Chapter 10 Adverse Drug Reactions and Drug Interactions

Chapter focus

The use of medications to alleviate ailments and to combat diseases has been common for centuries. Along with the development of therapeutic remedies came knowledge of the poisonous qualities of the ingredients. In modern times this evolved into the concept of adverse drug reactions and knowledge that in some instances these are dose-related (type A adverse drug reactions) but under other circumstances are unpredictable (type B adverse drug reactions). With the increasing use of multiple drug therapies, drug interactions are now a cause for concern because a drug interaction may result in loss of effi cacy (decreased effect) or the development of toxicity (enhanced effect). Many studies have confi rmed that adverse drug reactions and drug interactions are major clinical problems, accounting for a signifi cant number of hospital admissions, extended hospital stays and substantial costs to the health-care system. It is important that health-care professionals be aware of the adverse reaction and drug interaction profi les of drugs and be ever vigilant for the occurrence of adverse outcomes.

Key terms

adverse drug reaction
adverse drug event
adverse effect
drug interactions
pseudoallergic reaction
St John’s wort
type A adverse drug reaction
type B adverse drug reaction
type C adverse drug reaction
type D adverse drug reaction
type E adverse drug reaction
type F adverse drug reaction

Key abbreviations

ADE adverse drug event

ADR adverse drug reaction

DDI drug–drug interaction

HERBS and medicinal products have been used for treating ailments and diseases for centuries. Ebers Papyrus (1550 BC) lists more than 700 remedies and describes in detail the procedures for preparing and administering them. Some of the individuals treated would certainly have suffered an adverse reaction, especially with the use of lizard’s blood, domestic animal excreta, tortoise bile, cat uterus and perhaps with the more reasonable ingredients of castor oil, squill and opium.

The relationship between a remedy and a poison was entrenched in the early teachings of the Chinese and Mayans. People learnt to avoid medicinal substances that were poisonous. The ancient Greeks later described the concept of a medicine and a poison in scientific terms, and in 1758 William Withering described both the therapeutic benefits and the adverse effects of digitalis. Throughout the centuries, the use of medicinal products has gone hand in hand with reports of adverse drug reactions (ADRs).

Public concern about ADRs arose in the late 19th century because of the number of sudden deaths associated with the use of chloroform. This led to the development of investigative committees such as the Food and Drug Administration (FDA) in the USA, which establishes the safety of new drugs before marketing. Despite regulatory frameworks, there have been many notable incidences of ADRs that have resulted in withdrawal of the offending drug (Table 10-1). Public interest in the safety of drugs has increased as a result of better communication between consumers and health-care professionals.

Table 10-1 Notable incidences of adverse drug reactions necessitating withdrawal of the drug

image

Definitions

An adverse drug reaction (ADR) has been defined as ‘any response to a drug which is noxious, unintended, and which occurs at doses normally (and appropriately) used in man for the prophylaxis, diagnosis, or therapy of disease’ (World Health Organization 1984). This definition has been in use for the last 26 years and at various times has been modified slightly, because the word ‘noxious’ is perhaps not correct in the context of the definition. An alternative definition has been proposed: ‘an appreciably harmful or unpleasant reaction, resulting from an intervention related to the use of a medicinal product, which predicts hazard from future administration and warrants prevention or specific treatment, or alteration of the dosage regimen, or withdrawal of the product’ (Edwards & Aronson 2000). Clinical responses to an ADR include modifying the dose, discontinuing the drug, hospitalising the patient or providing supportive measures. This definition does not encompass the situations of drug overdose, drug withdrawal, drug abuse or error in administration. The latter is included within the definition of an adverse drug event.

An adverse drug event (ADE) is defined as an ‘injury resulting from medical intervention related to a drug’ (Bates et al 1995): simply stated, an adverse event occurs while a person is taking a drug, but it is not necessarily ‘due to the drug’. Examples of ADEs include under- or overmedication resulting from misuse or malfunction of infusion pumps or devices; aspiration pneumonia resulting from drug overdose; and errors in ordering, dispensing or administration. These incidents are usually investigated and may be attributed to simple mistakes such as picking up the wrong syringe, giving treatment to the wrong body site or giving the wrong treatment. Under WHO guidelines, these types of incidents are not classed as ADRs.

The term ‘side effect’ is often used by health-care professionals and often appears in drug advertisements and consumer information. Often this term is interpreted by individuals to mean that the adverse reaction is insignificant (non-deleterious) or medically trivial and in general acceptable. In fact a side effect occurs via a mechanism different from the pharmacological effect and may be dose-related or not dose-related. An example of a side effect is the dose-related anticholinergic effect of tricyclic antidepressants as this is not associated with the therapeutic effect. The term ‘adverse effect’ is preferable to side effect, and generally relates to an unwanted effect that occurs via a different mechanism to the pharmacological effect and may or may not be dose-related. As an example, anaphylaxis with penicillin is both an adverse effect and an adverse drug reaction. The difference is that an adverse effect is ‘seen from the point of view of the drug, whereas an adverse reaction is seen from the point of view of the patient’ (Edwards & Aronson 2000). Throughout the literature, however, despite a difference in their definitions, in the disciplines of pharmacology, pharmacy and clinical toxicology the terms adverse effect and adverse drug reaction are used interchangeably.

