Você está na página 1de 10

Pharmacokinetics

Pharmacokinetics (in Greek: pharmacon meaning drug and kinetikos meaning putting in motion, the study of time dependency; sometimes abbreviated as PK) is a branch of pharmacology dedicated to the determination of the fate of substances administered externally to a living organism. In practice, this discipline is applied mainly to drug substances, though in principle it concerns itself with all manner of compounds ingested or otherwise delivered externally to an organism, such as nutrients, metabolites, hormones, toxins, etc. Pharmacokinetics is often studied in conjunction with pharmacodynamics. Pharmacodynamics explores what a drug does to the body, whereas pharmacokinetics explores what the body does to the drug. Pharmacokinetics includes the study of the mechanisms of absorption and distribution of an administered drug, the rate at which a drug action begins and the duration of the effect, the chemical changes of the substance in the body (e.g. by enzymes) and the effects and routes of excretion of the metabolites of the drug.[1]

ADME
Pharmacokinetics is divided into several areas which includes the extent and rate of Absorption, Distribution, Metabolism and Excretion. This is commonly referred to as the ADME scheme. However recent understanding about the drug-body interactions brought about the inclusion of new term Liberation. Now Pharmacokinetics can be better described as LADME.

Liberation is the process of release of drug from the formulation. Absorption is the process of a substance entering the body. Distribution is the dispersion or dissemination of substances throughout the fluids and tissues of the body. Metabolism is the irreversible transformation of parent compounds into daughter metabolites. Excretion is the elimination of the substances from the body. In rare cases, some drugs irreversibly accumulate in a tissue in the body.

Pharmacokinetics describes how the body affects a specific drug after administration. Pharmacokinetic properties of drugs may be affected by elements such as the site of administration and the concentration in which the drug is administered. These may affect the absorption rate.[2]

Analysis
Pharmacokinetic analysis is performed by noncompartmental (model independent) or compartmental methods. Noncompartmental methods estimate the exposure to a drug by

estimating the area under the curve of a concentration-time graph. Compartmental methods estimate the concentration-time graph using kinetic models. Compartment-free methods are often more versatile in that they do not assume any specific compartmental model and produce accurate results also acceptable for bioequivalence studies.

Noncompartmental analysis
Noncompartmental PK analysis is highly dependent on estimation of total drug exposure. Total drug exposure is most often estimated by Area Under the Curve methods, with the trapezoidal rule (numerical differential equations) the most common area estimation method. Due to the dependence on the length of 'x' in the trapezoidal rule, the area estimation is highly dependent on the blood/plasma sampling schedule. That is, the closer your time points are, the closer the trapezoids are to the actual shape of the concentrationtime curve.

Compartmental analysis
Compartmental PK analysis uses kinetic models to describe and predict the concentrationtime curve. PK compartmental models are often similar to kinetic models used in other scientific disciplines such as chemical kinetics and thermodynamics. The advantage of compartmental over some noncompartmental analyses is the ability to predict the concentration at any time. The disadvantage is the difficulty in developing and validating the proper model. Compartment-free modeling based on curve stripping does not suffer this limitation. The simplest PK compartmental model is the one-compartmental PK model with IV bolus administration and first-order elimination. The most complex PK models (called PBPK models) rely on the use of physiological information to ease development and validation.

Bioanalytical methods
Bioanalytical methods are necessary to construct a concentration-time profile. Chemical techniques are employed to measure the concentration of drugs in biological matrix, most often plasma. Proper bioanalytical methods should be selective and sensitive. Mass spectrometry Pharmacokinetics is often studied using mass spectrometry because of the complex nature of the matrix (often blood or urine) and the need for high sensitivity to observe low dose and long time point data. The most common instrumentation used in this application is LCMS with a triple quadrupole mass spectrometer. Tandem mass spectrometry is usually employed for added specificity. Standard curves and internal standards are used for quantitation of usually a single pharmaceutical in the samples. The samples represent different time points as a pharmaceutical is administered and then metabolized or cleared from the body. Blank or t=0 samples taken before administration are important in determining background and insuring data integrity with such complex sample matrices. Much attention is paid to the linearity of the standard curve; however it is not uncommon to

use curve fitting with more complex functions such as quadratics since the response of most mass spectrometers is less than linear across large concentration ranges.[3][4][5] There is currently considerable interest in the use of very high sensitivity mass spectrometry for microdosing studies, which are seen as a promising alternative to animal experimentation.[6]

Absorption (pharmacokinetics)
In pharmacology (and more specifically pharmacokinetics), absorption is the movement of a drug into the bloodstream. Absorption involves several phases. First, the drug needs to be administered via some route of administration (oral, via the skin, etc.) and in a specific dosage form such as a tablet, capsule, and so on. In other situations, such as intravenous therapy, intramuscular injection, enteral nutrition and others, absorption is even more straight-forward and there is less variability in absorption and bioavailability is often near 100%. Absorption is a primary focus in drug development and medicinal chemistry, since the drug must be absorbed before any medicinal effects can take place. Moreover, the drug's pharmacokinetic profile can be easily and significantly changed by adjusting factors that affect absorption.

