Therapeutics and good prescribing



Therapeutics and good prescribing


S. Maxwell


Prescribing medicines is a major tool used by most doctors to restore or preserve the health of their patients. Medicines contain drugs (the specific chemical substances with pharmacological effects), either alone or in combination, in a formulation mixed with other ingredients. The beneficial effects of medicines must be weighed against their cost and the risks of adverse drug reactions and interactions, often caused by injudicious prescribing decisions and by prescribing errors. The modern prescriber must meet the challenges posed by an increasing number of drugs and formulations available and of indications for prescribing them, and the greater complexity of treatment regimens followed by individual patients (‘polypharmacy’, a particular challenge in the ageing population). The purpose of this chapter is to elaborate on the principles and practice that underpin good prescribing (Box 2.1).




Principles of clinical pharmacology


Prescribers need to understand what the drug does to the body (pharmacodynamics) and what the body does to the drug (pharmacokinetics) (Fig. 2.1). Although this chapter is focused on the most common drugs, which are synthetic small molecules, the same principles apply to the increasingly numerous ‘biological’ therapies (sometimes abbreviated to ‘biologics’) now in use, which include peptides, proteins, enzymes and monoclonal antibodies (p. 74).




Pharmacodynamics


Drug targets and mechanisms of action


Modern drugs are usually discovered by screening compounds for activity either to stimulate or to block the function of a specific molecular target, which is predicted to have a beneficial effect in a particular disease (Box 2.2). Other drugs have useful but less selective chemical properties, such as chelators (e.g. for treatment of iron or copper overload), osmotic agents (used as diuretics in cerebral oedema) or general anaesthetics (that alter the biophysical properties of lipid membranes). The following characteristics of the interaction of drugs with receptors illustrate some of the important determinants of the effects of drugs:



image 2.2   Examples of target molecules for drugs










































Drug target Description Examples
Receptors
Channel-linked receptors Ligand binding controls a linked ion channel, known as ‘ligand-gated’ (in contrast to ‘voltage-gated’ channels that respond to changes in membrane potential)
G-protein-coupled receptors (GPCRs) Ligand binding affects one of a family of ‘G-proteins’ that mediate signal transduction either by activating intracellular enzymes (such as adenylate or guanylate cyclase, producing cyclic AMP or GMP, respectively) or by controlling ion channels
Kinase-linked receptors Ligand binding activates an intracellular protein kinase that triggers a cascade of phosphorylation reactions
Transcription factor receptors Intracellular and also known as ‘nuclear receptors’; ligand binding promotes or inhibits gene transcription and hence synthesis of new proteins
Other targets
Voltage-gated ion channels Mediate electrical signalling in excitable tissues (muscle and nervous system)
Enzymes Catalyse biochemical reactions. Drugs interfere with binding of substrate to the active site or of co-factors
Transporter proteins Carry ions or molecules across cell membranes


image


(AMP = adenosine monophosphate; ATPase = adenosine triphosphatase; GMP = guanosine monophosphate; PPAR = peroxisome proliferator-activated receptor)




• Affinity describes the propensity for a drug to bind to a receptor and is related to the ‘molecular fit’ and the strength of the chemical bond. Some drug–receptor interactions are irreversible, either because the affinity is so strong or because the drug modifies the structure of its molecular target.


• Selectivity describes the propensity for a drug to bind to one target rather than another. Selectivity is a relative term, not to be confused with absolute specificity. It is common for drugs targeted at a particular subtype of receptor to exhibit some effect at other subtypes. For example, β-adrenoceptors can be subtyped on the basis of their responsiveness to the endogenous agonist noradrenaline (norepinephrine): the concentration of noradrenaline required to cause bronchodilatation (via β2-adrenoceptors) is ten times higher than that required to cause tachycardia (via β1-adrenoceptors). ‘Cardioselective’ β-blockers have anti-anginal effects on the heart (β1) but may still cause bronchospasm in the lung (β2) and are contraindicated for asthmatic patients.


• Agonists bind to a receptor to produce a conformational change that is coupled to a biological response. As agonist concentration increases, so does the proportion of receptors occupied, and hence the biological effect. Partial agonists activate the receptor, but cannot produce a maximal signalling effect equivalent to that of a full agonist even when all available receptors are occupied.