Incidence of adverse drug reactions

Over the period 2005–2006 in Australia the total expenditure on medications was $11,501 million, of which $10,551 million was on prescription pharmaceuticals (Australian Institute of Health and Welfare 2008). In most developed countries the elderly take on average 4–5 prescription drugs and 2 OTC drugs at any one time. Taking multiple types of drugs contributes to the incidence of ADRs. In addition to prescribed medications and those bought over the counter, Australians have embraced the use of complementary and alternative medicines. The trend has been strongest among women, and includes the use of herbal medicines, aromatherapy oils and ginseng. The estimated annual retail sales of complementary medicines in Australia are estimated to be in the range of $800 million to $1 billion.

ADRs occur in people of all ages and are twice as common in women. They are a major cause of morbidity and mortality, especially in the elderly. A survey conducted in a major New South Wales teaching hospital found that 30% of elderly patients were taking 6–10 types of medications and 13% took more than 10 types each day (Nair 1999). A review of Australian studies published over the period 1988–1996 found that 2.4%–3.6% of all hospital admissions were reported to be drug-related, and 6%–7% of emergency admissions, 12% of all admissions to medical wards and 15%–22% of all emergency admissions among the elderly were drug-related. Between 32% and 69% of drug-related admissions were reported as definitely or possibly preventable (Roughead et al 1998). The drugs most commonly implicated were antihypertensives, anticoagulants, cardiovascular drugs, cytotoxics and nonsteroidal anti-inflammatory drugs (NSAIDs). Cytotoxic and anticoagulant agents were also the drugs most often involved in serious ADRs in general practice settings in France (Lacoste-Roussillon et al 2001). Similarly the drugs most commonly implicated in fatal ADRs in Sweden are antithrombotic drugs, NSAIDs, antidepressants and cardiovascular drugs (Wester et al 2007).

Little attention has been paid to the incidence of ADRs in neonates, infants, children and adolescents. Before release of a new drug, few if any studies are undertaken in children because of questions of ethics, responsibility, cost and regulations. This often leads prescribers to estimate dosage and hence increases the risk of ADRs. A recent analysis of prospective paediatric studies from the UK, USA and Spain reported that the incidence of ADRs in hospitalised children was 9.53%. The overall rate of hospital admissions due to ADRs was 2.09%, of which 39.3% involved life-threatening reactions. In the outpatient setting the overall incidence of ADRs in children was 1.46% (Impicciatore et al 2001). These data clearly show ADRs as a significant health issue in children.

Classification of adverse drug reactions

The current classification system is not ideal, and not every ADR may fit perfectly into one of the categories. It is generally accepted that there are two main categories of ADR, type A (augmented) and type B (bizarre), and two subordinate categories, type C (chronic) and type D (delayed). A further two classes include end-of-use or withdrawal effects (type E) and unexpected failure of therapy (type F).

TYPE A (AUGMENTED, DOSE-RELATED) ADRs are characterised by:

predictability from the known pharmacology of the drug (often an exaggeration of effect)
relationship to dose
common occurrence (about 80% of ADRs)
usually mild
high morbidity and low mortality
reproducibility in animals.

Factors predisposing to type A reactions include the dose, pharmaceutical variation in drug formulation, pharmacokinetic variation (e.g. renal failure), pharmacodynamic variation (e.g. altered fluid and electrolyte balance) and drug–drug interactions (e.g. inhibition of metabolism of one drug by another concomitantly administered drug). Examples include:

sedation with the use of antihistamines
bleeding with anticoagulants
hypoglycaemia from the use of insulin
hypokalaemia with the use of diuretics.

TYPE B (BIZARRE, NON-DOSE-RELATED) ADRs are characterised by:

unpredictability
no relationship to dose
uncommon occurrence (about 20% of ADRs)
increased severity
high morbidity and high mortality
lack of reproducibility in animals.

These reactions are less common but often cause death. Factors contributing to type B reactions include pharmaceutical variation, receptor abnormalities, unmasking of a biological deficiency (e.g. glucose-6-phosphate dehydrogenase deficiency), abnormalities in drug metabolism (e.g. slow acetylators of the antituberculosis drug isoniazid), drug allergy (see next section) and drug–drug interactions (e.g. rare incidence of hepatitis) (Pirmohamed et al 1998). Examples include interstitial nephritis with the use of NSAIDs and eosinophilia with the use of anticonvulsants such as carbamazepine and phenytoin.

TYPE C (CHRONIC, DOSE-RELATED AND TIME-RELATED) ADRs are characterised by occurrence as a consequence of longterm use. Examples of reactions in this category include:

adaptive changes (e.g. development of drug tolerance and physical dependence)
appearance of tardive dyskinesia in persons treated long-term with neuroleptic drugs for schizophrenia
rebound phenomena (e.g. rebound tachycardia after the abrupt discontinuation of β-blockers and acute adrenal insufficiency after abrupt withdrawal of corticosteroids).

TYPE D (DELAYED, TIME-RELATED) ADRs are characterised by the appearance of delayed effects. These may be acceptable if the benefit of drug therapy outweighs the risk, as in the case of irreversible infertility in young persons receiving cytotoxic drugs for malignancies. In general, however, they are considered unacceptable. Examples include carcinogenesis (e.g. the association of lymphoma with immunosuppressive drugs) and teratogenesis.

TYPE E (END-OF-USE, WITHDRAWAL) ADRs are fortunately uncommon and are related to withdrawal of a drug. They include opiate withdrawal syndrome and myocardial ischaemia after abrupt cessation of β-blockers.

