• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/81

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

81 Cards in this Set

  • Front
  • Back
Why is drug biotransformation necessary?

Why is renal secretion of drugs slow if they have not be biotransformed?
Renal excretion plays a pivotal role in terminating the biologic activity of some drugs, particularly those that have small molecular volumes or possess polar characteristics, such as functional groups that are fully ionized at physiologic pH. However, many drugs do not possess such physicochemical properties. Pharmacologically active organic molecules tend to be lipophilic and remain un-ionized or only partially ionized at physiologic pH; these are readily reabsorbed from the glomerular filtrate in the nephron. Certain lipophilic compounds are often strongly bound to plasma proteins and may not be readily filtered at the glomerulus. Consequently, most drugs would have a prolonged duration of action if termination of their action depended solely on renal excretion.
What is the usually characteristic of drugs as far as solubility, ph, and whether then ionized or deionized?
Pharmacologically active organic molecules tend to be lipophilic and remain un-ionized or only partially ionized at physiologic pH; these are readily reabsorbed from the glomerular filtrate in the nephron. Certain lipophilic compounds are often strongly bound to plasma proteins and may not be readily filtered at the glomerulus. Consequently, most drugs would have a prolonged duration of action if termination of their action depended solely on renal excretion.
What results in regards to the a drug after it has been metabolized?
In general, lipophilic xenobiotics are transformed to more polar and hence more readily excreted products. The role that metabolism plays in the inactivation of lipid-soluble drugs can be quite dramatic. For example, lipophilic barbiturates such as thiopental and pentobarbital would have extremely long half-lives if it were not for their metabolic conversion to more water-soluble compounds.
What are the two major categories of biotransformation of drugs?
Most metabolic biotransformations occur at some point between absorption of the drug into the general circulation and its renal elimination. A few transformations occur in the intestinal lumen or intestinal wall. In general, all of these reactions can be assigned to one of two major categories called phase I and phase II reactions
What usually occurs in Phase I reactions?
Phase I reactions usually convert the parent drug to a more polar metabolite by introducing or unmasking a functional group (-OH, -NH2, -SH).

Often these metabolites are inactive, although in some instances activity is only modified or even enhanced.
What usually occurs in Phase II reactions?
However, many phase I products are not eliminated rapidly and undergo a subsequent reaction in which an endogenous substrate such as glucuronic acid, sulfuric acid, acetic acid, or an amino acid combines with the newly incorporated functional group to form a highly polar conjugate. Such conjugation or synthetic reactions are the hallmarks of phase II metabolism.
What needs to be characteristic of a Phase I metabolite to be excreted without further Phase II processing?
If phase I metabolites are sufficiently polar, they may be readily excreted.
With what endogenous substrates do Phase II reactions occur?
glucuronic acid, sulfuric acid, acetic acid, or an amino acid combines with the newly incorporated functional group to form a highly polar conjugate. Such conjugation or synthetic reactions are the hallmarks of phase II metabolism.
Do phase II reactions always happen before Phase I reactions?
For example, the hydrazide moiety of isoniazid is known to form an N-acetyl conjugate in a phase II reaction. This conjugate is then a substrate for a phase I type reaction, namely, hydrolysis to isonicotinic acid

Thus, phase II reactions may actually precede phase I reactions.
Where do drug biotransformations mostly occur?
Although every tissue has some ability to metabolize drugs, the liver is the principal organ of drug metabolism. Other tissues that display considerable activity include the gastrointestinal tract, the lungs, the skin, the kidneys, and the brain. A
What part of the digestions system can biotransformations aslo occur?
Other tissues that display considerable activity include the gastrointestinal tract, the lungs, the skin, the kidneys, and the brain.
Name some drugs metabolized more in the the small intestine than the liver.
Some orally administered drugs (eg, clonazepam, chlorpromazine, cyclosporine) are more extensively metabolized in the intestine than in the liver, whereas others (eg, midazolam) undergo significant (50%) intestinal metabolism. Thus, intestinal metabolism can contribute to the overall first-pass effect, and individuals with compromised liver function may rely increasingly on such intestinal metabolism for drug elimination.
What is the first-pass affect and how does it affect bioavailability of the drug?
After oral administration, many drugs (eg, isoproterenol, meperidine, pentazocine, morphine) are absorbed intact from the small intestine and transported first via the portal system to the liver, where they undergo extensive metabolism. This process is called the first-pass effect .
How are most biotransformations catalyzed?

Where are the enzymes located?
Although drug biotransformation in vivo can occur by spontaneous, noncatalyzed chemical reactions, most transformations are catalyzed by specific cellular enzymes.