• Antagonists bind to a receptor but do not produce the conformational change that initiates an intracellular signal. A competitive antagonist competes with endogenous ligands to occupy receptor binding sites, with the resulting antagonism depending on the relative affinities and concentrations of drug and ligand. Non-competitive antagonists inhibit the effect of an agonist by mechanisms other than direct competition for receptor binding with the agonist (e.g. by affecting post-receptor signalling).



Dose–response relationships


Plotting the logarithm of drug dose against drug response typically produces a sigmoidal dose–response curve (Fig. 2.2). Progressive increases in drug dose (which for most drugs is proportional to the plasma drug concentration) produce increasing response, but only within a relatively narrow range of dose; further increases in dose beyond this range produce little extra effect. The following characteristics of the drug response are useful in comparing different drugs:




• Efficacy describes the extent to which a drug can produce a target-specific response when all available receptors or binding sites are occupied (i.e. Emax on the dose–response curve). A full agonist can produce the maximum response of which the receptor is capable, while a partial agonist at the same receptor will have lower efficacy. Therapeutic efficacy describes the effect of the drug on a desired biological endpoint, and can be used to compare drugs that act via different pharmacological mechanisms (e.g. loop diuretics induce a greater diuresis than thiazide diuretics and therefore have greater therapeutic efficacy).


• Potency describes the amount of drug required for a given response. More potent drugs produce biological effects at lower doses, so they have a lower ED50. A less potent drug can still have an equivalent efficacy if it is given in higher doses.


The dose–response relationship varies between patients because of variations in the many determinants of pharmacokinetics and pharmacodynamics. In clinical practice, the prescriber is unable to construct a dose–response curve for each individual patient. Therefore, most drugs are licensed for use within a recommended range of doses that is expected to reach close to the top of the dose–response curve for most patients. However, it is sometimes possible to achieve the desired therapeutic efficacy at doses towards the lower end of, or even below, the recommended range.



Therapeutic index

The adverse effects of drugs are often dose-related in a similar way to the beneficial effects, although the dose–response curve for these adverse effects is normally shifted to the right (see Fig. 2.2). The ratio of the ED50 for therapeutic efficacy and for a major adverse effect is known as the ‘therapeutic index’. In reality, drugs have multiple potential adverse effects but the concept of therapeutic index is usually reserved for those requiring dose reduction or discontinuation. For most drugs, the therapeutic index is greater than 100 but there are some notable exceptions with therapeutic indices less than 10 (e.g. digoxin, warfarin, insulin, phenytoin, opioids). The doses of such drugs have to be titrated carefully for individual patients to maximise benefits but avoid adverse effects.



Desensitisation and withdrawal effects


Desensitisation refers to the common situation in which the biological response to a drug diminishes when it is given continuously or repeatedly. It may be possible to restore the response by increasing the dose of the drug but, in some cases, the tissues may become completely refractory to its effect.



• Tachyphylaxis describes desensitisation that occurs very rapidly, sometimes with the initial dose. This rapid loss of response implies depletion of chemicals that may be necessary for the pharmacological actions of the drug (e.g. a stored neurotransmitter released from a nerve terminal) or receptor phosphorylation.


• Tolerance describes a more gradual loss of response to a drug that occurs over days or weeks. This slower change implies changes in receptor numbers or the development of counter-regulatory physiological changes that offset the actions of the drug (e.g. accumulation of salt and water in response to vasodilator therapy).


• Drug resistance is a term normally reserved for describing the loss of effectiveness of an antimicrobial (p. 151) or cancer chemotherapy drug.


• In addition to these pharmacodynamic causes of desensitisation, reduced response may be the consequence of lower plasma and tissue drug concentrations as a result of altered pharmacokinetics (see below).


When drugs induce chemical, hormonal and physiological changes that offset their actions, discontinuation may allow these changes to cause ‘rebound’ withdrawal effects (Box 2.3).




Pharmacokinetics


Understanding ‘what the body does to the drug’ (Fig. 2.3) is extremely important for prescribers because this forms the basis on which the optimal route of administration and dose regimen are chosen and explains the majority of inter-individual variation in the response to drug therapy.