TYPE F (FAILURE, UNEXPECTED FAILURE OF THERAPY) ADRs are increasingly common and are often caused by a drug interaction (e.g. inadequate dose of the oral contraceptive when a drug that induces the metabolism of oestrogen is administered concomitantly).

Drug allergy

A drug allergy, or hypersensitivity, is a type B ADR. Drug allergies are characterised by:

occurrence in a small number of individuals
the requirement for previous exposure to either the same or a chemically related drug
the rapid development of an allergic reaction after re-exposure
the production of clinical manifestations of an allergic reaction (deShazo & Kemp 1997).

The diagnosis of a drug allergy is often difficult to establish because there are no reliable laboratory tests that can identify the relevant drug, and in some cases the symptoms can imitate infectious disease symptoms. The situation may be easier if the drug administered is notorious for producing an allergic reaction (e.g. penicillin), but it is difficult if the drug used is seldom reported to produce an allergic reaction.

Some drugs can produce a pseudoallergic reaction, i.e. one resembling an allergic reaction but for which there is no immunological basis. Usually these occur as a result of mast cell degranulation and subsequent release of histamine. Clinically they resemble the type I hypersensitivity reaction but they do not involve drug-specific IGE. An example of a pseudoallergic reaction is the release of histamine that occurs with opiates (e.g morphine), vancomycin and radiocontrast media.

Allergic reactions to drugs generally follow the type I–IV classification (see Chapter 47). Table 10-2 lists the types of reactions, the main clinical manifestation and examples of drugs commonly implicated.

Table 10-2 Allergic drug reactions

Type/reaction Clinical manifestations Examples of drugs
I Immediate hypersensitivity Urticaria, anaphylaxis, angio-oedema, bronchospasm Penicillins, streptomycin, local anaesthetics, neuromuscular blocking drugs, radiological contrast media
II Antibody-dependent cytotoxic Cytopenia, vasculitis, haemolytic anaemia Quinine, quinidine, rifampicin, metronidazole
III Complex-mediated Serum sickness, vasculitis, interstitial nephritis Anticonvulsants, antibiotics, hydralazine, diuretics
IV Cell-mediated or delayed hypersensitivity Contact sensitivity Local anaesthetic creams, antihistamine creams

Immune-modulating drugs and adverse drug reactions

The use of drugs that modulate the immune system is common for the treatment of diseases such as cancer, rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease and lupus. Although beneficial in many clinical settings the use of immune-modulating drugs is also associated with a significant number of ADRs (Table 10.3). Suppression of the immune response increases the risk of infection and cancer and paradoxically some of these drugs can also induce hypersensitivity (type B reactions) and/or autoimmune syndromes (Uetrecht 2009).

Table 10-3 Commonly used immune-modulating drugs and associated adrs

Immune-modulating Drug Therapeutic effect Adverse Drug Reactions
Corticosteroids Immune suppression; decreased expression of nitric oxide synthase; decreased expression of intracellular adhesion molecules Increased risk of infections such as Pneumocystis carinii and systemic fungal infections
Cytotoxic drugs Inhibition of cell proliferation Increased risk of infections (e.g. herpes zoster, cytomegalolvirus) and life-threatening infections; increased risk of malignancies
Calcineurin inhibitors Immune suppression Increased risk of lymphomas and nonmelanoma skin cancers
Protein biological agents e.g. antibodies, interferons, antiadhesion molecules Immune-modulating Formation of autoantibodies; increased risk of infection and malignancies e.g. lymphomas (infliximab); progressive multifocal leukoencephalopathy (rituximab)
Sphingosine agonists Immune suppression Increased risk of infections

Risk factors for developing an adverse drug reaction

Risk factors for ADRs are specific to both the person and the drug.

Factors relating to the person include:

age—the elderly and neonates have a higher incidence
gender—women appear to be more susceptible
concurrent disease—an association between viral infections, drug use and skin reactions has been described
genetic factors—deficiency of an enzyme involved in the metabolism of a drug may increase the risk of an ADR
history of prior drug reaction—some individuals appear to be more susceptible to allergic drug reactions, including people with a history of atopic disease (e.g. asthma, hay fever, eczema).

Factors specific to the drug include:

chemical characteristics—large molecules such as heparin can themselves be immunogenic, and smaller drug molecules or their metabolites can combine with body proteins to form antigens that elicit an allergic response
route of drug administration—topical and oral routes generally involve a lower incidence of drug allergy; topical application tends to cause a delayed hypersensitivity reaction
dose—many ADRs are dose-related
duration and frequency—prolonged and frequent therapy can increase the likelihood of an ADR.

Pharmacogenomics and adverse drug reactions

The role of genetics in predisposing individuals to ADRs has been known since the late 1950s through the discovery of enzyme deficiencies such as pseudocholinesterase and the link to succinylcholine apnoea. This easily recognised and diagnosed enzyme deficiency led many to conclude that we would be able to identify using genetic tests those individuals that were more likely to develop ADRs to some drugs. This concept although attractive has been replaced by the realisation that pharmacogenomics will aid in the prediction and prevention of only a limited number of ADRs, and as such the uptake of pharmacogenetic testing has been rather variable and limited (Alfirevic & Pirmohamed 2008). The era of pharmacogenomics and the prescribing of medications tailored to individual genotypes is on the horizon but genotyping would remove only one of the factors contributing to the risk of ADRs, leaving age, a history of atopy, polypharmacy and inappropriate prescribing to deal with. Examples of pharmacogenetic tests for the prevention of ADRs include:

testing for the human leucocyte antigen HLA-B*5701 to identify individuals at risk of developing hypersensitivity reaction to the antiretroviral drug abacavir
testing for glucose-6-phosphate dehydrogenase (G6PD) deficiency, an essential erythrocyte enzyme that protects erythrocytes from oxidative stress-induced haemolysis. Drugs that induce haemolytic anaemia in G6PD deficient individuals include primaquine, sulfamethoxazole, dapsone, nalidixic acid and nitrofurantoin
testing for thiopurine methyl transferase deficiency to identify individuals at risk of azathioprine and 6-mercaptopurine toxicity
testing for UGT1A1 deficiency to identify individuals at risk of developing irinotecan-induced diarrhoea and leucopenia.

