term test 2 Flashcards

1
Q

Describe the steps before writing a prescription.

A
  1. Assess patient + decide if drug therapy is required
  2. Choose the best drug therapy after consideration.
  3. Choose the most appropriate route of administration.
  4. Determine appropriate dosage for the patient. (dose+frequency)
  5. Decide if follow-up or monitoring is required.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Drug administration general steps.

A
  1. Figure out your drug target (receptor) and agent.

2. Figure out how to get your desired drug to your target.

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

How is a dosage determined?

A

Follow recommended guidelines and consider patient factors.

propertied of the drug+target drug concentration from lit and use a formula

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

How do you identify the target concentration?

A

Based on published literature or product monograph
+ Patient’s sign and symptoms or a surrogate marker
(recommended drug dosage is based on a target concentration)

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

Does the plasma drug concentrations indicate the drug concentration at the site of action or the action of the drug?

A

No, not always.

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

Define dose

A

Amount of drug

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

Define dosing interval (τ)

A

Time between doses or frequency of dosing

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

Define dosage

A

Amount of drug to be administered in a defined time interval

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

Formula for calculating concentrations, define variables

A

Ct = C0e^(-ket)
or ln C = ln C0 – ket

t= time
Ct = plasma concentration at given time
C0 = initial plasma concentration 
ke = elimination rate constant
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Formula for calculating a single dose.

A

Dose = (target drug concentration)(Vd)

if rearranging for Vd use C0 instead of Cplasma

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

Assumptions made about a single dose.

A
  • all of the dose is distributed before any chance of elimination
  • use initial concentration because drug will be eliminated over time
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Formula for single dose through oral administration.

A

Dose = (C0)(Vd)/F

F - bioavailability

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

What is steady-state plasma concentration?

A

The concentration at which the rate of drug entry is equal to the rate of drug exit. Looks like a plateau on a graph
IN=OUT

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

How long does steady-state plasma concentration take to achieve?

A

5 half-lives

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

Formula for dosing for constant infusions.

A

Css = Q/Cl

steady-state plasma concentration = rate of infusion / rate of clearance

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

What is a loading dose?

A

A loading dose is an initial does given to reach steady state faster than 5 half-lives.

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

Formula for loading dose.

A

loading dose = (Css)(Vd)

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

Formula for repeated IV dosing

A

Css = Dm / (Cl)(Tm)

Dm = maintenance dose
Tm = time between doses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Formula for repeated oral dosing.

A

Css = (Dm)(F) / (Cl)(Tm) OR = (Dm)(F)/(ke)(Vd)(Tm)

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

Formula for ORAL loading dose

A

LD= (C)(Vd)/F

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

What is considered when determine the maintenance dose and interval?

A
  • available products
  • safety
  • convenience of drug administration
  • concentrations fluctuate more if interval is less frequent
  • dose is proportional to interval
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is the common maintenance interval?

A

the half-life

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

What is bioequivalence?

A

Comparable plasma concentration versus time profiles between drug products

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

What is therapeutic equivalence?

A

Comparable clinical effectiveness (and safety) between related drug products

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

How do you achieve bioequivalence?

A
  • bioavailability must be the same
  • Cmax and AUC must have no more than a 20% difference between drugs
  • confidence intervals must be within 80-125% of the original drug
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

List some factors affecting drug dosing.

A

age, body mass, sex, adherence, genetics, administration, tolerance, psychological factors, diet, cost, immune responses, etc.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What are the special populations we must consider in drug dosing?

A

Breastfeeding and pregnant women, infant/children, elderly, the very ill and those with genetic variants of drug targets, enzymes, transporters, etc.

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

What are current prescribing issues?

A
  • inappropriate prescribing (e.g wrong drug)
  • over-prescribing
  • under-prescribing
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

How do we balance and meausure safety and efficacy of dosing?

A

Efficacy is based on population average
Toxicity - individual risk

Prioritize the type, quantity, quality of evidence , and time to both

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

Define drug biotransformation.

A

Conversion of a foreign chemical from one form into another within a living organism, usually be an enzyme-catalyzed rxn

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

How are the drug metabolism system and immune system analogous?