Drug absorption and routes of administration


Absorption is the process by which drug molecules gain access to the blood stream. The rate and extent of drug absorption depend on the route of administration (see Fig. 2.3).



Enteral administration

These routes involve administration via the gastrointestinal tract:



• Oral. This is the commonest route of administration because it is simple, convenient and readily used by patients to self-administer their medicines. Absorption after an oral dose is a complex process that depends on the drug being swallowed, surviving exposure to gastric acid, avoiding unacceptable food binding, being absorbed across the small bowel mucosa into the portal venous system, and surviving metabolism by gut wall or liver enzymes (‘first-pass metabolism’). As a consequence, absorption is frequently incomplete following oral administration. The term ‘bioavailability’ describes the proportion of the dose that reaches the systemic circulation intact.


• Buccal, intranasal and sublingual (SL). These routes have the advantage of enabling rapid absorption into the systemic circulation without the uncertainties associated with oral administration (e.g. organic nitrates for angina pectoris, triptans for migraine, opioid analgesics).


• Rectal (PR). The rectal mucosa is occasionally used as a site of drug administration when the oral route is compromised because of nausea and vomiting or unconsciousness (e.g. diazepam in status epilepticus).



Parenteral administration

These routes avoid absorption via the gastrointestinal tract and first-pass metabolism in the liver:



• Intravenous (IV). The IV route enables all of a dose to enter the systemic circulation reliably, without any concerns about absorption or first-pass metabolism (i.e. the dose is 100% bioavailable), and rapidly achieve a high plasma concentration. It is ideal for very ill patients when a rapid, certain effect is critical to outcome (e.g. benzylpenicillin for meningococcal meningitis).


• Intramuscular (IM). IM administration is easier to achieve than the IV route (e.g. adrenaline (epinephrine) for acute anaphylaxis) but absorption is less predictable and depends on muscle blood flow.


• Subcutaneous (SC). The SC route is ideal for drugs that have to be administered parenterally because of low oral bioavailability, are absorbed well from subcutaneous fat, and might ideally be injected by patients themselves (e.g. insulin, heparin).


• Transdermal. A transdermal patch can enable a drug to be absorbed through the skin and into the circulation (e.g. oestrogens, testosterone, nicotine, nitrates).



Other routes of administration



• Topical application of a drug involves direct administration to the site of action (e.g. skin, eye, ear). This has the advantage of achieving sufficient concentration at this site while minimising systemic exposure and the risk of adverse effects elsewhere.


• Inhaled (INH) administration allows drugs to be delivered directly to a target in the respiratory tree, usually the small airways (e.g. salbutamol, beclometasone). However, a significant proportion of the inhaled dose may be absorbed from the lung or is swallowed and can reach the systemic circulation. The most common mode of delivery is the metered-dose inhaler but its success depends on some degree of manual dexterity and timing (see Fig. 19.23, p. 670). Patients who find these difficult may use a ‘spacer’ device to improve drug delivery. A special mode of inhaled delivery is via a nebulised solution created by using pressurised oxygen or air to break up solutions and suspensions into small aerosol droplets that can be directly inhaled from the mouthpiece of the device.



Drug distribution


Distribution is the process by which drug molecules transfer into and out of the blood stream. This is influenced by the drug’s molecular size and lipid solubility, the extent to which it binds to proteins in plasma, its susceptibility to drug transporters expressed on cell surfaces, and its binding to its molecular target and to other cellular proteins (which can be irreversible). Most drugs diffuse passively across capillary walls down a concentration gradient into the interstitial fluid until the concentration of free drug molecules in the interstitial fluid is equal to that in the plasma. As drug molecules in the blood are removed by metabolism or excretion, the plasma concentration falls and drug molecules diffuse back from the tissue compartment into the blood, and eventually all will be eliminated. Note that this reverse movement of drug away from the tissues will be prevented if further drug doses are administered and absorbed into the plasma.