Drug–drug interactions

A drug–drug interaction (commonly shortened to ‘drug interaction’ or DDI) occurs when the drug’s pharmacological effect is altered by another drug: that is, an increased therapeutic and/or adverse effect or a decreased therapeutic and/or adverse effect. Moreover, drug interactions often are unanticipated and go unrecognised and the clinical and economic importance is frequently underestimated.

Frequency of drug interactions

The exact frequency of drug interactions is unknown, although anecdotal evidence suggests that they are relatively common and result in a significant number of hospital admissions. The possibility of a drug interaction exists whenever two or more medications are prescribed to an individual, and the likelihood of an interaction will grow as the number of medications used increases. For a person taking two drugs the estimated incidence of a DDI is 5.6% while a person taking six drugs has an estimated incidence of a DDI of 56%. One of the problems in identifying a DDI is that the physiological/biochemical changes resulting from a DDI may be masked by, or confused with, the clinical signs and symptoms of the illness or other comorbidities. Additionally, it may be difficult to identify which ‘drugs’ are involved in a DDI especially if the prescriber is unaware of other medications the patient is taking, for example OTC drugs and/or complementary/alterative medications.

Individuals at greatest risk of a drug interaction are:

the severely ill, who typically receive multiple drugs
individuals receiving chronic therapy, often comprising a cocktail of drugs (e.g. in the treatment of either HIV infection or cancer)
the elderly, who tend to have multiple pathologies and often receive multiple drugs concurrently.

Drug interactions are of greatest concern with drugs that have a narrow therapeutic index. Even a small change in the concentration of the drug available at the target site (e.g. receptor, enzyme) can lead to a major alteration in response. For example:

enhanced anticoagulation (bleeding) with warfarin resulting from concomitant use of the antiarrhythmic drug amiodarone, which inhibits the metabolism of warfarin
bradycardia with digoxin resulting from concomitant administration of the antiarrhythmic drug quinidine, which decreases renal or biliary excretion of digoxin.

Interactions involving drugs with a wide therapeutic index (e.g. penicillin antibiotics, β-adrenoceptor antagonists) cause fewer problems. Knowledge of the mechanisms of drug interactions is essential to enable health professionals to prevent interactions occurring (wherever possible) and to systematically analyse potentially new drug interactions. Indeed, analysis of known and potential interactions is critical in the planning of a therapeutic regimen.

Although drug interactions may have deleterious effects, they may also be used to advantage. For example, antimicrobial drugs with different mechanisms of action are commonly used in combination for increased effectiveness in treating bacterial infections. Similarly, combinations of drugs are commonly used in cancer chemotherapy and in the treatment of tuberculosis.

Classification of drug interactions

Drug interactions are broadly classified, according to their pharmacological mechanism, into either pharmacodynamic or pharmacokinetic interactions.

Pharmacodynamic drug interactions

Pharmacodynamic drug interactions may be ‘direct’ or ‘indirect’. Direct pharmacodynamic interactions involve effects at a common target, additively (and possibly potentiation) or antagonism due to actions at different sites in an organ. An example of antagonism at a common receptor site is the concurrent use of a β2-adrenoceptor agonist (used in the treatment of asthma, e.g. salbutamol) and a non-selective β-adrenoceptor antagonist (used in the treatment of hypertension, e.g. propranolol). Both drugs have opposing effects at the same receptor (i.e. the β2-adrenoceptor). Unintentional interactions of this type should not occur because they are so obvious from the known pharmacology of the drugs.

Examples of direct pharmacodynamic interactions involving drugs with different mechanisms of action include the following:

Monoamine oxidase (MAO) inhibitors, which are used in the treatment of depression and which increase the amount of noradrenaline stored in nerve terminals, interact dangerously (to cause marked hypertension) with ‘sympathomimetic’ drugs such as ephedrine that cause the release of stored noradrenaline. The tyramine present in foods such as cheese, yeast extracts and Chianti-type wines produces a similar response in patients treated with MAO inhibitors.
Warfarin is an anticoagulant that inhibits vitamin K-mediated synthesis of clotting factors. The risk of bleeding is increased by coadministration of aspirin, which decreases platelet aggregation by inhibiting the synthesis of thromboxane A2.
Combinations of CNS depressants enhance drowsiness, even when they act at different sites within the CNS, e.g. combinations of alcohol (ethanol), ‘sedating’ histamine-1-receptor antagonists (antihistamines), benzodiazepine hypnosedatives/anxiolytics, tricyclic antidepressants and antipsychotic drugs.

