Pharm 1 Flashcards
Pharmacodynamics
study of how target cell responds to delivered concentration of drug.
what are the pharmacokinetic factors?
Absorption Distribution Metabolism Excretion -factors affect concentration of drug at active site as function of time.
Lipid soluble drugs:
what do they regulate?
How long to take effect?
How long do effects last?
Modify gene expression
Lag period of 30 min - hours before effect
Effect lasts hours to days
mechanism of cytokine receptor action and receptor examples
JAK / STAT kinase activity
non-intrinsic
Growth Hormone, Erythropoietin, Interferon
Transmembrane receptor activity and examples.
Intrinsic kinase activity (Tyrosine kinase, serine, guanylyl cyclase)
EGF, PDGF, ANP, TGFb
Kd = ?
Kd = [D][R]/[DR] = k(off)/k(on)
Kd is dissociation constant
What is physiological antagonism?
Agonist and antagonist work at independent sites, but have opposite effects.
What is chemical antagonism?
combination of agonist with antagonist -> inactivation of the agonist.
heparin + protamine, dimercaprol + mercuric ion
What is volume of distribution (Vd)
Relates blood concentration to total drug in body.
Vd= amount administered / concentration at t=0
Vd is the apparent volume of plasma that would have yielded the extrapolated concentration at t=0
Low Vd: drug trapped in blood
High Vd: drug distributes readily
What does Vd say about a drug whose target is w/in CNS?
Drugs w/ high lipid solubility (Vd>40) needed to cross BBB.
These drugs are eliminated via hepatic metabolism rather than renal excretion.
probenecid
competitive inhibitor of tubular secretion of penecillins - prolongs effect
Define drug metabolism
The process of converting lipophilic chemicals to hydrophobic chemicals that are readily excreted in the urine or bile.
What is the most common cause of acute liver failure in US?
Acetaminophen OD
Briefly describe Acetaminophen metabolism
2 main pathways -> nontoxic glucuronide (glucuronidation) and sulfate (sulfation)
When saturated, P450 enzymes -> toxic NAPQI
NAPQI can be detoxified by glutathione conjugation.
If saturated, Rxn w/ cell macromolecules -> liver cell death
What is MPTP?
Related to Demerol. Metabolized by MAO-B then auto-oxidized to MPP+ which interferes w/ mitochondrial respiration in DA producing cells -> Parkinson’s like symptoms in users.
What is responsible for grapefruit juice’s drug interactions?
grapefruit juice contains furanocoumarins that inhibit CYP3A4 in the intestine
What is terfenidine?
Marketed as Seldane in 1985 - prodrug metabolized to fexofenadine (allegra) by CYP3A4
Inhibition of CYP3A4 -> terfenadine accumulation -> cardiotoxicity (torsades de pointes)
What pathophysiological conditions can impact drug metabolism?
Liver disease Cardiac disease (reduced CO, reduced metabolism of "flow limited drugs) Metabolic abnormalities (obesity, infection, inflammation) -> alteration in expression of drug metabolizing enzymes.
What is Phase I drug metabolism?
Oxidation, Reduction, Hydrolysis
*expose or introduce a functional group
What is Phase II metabolism?
Conjugation of a functional group on an endogenous molecule
What is Phase III metabolism?
term not often used.
refers to transporters that “pump” xenobiotics or conjugates out of cells.
What enzymes are most responsible for metabolism of oral medications?
CYP2D6 and CYP3A4
over 50% of orally effective meds.
What organs contain the most xenobiotic metabolizing enzymes and transporters?
Liver and small intestine
What is a first pass effect?
metabolism in sm. intestine or liver limiting bioavailability of drug. Seen w/ orally administered meds.
Name 3 xenobiotic sensing receptors, their ligands and describe the concept.
Aryl hydrocarbon (Ah) receptor: aromatic and polychlorinated hydrocarbons
Pregnane X receptor (PXR): hyperforin (st. johns wort), rifampicin and others
Constitutive androstane receptor (CAR): phenobarbital and others
All receptors induce expression of metabolizing enzymes
What kind of reaction does the Cytochrome P450 family of enzymes assist with?