At the subcellular level, these enzymes may be located in the endoplasmic reticulum (ER), mitochondria, cytosol, lysosomes, or even the nuclear envelope or plasma membrane.
In what part of the cell are many drug metabolizing enzymes located?
Many drug-metabolizing enzymes are located in the lipophilic endoplasmic reticulum membranes of the liver and other tissues.
What are Microsomes? From which part of the cell are they made from?
MICROSOMAL MIXED FUNCTION OXIDASE SYSTEM & PHASE I REACTIONS
Many drug-metabolizing enzymes are located in the lipophilic endoplasmic reticulum membranes of the liver and other tissues. When these lamellar membranes are isolated by homogenization and fractionation of the cell, they re-form into vesicles called microsomes. Microsomes retain most of the morphologic and functional characteristics of the intact membranes, including the rough and smooth surface features of the rough (ribosome-studded) and smooth (no ribosomes) endoplasmic reticulum.
What do rough microsomes generally do?
Whereas the rough microsomes tend to be dedicated to protein synthesis.
What do smooth microsomes do?
the smooth microsomes are relatively rich in enzymes responsible for oxidative drug metabolism.
What important enzymes do Smooth Microsomes contain?
the smooth microsomes are relatively rich in enzymes responsible for oxidative drug metabolism. In particular, they contain the important class of enzymes known as the mixed function oxidases (MFOs), or monooxygenases.
What is required for the activity of Mixed function oxidazes (Monooxygenases)?
The activity of these enzymes requires both a reducing agent (nicotinamide adenine dinucleotide phosphate [NADPH]) and molecular oxygen; in a typical reaction, one molecule of oxygen is consumed (reduced) per substrate molecule, with one oxygen atom appearing in the product and the other in the form of water.
What is Cytochrome P450?
The second microsomal enzyme is a hemoprotein called cytochrome P450, which serves as the terminal oxidase.
In terms of drug metabolizing, act in make of what is considered the rate determining step?
The relative abundance of P450s, compared with that of the reductase in the liver, contributes to making P450 heme reduction a rate-limiting step in hepatic drug oxidations.
What is the substrate specificity of P450s?
The potent oxidizing properties of this activated oxygen permit oxidation of a large number of substrates. Substrate specificity is very low for this enzyme complex. High lipid solubility is the only common structural feature of the wide variety of structurally unrelated drugs and chemicals that serve as substrates in this system
How fast are P450 reactions?
However, compared with many other enzymes including phase II enzymes, P450s are remarkably sluggish catalysts, and their drug biotransformation reactions are slow.
Which form of the P450 enzyme is most important?

What is the second most important one?
Of these, CYP1A2, CYP2A6, CYP2B6, CYP2C9, CYP2D6, CYP2E1, and CYP3A4 appear to be the most important forms, accounting for approximately, 15%, 4%, 1%, 20%, 5%, 10%, and 30%, respectively, of the total human liver P450 content. Together, they are responsible for catalyzing the bulk of the hepatic drug and xenobiotic metabolism
Describe the oxidative cycle of P450 and a drug?
Microsomal drug oxidations require P450, P450 reductase, NADPH, and molecular oxygen.

Briefly, oxidized (Fe3+) P450 combines with a drug substrate to form a binary complex (step 1). NADPH donates an electron to the flavoprotein P450 reductase, which in turn reduces the oxidized P450-drug complex (step 2). A second electron is introduced from NADPH via the same P450 reductase, which serves to reduce molecular oxygen and to form an "activated oxygen"-P450-substrate complex (step 3). This complex in turn transfers activated oxygen to the drug substrate to form the oxidized product (step 4).
What isoform of P450 accounts for nearly 50% of prescription drug metabolism?
It is noteworthy that CYP3A4 alone is responsible for the metabolism of over 50% of the prescription drugs metabolized by the liver. The involvement of individual P450s in the metabolism of a given drug may be screened in vitro by means of selective functional markers, selective chemical P450 inhibitors, and P450 antibodies. In vivo, such screening may be accomplished by means of relatively selective noninvasive markers, which include breath tests or urinary analyses of specific metabolites after administration of a P450-selective substrate probe.
What drugs go through oxidiation via P450 Hydroxylation ?
Acetanilide, propranolol, phenobarbital, phenytoin, phenylbutazone, amphetamine, warfarin, 17-ethinyl estradiol, naphthalene, benzpyrene
What drugs go through oxidiation via P450 N-dealkylation?
Morphine, ethylmorphine, benzphetamine, aminopyrine, caffeine, theophylline
What drugs go through oxidiation via P450 O-dealkylation?
Codeine, p-nitroanisole
What drugs go through oxidiation via P450 S-Dealklation?
6-Methylthiopurine, methitural
What drug goes through Amine Oxidation independent of P450?
Phenylethylamine, epinephrine
What drug goes through dehydrogenation independent of P450?
Ethanol (Hence why clearance is based on time scale)
What drugs are reduced?
Nitrobenzene, chloramphenicol, clonazepam, dantrolene
What drugs are hydrolyzed by esterification?
Procaine, succinylcholine, aspirin, clofibrate, methylphenidate
What drugs are hydrolyzed via Amide reaction?
Procainamide, lidocaine, indomethacin
What is enzyme Induction?

How does it occur?

Is quicker metabolism the only effect this can have?
Some of the chemically dissimilar P450 substrate drugs, on repeated administration, induce P450 expression by enhancing the rate of its synthesis or reducing its rate of degradation (Table 4–2). Induction results in accelerated substrate metabolism and usually in a decrease in the pharmacologic action of the inducer and also of coadministered drugs. However, in the case of drugs metabolically transformed to reactive metabolites, enzyme induction may exacerbate metabolite-mediated toxicity.
How can environmental factors affect P450 induction?