Dissolution
In the most standard situation, a tablet is ingested and passes through the esophagus to the stomach. Because the stomach is an aqueous environment, this is the first place where a tablet will dissolve. The rate of dissolution is a key target for controlling the duration of a drug's effect, and as such, several dosage forms that contain the same active ingredient may be available, differing only in the rate of dissolution. If a drug is supplied in a form that is not readily dissolved, the drug may be released more gradually over time with a longer duration of action. Having a longer duration of action may improve compliance since the medication will not have to be taken as often. Additionally, slow-release dosage forms may maintain concentrations within an acceptable therapeutic range over a long period of time, as opposed to quick-release dosage forms which may result in sharper peaks and troughs in serum concentrations.

The rate of dissolution is described by the Noyes-Whitney equation as shown below: Where:

is the rate of dissolution. A is the surface area of the solid. C is the concentration of the solid in the bulk dissolution medium. Cs is the concentration of the solid in the diffusion layer surrounding the solid. D is the diffusion coefficient. L is the diffusion layer thickness.

As can be inferred by the Noyes-Whitney equation, the rate of dissolution may be modified primarily by altering the surface area of the solid. The surface area may be adjusted by altering the particle size (e.g. micronization). The rate of dissolution may also be altered by choosing a suitable polymorph of a compound. Specifically, crystalline forms dissolve slower than amorphous forms. Also, coatings on a tablet or a pellet may act as a barrier to reduce the rate of dissolution. Coating may also be used to modify where dissolution takes place. For example, enteric coatings may be applied to a drug, so that the coating only dissolves in the basic environment of the intestines. This will prevent release of the drug before reaching the intestines. Since solutions are already dissolved, they do not need to undergo dissolution before being absorbed.

Ionization
The gastrointestinal tract is lined with epithelial cells. Drugs must pass through these cells in order to be absorbed into the circulatory system. One particular cellular barrier that may prevent absorption of a given drug is the cell membrane. Cell membranes are essentially lipid bilayers which form a semipermeable membrane. Pure lipid bilayers are generally permeable only to small, uncharged solutes. Hence, whether or not a molecule is ionized will affect its absorption, since ionic molecules are considered charged molecules by definition. The Henderson-Hasselbalch equation offers a way to determine the proportion of a substance that is ionized at a given pH. In the stomach, drugs that are weak acids (such as aspirin) will be present mainly in their non-ionic form, and weak bases will be in their ionic form. Since non-ionic species diffuse more readily through cell membranes, weak acids will have a higher absorption in the highly-acidic stomach. However, the reverse is true in the basic environment of the intestines-- weak bases (such as caffeine) will diffuse more readily since they will be non-ionic.

This aspect of absorption has been targeted by medicinal chemistry. For example, a suitable analog may be chosen so that the drug is more likely to be in a non-ionic form. Also, prodrugs of a compound may be developed by medicinal chemists-- these chemical variants may be more readily absorbed and then metabolized by the body into the active compound. However, changing the structure of a molecule is less predictable than altering dissolution properties, since changes in chemical structure may affect the pharmacodynamic properties of a drug.

Distribution (pharmacology)
Distribution in pharmacology is a branch of pharmacokinetics which describes the reversible transfer of drug from one location to another within the body. The distribution of a drug between tissues is dependent on permeability between tissues (between blood and tissues in particular), blood flow and perfusion rate of the tissue and the ability of the drug to bind plasma proteins and tissue. pH parturition plays a major role as well. Once a drug enters into systemic circulation by absorption or direct administration, A drug has to be distributed into interstitial and intracellular fluids.The lipid solubility, pH of compartment, extent of binding with plasma protein and tissue proteins, cardiac output, regional blood flow, capillary permeability are associated for distribution of the drug through tissues.The drug is easily distributed in highly perfused organs like liver, heart, kidney etc in large quantities & in small quantities it is distributed in low perfused organs like muscle, fat, peripheral organs etc. The drug can be moved from the plasma to the tissue until the equilibrium is established (for unbound drug present in plasma). The volume of distribution (VD) of a drug is a property that quantifies the extent of distribution.