Oxidation
usually monooxidation with reducing equivalents from NADPH
R (Substrate) + O2 + NADH -> ROH + H20 + NADP+
Where are CYP enzymes located?
Mostly liver ER.
to some extent in most tissues, some in mitochondria (steroidogenic)
How many CYPs do humans express?
57
18 families, 43 subfamilies
12 perform most drug metabolism
3 metabolize most : CYP3A4/5 and CYP2D6 and CYP2C8/9
Describe Cytochrome P450 nomenclature
CYP = P450 superfamily followed by number, letter, number 1st number: family: >40% similarity letter: >50% similarity 2nd number: gene #
List reactions carried out by P450 enzymes
Hydroxylation: aliphatic carbon (CYP2C9)
Hydroxylation: aromatic carbon (CYP2C19) same fam/subfam
Epoxidation
Heteroatom oxidation (S- oxidation, N-oxidation)
Heteroatom dealkylation (CYP2D6)
1. hydroxylation of methoxy carbon 2. rearrangement -> loss of carbon as formaldehyde (HCHO)
What are the 3 most prevalent CYP enzymes?
CYP3A4/5
CYP2D6
CYP2C8/9
How are azo and nitro reductions usually carried out?
Via intestinal flora
Occasionally P450 or others.
Protonosil : why important and how metabolized
Discovery led to first sulfa drug (sulfanilamide) and effective antimicrobial therapies
Azo reduction of protonosil -> sulfanilamide
What are two important enzymes that participate in hydrolysis of xenobiotics?
Carboxylesterases (CE1, CE2) Epoxide hydrolases (microsomal mEH, soluble sEH) -detoxify electrophilic epoxides that could otherwise interact with proteins and nucleic acids
why are mEH and sEH often expressed in same cells as P450?
detoxify esters produced by oxidative metabolism
What are flavin monooxygenases? How are they similar to P450?
catalyze oxidation of N, S, and P heteroatoms
Similar to CYP: require NADPH and O2, but do not need additional protein
5 mammalian flavin monooxygenases.
What reaction do epoxide hydrolases carry out?
trans addition of water to alkyl epoxides and arene oxides
-> trans 1,2 dihydrodiols
In general, are conjugated metabolites more or less polar than original substrate?
Usually more polar
Usually pharmacologically inactive and easily excreted, but not always.
What are 6 major conjugation reactions?
Glucuronidation Sulfation Glutathione conjugation Acetylation Methylation Amino Acid conjugation
Describe glucouronidation
Transfer of glucuronic acid from co-factor UDP-glucuronic acid to nucleophilic heteroatom on substrate.
Enzyme: UGT
Where are UGTs located?
Microsomal (ER)
What enzyme metabolizes morphine?
UGT2B7
morphine -> morphine 6-glucuronide (active metabolite)
What is a sulfonation reaction? What enzyme?
Transfer of sulfonate group from co-factor 3-phosphoadenosine-5’-phosphosulfate (PAPS) to available substrate group
substrates: aliphatic alchol, phenol, amine, N-oxides, N-hydroxyls, NOT CARBOXYLIC ACIDS!!
Enzyme: cytosolic sulfotransferases
Describe acetylation reaction and enzyme involved
Transfer of acetyl group from acetyl co-A to amino group on amine substrate
Enzyme: N-acetyltransferases (NAT-1 and NAT-2 in cytosol)
Describe methylation reaction and enzyme involved.
Transfer of methyl group from cofactor to substrate
Cofactor: S-adenosylmethionine (SAM)
Substrates: phenol, catechol, aliphatic and aromatic amines, sulfhydryl compunds, some metals
Enzyme: methyl transferases : COMT, POMT, PNMT, HNMT, NNMT, etc.
What are first order and zero order processes?
rate = constant * [drug]^n (n=kinetic order)
First order: are directly dependent on drug concentration : elimination - constant fraction or percent lost per unit time
Zero order: not dependent on concentration - constant rate
Is normal drug metabolism zero or first order?