What are some factors?
Environmental chemicals and pollutants are also capable of inducing P450 enzymes. As previously noted, exposure to benzo[a]pyrene and other polycyclic aromatic hydrocarbons, which are present in tobacco smoke, charcoal-broiled meat, and other organic pyrolysis products, is known to induce CYP1A enzymes and to alter the rates of drug metabolism. Other environmental chemicals known to induce specific P450s include the polychlorinated biphenyls (PCBs), which were once used widely in industry as insulating materials and plasticizers, and 2,3,7,8-tetrachlorodibenzo-p-dioxin (dioxin, TCDD), a trace byproduct of the chemical synthesis of the defoliant 2,4,5-T
What is mechanism that causes P450 enzyme inductions to create more of it?
Increased P450 synthesis requires enhanced transcription and translation along with increased synthesis of heme, its prosthetic cofactor. A cytoplasmic receptor (termed AhR) for polycyclic aromatic hydrocarbons (eg, benzo[a]pyrene, dioxin) has been identified. The translocation of the inducer-receptor complex into the nucleus, followed by ligand-induced dimerization with Arnt, a closely related nuclear protein, leads to subsequent activation of regulatory elements of CYP1A genes, resulting in their induction. This is also the mechanism of CYP1A induction by cruciferous vegetables, and the proton pump inhibitor, omeprazole. A pregnane X receptor (PXR), a member of the steroid-retinoid-thyroid hormone receptor family, has recently been shown to mediate CYP3A induction by various chemicals (dexamethasone, rifampin, mifepristone, phenobarbital, atorvastatin, and hyperforin, a constituent of St. John's wort) in the liver and intestinal mucosa. A similar receptor, the constitutive androstane receptor (CAR) has been identified for the relatively large and structurally diverse phenobarbital class of inducers of CYP2B6, CYP2C9 and CYP3A4. Peroxisome proliferator receptor (PPAR) is yet another nuclear receptor highly expressed in liver and kidneys, which uses lipid-lowering drugs (eg, fenofibrate and gemfibrozil) as ligands. Consistent with its major role in the regulation of fatty acid metabolism, PPAR mediates the induction of CYP4A enzymes, responsible for metabolism of fatty acids such as arachidonic acid and its physiologically relevant derivatives. It is noteworthy, that on binding of its particular ligand, PXR, CAR and PPAR, each forms heterodimers with another nuclear receptor, the retinoid X-receptor (RXR). This heterodimer in turn binds to response elements within the promoter regions of specific P450 genes to induce gene expression.
What changes to the substrate may induce P450 induction?
P450 enzymes may also be induced by substrate stabilization, eg, decreased degradation, as is the case with troleandomycin- or clotrimazole-mediated induction of CYP3A enzymes, the ethanol-mediated induction of CYP2E1, and the isosafrole-mediated induction of CYP1A2.
Which substrates does CY1A2 work on?

What induces it? Inhibits it?
Acetaminophen, antipyrine, caffeine, clomipramine, phenacetin, tacrine, tamoxifen, theophylline, warfarin Smoking, charcoal-broiled foods, cruciferous vegetables, omeprazole

Galangin, furafylline, fluvoxamine
Which substrates does CY2A6 work on?

What induces it? Inhibits it?
Coumarin, tobacco nitrosamines, nicotine (to cotinine and 2'-hydroxynicotine)

Rifampin, phenobarbital


Tranylcypromine, menthofuran, methoxsalen
Which substrates does CY2B6 work on?

What induces it? Inhibits it?
Artemisinin, bupropion, cyclophosphamide, efavirenz, ifosfamide, ketamine, S-mephobarbital, S-mephenytoin (N-demethylation to nirvanol), methadone, nevirapine, propofol, selegiline, sertraline, ticlopidine

Phenobarbital, cyclophosphamide


Ticlopidine, clopidogrel
Which substrates does CY2C8 work on?

What induces it? Inhibits it?
Taxol, all-trans-retinoic acid

Rifampin, barbiturates

Trimethoprim
Which substrates does CY2C9 work on?

What induces it? Inhibits it?
Celecoxib, flurbiprofen, hexobarbital, ibuprofen, losartan, phenytoin, tolbutamide, trimethadione, sulfaphenazole, S-warfarin, ticrynafen

Barbiturates, rifampin

Tienilic acid, sulfaphenazole
Which substrates does CY2C18 work on?

What induces it? Inhibits it?
Tolbutamide, phenytoin

Phenobarbital
Which substrates does CY2C19 work on?

What induces it? Inhibits it?
Diazepam, S-mephenytoin, naproxen, nirvanol, omeprazole, propranolol Barbiturates, rifampin N3-benzylnirvanol, N3-benzylphenobarbital, fluconazole
Which substrates does CY2D6 work on?