Drug metabolism
Drug metabolism is the metabolism of drugs, their biochemical modification or degradation, usually through specialized enzymatic systems. This is a form of xenobiotic metabolism. Drug metabolism often converts lipophilic chemical compounds into more readily excreted polar products. Its rate is an important determinant of the duration and intensity of the pharmacological action of drugs. Drug metabolism can result in toxication or detoxication - the activation or deactivation of the chemical. While both occur, the major metabolites of most drugs are detoxication products.

Drugs are almost all xenobiotics. Other commonly used organic chemicals are also xenobiotics, and are metabolized by the same enzymes as drugs. This provides the opportunity for drug-drug and drug-chemical interactions or reactions.

Phase I vs. Phase II


Phase I and Phase II reactions are biotransformations of chemicals that occur during drug metabolism. Phase I reactions usually precede Phase II, though not necessarily. During these reactions, polar bodies are either introduced or unmasked, which results in (more) polar metabolites of the original chemicals. In the case of pharmaceutical drugs, Phase I reactions can lead either to activation or inactivation of the drug. Phase I reactions (also termed nonsynthetic reactions) may occur by oxidation, reduction, hydrolysis, cyclization, and decyclization reactions. Oxidation involves the enzymatic addition of oxygen or removal of hydrogen, carried out by mixed function oxidases, often in the liver. These oxidative reactions typically involve a cytochrome P450 monooxygenase (often abbreviated CYP), NADPH and oxygen. The classes of pharmaceutical drugs that utilize this method for their metabolism include phenothiazines, paracetamol, and steroids. If the metabolites of phase I reactions are sufficiently polar, they may be readily excreted at this point. However, many phase I products are not eliminated rapidly and undergo a subsequent reaction in which an endogenous substrate combines with the newly incorporated functional group to form a highly polar conjugate. A common Phase I oxidation involves conversion of a C-H bond to a C-OH. This reaction sometimes converts a pharmacologically inactive compound (a prodrug) to a pharmacologically active one. By the same token, Phase I can turn a nontoxic molecule into a poisonous one (toxification). A famous example is acetonitrile, CH3CN. Simple hydrolysis in the stomach transforms acetonitrile into acetate and ammonia, which are comparatively innocuous. But Phase I metabolism converts acetonitrile to HOCH2CN, which rapidly dissociates into formaldehyde and hydrogen cyanide, both of which are toxic. Phase I metabolism of drug candidates can be simulated in the laboratory using nonenzyme catalysts.[1] This example of a biomimetic reaction tends to give a mixture of products that often contains the Phase I metabolites, and Alpha Chimica's approach to preparing prospective drug candidates makes use of this in vitro chemistry. Phase II reactions usually known as conjugation reactions (e.g., with glucuronic acid, sulfonates (commonly known as sulfation) , glutathione or amino acids) are usually detoxication in nature, and involve the interactions of the polar functional groups of phase I metabolites. Sites on drugs where conjugation reactions occur include carboxyl (-COOH), hydroxyl (-OH), amino (NH2), and sulfhydryl (-SH) groups. Products of conjugation

reactions have increased molecular weight and are usually inactive unlike Phase I reactions which often produce active metabolites.

Sites
Quantitatively, the smooth endoplasmic reticulum of the liver cell is the principal organ of drug metabolism, although every biological tissue has some ability to metabolize drugs. Factors responsible for the liver's contribution to drug metabolism include that it is a large organ, that it is the first organ perfused by chemicals absorbed in the gut, and that there are very high concentrations of most drug-metabolizing enzyme systems relative to other organs. If a drug is taken into the GI tract, where it enters hepatic circulation through the portal vein, it becomes well-metabolized and is said to show the first pass effect. Other sites of drug metabolism include epithelial cells of the gastrointestinal tract, lungs, kidneys, and the skin. These sites are usually responsible for localized toxicity reactions.

Major enzymes and pathways


Several major enzymes and pathways are involved in drug metabolism, and can be divided into Phase I and Phase II reactions:

Phase I
Oxidation

Cytochrome P450 monooxygenase system Flavin-containing monooxygenase system Alcohol dehydrogenase and aldehyde dehydrogenase Monoamine oxidase Co-oxidation by peroxidases

Reduction

NADPH-cytochrome P450 reductase Reduced (ferrous) cytochrome P450

It should be noted that during reduction reactions, a chemical can enter futile cycling, in which it gains a free-radical electron, then promptly loses it to oxygen (to form a superoxide anion). Hydrolysis