Volume of distribution

The apparent volume of distribution (Vd) is the volume into which a drug appears to have distributed following intravenous injection. It is calculated from the equation


Vd=D/Co


image

where D is the amount of drug given and C0 is the initial plasma concentration (Fig. 2.4A). Drugs that are highly bound to plasma proteins may have a Vd below 10 L (e.g. warfarin, aspirin), while those that diffuse into the interstitial fluid but do not enter cells because they have low lipid solubility may have a Vd between 10 and 30 L (e.g. gentamicin, amoxicillin). It is an ‘apparent’ volume because those drugs that are lipid-soluble and highly tissue-bound may have a Vd of greater than 100 L (e.g. digoxin, amitriptyline). Drugs with a larger Vd are eliminated more slowly from the body.




Drug elimination



Drug metabolism

Metabolism is the process by which drugs are chemically altered from a lipid-soluble form suitable for absorption and distribution to a more water-soluble form that is necessary for excretion. Some drugs, known as ‘pro-drugs’, are inactive in the form in which they are administered, but are converted to an active metabolite in vivo.


Phase I metabolism involves oxidation, reduction or hydrolysis to make drug molecules suitable for phase II reactions or for excretion. Oxidation is much the commonest form of phase I reaction and chiefly involves members of the cytochrome P450 family of membrane-bound enzymes in the endoplasmic reticulum of hepatocytes.


Phase II metabolism involves combining phase I metabolites with an endogenous substrate to form an inactive conjugate that is much more water-soluble. Reactions include glucuronidation, sulphation, acetylation or methylation, and conjugation with glutathione. This is necessary to enable renal excretion because lipid-soluble metabolites will simply diffuse back into the body after glomerular filtration (p. 430).



Drug excretion

Excretion is the process by which drugs and their metabolites are removed from the body.


Renal excretion is the usual route of elimination for drugs or their metabolites that are of low molecular weight and sufficiently water-soluble to avoid reabsorption from the renal tubule. Drugs bound to plasma proteins are not filtered by the glomeruli. The pH of the urine is more acidic than that of plasma, so that some drugs (e.g. salicylates) become un-ionised and tend to be reabsorbed. Alkalination of the urine can hasten excretion (e.g. after a salicylate overdose). For some drugs, active secretion into the proximal tubule lumen, rather than glomerular filtration, is the predominant mechanism of excretion (e.g. methotrexate, penicillin).


Faecal excretion is the predominant route of elimination for drugs with high molecular weight, including those that are excreted in the bile after conjugation with glucuronide in the liver, and any drugs that are not absorbed after enteral administration. Molecules of drug or metabolite that are excreted in the bile enter the small intestine, where they may, if they are sufficiently lipid-soluble, be reabsorbed through the gut wall and return to the liver via the portal vein (see Fig. 2.3). This recycling between the liver, bile, gut and portal vein is known as ‘enterohepatic circulation’ and can significantly prolong the residence of drugs in the body.



Elimination kinetics

The net removal of drug from the circulation results from a combination of drug metabolism and excretion, and is usually described as ‘clearance’, i.e. the volume of plasma that is completely cleared of drug per unit time.


For most drugs, elimination is a high-capacity process that does not become saturated, even at high dosage. The rate of elimination is therefore directly proportional to the drug concentration because of the ‘law of mass action’, whereby higher drug concentrations will drive faster metabolic reactions and support higher renal filtration rates. This results in ‘first-order’ kinetics, when a constant fraction of the drug remaining in the circulation is eliminated in a given time and the decline in concentration over time is exponential (see Fig. 2.4A). This elimination can be described by the drug’s half-life (t1/2), i.e. the time taken for the plasma drug concentration to halve, which remains constant throughout the period of drug elimination. The significance of this phenomenon for prescribers is that the effect of increasing doses on plasma concentration is predictable – a doubled dose leads to a doubled concentration at all time points.


For a few drugs in common use (e.g. phenytoin, alcohol), elimination capacity is exceeded (saturated) within the usual dose range. This is called ‘zero-order’ kinetics. Its significance for prescribers is that, if the rate of administration exceeds the maximum rate of elimination, the drug will accumulate progressively, leading to serious toxicity.



