent in African populations
23
Q

VKORC1 allele and warfarin

A
  • target of anticoagulant warfarin and a key enzyme in vitamin K recycling process
  • the most important polymorphism leads to inc. sensitivity to warfarin and occurs most frequently in Asians and least frequently in Africans (VKORC1-1639G>A polymorphism)
24
Q

What is a Lead Compound?

A
  • chemical compound that has pharmacological/biological activity and a structure thats used as a starting point for chemical modifications to improve potency, selectivity, pharmacokinetic parameters
25
Q

What is a Leading Candidate?

A
  • lead compound that has undergone modification after animal and cell screenings
  • will undergo preclinical and toxicity testing before human evaluation begins
26
Q

Why is preclinical testing preformed?

A

to evaluate a leading compounds safety and toxicity

  • identifies human toxicities, designs tests to further study toxic mechanisms, and predicts the most relevant ones to be monitored in clinical trials
  • want to test compound in a cellular system that best represents the disease state it will be used on
27
Q

What is the No-Effect dose, the Minimum Lethal dose (LDmin), and the Median Lethal dose (LD50)?

A

NE = max dose at which toxic effect is NOT seen
- lower than threshold of harmful effect

Min Leth = smallest dose that kills an animal

Med Leth = does that kills 50% of the animals

28
Q

What is Phase 0 of clinical testing?

A
  • “microdosing” or subpharmaceutical doses given to human volunteers
  • can offer supportive/alternative data that allows selection of suitable drug candidates when animal and in vitro testing isn’t reliable
  • financially advantageous
29
Q

What is Phase I of clinical testing?

A
  • determines if humans and animals show different responses to investigational drug and determine limits of safe dosage range
  • 25-50 healthy volunteers, though in cases of significant toxicity, subjects with disease state may be included (AIDS/chemo)
  • performed in inpatient clinic
30
Q

What data is reported from Phase I testing? (H/A/M)

A

absorption, half-life, and metabolism

31
Q

What is Phase II of clinical testing?

A
  • 100-200 pts with the target disease are used
  • usually single-blind design; performed in hospitals
  • drug failure typically occurs during this phase
32
Q

What data is reported from Phase II testing? (E/D/T)

A

efficacy, dosing requirements, and toxicities

33
Q

What is Phase III of clinical testing?

A
  • 300-3000 or more pts with target disease are used
  • usually cross-over or double-blind design
  • determine overall benefit-risk relationship of drug
  • most expensive phase of clinical testing
34
Q

What phase would the FDA grant approval of a drug being used to treat serious diseases and life-threatening diseases?

A
  • granted approval during Phase III trials if used to treat serious diseases
  • granted during Phase II trials in a controlled-market setting if used to treat life-threatening diseases
35
Q

What is Phase IV of clinical testing?

A
  • only begins after approval to market of new drug
  • continues to monitor safety of the new drug
  • feedback essential for drugs that have side effects occuring in every 1 of 10,000 patients
  • no fixed duration