What induces it? Inhibits it?
Acetaminophen, chlorzoxazone, enflurane, halothane, ethanol (a minor pathway)

Ethanol, isoniazid

4-Methylpyrazole, disulfiram
Which substrates does CY3A4 work on?

What induces it? Inhibits it?

***Know this one***
Acetaminophen, alfentanil, amiodarone, astemizole, cisapride, cocaine, cortisol, cyclosporine, dapsone, diazepam, dihydroergotamine, dihydropyridines, diltiazem, erythromycin, ethinyl estradiol, gestodene, indinavir, lidocaine, lovastatin, macrolides, methadone, miconazole, midazolam, mifepristone, nifedipine, paclitaxel, progesterone, quinidine, rapamycin, ritonavir, saquinavir, spironolactone, sulfamethoxazole, sufentanil, tacrolimus, tamoxifen, terfenadine, testosterone, tetrahydrocannabinol, triazolam, troleandomycin, verapamil

Barbiturates, carbamazepine, glucocorticoids, macrolide antibiotics, pioglitazone, phenytoin, rifampin, St. John's wort


Azamulin, diltiazem, erythromycin, fluconazole, grapefruit juice (furanocoumarins), itraconazole, ketoconazole, ritonavir, troleandomycin

CYP3A5 has similar substrate and inhibitor profiles, but is generally less active than 3A4.
Explain how P450 Enzyme Inhibition works?
Certain drug substrates inhibit cytochrome P450 enzyme activity (Table 4–2). Imidazole-containing drugs such as cimetidine and ketoconazole bind tightly to the P450 heme iron and effectively reduce the metabolism of endogenous substrates (eg, testosterone) or other coadministered drugs through competitive inhibition. Macrolide antibiotics such as troleandomycin, erythromycin, and erythromycin derivatives are metabolized, apparently by CYP3A, to metabolites that complex the cytochrome P450 heme iron and render it catalytically inactive. Another compound that acts through this mechanism is the inhibitor proadifen (SKF-525-A, used in research), which binds tightly to the heme iron and quasi-irreversibly inactivates the enzyme, thereby inhibiting the metabolism of potential substrates.

Some substrates irreversibly inhibit P450s via covalent interaction of a metabolically generated reactive intermediate that may react with the P450 apoprotein or heme moiety or even cause the heme to fragment and irreversibly modify the apoprotein. The antibiotic chloramphenicol is metabolized by CYP2B1 to a species that modifies the P450 protein and thus also inactivates the enzyme. A growing list of such suicide inhibitors —inactivators that attack the heme or the protein moiety—includes certain steroids (ethinyl estradiol, norethindrone, and spironolactone); fluroxene; allobarbital; the analgesic sedatives allylisopropylacetylurea, diethylpentenamide, and ethchlorvynol; carbon disulfide; grapefruit furanocoumarins; selegiline; phencyclidine; ticlopidine and clopidogrel; ritonavir, and propylthiouracil. On the other hand, the barbiturate secobarbital is found to inactivate CYP2B1 by modification of both its heme and protein moieties. Other metabolically activated drugs whose P450 inactivation mechanism is not fully elucidated are mifepristone, troglitazone, raloxifene, and tamoxifen.
What are suicide inhibitors?
Some substrates irreversibly inhibit P450s via covalent interaction of a metabolically generated reactive intermediate that may react with the P450 apoprotein or heme moiety or even cause the heme to fragment and irreversibly modify the apoprotein.

The antibiotic chloramphenicol is metabolized by CYP2B1 to a species that modifies the P450 protein and thus also inactivates the enzyme.

A growing list of such suicide inhibitors - inactivators that attack the heme or the protein moiety - includes certain steroids (ethinyl estradiol, norethindrone, and spironolactone); fluroxene; allobarbital; the analgesic sedatives allylisopropylacetylurea, diethylpentenamide, and ethchlorvynol; carbon disulfide; grapefruit furanocoumarins; selegiline; phencyclidine; ticlopidine and clopidogrel; ritonavir, and propylthiouracil.

On the other hand, the barbiturate secobarbital is found to inactivate CYP2B1 by modification of both its heme and protein moieties.

Other metabolically activated drugs whose P450 inactivation mechanism is not fully elucidated are mifepristone, troglitazone, raloxifene, and tamoxifen.
What is product of Phase II reactions?
Parent drugs or their phase I metabolites that contain suitable chemical groups often undergo coupling or conjugation reactions with an endogenous substance to yield drug conjugates
What is most dominant Phase II enzyme?
Conjugate formation involves high-energy intermediates and specific transfer enzymes. Such enzymes (transferases ) may be located in microsomes or in the cytosol. Of these, uridine 5'-diphosphate [UDP]-glucuronosyl transferases [UGTs ] are the most dominant enzymes.

These microsomal enzymes catalyze the coupling of an activated endogenous substance (such as the UDP derivative of glucuronic acid) with a drug (or endogenous compound such as bilirubin, the end product of heme metabolism)
What enzymes are used for Glucuronidation?