First order
Plasma concentrations of most drugs are below Km, so rate is dependent on concentration.
What order kinetics is alcohol metabolism?
Zero order
consumed in large quantity (g vs mg) so metabolizing system is easily saturated.
First order when when blood levels <10mg/dl.
10mg/dl -100mg dl is transitional bet. zero and first.
What is the relationship between ke and t(1/2)?
Inverse
ke=0.7/t(1/2)
ke=elimination rate constant =~amount of drug lost / unit time
How do you calculate the rate of drug output?
rate of output = X(ke)
X = amount of drug in body, ke= elimination rate constant.
BUT - X changes. X = C(Vd) C=serum conc.
So: **rate of output = C * Vd * ke
What is the steady state equation?
Input = output f(D/T) = Css * Vd * ke = Css * Vd * (0.7/t(1/2))
f(D/T)=Css * Clearance
How long does it take for a drug concentration to reach steady-state?
4t(1/2)
What is a loading dose, how is it calculated and how is it administered?
Dose needed to immediately reach steady state
Loading Dose = Css * Vd
Usually given in two doses separated by time to avoid idiosyncratic adverse responses.
What is a maintenance dose and how is it calculated?
Dose needed at regular intervals to maintain steady state.
Maintenance Dose = Clearance * Css
How is drug clearance calculated?
Clearance = Vd * ke = Vd * (0.7/t(1/2)) Clearance = (metabolism + excretion)/[drug]plasma
Clearance is additive across systems. Total body clearance = CL(liver) + CL(kidney) + CL(other)
What classes of drugs are most often involved in predictable adverse events?
Cardiovascular agents, Analgesics, hypoglycemics
accounted for 86.5% of adverse effects in quoted study.
What is an example of an direct chemical interaction?
Ca++ in dairy along with Tetracycline inhibiting tetracycline absorption
Tetracycline is a Ca++ chelator and is not absorbed w/ bound Ca++
cimetidine inhibiting CYP3A4 leading to toxic accumulation of CNS drugs is what kind of drug-drug interaction?
parmacokinetic
A patient taking OTC decongestant that causes vasoconstriction while taking a antihypertensive vasodilator is what kind of drug-drug interaction?
negative pharmacodynamic
What is meant by additivity in terms of drug-drug interactions?
2 drugs given together have additive effects
A -> increases HR by 10 bpm
B -> increases HR by 9 bpm
A+B -> increases HR by 19bpm
What is meant by potentiation in terms of drug-drug interactions?
use of two drugs together has an enhanced effect. AKA supra-additivity
A -> increased HR by 10bpm
B -> increased HR by 9bpm
A + B -> increased HR by 25 bpm
What is a subadditive effect in terms of drug-drug interactions?
2 drugs given together have a less than additive effect
A -> increased HR by 10bpm
B -> increased HR by 9 bpm
A + B -> increased HR by 13 bpm
What is synergism in terms of drug-drug interaction?
drugs work together to common effect. Often used interchangeably with additive and supra-additive
What is a quantal response?
All or nothing response.
Relief of headache. Partial relief would be non-response.
What is a therapeutic index?
TI = LD50 / ED50
Estimates separation of dose between effective and toxicity
What is the certainty of safety factor?
TD1 / ED99
Larger ratio = less toxicity
What is a polymorphism?
Variation in DNA sequence present in at least 1% of population
-some polymorphisms can impact drug metabolism and -> adverse effects and death.
6-mercaptopurine: what and what is a relevant genetic variation?
6-MP: used to treat childhood acute lymphoblastic leukemia
1: 300 kids has mutation in thiopurine methyltransferase -> inability to metabolize 6-MP -> drug accumulation -> potential fatality
* * patients always tested before being given 6-MP**
What are two ways to test for CYP2C19 poor metabolizers?