A

Both are meant to protect the organism from foreign substances, but they can be harmful if not well-regulated and well-balanced.
ex. transformation into a more active/dangerous form

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

What are the consequences of drug biotransformation?

A
  • production of stable metabolites that have DECREASED, increased, or maintained activity
  • production of chemically reactive metabolites
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

Example of biotransformation that decreases pharmacologic activity.

A

Acetaminophen (active parent drug)

  • by UDP-glucuronosyl-transferase becomes Acetaminophen glucuronide (inactive)
  • by phenolsulfotransferase becomes Acetaminophen sulfate (inactive)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

Example of biotransformation of a pro-drug into an active metabolite (increases pharmacologic activity)

A

Cyclophosphamide (nitrogen mustards)

  • with CYP3A4 and CYP2B6, stable to travel through body
  • in tumor becomes phosphoramide mustard which is the main cytotoxic metabolite that acts to kill cancer cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

Example of biotransformation that maintains pharmacologic activity.

A

Propafenone (anti-arrhythmic drug)

  • add O becomes 5-hydroxypropafenone (anti-arrhythmic drug)
  • remove propyl in chain becomes N-Depropylpropafenone
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What happens with chemically-reactive metabolites?

A

They covalently bind to cellular macromolecules and have toxic consequences including hepatic toxicity, carcinogenic, teratogenicity, etc.

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

Example of biotransformation into chemically-reactive metabolite

A

Aromatic anticonvulsant drugs (used to treat seizures) ex.phenytoin

  • mediated by cytochrome P450
  • arene oxide is formed, covalently binds cellular macromolecule

body now recognizes drug-macromolecule complex as foreign - immune response
which can lead to rejection of own skin or liver

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

Where does biotransformation take place?

A

almost every tissue but mostly the LIVER especially for oral administration
GI tract, lungs, kidneys, skin and eyes are also important (entry/exit portals for drugs)

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

What may cause tissue-specific toxicity?

A

Enzyme expression in afflicted tissues.

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

What is the first-pass effect?

A

Biotransformation by liver and GI tract before reaching systemic circulation
- alters bioavailability

41
Q

How do we biotransform so many different foreign chemicals?

A
  1. Enzyme multiplicity

2. Broad substrate selectivity - one enzyme can accept many substrates

42
Q

What is Phase 1 of biotransformation?

A

exposes or introduces FUNCTIONAL GROUPS

  • oxidation, reduction, hydrolysis
  • drug becomes metabolite
43
Q

What is Phase 2 of biotransformation?

A

synthesis ran, conjugation

results in conjugated metabolite

44
Q

What are the possible pathways of biotransformation?

A

Drug - phase 1 - phase 2
Drug - phase 2

Excreted unchanged or after phase 1 or if water-soluble

45
Q

Example of phase 1 then phase 2 biotransformation

A

Phase 1: codeine (prodrug) with P450 becomes morphine
Phase 2: morphine with UGT becomes morphine-3-glucuronide
OR
1: heroin is hydrolyzed(deacetylated) becomes morphine
2: morphine UGT - morphine-3-glucuronide

46
Q

Example of direct conjugation biotransformation

A

morphine with UGT in phase 2 becomes morphine-3-glucuronide

47
Q

List the major enzymes involved in Phase 1.

A
  • Monooxygenases ex. cytochromes P450
  • Oxidases and Dehydrogenases
  • Reductases
  • Hydrolases
48
Q

List the major phase 2 conjugating enzymes.

A
  • **TRANSFERASES
  • UDP-glucuronosyltransferases
  • Methyltransferases
  • Sulfotransferases
49
Q

Define microsomes.

A

A subcellular fraction NOT found in intact living cells

50
Q

Who discovered cytochrome P450 and who was the first to show that it is an enzyme that metabolizes steroids and drugs?

A
  • Tsuneo Omura and Ryo Sato

- Ronald Estabrook

51
Q

What is similar between all reactions catalyzed by cytochrome P450?

A

An oxygen is being inserted.

52
Q

What is the general formula that cytochrome P450 mediates?