List Endogenous Reactant, Location of enzyme and type of substrates it works on and names of some drugs.
UDP glucuronic acid

UDP glucuronosyltransferase (microsomes)

Phenols, alcohols, carboxylic acids, hydroxylamines, sulfonamides


Nitrophenol, morphine, acetaminophen, diazepam, N-hydroxydapsone, sulfathiazole, meprobamate, digitoxin, digoxin
What enzymes are used for Acetylation?

List Endogenous Reactant, Location of enzyme and type of substrates it works on and names of some drugs.
Acetyl-CoA

N-Acetyltransferase (cytosol)

Amines

Sulfonamides, isoniazid, clonazepam, dapsone, mescaline
What enzymes are used for Glutathione conjungation?

List Endogenous Reactant, Location of enzyme and type of substrates it works on and names of some drugs.
Glutathione (GSH)

GSH-S-transferase (cytosol, microsomes)

Epoxides, arene oxides, nitro groups, hydroxylamines

Acetaminophen, ethacrynic acid, bromobenzene
What enzymes are used for Sulfation?

List Endogenous Reactant, Location of enzyme and type of substrates it works on and names of some drugs.
Phosphoadenosyl phosphosulfate

Sulfotransferase (cytosol)

Phenols, alcohols, aromatic amines Estrone, aniline, phenol, 3-hydroxy-coumarin, acetaminophen, methyldopa

Acyl-CoA derivatives of carboxylic acids

Salicylic acid, benzoic acid, nicotinic acid, cinnamic acid, cholic acid, deoxycholic acid
What enzymes are used for Methylation?

List Endogenous Reactant, Location of enzyme and type of substrates it works on and names of some drugs.
S-Adenosylmethionine

Transmethylases (cytosol)

Catecholamines, phenols, amines

Dopamine, epinephrine, pyridine, histamine, thiouracil
What enzymes are used for Water conjugation?

List Endogenous Reactant, Location of enzyme and type of substrates it works on and names of some drugs.
Water

Epoxide hydrolase (microsomes) Arene oxides, cis-disubstituted and mono-substituted oxiranes

Benzopyrene 7,8-epoxide, styrene 1,2-oxide, carbamazepine epoxide
(cytosol) Alkene oxides, fatty acid epoxides Leukotriene A4
Glycine in Type II reaction.
Glycine

Acyl-CoA glycinetransferase (mitochondria)

Acyl-CoA derivatives of carboxylic acids

Salicylic acid, benzoic acid, nicotinic acid, cinnamic acid, cholic acid, deoxycholic acid
Explain how the metabolism of drugs can lead to toxic products?
Metabolism of drugs and other foreign chemicals may not always be an innocuous biochemical event leading to detoxification and elimination of the compound. Indeed, as previously noted, several compounds have been shown to be metabolically transformed to reactive intermediates that are toxic to various organs. Such toxic reactions may not be apparent at low levels of exposure to parent compounds when alternative detoxification mechanisms are not yet overwhelmed or compromised and when the availability of endogenous detoxifying cosubstrates (GSH, glucuronic acid, sulfate) is not limited. However, when these resources are exhausted, the toxic pathway may prevail, resulting in overt organ toxicity or carcinogenesis. The number of specific examples of such drug-induced toxicity is expanding rapidly.
Explain how Acetaminophen can follow Phase I and Phase II pathways.

Why is Phase I toxic?
An example is acetaminophen (paracetamol)-induced hepatotoxicity (Figure 4–5). Acetaminophen, an analgesic antipyretic drug, is quite safe in therapeutic doses (1.2 g/d for an adult). It normally undergoes glucuronidation and sulfation to the corresponding conjugates, which together make up 95% of the total excreted metabolites. The alternative P450-dependent GSH conjugation pathway accounts for the remaining 5%. When acetaminophen intake far exceeds therapeutic doses, the glucuronidation and sulfation pathways are saturated, and the P450-dependent pathway becomes increasingly important. Little or no hepatotoxicity results as long as hepatic GSH is available for conjugation. However, with time, hepatic GSH is depleted faster than it can be regenerated, and a reactive, toxic metabolite accumulates. In the absence of intracellular nucleophiles such as GSH, this reactive metabolite (N-acetylbenzoiminoquinone) reacts with nucleophilic groups of cellular proteins, resulting in hepatotoxicity.
How do genetic factors influence drug metabolism?
Genetic factors that influence enzyme levels account for some of these differences. Succinylcholine, for example, is metabolized only half as rapidly in persons with genetically determined defects in pseudocholinesterase as in persons with normally functioning pseudocholinesterase. Analogous pharmacogenetic differences are seen in the acetylation of isoniazid and the hydroxylation of warfarin. The defect in slow acetylators (of isoniazid and similar amines) appears to be caused by the synthesis of less of the NAT2 enzyme rather than of an abnormal form of it. Inherited as an autosomal recessive trait, the slow acetylator phenotype occurs in about 50% of blacks and whites in the USA, more frequently in Europeans living in high northern latitudes, and much less commonly in Asians and Inuits (Eskimos). The slow acetylator phenotype is also associated with a higher incidence of drug-induced autoimmune disorders and bicyclic aromatic amine-induced bladder cancer.
What are genetic polymorphisms in Phase II UGTs and GSTs associated with?
Genetic polymorphisms in the expression of other phase II enzymes (UGTs and GSTs) also occur. Thus, UGT-polymorphisms are associated with hyperbirubinemic diseases as well as impaired drug conjugation and/or elimination. Similarly, genetic polymorphisms in GST expression can lead to significant adverse effects and toxicities of drugs dependent on GSH conjugation for elimination.