Genotype: test DNA for presence of specific polymorphisms
Phenotype: administer probe that is degraded by CYP2C19 (s-mephenytoin) and measure for metabolites
What are two methods of determining the fraction of phenotypic variability in drug metabolism attributable to genetics?
Twin studies: monozygotic vs. dizygotic
Multigenerational kindred studies: interfamily vs. intrafamily
What have twin studies revealed about drug metabolism?
Drug metabolism is highly heritable.
more drug metabolism similarities between monozygotic twins than dizygotic twins.
Poor metabolism by CYP2D6 is what kind of trait?
Autosomal recessive.
Poor metabolism by CYP2C19 is what kind of trait?
codominant
herozygotes exhibit intermediate phenotype.
What are 3 major types of polymorphisms?
SNP (single nucleotide polymorphism) - ~1 every few hundred - 1,000 bps. ~10 million in genome.
InDel (insertion/deletion) - less common than SNP
CNV (copy number variation) - caused by genetic rearrangement - duplication, deletion, inversion. occurs in ~10% of human genome
-CYP2D6 ultra-rapid metabolism
3 types of SNPs
- nonsynonymous (missense) - change -> different AA
- synonymous (sense) - change -> same AA
- may contribute to phenotypic trait: slower translation -> altered folding -> altered membrane insertion (C3435T SNP in ABCB1 transporter: P-glycoprotein efflux pump for many drugs)
- nonsense - change -> stop codon
What effects can SNPs in non-coding regions of DNA produce?
- alteration of elements that determine mRNA translatability
- promoter/ enhancer regions - may change transcription
- near exon/intron boundary - may change splicing or introduce a premature stop codon
- intergenic regions - may change DNA tertiary structure interaction with chromatin, topoisomerase, or DNA replication
What is gene linkage?
The extent to which two genotypes found at two loci are indepent of each other:
- linkage equilibrium: independence
- linkage disequilibrium: not independent, varying degrees - in complete disequilibrium, two genotypes always occur together - SNP at one point -> SNP in second Locus
What factors influence the clinical importance of gene polymorphisms?
- frequency and penetrance of allele
- narrowness of TI / sharpness of dose response curve - will small change -> toxicity or ineffectiveness of drug?
- limited availability of alternate clearance pathways
- availability of alternative drugs.
Polymorphisms in UGT1A1 cause what?
hyperbilirubinemia
UGT1A1 involved in metabolizing bilirubin. >50 polymorphisms -> inheritable hyperbilirubinemia
Crigler-Najjar Syndrome types I and II
Gilbert’s Syndrome
Acetylation polymorphism
Alteration in rate of acetylation of drugs (n-acetyltransferases)
NAT-1 and NAT-2 (2 is primarily polymorphic - metabolism of procainamide, caffeine, 4-aminobiphenyl (bladder carcinogen))
ethnic variation: 90% of others.
Coded @ 4 alleles @ 1 locus on chrom 8. 3 slow, 1 fast. Fast dominant. Must have 2 slow to display slow acetylation.
Effects of acetylation polymorphism
Slow: 1. prone to polyneuropathy during isoniazid treatment
2. likelihood of hemolytic anemia w/ sulfa drugs
3. elevated incidence of bladder cancer w/ arylamine carcinogens
Fast: need higher doses for effect.
Outline ETOH metabolism
ETOH -(ADH)-> acetaldehyde -(ALDH)-> acetic acid -> acetyl CoA
acetaldehyde is toxic - accumulation -> flushing
Alcohol Dehydrogenase polymorphism
ADH: 7 enzymes, 5 classes
faster action -> rapid accumulation of acetaldehyde -> increased flushing
ADH21 - B1B2 - 95% caucasians Vmax = 9
ADH22 - B2B2 - most Chinese, Japanese, Korean Vmax = 400
ADH2*3 - B3B3 - 15% Africans Vmax = 300
Length of time before a poison’s effects are seen is determined by:
Dose
Age
Personal habits
Genetics
What is meant by acute, subacute, and chronic exposure?