A

RH (substrate) + O2 + NADPH + H+ -> ROH (product) +H2O + NADP+

53
Q

Describe the process of the P450 reaction.

A
  1. P450 with ferric (oxidized) iron binds substrate
  2. NADPH- cytochrome P450 reductase transfers electron to reduce iron to ferrous state.
  3. Now ferrous iron can bind molecular oxygen.
  4. Rearrangements
  5. Transfer of second electron from P450 reductase (or b5 reductase)
  6. Oxygen now activated so oxygen-oxygen bond splits. One forms water.
  7. Other oxygen is highly reactive and abstracts hydrogen.
  8. Monooxygenase - carbon radical and hydroxyl radical recombine.
  9. Hydroxylated product is formed and ferric iron is regenerated.
54
Q

What is a rate-limiting step in P450 reactions?

A

The NADPH-cytochrome P450 reductase (POR) due to its abundance compared to P450

55
Q

Where are P450 and POR located?

A

On the cytosolic side of the membrane of ER

56
Q

How did we obtain so many P450s?

A

they evolved by duplication of ancestral P450 genes and accumulation of mutation that change structure over a long period of time.

57
Q

What are the main roles of the families within the human P450 gene superfamily?

A

1-3 are the major P450s for xenobiotic metabolism

4-51 are involved in biosynthetic reactions plus metabolism of endogenous physiologic substrates

58
Q

How are the human P450 genes classified into families and subfamilies?

A

Based on extent of similar gene sequences
ie. to be in the same family: >40% amino acid identity
same subfamily: >55%

59
Q

CYP3A4

A
  • most abundant P450 in human liver and small intestine
  • CYP3A4/5 metabolize 50% of known drugs ie. erythromycin, codeine, lovastatin, diazepam, taxol
  • important in drug-drug interactions and bioavailability
  • many substrates
  • inducible by many xenobiotics
  • CYP3A7: major P450 in human fetal liver
60
Q

CYP1A2

A
  • expressed only in the liver
  • close relative 1A1 is in tissues other than the liver
  • induced by polycyclic aromatic hydrocarbons (PAHs found in cigarette smoke) therefore CYP1A1 involved in lung cancer
  • biotransforms caffeine, acetaminophen, aromatic amine carcinogens, aflatoxin (liver carcinogen)
61
Q

CYP2C9

A
  • main 2C in liver
  • polymorphic in humans
  • catalyzes hydroxylation of tolbutamide (hypoglycemic agent), phenytoin (anticonvulsant), warfarin (anti-coagulant)
62
Q

CYP2C19

A
  • mainly in liver
  • polymorphic
  • catalyzes hydroxylation of S-mephenytoin (anticonvulsant)
63
Q

CYP2D6

A
  • highly polymorphic
  • low abundance in liver
  • BUT catalyzes biotransformation of drugs of many therapeutic categories:
    antiarrhythmics, antidepressants, beta-blockers, analgesics
64
Q

CYP2E1

A
  • originally discovered as the “microsomal ethanol oxidizing system” (MEOS)
  • induced by heavy alcohol intake
  • expressed in liver, kidney, lung and some other extra hepatic tissues
  • catalyzes biotransformation of ethanol and many other small organic molecules
    ie. halogenated alkanes;converts benzene and chloroform into hepatotoxic metabolites
65
Q

Describe the detoxification of the anticancer drug, doxorubicin, reduction formula.

A
  • mediated by NAD(P)H: quinone oxidoreductase
  • quinone
    Quinone by POR gains an electron to become Semiquinone (unstable), can be reversed in the presence of oxygen but
    produces hydroxyl radical leading to oxidative stress and cytotoxicity

detoxification pathway:
Quinone by quinone reductase gains 2 electrons to form hydroquinone

66
Q

What are epoxides?

A

chemically-reactive electrophilic molecules that can bind covalently to proteins and nucleic acids leading to cytotoxicity/cancer

67
Q

What does epoxide hydrolase do and where are its enzymes?

A

conjugates the expocide with water (acts like phase 2 enzyme)
in nearly all tissues: microsomal forms and cytosolic forms

68
Q

Describe the epoxide hydrolase formula.