Genetically determined defects in the phase I oxidative metabolism of debrisoquin, phenacetin, guanoxan, sparteine, phenformin, warfarin, and others have been reported.

The defects are apparently transmitted as autosomal recessive traits and may be expressed at any one of the multiple metabolic transformations that a chemical might undergo.
Explain the genetic variances due to Debrisoquin-spartein oxidation.
Of the several recognized genetic varieties of phase I drug metabolism polymorphisms, the following have been particularly well characterized and afford some insight into possible underlying mechanisms. First is the debrisoquin-sparteine oxidation type of polymorphism, which apparently occurs in 3–10% of whites and is inherited as an autosomal recessive trait. In affected individuals, the CYP2D6-dependent oxidations of debrisoquin and other drugs (Table 4–2; Figure 4–6) are impaired. These defects in oxidative drug metabolism are probably coinherited. The precise molecular basis for the defect appears to be faulty expression of the P450 protein, resulting in little or no isoform-catalyzed drug metabolism.
Explain the genetic variances in CYP3A4.
Allelic variants of CYP3A4 have also been reported, but their contribution to its well-known interindividual variability in drug metabolism apparently is limited. On the other hand, the expression of CYP3A5, another human liver isoform, is markedly polymorphic, ranging from 0% to 100% of the total hepatic CYP3A content. This CYP3A5 protein polymorphism is now known to result from a single nucleotide polymorphism (SNP) within intron 3, which enables normally spliced CYP3A5 transcripts in 5% of Caucasians, 29% of Japanese, 27% of Chinese, 30% of Koreans, and 73% of African Americans. Thus, it can significantly contribute to interindividual differences in the metabolism of preferential CYP3A5 substrates such as midazolam.
What is ultrarapid metabolism in genetic issues?
More recently, however, another polymorphic genotype has been reported that results in ultrarapid metabolism of relevant drugs due to the presence of 2D6 allelic variants with up to 13 gene copies in tandem. This genotype is most common in Ethiopians and Saudi Arabians, populations that display it in up to one third of individuals. As a result, these subjects require twofold to threefold higher daily doses of nortriptyline (a 2D6 substrate) to achieve therapeutic plasma levels. Conversely, in these ultrarapid-metabolizing populations, the prodrug codeine (another 2D6 substrate) is metabolized much faster to morphine, often resulting in undesirable adverse effects of morphine, such as abdominal pain.
Explain genetic variances in CYP2C19.
A second well-studied genetic drug polymorphism involves the stereoselective aromatic (4)-hydroxylation of the anticonvulsant mephenytoin, catalyzed by CYP2C19. This polymorphism, which is also inherited as an autosomal recessive trait, occurs in 3–5% of Caucasians and 18–23% of Japanese populations. It is genetically independent of the debrisoquin-sparteine polymorphism. In normal "extensive metabolizers ," (S )-mephenytoin is extensively hydroxylated by CYP2C19 at the 4 position of the phenyl ring before its glucuronidation and rapid excretion in the urine, whereas (R )-mephenytoin is slowly N -demethylated to nirvanol, an active metabolite. "Poor metabolizers ," however, appear to totally lack the stereospecific (S)-mephenytoin hydroxylase activity, so both (S)- and (R)-mephenytoin enantiomers are N-demethylated to nirvanol, which accumulates in much higher concentrations. Thus, poor metabolizers of mephenytoin show signs of profound sedation and ataxia after doses of the drug that are well tolerated by normal metabolizers. The molecular basis for this defect is a single base-pair mutation in exon 5 of the CYP2C19 gene that creates an aberrant splice site, a correspondingly altered reading frame of the mRNA, and, finally, a truncated, nonfunctional protein. It is clinically important to recognize that the safety of a drug may be severely reduced in persons who are poor metabolizers.
Explain why CYP2C9 variance leads to warfarin problems.
