metabolism and excretion of drugs pt.pptx

ElKlarens 25 views 34 slides Oct 17, 2024
Slide 1
Slide 1 of 34
Slide 1
1
Slide 2
2
Slide 3
3
Slide 4
4
Slide 5
5
Slide 6
6
Slide 7
7
Slide 8
8
Slide 9
9
Slide 10
10
Slide 11
11
Slide 12
12
Slide 13
13
Slide 14
14
Slide 15
15
Slide 16
16
Slide 17
17
Slide 18
18
Slide 19
19
Slide 20
20
Slide 21
21
Slide 22
22
Slide 23
23
Slide 24
24
Slide 25
25
Slide 26
26
Slide 27
27
Slide 28
28
Slide 29
29
Slide 30
30
Slide 31
31
Slide 32
32
Slide 33
33
Slide 34
34

About This Presentation

Pharmacology


Slide Content

Metabolism and Excretion of Drugs

Metabolism and Excretion of Drugs Both metabolism and excretion can be viewed as processes responsible for elimination of drug (parent and metabolite) from the body. Drug metabolism changes the chemical structure of a drug to produce a drug metabolite, which is frequently but not universally less pharmacologically active . Metabolism also renders the drug compound more water soluble and therefore more easily excreted.

Drug metabolism reactions are carried out by enzyme systems that evolved over time to protect the body from exogenous chemicals . The kidney cannot efficiently eliminate lipophilic drugs that readily cross cell membranes and are reabsorbed in the distal convoluted tubules . Therefore , lipid-soluble agents are first metabolized into more polar (hydrophilic) substances in the liver via two general sets of reactions ,

called phase I and phase II phase I oxidative or reductive enzymes and phase II conjugative enzymes

Phase I Phase I reactions convert lipophilic drugs into more polar molecules by introducing or unmasking a polar functional group, such as –OH or –NH2. Phase I reactions usually involve reduction, oxidation, or hydrolysis. Phase I metabolism may increase, decrease, or have no effect on pharmacologic activity.

The phase I reactions most frequently involved in drug metabolism are catalyzed by microsomal mixed-function oxidases (the cytochrome P450 system ). Phase I reactions not involving the microsomal mixed-function oxidases

Phase I reactions utilizing the P450 system The P450 system is important for the metabolism of many endogenous compounds (such as steroids, lipids) and for the biotransformation of exogenous substances ( xenobiotics ).

Cytochrome P450 is designated as CYP, is a superfamily of heme -containing isozymes are located in most cells, but primarily in the liver and GI tract.

Nomenclature: The family name is indicated by the Arabic number that follows CYP , and the capital letter designates the subfamily, for example, CYP3A . A second number indicates the specific isozyme , as in CYP3A4 . multiple enzymes exits

Specificity there are many different genes that encode multiple enzymes , there are as well many different P450 isoforms . These enzymes have the capacity to modify a large number of structurally diverse substrates . In addition, an individual drug may be a substrate for more than one isozyme

Four isozymes are responsible for the vast majority of P450-catalyzed reactions They are CYP3A4/5 , CYP2D6 , CYP2C8/9, and CYP1A2. Considerable amounts of CYP3A4 are found in intestinal mucosa, accounting for first-pass metabolism of drugs such as chlorpromazine and clonazepam

Genetic variability P450 enzymes exhibit considerable genetic variability among individuals and racial groups . Variations in P450 activity may alter drug efficacy and the risk of adverse events . CYP2D6 , in particular, has been shown to exhibit genetic polymorphism. CYP2D6 mutations result in very low capacities to metabolize substrates.

Some individuals, for example, obtain no benefit from the opioid analgesic codeine, because they lack the CYP2D6 enzyme that activates the drug. Similar polymorphisms have been characterized for the CYP2C subfamily of isozymes .

For instance, c lopidogrel carries a warning that patients who are poor CYP2C19 metabolizers have a higher incidence of cardiovascular events (for example, stroke or myocardial infarction) when taking this drug. Clopidogrel is a prodrug , and CYP2C19 activity is required to convert it to the active metabolite.

At this point , we should know The CYP450-dependent enzymes are an important target for pharmacokinetic drug interactions Namely Induction of selected CYP isozymes Inhibition of CYP isozyme activity

Inducers: Xenobiotics (chemicals not normally produced or expected to be present in the body, for example, drugs or environmental pollutants) may induce the activity of these enzymes . Certain drugs (for example, phenobarbital , rifampin , and carbamazepine ) are capable of increasing the synthesis of one or more CYP isozymes .

This results in increased biotransformation of drugs and can lead to significant decreases in plasma concentrations of drugs metabolized by these CYP isozymes , with concurrent loss of pharmacologic effect . For example, rifampin , an antituberculosis drug , significantly decreases the plasma concentrations of human immunodeficiency virus (HIV) protease inhibitors, thereby diminishing their ability to suppress HIV replication.