A

epoxide (arene oxide) +H2O -> dihydrodiol

become s safe and not chemically reactive

69
Q

What are the enzyme, substrates and cofactor of glucuronidation(phase2)?

A

Enzyme: UDP-glucuronosyltransferase (UGT)
- microsomal (ER lumen side)
Substrates: hydroxyl, carboxyl, amine, sulfhydryl functional groups ie. morphine, acetaminophen
Cofactor: uridine diphosphate-glucuronic acid (UDPGA)
- donates a functional group

70
Q

What are the genetic deficiencies in glucuronidation?

A
  • Gilberts syndrom
  • Crigler-Najar syndrome: may lead to hyperbilirubinemia and impaired drug clearance
    possible aplastic anemia from exposure to chloramphenicol
  • newborns are deficient in glucuronidation so may get grey baby syndrome from chloramphenicol
71
Q

What are the enzyme, substrates, and cofactors involved in glutathione conjugation (phase2)?

A

Enzyme: glutathione S-transferase (GST)
- cytosolic, microsomal, mitochondrial

Substrates: electrophilic centres such as epoxides, arena oxides, nitro groups, and hydroxylamines ie. ethacrynic acid, bromobenzene

Cofactor: reduced glutathione

72
Q

What are the enzyme, substrates, and cofactors involved in sulfation (phase2)?

A

Enzyme: sulfotransferase (SULT)
- cytosolic

Substrates: phenols and aliphatic alcohols ie. acetaminophen, steroid hormones

Cofactor: 3’-phosphosadenosine-5’phospholsulfate (PAPS)

73
Q

What is an example of integration of multiple enzyme pathways?

A

Acetaminophen hepatotoxicity: high dose acetaminophen causes death of hepatocytes

74
Q

Describe the integrated acetaminophen pathways.

A

parent drug is usually undergo glucuronidation or sulfation to form nontoxic compounds.

CYP2E1 and CYP3A4 can mediate formation of NAPQI (a reactive toxic intermediate) which can potentially bind cellular macromolecules that may lead to cellular cell death

N-acetylcysteine (NAC) can be life saving as it restores depleted GSH pools. 
adds glutathione (GSH-conjugation) to NAPQI.
75
Q

What are the enzyme, substrates, and cofactors involved in acetylation (phase2)?

A

Enzyme: N-acetyltransferase (NAT)

  • cytosolic
  • fast and slow acetylators (ie. NAT2 and aromatic amine-inducer bladder cancer)

Substrates: aromatic amine and hydrazine groups ie. isoniazid, sulfamethoxazole

Cofactor: acetyl-CoA

76
Q

What are the enzyme, substrates, and cofactors involved in methylation (phase 2)?

A

Enzyme: methyltransferase (MT)
- mostly cytosolic

Substrates: phenol, catechol, aliphatic and aromatic amine, N-hetericyclic, sulfhydryl ie. levodopa, 6- mercaptopurine

Cofactor: S-adenosylmethionine (SAM)

77
Q

What are the potential effects of induction or inhibition of drug-metabolizing enzymes?

A
  1. Alter the efficacy of a therapeutic agent

2. Alter the toxicity of drugs or environmental chemicals

78
Q

Are P450s inducible?

A

Majority are non-inducible or weakly

2E1 and 1A2 are strongly inducible

79
Q

What are some chemicals that activate the aryl hydrocarbon receptor and are inducers of drug-metabolizing enzymes?

A

TCDD (environmental toxicant), MC, benzo a pyrene (carcinogen), and carbazole (cabbage family)

80
Q

How does benzo[a]pyrene induce its own biotransformation?

A

Prior exposure will increase the levels of certain drug-metabolizing enzymes, which increases the capacity of that organism to biotransform benzo[a]pyrene

81
Q

What induces CYP1A1 activity?

A

Cigarette smoke

82
Q

At what stages are P450 enzymes regulated?

A

Mostly in gene transcription

but also during processing, mRNA stabilization, translation and enzyme stabilization/ degradation

83
Q

Describe the aryl hydrocarbon receptor (AHR) response pathway.