An indirect pharmacodynamic interaction occurs when the pharmacological effects of one drug alter the response to another drug, even though the two effects are not themselves directly related. Common examples include the following:

Certain diuretics (e.g. frusemide or hydrochlorothiazide) lower the blood potassium concentration. This will enhance the toxic effects of the cardiac glycoside digoxin, which is used in the treatment of atrial fibrillation and cardiac failure, and of type III antiarrhythmic drugs (e.g. amiodarone) that prolong the cardiac action potential.
Apart from reducing renal blood flow, NSAIDs promote the retention of salt and water by reducing prostaglandin-mediated effects on the action of antidiuretic hormone and the reabsorption of chloride ions. This, in turn, may reduce the effectiveness of antihypertensive drugs.

Pharmacokinetic drug interactions

The plasma concentration of a drug may be altered by interactions occurring during absorption, distribution, metabolism and excretion:

Absorption. Absorption interactions involve a change in either the rate or the extent of absorption. Drugs that change the rate of gastric emptying (i.e. the time it takes for the contents of the stomach to empty into the small bowel) will alter the rate of absorption of coadministered drugs. Muscarinic receptor antagonists (e.g. hyoscine) delay gastric emptying and gastrointestinal motility. This combination of effects delays drug absorption from the gastrointestinal tract. Many drugs, including tricyclic antidepressants and histamine-1-receptor antagonists, that possess antimuscarinic properties (sometimes referred to as ‘anticholinergic adverse effects’) delay the absorption of coadministered drugs. Gastric emptying rate is slowed by opioid drugs, including morphine and pethidine, and hence the time to peak plasma concentration is generally increased for a drug coadministered with an opioid.

Coadministered drugs may also decrease the extent of drug absorption. Whereas changes in the rate of absorption generally affect only the time for onset of action, changes in extent of absorption can alter response. For example, cholestyramine is a bile acidbinding resin used in the treatment of hypercholesterolaemia. Unfortunately, cholestyramine also binds other drugs, reducing the amount of drug that is absorbed. As cholestyramine reduces the absorption of corticosteroids, digoxin, thyroxine and warfarin (and probably other drugs), these drugs should be administered either several hours before or after the cholestyramine dose.

Distribution. As many drugs circulate in the blood bound (at least in part) to the proteins albumin and α1-acid glycoprotein, they may compete for the same binding sites. Displacement from plasma protein of one drug by another is common, and this leads to an increase in the unbound, pharmacologically active, concentration of the drug in the blood. Although it is still widely believed that the increase in unbound concentration arising from ‘displacement interactions’ may precipitate drug toxicity, this is rarely the case. Following a drug displacement interaction, the concentration of unbound drug in blood does indeed increase. However, the unbound drug is available for distribution into tissues, leading to an increase in the volume of distribution, hepatic clearance and renal excretion. There is, however, a decrease in total drug concentration (i.e. bound plus unbound drug) because of the higher clearance.
Metabolism. As discussed in Chapter 6, administration of some drugs can lead to decreased (inhibited) or increased (induced) activity of drugmetabolising enzymes such as cytochrome P450 (CYP). Many important drug interactions arise from altered metabolism, and the clinical importance of the interaction will depend on the change in clearance and the therapeutic index of the altered drug. A 10% change in clearance is unlikely to be important, but a 30% change in the clearance of a narrow-therapeutic-index drug like warfarin can have serious implications. Importantly, just as there is considerable interindividual variability in the clearance of metabolised drugs, there is significant variability in the magnitude of the change in clearance associated with any metabolic drug interaction.

Induction results in increased enzyme activity and drug metabolism, hence the steady-state blood concentration will decrease with the possibility of therapeutic failure (see Chapters 6 and 8)

Drugs known to cause induction are generally nonselective in their effects on CYP enzymes. Examples include:

the antituberculosis drug rifampicin, which appears to induce all CYP- and UDPglucuronosyl transferase (UGT) isoforms, therefore potentially decreasing the blood concentration of all coadministered drugs that are metabolised by these enzymes
the anticonvulsant drugs phenobarbitone, phenytoin and carbamazepine, which induce CYP2C9 and CYP3A4, and possibly other isoforms of CYP and UGT. Epileptic patients receiving these drugs are prone to drug interactions and their consequences (e.g. unwanted pregnancy due to enhanced metabolism of oral contraceptive steroids)
chronic consumption of ethanol (alcohol), which induces CYP2E1, although there are relatively few clinically used drugs that are metabolised by this enzyme.

Inhibitory drug interactions are relatively common, and inhibition of metabolism increases steady-state blood concentration and the likelihood of drug toxicity. Some drugs, notably the H2 antagonist cimetidine, inhibit the activity of most CYP enzymes (although UGT is unaffected). Conversely, probenecid inhibits most UGT enzymes (without affecting CYP). Most inhibitory interactions are relatively selective for one or a limited number of drug-metabolising enzymes, as they most commonly arise from competition for metabolism at the enzyme active site. It is generally not correct to refer to a drug as ‘an inhibitor of drug metabolism’. Rather, a drug will normally selectively inhibit the metabolism of other drugs for a limited number of enzymes, and this specificity of interaction is used to predict and interpret metabolic drug interactions. Some selective inhibitors of CYP enzymes are shown in Table 10-4. As an example, fluoxetine causes interactions with many drugs metabolised by CYP2D6 (e.g. other antidepressants and perhexiline), which generally requires a reduction of the dose. The clearances of drugs metabolised by CYP3A4 (see Clinical Interest Box 10-1) are similarly decreased by the commonly used antibiotic erythromycin, again generally requiring a dose reduction. Examples of CYP metabolic drug interactions include:

Amiodarone and its active metabolite desethlyamiodarone inhibit CYP2C9, which metabolises S-warfarin. This DDI increases the risk of major bleeding.
Paroxetine, a CYP2D6 inhibitor, reduces the plasma concentration of endoxifen, an active metabolite of tamoxifen used in breast cancer treatment.
Azole antifungals inhibit CYP3A decreasing clearance of some statins, which increases the risk of statin-induced myopathy.
Currently marketed antiretroviral protease inhibitors (e.g. ritonavir) inhibit CY3A4 and decrease hepatic clearance of CYP3A4 substrates (e.g. amlodipine) increasing the plasma concentration.