Esterases and amidases Epoxide hydrolase

Phase II
Methylation

methyltransferase

Sulphation

Glutathione S-transferases Sulfotransferases

Acetylation

N-acetyltransferases Amino acid N-acyl transferases

Glucuronidation

UDP-glucuronosyltransferases o Mercapturic acid biosynthesis

Factors that affect Drug Metabolism


The duration and intensity of pharmacological action of most lipophilic drugs are determined by the rate they are metabolized to inactive products. The Cytochrome P450 monooxygenase system is the most important pathway in this regard. In general, anything that increases the rate of metabolism (e.g., enzyme induction) of a pharmacologically active metabolite will decrease the duration and intensity of the drug action. The opposite is also true (e.g., enzyme inhibition). Various physiological and pathological factors can also affect drug metabolism. Physiological factors that can influence drug metabolism include age, individual variation (e.g., pharmacogenetics), enterohepatic circulation, nutrition, intestinal flora, or sex differences. In general, drugs are metabolized more slowly in fetal, neonatal and elderly humans and animals than in adults. Genetic variation (polymorphism) accounts for some of the variability in the effect of drugs. With N-acetyltransferases (involved in Phase II reactions), individual variation creates a group of people who acetylate slowly (slow acetylators) and those who acetylate quickly, split roughly 50:50 in the population of Canada. This variation may have dramatic consequences, as the slow acetylators are more prone to dose-dependent toxicity. Cytochrome P450 monooxygenase system enzymes can also vary across individuals, with deficiencies occurring in 1 - 30% of people, depending on their ethnic background.

Pathological factors can also influence drug metabolism, including liver, kidney, or heart diseases. In silico modelling and simulation methods allow drug metabolism to be predicted in virtual patient populations prior to performing clinical studies in human subjects.[2] This can be used to identify individuals most at risk from adverse reaction.

Excretion
Excretion is the process of eliminating waste products of metabolism and other non-useful materials.[1] It is an essential process in all forms of life. It contrasts secretion, where the substance may have specific tasks after leaving the cell. In single-celled organisms, waste products are discharged directly through the surface of the cell. Multicellular organisms utilize more complex excretory methods. Higher plants eliminate gases through the stomata, or pores, on the surface of leaves. Animals have special excretory organs.

Mammalian excretion
In mammals, the excretory processes are the formation of urine in the kidneys and the formation of carbon dioxide (a mammal's abundant metabolic waste) molecules as a result of respiration, which is then exhaled from the lungs. These waste products are eliminated by urination and exhalation respectively. In urination, hormonal control over excretion occurs in the distal tubules of the kidneys as directed by the hypothalamus.

In kidney
in mammals the main organs of excretion are the kidneys and accessory urinary organs, through which urine is eliminated,[2] and the large intestines, from which solid wastes are expelled. In strict biological terminology, undigested food expelled in the feces is not considered to be excretion, since it is not metabolic waste. Substances secreted into the bile and then eliminated in the feces are considered to be excreted, however. The skin and lungs also have excretory functions: the skin eliminates metabolic wastes like urea and lactic acid through sweating,[3] and the lungs expel carbon dioxide.

Other

Mucociliary excretion is the excretion of mucus in the respiratory system. Biliary excretion occurs via the bile which is delivered to the duodenum and removed in the feces.

Perspiration is another excretory process which removes salts and water from the body, although the primary purpose is cooling. Breast milk

Non-mammalian
Chemical structure of uric acid. In plants, breakdown of substances is much slower than in animals. Hence accumulation of waste is much slower and there are no special organs of excretion. Green plants in darkness or plants that do not contain chlorophyll produce carbon dioxide and water as respiratory waste products. Carbon dioxide released during respiration gets utilized during photosynthesis.Oxygen itself can be thought of as a waste product generated during photosynthesis. Plants can get rid of excess water by transpiration. Waste products may be stored in leaves that fall off. Other waste materials that are exuded by some plants resins, saps, latexes, etc. are forced from the interior of the plant by hydrostatic pressures inside the plant and by absorptive forces of plant cells. Plants also excrete some waste substances into the soil around them.[4] Aquatic animals usually excrete ammonia directly into the external environment, as this compound has high solubility and there is ample water available for dilution. In terrestrial animals ammonia-like compounds are converted into other nitrogenous materials as there is less water in the environment and ammonia itself is toxic. White cast of Uric acid defecated with the dark feces from a lizard. Insects, birds and some other reptiles also undergo a similar mechanism Birds excrete their nitrogenous wastes as uric acid in the form of a paste. This is metabolically more expensive, but allows more efficient water retention and it can be stored more easily in the egg. Many avian species, especially seabirds, can also excrete salt via specialized nasal salt glands, the saline solution leaving through nostrils in the beak. In insects, a system involving Malpighian tubules is utilized to excrete metabolic waste. Metabolic waste diffuses or is actively transported into the tubule, which transports the wastes to the intestines. The metabolic waste is then released from the body along with fecal matter. Many people misuse the term excretion as a euphemism for defecation, and use excrement for feces, but this is biologically incorrect.[1]

Você também pode gostar