The third relatively well-characterized genetic polymorphism is that of CYP2C9. Two well-characterized variants of this enzyme exist, each with amino acid mutations that result in altered metabolism. The CYP2C9*2 allele encodes an Arg144Cys mutation, exhibiting impaired functional interactions with P450 reductase. The other allelic variant, CYP2C9*3, encodes an enzyme with an Ile359Leu mutation that has lowered affinity for many substrates. For example, individuals displaying the CYP2C9*3 phenotype have greatly reduced tolerance for the anticoagulant warfarin. The warfarin clearance in CYP2C9*3-homozygous individuals is about 10% of normal values, and these people have a much lower tolerance for the drug than those who are homozygous for the normal wild-type allele. These individuals also have a much higher risk of adverse effects with warfarin (eg, bleeding) and with other CYP2C9 substrates such as phenytoin, losartan, tolbutamide, and some nonsteroidal anti-inflammatory drugs.
Explain the genetic variances in CYP2A6.
Polymorphisms in the CYP2A6 gene have also been recently characterized, and their prevalence is apparently racially linked. CYP2A6 is responsible for nicotine oxidation, and tobacco smokers with low CYP2A6 activity consume less and have a lower incidence of lung cancer. CYP2A6 1B allelic variants associated with faster rates of nicotine metabolism have been recently discovered. It remains to be determined whether patients with these faster variants will fall into the converse paradigm of increased smoking behavior and lung cancer incidence.
Explain the genetic variances in CYP2B6?
Additional genetic polymorphisms in drug metabolism (eg, CYP2B6) that are inherited independently from those already described are being discovered. For instance, a 20- to 250-fold variation in interindividual CYP2B6 expression partly due to genetic polymorphisms has been reported. This may significantly impact the metabolism of several clinically relevant drugs such as cyclophosphamide, methadone, efavirenz, selegiline, and propofol. Studies of theophylline metabolism in monozygotic and dizygotic twins that included pedigree analysis of various families have revealed that a distinct polymorphism may exist for this drug and may be inherited as a recessive genetic trait. Genetic drug metabolism polymorphisms also appear to occur for aminopyrine and carbocysteine oxidations. Regularly updated information on human P450-polymorphisms is available at http://www.imm.ki.se/CYPalleles/.
Can genetic variations occur in other enzymes besides P450 enzymes.
Although genetic polymorphisms in drug oxidations often involve specific P450 enzymes, such genetic variations can also occur in other enzymes. Recent descriptions of a polymorphism in the oxidation of trimethylamine, believed to be metabolized largely by the flavin monooxygenase (Ziegler's enzyme), result in the "fish-odor syndrome" in slow metabolizers, thus suggesting that genetic variants of other non–P450-dependent oxidative enzymes may also contribute to such polymorphisms.
How does diet and environmental factors affect drug metabolism.
Diet and environmental factors contribute to individual variations in drug metabolism. Charcoal-broiled foods and cruciferous vegetables are known to induce CYP1A enzymes, whereas grapefruit juice is known to inhibit the CYP3A metabolism of coadministered drug substrates (Table 4–2). Cigarette smokers metabolize some drugs more rapidly than nonsmokers because of enzyme induction (see previous section). Industrial workers exposed to some pesticides metabolize certain drugs more rapidly than unexposed individuals. Such differences make it difficult to determine effective and safe doses of drugs that have narrow therapeutic indices.
How does age and sex affect drug metabolism?
Increased susceptibility to the pharmacologic or toxic activity of drugs has been reported in very young and very old patients compared with young adults (see Chapters 59 and 60). Although this may reflect differences in absorption, distribution, and elimination, differences in drug metabolism also play a role. Slower metabolism could be due to reduced activity of metabolic enzymes or reduced availability of essential endogenous cofactors.