A

AHR is in the cytosol in a complex with HSP90 and AIP
inducing agent (TCDD) diffuses through membrane and directly binds to AHR, making it more active
chaperone proteins are lost
AHR-TCDD enter nucleus and find AH receptor nuclear translocator forming a DNA binding factor
Leads to upregulation of CYP1A1,1A2,1B1,2S1 and phase 2 enzymes, GST and UGT

homeostatic mechanism -> enhances biotransformation to eliminate drug

84
Q

Describe the relationship between St. John wort and Indlnavlr.

A

St. John’s wort can speed up metabolism of Indlnavlr, which compromises its therapeutic effect

85
Q

Describe the pathway of pregnane X receptor.

A

pregnane x receptor resides in cytosol

inducer ie. hyperforin diffuses into cytosol binds PXR, activating it and removing chaperone proteins
they enter the nucleus and bind to RXR (its nuclear dimerization partner) (retinoid X receptor)
then bind to specific DNA sequences
ex. may induce CYP3A4 which increases metabolism of many drugs

86
Q

What is a therapeutic benefit of inducing drug-metabolizing enzymes?

A

treatment: treating neonates with hyperbilirubinemia with phenobarbital
action: induces UDP-glucuronosyltransferase
goal: increase bilirubin conjugation and clearance by elevating the rate of glucuronidation

87
Q

What is a possible prophylactic benefit of inducing drug-metabolizing enzymes?

A

induce CYP1A2 in liver of premenopausal women with high risk for breast cancer by introducing indole-3-carbinol
goal: reduce risk of breast cancer by reducing circulating levels of 17beta-estradiol (biotransformed by CYP1A2

88
Q

Describe the metabolism of estradiol to catechol estrogens.

A

AHR induces CYP1A1, 1A2, and 1B1

protective pathway in breast cancer:
17beta-estradiol form a less active estrogen (2-OH) through mediation of 1A1-1A2 dimer

but if mediated by CYP1B1 4-OH is formed which is a DNA damaging agent

89
Q

What is a protection mechanism of the potent carcinogen, DMBA?

A

pretreatment with the pesticide, DDT, enhances first-pass clearance of DMBA

90
Q

What is the relationship between the antihistamine, seldane ie.terfenadine, and the antifungal ketoconazole?

A

When taken at the same, it results in dangerous heart rhythm abnormalities

91
Q

Describe the mechanism of terfenadine.

A

blocks cardiac hERG K+ channels and results in a long Q-T interval - can be fatal

prodrug that relies on metabolism of CYP3A4 but drugs such as erythromycin and ketoconazole are inhibitors of CYP3A4 so no anti-histaminic benefits

92
Q

What is the metabolic bottleneck?

A

when too many compounds compete for the same enzyme

93
Q

What is an example of a dietary agent?

A

Grapefruit - inhibits metabolism of many medications especially those whose main route of metabolism is via CYP3A4

94
Q

How do we determine which human enzymes metabolize a particular drug?

A
  1. in vitro tests with human tissues
  2. in vitro tests with an individual cloned human enzyme expressed in a heterologous system (note:p450s also need reductase)`
95
Q

What is the XenoTech approach to matching substrate to enzyme?

A
  1. pure enzyme expressed from cDNA
  2. correlation analysis in liver microsome bank
  3. inhibition of the candidate enzyme with selective antibodies or selective chemical inhibitors
96
Q

How can you ensure correct enzyme to substrate matching if in vitro and not in vivo?

A
  • appropriate substrate concentration was tested in vitro
  • all competing pathways are considered
  • individual enzyme variation is considered

BUT in vitro only ever shows potential

97
Q

Which enzymes would pharmaceutical companies prefer are NOT the primary route of metabolism for a new drug?

A
  1. CYP3A4 - too much competition so increased potential drug-drug interactions
  2. CYP2D6 - high degree of genetic polymorphism leads to some individuals being “rapid” metabolizers while others are slow
98
Q

How is caffeine used as a probe drug?

A

Safe for intake and can tell you about the levels and activity of multiple drug-metabolizing enzymes.
- CYP1A2, N-acetyltransferase (NAT2) and XO