Table 10-4 Inhibitors of CYP enzymes

CYP Inhibitors
CYP1A2 Fluvoxamine, propranolol
CYP2C8 Gemfibrozil, trimethoprim
CYP2C9 Amiodarone, fluconazole, gemfibrozil, sulfonamide antibiotics
CYP2C19 Fluvoxamine, moclobemide
CYP2D6 Fluoxetine, paroxetine, perphenazine, quinidine, quinine, thioridazine
CYP3A4 Clarithromycin, diltiazem, erythromycin, indinavir, itraconazole, ketoconazole, nelfinavir, ritonavir, roxithromycin, verapamil

Clinical interest Box 10-1 Colchicine and CYP3a4 inhibitors

Colchicine is used in the treatment of acute gouty arthritis (see Chapter 47) and has a narrow therapeutic index and dose-limiting severe adverse effects that include nausea, vomiting, diarrhoea and abdominal pain. Metabolised by CYP3A4, colchicine is subject to multiple DDIs involving drugs that inhibit CYP3A4 and hence increase the plasma concentration of colchicine and risk of ADRs. Four cases of colchicine toxicity have been reported to the TGA, three of which were fatal. One of the fatal cases involved the concomitant use of clarithromycin (a CYP3A4 inhibitor) as part of therapy for Helicobacter pylori eradication. The patient developed multi-organ failure and massive myelosuppression.

Source: Australian Adverse Drug Reactions Bulletin, Volume 27, No. 5, October 2008.

Up-to-date information on CYP DDIs can be found at the Indiana University, Division of Clinical Pharmacology website (http://medicine.iupui.edu/clinpharm/ddis/).

Metabolic drug interactions also occur with other drug-metabolising enzymes. Probenecid is a ‘universal’ inhibitor of drug glucuronidation, and there is evidence to suggest that rifampicin, phenobarbitone, phenytoin and carbamazepine may induce numerous UGT enzymes. For example, fluconazole appears to inhibit only UGT2B7 (which metabolises morphine and zidovudine). In the case of morphine, a rise in the plasma concentration may result in respiratory depression.

A potentially fatal interaction occurs when azathioprine and allopurinol are coadministered. Allopurinol is an inhibitor of the enzyme xanthine oxidase, and is used in the treatment of gout and gouty arthritis. Azathioprine (used mainly in the treatment of cancer) is converted to an active metabolite, 6-mercaptopurine, which is subsequently cleared by xanthine oxidase. Coadministration of azathioprine and allopurinol leads to accumulation of 6-mercaptopurine, resulting in potentially life-threatening bone marrow suppression.

Excretion. Interactions may occur between drugs that are excreted by active transport systems in the kidney. The mechanism of such interactions is simply ‘competition’ for the same transporter. For example, the renal clearance of methotrexate is impaired by coadministration of probenecid or salicylates (and possibly other NSAIDs).

Metabolic drug interactions involving nutrients and herbal medicines

Although there is wide appreciation of drug interactions, interactions between nutrients and/or food components and herbal remedies are often not considered and, in fact, may be discounted. Chemicals present in food may alter the activity of drug-metabolising enzymes. Notable in this regard are chemicals present in grapefruit (but not orange) juice that inhibit the activity of CYP3A4 present in the gastrointestinal tract. (CYP3A4 is localised in both liver and small bowel.) The enzyme present in the small bowel appears to contribute significantly to the first-pass metabolism of numerous CYP3A4 substrates. Thus, the bioavailability of a number of drugs, such as cyclosporin, felodipine, midazolam, triazolam and verapamil, increases significantly when they are taken with grapefruit juice, which enhances the potential for toxicity (see Clinical Interest Box 10-2).

Clinical interest Box 10-2 Grapefruit juice–drug interactions

Food–drug interactions are potentially an everyday occurrence. Many scientific findings have been serendipitous and perhaps none more so than an unexpected observation during a clinical study that the grapefruit juice that was used to mask the taste of ethanol led to an increase in the oral bioavailability of the drug being studied. Subsequent research indicated that the effect was due to the inhibition of CYP3A4 activity in the intestine, which resulted in a reduction of presystemic metabolism and subsequent increased bioavailability. This interaction is most clinically relevant with certain calcium channel blockers (e.g. verapamil), saquinavir, cyclosporin, midazolam and triazolam, and may be important with cisapride.

Although the active components in grapefruit juice respon sible for the inhibition of enzyme activity have not yet been fully identified, the effect is observed with a single glass of juice and can persist for 24 hours: ‘Since grocers do not take a drug history, physicians, pharmacists and other health profes sionals should educate patients about consumption of grapefruit juice with medications’ (Bailey et al 1998).