Acute : single or multiple exposures over 24 hrs
Subacute : multiple exposures 24 hrs - 3 mos
Chronic: multiple exposures over 3 mos
What are the differences between non-cumulative and cumulative poisons?
Non-cumulative: total dose doesn’t matter w/ small individual doses. Easily excreted, cleared by body. No permanent irreversible damage at low dose.
Cumulative: Total dose important. Accumulates or causes irreversible damage.
In the case of OD or poisoning is drug elimination 1st or zero order?
Zero order - some process is saturated.
** increase in dose -> disproportionate increase in blood level**
Aspirin metabolism
Acetylsalicylic acid -> salicylate by plasma esterases (t1/2 = 15min
Salicylate -> conjugated to glycine -> salicyluric acid
** this step saturates @ ~1g aspirin. Saturated w/ 3 tabs.
Very high dose : zero order
Lower dose: mixed
Low dose: first order
How do heavy metals exert toxicity?
Combine w/ key amino acid residues on proteins
- active sites
- key residues of structural proteins
Heavy metals are not metabolized - may persist for long periods.
Symptoms of acute inorganic Pb poisoning
Acute GI distress
Progression to CNS abnormalities
Often looks like pancreatitis, ulcer, appendicitis
What is the basis for lead’s toxicity?
It is a bivalent cation and interferes with Ca++ dependent processes.
Symptoms of chronic inorganic Pb poisoning
Weakness, nervousness, tremor, GI distress, anorexia, weight loss, headache
Recurrent abdominal pain, extensor muscle weakness w/o sensory deficit (wrist drop is characteristic)
What is the usual cause of organic lead poisoning? What are the symptoms?
tetraethyl or tetramethyl Pb in gasoline - highly volatile - easily inhaled, lipid soluble - easily absorbed
Acute CNS disorders, few hematologic abnormalities
rapid progression - hallucinations, insomnia, headache, irritability
How is Pb poisoning tested for?
Standard screening: FEP - free erythrocyte protoporphyrin test Also: Pb in blood 24hr urine test for Pb Abnormalities of porphyrin production: delta-ALA and Coproporphyrin III.
What is the t1/2 of Pb in blood and bone?
Blood: 1-2 mos. Steady state in 6 mos.
Bone: 20 yrs.
How does Pb distribute in the body?
First to soft tissues - kidney and liver, then to teeth, hair and bones
Pb in blood is assoc. w/ RBCs
Eventually 95% will deposit in bone
Excreted in urine and feces
What are some biochemical effects of Pb?
Inhibition of SH containing enzymes in heme biosynthesis
Inhibition of ferrochelatase (heme production)
produces hypochromic microcytic anemia
Pb poisoning treatment
Remove source
Treat seizures w/ diazepam
Treat cerebral edema with mannitol and dexamethosone
Maintain fluid and electrolyte balance
Chelation therapy: Dimercaprol w/ Edetate Calcium Disodium (CaNa2 EDTA) then D-Penecillamine for long-term
3 forms of mercury
Elemental (Hg) Inorganic salts (HgCl2) Organic mercuruials (methyl mercuric chloride)
After absorption where are the highest concentrations of Hg found?
Renal proximal tubules
w/in a few hours of absorption
What is the basis of Hg’s toxicity?
Binds SH groups (cysteinyl residues of structural proteins and enzymes, SH of glutathione)
Precipitates protein on contact -> corrosive
How is Hg excreted?
Usually through urine - most over 1 week period.
kidneys and brain retain for longer
Symptoms of acute Hg intoxication
Acute: Chest pain, SOB, metallic taste, nausea, vomiting, acute kidney damage 2nd, severe gingivitis and gastroenteritis on 3rd-4th days. Most severe cases - muscle tremor and psychopathology
Symptoms of chronic Hg intoxication
GI complaints, renal insufficiency, gingivitis, loose teeth, discolored gums, enlarged salivary glands, tremors in fingers, arms and legs, ocular changes (deposition of Hg in lens)
Personality changes - fearfulness, inability to concentrate, irritability,