Inhibitors: Inhibition of CYP isozyme activity is an important source of drug interactions that lead to serious adverse events. The most common form of inhibition is through competition for the same isozyme .

Some drugs, however, are capable of inhibiting reactions for which they are not substrates (for example, ketoconazole ), leading to drug interactions. Numerous drugs have been shown to inhibit one or more of the CYP-dependent biotransformation pathways of warfarin For example, omeprazole is a potent inhibitor of three of the CYP isozymes responsible for warfarin metabolism

Phase I reactions not involving the P450 system These include amine oxidation (for example, oxidation of catecholamines or histamine), alcohol dehydrogenation (for example, ethanol oxidation ), esterases (for example, metabolism of aspirin in the liver), and hydrolysis (for example, of procaine ).

Phase II conjugative enzymes This phase consists of conjugation reactions many phase I metabolites are still too lipophilic to be excreted

A subsequent conjugation reaction with an endogenous substrate, such as glucuronic acid, sulfuric acid, acetic acid, or an amino acid, results in polar, usually more water-soluble compounds that are often therapeutically inactive A notable exception is morphine-6-glucuronide , which is more potent than morphine.

Glucuronidation is the most common and the most important conjugation reaction . [Note: Drugs already possessing an –OH, –NH2, or –COOH group may enter phase II directly and become conjugated without prior phase I metabolism.] The highly polar drug conjugates are then excreted by the kidney or in bile

FACTORS INFLUENCING DRUG DISTRIBUTION Distribution is the delivery of drug from the systemic circulation to tissues. Once a drug has entered the blood compartment, the rate at which it penetrates tissues and other body fluids depends on several factors. These include

(1) capillary permeability, (2) blood flow–tissue mass ratio (i.e., perfusion rate), (3) extent of plasma protein and specific organ binding, (4) regional differences in pH, (5) transport mechanisms available, and (6) the permeability characteristics of specific tissue membranes

BINDING OF DRUGS TO PLASMA PROTEINS Most drugs found in the vascular compartment are bound reversibly with one or more of the macromolecules in plasma. Although some drugs simply dissolve in plasma water, most are associated with plasma components such as albumin, globulins, transferrin , ceruloplasmin , glycoproteins , and ᾳ and β lipoproteins. While many acidic drugs bind principally to albumin, basic drugs frequently bind to other plasma proteins, such as lipoproteins and ᾳ 1-acid glycoprotein (ᾳ 1-AGP), in addition to albumin.

The extent of this binding will influence the drug’s distribution and rate of elimination because only the unbound drug can diffuse through the capillary wall, produce its systemic effects, be metabolized, and be excreted . Since only the unbound (or free) drug diffuses through the capillary walls, extensive binding may decrease the intensity of drug action. The magnitude of this decrease is directly proportional to the fraction of drug bound to plasma protein.

At low drug concentrations, the stronger the affinity between the drug and protein, the smaller the fraction that is free. As drug dosage increases, eventually the binding capacity of the protein becomes saturated and any additional drug will remain unbound.

The binding of a drug to plasma proteins will decrease its effective plasma to tissue concentration gradient, that is, the force that drives the drug out of the circulation, thereby slowing the rate of transfer across the capillary. As the free drug leaves the circulation, the protein–drug complex begins to dissociate and more free drug becomes available for diffusion.

Thus, binding does not prevent the drug from reaching its site of action but only retards the rate at which this occurs. Extensive plasma protein binding may prolong drug availability and duration of action. Protein binding also plays a role in the distribution of drugs and thus the volume of distribution.

Drugs that are highly bound to plasma proteins may distribute less widely because they remain trapped in the peripheral vasculature, since the plasma proteins themselves cannot traverse into the extravascular space. However, if the affinity of a drug for tissues (e.g., fat, muscle) is greater than the affinity for plasma proteins, widespread distribution can occur despite a high degree of plasma protein binding

The binding of drugs to plasma proteins is usually nonspecific; that is, many drugs may interact with the same binding site. A drug with a higher affinity may displace a drug with weaker affinity. Increases in the non–protein-bound drug fraction (i.e., free drug) can theoretically result in an increase in the drug’s intensity of pharmacological response, side effects, and potential toxicity.

Some disease states (e.g., hyperalbuminemia , hypoalbuminemia , uremia, hyperbilirubinemia ) have been associated with changes in plasma protein binding of drugs. For example, in uremic patients the plasma protein binding of certain acidic drugs (e.g., penicillin, sulfonamides, salicylates , and barbiturates) is reduced.

References Lippincott Illustrated Reviews: Pharmacology Sixth Edition byKaren Whalen, Ph The Pharmacological Basis of therapeutics (Goodman and Gilman,s ) Rang and Dale’s Pharmacology Katzung et al Basic and Clinical Pharmacology
Tags