Sex-dependent variations in drug metabolism have been well documented in rats but not in other rodents. Young adult male rats metabolize drugs much faster than mature female rats or prepubertal male rats. These differences in drug metabolism have been clearly associated with androgenic hormones. Clinical reports suggest that similar sex-dependent differences in drug metabolism also exist in humans for ethanol, propranolol, some benzodiazepines, estrogens, and salicylates.
Why are drug-drug interactions during metabolism important?
Many substrates, by virtue of their relatively high lipophilicity, are not only retained at the active site of the enzyme but remain nonspecifically bound to the lipid endoplasmic reticulum membrane. In this state, they may induce microsomal enzymes, particularly after repeated use. Acutely, depending on the residual drug levels at the active site, they also may competitively inhibit metabolism of a simultaneously administered drug.

Enzyme-inducing drugs include various sedative-hypnotics, antipsychotics, anticonvulsants, the antitubercular drug rifampin, and insecticides (Table 4–5). Patients who routinely ingest barbiturates, other sedative-hypnotics, or certain antipsychotic drugs may require considerably higher doses of warfarin to maintain a therapeutic effect. On the other hand, discontinuance of the sedative inducer may result in reduced metabolism of the anticoagulant and bleeding—a toxic effect of the ensuing enhanced plasma levels of the anticoagulant. Similar interactions have been observed in individuals receiving various combinations of drug regimens such as rifampin, antipsychotics, or sedatives with contraceptive agents, sedatives with anticonvulsant drugs, and even alcohol with hypoglycemic drugs (tolbutamide).
How does the simultaneous administration of two or more drugs affect the metabolism of each?
Conversely, simultaneous administration of two or more drugs may result in impaired elimination of the more slowly metabolized drug and prolongation or potentiation of its pharmacologic effects (Table 4–6). Both competitive substrate inhibition and irreversible substrate-mediated enzyme inactivation may augment plasma drug levels and lead to toxic effects from drugs with narrow therapeutic indices. Indeed, such acute interactions of terfenadine (a second-generation antihistamine) with a CYP3A4 substrate-inhibitor (ketoconazole, erythromycin, or grapefruit juice) resulted in fatal cardiac arrhythmias (torsade de pointes) requiring its withdrawal from the market. Similar drug-drug interactions with CYP3A4 substrate-inhibitors (such as the antibiotics erythromycin and clarithromycin, the antidepressant nefazodone, the antifungals itraconazole and ketoconazole, and the HIVprotease inhibitors indinavir and ritonavir), and consequent cardiotoxicity led to withdrawal or restricted use of the 5-HT4 agonist, cisapride. Similarly, allopurinol both prolongs the duration and enhances the chemotherapeutic and toxic actions of mercaptopurine by competitive inhibition of xanthine oxidase. Consequently, to avoid bone marrow toxicity, the dose of mercaptopurine must be reduced in patients receiving allopurinol. Cimetidine, a drug used in the treatment of peptic ulcer, has been shown to potentiate the pharmacologic actions of anticoagulants and sedatives. The metabolism of the sedative chlordiazepoxide has been shown to be inhibited by 63% after a single dose of cimetidine; such effects are reversed within 48 hours after withdrawal of cimetidine.
Explain one way that one drug may affect the metabolism of another drug.
Impaired metabolism may also result if a simultaneously administered drug irreversibly inactivates a common metabolizing enzyme. These inhibitors, in the course of their metabolism by cytochrome P450, inactivate the enzyme and result in impairment of their own metabolism and that of other cosubstrates. This is indeed the case of the furanocoumarins in grapefruit juice that inactivate CYP3A4 in the intestinal mucosa and consequently enhance its proteolytic degradation. This impairment of their intestinal first-pass CYP3A4-dependent metabolism significantly enhances the bioavailability of drugs, such as felodipine, nifedipine, terfenadine, verapamil, ethinylestradiol, saquinavir, and cyclosporine A, and is associated with clinically relevant drug-drug interactions.

Recovery from this potential for interactions is dependent on CYP3A4 resynthesis and thus may be slow.
Explain how drug may compete for the same endogenous compounds.
Some drugs require conjugation with endogenous substrates such as GSH, glucuronic acid, or sulfate for their inactivation. Consequently, different drugs may compete for the same endogenous substrates, and the faster-reacting drug may effectively deplete endogenous substrate levels and impair the metabolism of the slower-reacting drug. If the latter has a steep dose-response curve or a narrow margin of safety, potentiation of its pharmacologic and toxic effects may result.
How does disease effect drug metabolism?
Acute or chronic diseases that affect liver architecture or function markedly affect hepatic metabolism of some drugs. Such conditions include alcoholic hepatitis, active or inactive alcoholic cirrhosis, hemochromatosis, chronic active hepatitis, biliary cirrhosis, and acute viral or drug-induced hepatitis. Depending on their severity, these conditions may significantly impair hepatic drug-metabolizing enzymes, particularly microsomal oxidases, and thereby markedly affect drug elimination. For example, the half-lives of chlordiazepoxide and diazepam in patients with liver cirrhosis or acute viral hepatitis are greatly increased, with a corresponding prolongation of their effects. Consequently, these drugs may cause coma in patients with liver disease when given in ordinary doses.

Some drugs are metabolized so readily that even marked reduction in liver function does not significantly prolong their action. However, cardiac disease, by limiting blood flow to the liver, may impair disposition of those drugs whose metabolism is flow-limited (Table 4–7). These drugs are so readily metabolized by the liver that hepatic clearance is essentially equal to liver blood flow. Pulmonary disease may also affect drug metabolism, as indicated by the impaired hydrolysis of procainamide and procaine in patients with chronic respiratory insufficiency and the increased half-life of antipyrine in patients with lung cancer. The impaired enzyme activity or defective formation of enzymes associated with heavy metal poisoning or porphyria also results in reduced hepatic drug metabolism.
Which drugs are metabolized os rapidly that there clearance is blood-flow limited.
Alprenolol Lidocaine
Amitriptyline Meperidine
Clomethiazole Morphine
Desipramine Pentazocine
Imipramine Propoxyphene
Isoniazid Propranolol
Labetalol Verapamil
How does endocrine dysfunction affect drug metabolism?
Although the effects of endocrine dysfunction on drug metabolism have been well explored in experimental animal models, corresponding data for humans with endocrine disorders are scanty. Thyroid dysfunction has been associated with altered metabolism of some drugs and of some endogenous compounds as well. Hypothyroidism increases the half-life of antipyrine, digoxin, methimazole, and some blockers, whereas hyperthyroidism has the opposite effect. A few clinical studies in diabetic patients indicate no apparent impairment of drug metabolism, although impairment has been noted in diabetic rats. Malfunctions of the pituitary, adrenal cortex, and gonads markedly reduce hepatic drug metabolism in rats. On the basis of these findings, it may be supposed that such disorders could significantly affect drug metabolism in humans. However, until sufficient evidence is obtained from clinical studies in patients, such extrapolations must be considered tentative.

Finally, the release of inflammatory mediators, cytokines, and nitric oxide associated with bacterial or viral infections, cancer, or inflammation are known to impair drug metabolism by inactivating P450s and enhancing their degradation.