Repeated dose regimens


The goal of therapy is usually to maintain drug concentrations within the therapeutic range (see Fig. 2.2) over several days (e.g. antibiotics) or even for months or years (e.g. antihypertensives, lipid-lowering drugs, thyroid hormone replacement therapy). This goal is rarely achieved with single doses, so prescribers have to plan a regimen of repeated doses. This involves choosing the size of each individual dose and the frequency of dose administration.


As illustrated in Figure 2.4B, the time taken to reach drug concentrations within the therapeutic range depends on the half-life of the drug. Typically, with doses administered regularly, it takes approximately 5 half-lives to reach a ‘steady state’ in which the rate of drug elimination is equal to the rate of drug administration. This applies when starting new drugs and when adjusting doses of current drugs. With appropriate dose selection, steady state drug concentrations will be maintained within the therapeutic range. This is important for prescribers because it means that the effects of a new prescription, or dose titration, for a drug with a long half-life (e.g. digoxin – 36 hours) may not be known for a few days. In contrast, drugs with a very short half-life (e.g. dobutamine – 2 minutes) have to be given continuously by infusion but reach a new steady state within minutes.


For drugs with a long half-life, if it is unacceptable to wait for 5 half-lives until concentrations within the therapeutic range are maintained, then an initial ‘loading dose’ can be given that is much larger than the maintenance dose, and equivalent to the amount of drug required in the body at steady state. This achieves a peak plasma concentration close to the plateau concentration, which can then be maintained by successive maintenance doses.


‘Steady state’ actually involves fluctuations in drug concentrations, with peaks just after administration followed by troughs just prior to the next administration. The manufacturers of medicines recommend dosing regimens that predict that, for most patients, these oscillations result in troughs within the therapeutic range and peaks that are not high enough to cause adverse effects. The optimal dose interval is a compromise between convenience for the patient and a constant level of drug exposure. More frequent administration (e.g. 25 mg 4 times daily) achieves a smoother plasma concentration profile than 100 mg once daily but is much more difficult for patients to sustain. A solution to this need for compromise in dosing frequency for drugs with half-lives of less than 24 hours is the use of ‘modified-release’ formulations. These allow drugs to be absorbed more slowly from the gastrointestinal tract and reduce the oscillation in plasma drug concentration profile, which is especially important for drugs with a low therapeutic index (e.g. levodopa).



Inter-individual variation in drug responses


Prescribers have numerous sources of guidance about how to use drugs appropriately (e.g. dose, route, frequency, duration) for many conditions. However, this advice is based on average dose–response data derived from observations in many individuals. When applying this information to an individual patient, prescribers must take account of inter-individual variability in response. Some of this variability is predictable and good prescribers are able to anticipate it and adjust their prescriptions accordingly to maximise the chances of benefit and minimise harm. Inter-individual variation in responses also mandates that effects of treatment should be monitored (p. 39).


Some inter-individual variation in drug response is accounted for by differences in pharmacodynamics. For example, the beneficial natriuresis produced by the loop diuretic furosemide is often significantly reduced at a given dose in patients with renal impairment, while confusion caused by opioid analgesics is more likely in the elderly. However, differences in pharmacokinetics more commonly account for different drug responses. Examples of factors influencing the absorption, metabolism and excretion of drugs are shown in Box 2.4.



image 2.4   Patient-specific factors that influence pharmacokinetics












It is hoped that a significant proportion of the inter-individual variation in drug responses can be explained by studying genetic differences in single genes (‘pharmacogenetics’) (Box 2.5) or the effects of multiple gene variants (‘pharmacogenomics’). The aim is to identify those patients most likely to benefit from specific treatments and those most susceptible to adverse effects. In this way, it may be possible to select drugs and dose regimens for individual patients to maximise the benefit:hazard ratio (‘personalised medicine’).