Currently there is intense interest in the effects of herbal medicines on drug metabolism and the consequences of herb–drug interactions, which are based on the same pharmacokinetic and pharmacodynamic mechanisms as DDIs. Herbal medicines are used widely, particularly by women, given the perception that ‘natural’ products are a safe and effective alternative to pharmaceuticals. As plant products, herbal medicines typically contain hundreds of different chemicals; it is not surprising that some of these will alter the activity of drug-metabolising enzymes. In the USA the seven top-selling herbal medicines in descending order are ginkgo, St John’s wort, ginseng, kava, saw palmetto, garlic and echinacea (Bressler 2005).

Important in terms of drug interaction is St John’s wort, which is taken to treat the symptoms of depression. St John’s wort contains chemicals called hyperforins, which mimic the effects of rifampicin as an inducer of CYP enzymes. Consumption for two weeks significantly induces the activity of both hepatic and intestinal CYP3A4 and the intestinal transporter P-glycoprotein (see Chapter 6). Thus the clearances of amitriptyline, carbamazepine, cyclosporin, HIV protease inhibitors, warfarin and several other drugs have been shown to be increased in subjects taking St John’s wort, with risk of therapeutic failure. Similarly, the plasma concentrations of alprazolam and midazolam have been shown to be reduced in healthy volunteers taking St John’s wort. Furthermore, oral contraceptives contain ethinyloestradiol, which is metabolised by CYP3A4. Concomitant administration of St John’s wort increases the metabolism of ethinyloestradiol and unplanned pregnancy has been reported as an issue in women who use oral contraceptive steroids and St John’s wort. Not surprisingly given its use in depression, interactions with ‘synthetic’ antidepressants have been reported. The combination of St John’s wort and the serotonin re-uptake inhibitors (sertraline, paroxetine, nefazadone and venlafaxine) may result in headaches, changes in mental state, tremors, autonomic instability, GI upset, myalgias and motor restlessness (symptoms similar to central serotonin excess). These symptoms may be explained by inhibition of serotonin reuptake in the brain by St John’s wort.

There is evidence to suggest that multiple herbal products interact (pharmacodynamically or pharmacokinetically) with ‘pharmaceutical’ drugs, and studies investigating the mechanisms involved and quantifying the magnitudes of any interactions are ongoing. Interactions between herbal medicines and conventional drugs are no longer just a theoretical possibility, and it is likely that their incidence is more common than anticipated initially. It is essential that a patient’s exposure to herbal medicines be determined when assessing the potential for drug interactions (refer to Appendix 3).

Strategies for limiting adverse drug reactions and drug interactions

Hospital admissions for ADRs are a significant and expensive health problem worldwide. The evidence from numerous studies indicates that ADRs may be impossible to avoid completely, but given the high predictability of many ADRs, there is significant room for improvement in reducing their incidence. Strategies for reducing/limiting ADRs include:

careful history-taking, including drug allergies, types of allergies, and use of prescribed, over-the-counter and complementary/alternative medications
considering non-drug treatment options
correct and appropriate dosing using the lowest effective dose
frequent review of therapeutic goals and drug regimens
being familiar with potential ADRs of a drug to avoid misinterpretation of an ADR as a symptom of a new medical condition
avoiding polypharmacy and keeping the drug regimen simple
careful communication with the patient and/or carer to inform them of the risks and benefits of the proposed treatment and the need for adherence to the recommended dosing schedule.

There is an overwhelming amount of information published on drug interactions, and sources such as the Australian Medicines Handbook, CARM (see Clinical Interest Box 10-3) and the website of Professor D Flockhart, School of Medicine, Indiana University, are excellent. Continued education of health professionals is essential, as new drugs enter the marketplace on an annual basis. Specific strategies for limiting drug interactions (in addition to those listed above) include:

if possible avoiding drug combinations
spacing the dosing time to avoid the interaction
close laboratory or clinical monitoring for early evidence of the interaction
improving patient education on drug interactions.

Clinical interest Box 10-3 Centre for adverse reactions monitoring

image

Pharmacovigilance activities in New Zealand include the reporting of adverse drug reactions. Based in Dunedin, the Centre for Adverse Reactions Monitoring (CARM) collects and evaluates adverse reaction reports from health professionals. These are reported for medicines, herbal products and dietary supplements, and are held in a database of over 50,000 reports. Individual patients who suffer from lifethreatening adverse reactions have CARM warnings or danger alerts recorded against their National Health Index (NHI) numbers; the information is accessible through hospital systems and contributes to the safe use of medicines.

Anonymised data from various pharmacovigilance sources in New Zealand are fed into the database of the World Health Organization’s International Drug Monitoring Programme based in Uppsala, Sweden. CARM also provides feedback to the Medicines Adverse Reactions Committee which in turn makes recommendations to Medsafe, the regulatory medicines authority in New Zealand. Although the activities of CARM are focused on spontaneous reports, the Intensive Medicines Monitoring Programme (IMMP) monitors new medicines using a method called prescription event monitoring. IMMP operates in close collaboration with the spontaneous reporting programme and together they supplement the adverse reactions database.

Adapted from: http://carm.otago.ac.nz/CARM.asp [27 January 2010].

Our knowledge of adverse drug reactions and drug interactions continues to advance but our ability to apply this knowledge to patient populations seems to lag behind. A multidisciplinary approach involving the consumers, the prescribers and the carers is essential if we are to minimise both the personal and societal impact of ADRs and DDIs.