image 2.5   Examples of pharmacogenetic variations that influence drug response













































































Genetic variant Drug affected Clinical outcome
Pharmacokinetic
Aldehyde dehydrogenase-2 deficiency Ethanol Elevated blood acetaldehyde causes facial flushing and increased heart rate in ~50% of Japanese, Chinese and other Asian populations
Acetylation Isoniazid, hydralazine, procainamide Increased responses in slow acetylators, up to 50% of some populations
Oxidation (CYP2D6) Nortriptyline Increased risk of toxicity in poor metabolisers
Codeine Reduced responses with slower conversion of codeine to more active morphine in poor metabolisers, 10% of European populations
Increased risk of toxicity in ultra-fast metabolisers, 3% of Europeans but 40% of North Africans
Oxidation (CYP2C18) Proguanil Reduced efficacy with slower conversion to active cycloguanil in poor metabolisers
Oxidation (CYP2C9) Warfarin Polymorphisms known to influence dosages
Oxidation (CYP2C19) Clopidogrel Reduced enzymatic activation results in reduced antiplatelet effect
Sulphoxidation Penicillamine Increased risk of toxicity in poor metabolisers
HLA-B*1502 Carbamazepine Increased risk of serious dermatological reactions (e.g. Stevens–Johnson syndrome) for 1 in 2000 in Caucasian populations (much higher in some Asian countries)
Pseudocholinesterase deficiency Suxamethonium (succinylcholine) Decreased drug inactivation leads to prolonged paralysis and sometimes persistent apnoea requiring mechanical ventilation until the drug can be eliminated by alternate pathways in 1 in 1500 people
Pharmacodynamic
Glucose-6-phosphate dehydrogenase (G6PD) deficiency Oxidant drugs including antimalarials (e.g. chloroquine, primaquine) Risk of haemolysis in G6PD deficiency
Acute intermittent porphyria Enzyme-inducing drugs Increased risk of an acute attack
SLC01B1 polymorphism Statins Increased risk of rhabdomyolysis
HLA-B*5701 polymorphism Abacavir Increased risk of skin hypersensitivity reactions
HLA-B*5801 polymorphism Allopurinol Increased risk of rashes in Han Chinese
HLA-B*1502 polymorphism Carbamazepine Increased risk of skin hypersensitivity reactions in Han Chinese


image


(HLA = human leucocyte antigen)




Adverse outcomes of drug therapy


The decision to prescribe a drug always involves a judgement of the balance between therapeutic benefits and risk of an adverse outcome. Both prescribers and patients tend to be more focused on the former but a truly informed decision requires consideration of both.



Adverse drug reactions


Some important definitions for the adverse effects of drugs are:



• Adverse event. A harmful event that occurs while a patient is taking a drug, irrespective of whether the drug is suspected of being the cause.


• Adverse drug reaction (ADR). An unwanted or harmful reaction that is experienced following the administration of a drug or combination of drugs under normal conditions of use and is suspected to be related to the drug. An ADR will usually require the drug to be discontinued or the dose reduced.


• Side-effect. Any effect caused by a drug other than the intended therapeutic effect, whether beneficial, neutral or harmful. The term ‘side-effect’ is often used interchangeably with ‘ADR’, although the former usually implies an effect that is less harmful, is predictable and may not require discontinuation of therapy (e.g. ankle oedema with vasodilators).


• Drug toxicity. Adverse effects of a drug that occur because the dose or plasma concentration has risen above the therapeutic range, either unintentionally or intentionally (drug overdose, see Fig. 2.2, p. 19).


• Drug abuse. The misuse of recreational or therapeutic drugs that may lead to addiction or dependence, serious physiological injury (such as liver damage), psychological harm (abnormal behaviour patterns, hallucinations, memory loss) or death (p. 240).




Prevalence of ADRs

ADRs are a common cause of illness, accounting in the United Kingdom (UK) for approximately 3% of consultations in primary care and 7% of emergency admissions to hospital, and affecting around 15% of hospital inpatients. Many ‘disease’ presentations are eventually attributed to ADRs, emphasising the importance of always taking a careful drug history (Box 2.6). Factors accounting for the rising prevalence of ADRs are the increasing age of patients, polypharmacy (higher risk of drug interactions), increasing availability of over-the-counter medicines, increase in use of herbal or traditional medicines, and increase in medicines available via the Internet. Risk factors for ADRs are shown in Box 2.7.



Apr 9, 2017 | Posted by in GENERAL SURGERY | Comments Off on Therapeutics and good prescribing

Full access? Get Clinical Tree

Get Clinical Tree app for offline access