Key points

image

An adverse drug reaction is defined as any response to a drug that is noxious and unintended, and that occurs at doses normally (and appropriately) used for the prophylaxis, diagnosis or therapy of disease.
Public interest in the safety of drugs has increased as a result of better communication between patients and health-care professionals.
Taking multiple drugs contributes to the incidence of ADRs.
Adverse drug reactions occur in people of all ages and are twice as common in women. They are a major cause of morbidity and mortality, especially in the elderly.
There are two main categories of adverse drug reactions, type A (predictable) and type B (unpredictable), and subordinate categories, type C (chronic use), type D (delayed reactions), type E (end-of-use or withdrawal effects) and type F (unexpected failure of therapy).
The type B adverse drug reactions include immunological reactions such as drug allergy and hypersensitivity.
The diagnosis of a drug allergy can often be difficult to establish because there are no reliable laboratory tests that can identify the relevant drug, and the symptoms can sometimes imitate infectious disease symptoms (e.g. fever).
Risk factors for developing an ADR include age, gender, presence of concurrent disease, genetics, a history of prior drug reaction, the drug dose and the duration and frequency of drug use.
Drug interactions are broadly classified according to their pharmacological mechanisms: that is, pharmacodynamic or pharmacokinetic.
Pharmacodynamic drug interactions may be ‘direct’ or ‘indirect’.
Direct pharmacodynamic interactions involve effects at a common target, additively (and possibly potentiation) or antagonism due to actions at different sites in an organ.
A pharmacodynamic interaction occurs when the pharmacological effects of one drug alter the response to another drug, even though the two types of effects are not themselves directly related.
A pharmacokinetic drug interaction can alter the concentration of drug in the systemic circulation through interactions occurring at any stage: that is, during absorption, distribution, metabolism or excretion.
Strategies for reducing the incidence of ADRs and drug interactions include careful history-taking, considering nondrug treatment, correct and appropriate dosing, frequent review of therapeutic goals and drug regimens, avoiding polypharmacy and careful communication with the patient or carer.

Review exercises

1. Define an adverse drug reaction.
2. Discuss the six categories of adverse drug reactions and provide examples of drugs implicated.
3. Discuss the mechanistic bases of the four types of drug allergies.
4. Discuss the clinical implications of an inhibitory drug interaction involving a drug with a narrow therapeutic index.
5. Discuss the impact of consumption of St John’s wort on the pharmacokinetics of antiretroviral protease inhibitors.

References and further reading

Alfirevic A., Pirmohamed M. Adverse drug reactions and pharmacogenomics: recent advances. Personalized Medicine. 2008;5:11-23.

Australian Institute of Health and Welfare. Australia’s Health 2008. Cat. no. AUS 99. Canberra: AIHW, 2008.

Bailey D.G., Arnold M.O., Spence J.D. Grapefruit juice–drug interactions. British Journal of Clinical Pharmacology. 1998;46:101-110.

Bates D.W., Cullen D.J., Laird N., Petersen L.A., Small S.D., et al. Incidence of adverse drug events and potential adverse drug events: implications for prevention. Journal of the American Medical Association. 1995;274:29-34.

Bressler R. Herb–drug interactions: interactions between kava and prescription medications. Geriatrics. 2005;60:24-65.

deShazo R.D., Kemp S.F. Allergic reactions to drugs and biologic agents. Journal of the American Medical Association. 1997;278:1895-1906.

Edwards I.R., Aronson J.K. Adverse drug reactions: definitions, diagnosis, and management. Lancet. 2000;356:1255-1259.

Flockhart DA. Drug Interactions: Cytochrome P450 Drug Interaction Table. Indiana University School of Medicine. 2009. Available: http://medicine.iupui.edu/clinpharm/ddis/table.asp [27 January 2010].

Hansten P.D. Drug interaction management. Pharmacy World Science. 2003;25:94-97.

Impicciatore P., Choonara I., Clarkson A., Provasi D., Pandolfini C., Bonati M. Incidence of adverse drug reactions in paediatric in/out-patients: a systematic review and metaanalysis of prospective studies. British Journal of Clinical Pharmacology. 2001;52:77-83.

Izzo A.A. Herb–drug interactions: an overview of the clinical evidence. Fundamental and Clinical Pharmacology. 2004;19:1-16.

Lacoste-Roussillon C., Pouyanne P., Haramburu F., Miremont G., Begaud B. Incidence of serious adverse drug reactions in general practice: a prospective study. Clinical Pharmacology and Therapeutics. 2001;69:458-462.

Nair B. Older people and medications: what is the right prescription? [editorial]. Australian Prescriber. 1999;22:130-131.

Pirmohamed M., Breckenridge A.M., Kitteringham N.R., Park B.K. Adverse drug reactions. British Medical Journal. 1998;316:1295-1298.

Riedl M., Casillas A.M. Adverse drug reactions: types and treatment options. American Family Physician. 2003;68:1781-1790.

Roughead E.E., Gilbert A.L., Primrose J.G., Sansom L.N. Drugrelated hospital admissions: a review of Australian studies published 1988–1996. Medical Journal of Australia. 1998;168(8):405-408.

Uetrecht J. Immune-mediated adverse drug reactions. Chemical Research Toxicology. 2009;22:24-34.

Wester K., Jonsson A.K., Spigset O., Druid H., Hagg S. Incidence of fatal adverse drug reactions: a population based study. British Journal of Clinical Pharmacology. 2007;65:573-579.

World Health Organization. Collaborating Centers for International Drug Monitoring. Geneva: WHO, 1984 (WHO publication DEM/NC/84:153.(E)).