Key Points

  • Drug–drug interactions refer to alterations in drug levels or drug effects (or both) related to the administration of two or more prescribed, recreational, or over-the-counter agents in close temporal proximity.

  • Although some drug–drug interactions involving psychotropic medications are life-threatening, most interactions manifest in more subtle ways through increased side-effect burden, aberrant drug levels, or diminished efficacy.

  • Pharmacokinetic drug–drug interactions involve a change in the plasma level or tissue concentration of one drug following co-administration of one or more other drugs due to an action of the co-administered agents on one of four key pharmacokinetic processes: absorption, distribution, metabolism, or excretion.

  • Pharmacodynamic drug–drug interactions involve an effect of one or more drugs on another drug at biological receptor sites of action and do not involve a change in plasma level or tissue concentration.

  • The potential for drug–drug interactions should be carefully considered whenever prescribing medications associated with interactions that are uncommon but catastrophic (e.g., hypertensive crises, Stevens-Johnson syndrome, or cardiac arrhythmias) and medications with low therapeutic indices (e.g., warfarin) or narrow therapeutic windows (e.g., cyclosporine), and when prescribing for frail or clinically brittle patients for whom small variations in side effects or efficacy may be particularly troublesome.

Overview

An understanding of drug–drug interactions is essential to the practice of psychopharmacology. As in other areas of medicine, polypharmacy has become an increasingly accepted approach in psychiatry for addressing difficult-to-treat disorders. Moreover, general medical co-morbidity is common among patients with psychiatric disorders, elevating the likelihood of complex medication regimens. Similarly, the widespread use of over-the-counter (OTC) supplements by patients receiving treatment for psychiatric disorders may invite additional risk of drug–drug interactions. When they occur, drug–drug interactions may manifest in myriad ways, from perplexing laboratory tests to symptoms that are difficult to distinguish from the underlying psychiatric and physical conditions under treatment. The comprehensive evaluation of patients with psychiatric disorders therefore requires a careful assessment of potential drug–drug interactions.

Drug–drug interactions refer to alterations in drug levels or drug effects (or both) attributed to the administration of two or more prescribed, illicit, or OTC agents in close temporal proximity. Although many drug–drug interactions involve drugs administered within minutes to hours of each other, some drugs may participate in interactions days or even weeks after their discontinuation because of prolonged elimination half-lives (e.g., fluoxetine) or due to their long-term impact on metabolic enzymes (e.g., carbamazepine). Some drug–drug interactions involving psychotropic medications are life-threatening, such as those involving the co-administration of monoamine oxidase inhibitors (MAOIs) and drugs with potent serotonergic (e.g., meperidine) or sympathomimetic (e.g., phenylpropanolamine) effects. These combinations are therefore absolutely contraindicated. However, most drug–drug interactions in psychopharmacology manifest in somewhat more subtle ways, often leading to poor medication tolerability and compliance due to adverse events (e.g., orthostatic hypotension, sedation, or irritability), diminished medication efficacy, or puzzling manifestations (such as altered mental status or unexpectedly high or low drug levels). Drug combinations that can produce these often less than catastrophic drug–drug interactions are usually not absolutely contraindicated. Some of these combinations may, indeed, be valuable in the treatment of some patients while wreaking havoc for other patients. The capacity to anticipate and to recognize both the major, but rare, and the more subtle, but common, potential drug–drug interactions allows the practitioner to minimize the impact of these interactions as an obstacle to patient safety and to therapeutic success. This is both an important goal and a considerable challenge in psychopharmacology.

While drug–drug interactions are ubiquitous, few studies have systematically assessed in vivo drug–drug interactions of most interest to psychiatrists. Fortunately, well-designed studies of drug–drug interactions are an increasingly integral part of drug development. Beyond these studies, however, the literature on drug–drug interactions remains a patchwork of case reports, post-marketing analyses, extrapolation from animal and in vitro studies, and extrapolation from what is known about other drugs with similar properties. While these studies often shed some light on the simplest case of a single drug (drug B) exerting an effect on another (drug A), they rarely consider the common clinical scenario in which multiple drugs with numerous potential interactions among them are co-administered. Under these circumstances, the range of possible, if not well-delineated, drug–drug interactions often seems overwhelming.

Fortunately, an increasing range of resources are available (including prescribing software packages and regularly updated websites, such as www.drug-interactions.com ) that allow for the prevention and detection of potential interactions. In addition, it is important to recall that numerous factors contribute to inter-individual variability in drug response. These factors include treatment adherence, age, gender, nutritional status, disease states, and genetic polymorphisms that may influence risk of adverse events and treatment resistance ( Figure 11-1 ). Drug–drug interactions are an additional factor that influence how patients react to drugs. The importance of these interactions depends heavily on the clinical context. In many cases, the practical impact of drug–drug interactions is likely to be very small compared with other factors that affect treatment response, drug levels, and toxicity. It is reasonable, therefore, to focus special attention on the contexts in which drug–drug interactions are most likely to be clinically problematic.

Figure 11-1, Factors contributing to inter-individual variability in drug response.

First, it is crucial to be familiar with the small number of drug–drug interactions in psychopharmacology that, though uncommon, are associated with potentially catastrophic consequences. These include drugs associated with ventricular arrhythmias, hypertensive crisis, serotonin syndrome, Stevens-Johnson syndrome, seizures, and severe bone marrow suppression. In addition, drug–drug interactions are important to consider when a patient's drugs include those with a low therapeutic index (e.g., lithium, digoxin, or warfarin) or a narrow therapeutic window (e.g., indinavir, nortriptyline, or cyclosporine) such that relatively small alterations in pharmacokinetic or pharmacodynamic behavior may jeopardize a patient's well-being. In addition, it is worthwhile to consider potential drug–drug interactions whenever evaluating a patient whose drug levels are unexpectedly variable or extreme, or a patient with a confusing clinical picture (such as clinical deterioration), or with unexpected side effects. Finally, drug–drug interactions are likely to be clinically salient for a patient who is medically frail or elderly, owing to altered pharmacokinetics and vulnerability to side effects, as well as for a patient heavily using alcohol, cigarettes, or illicit drugs, or being treated for a drug overdose.

Classification

Drug–drug interactions may be described as pharmacokinetic, pharmacodynamic, idiosyncratic, or mixed, depending on the presumed mechanism underlying the interaction ( Box 11-1 ). Pharmacokinetic interactions are those that involve a change in the plasma level or tissue distribution (or both) of one drug by virtue of co-administration of another drug. These interactions occur due to effects at one or more of the four pharmacokinetic processes by which drugs are acted on by the body: absorption, distribution, metabolism, and excretion. Because of the importance of these factors, particularly metabolism, in drug–drug interactions, a more detailed description of pharmacokinetic processes follows. An example of a pharmacokinetic drug–drug interaction is the inhibition of the metabolism of lamotrigine by valproic acid, thereby raising lamotrigine levels and increasing the risk of potentially serious adverse events, including hypersensitivity reactions (such as Stevens-Johnson syndrome). In contrast, pharmacodynamic interactions are those that involve a known pharmacological effect at biologically-active (receptor) sites. These interactions occur due to effects on the mechanisms through which the body is acted on by drugs and do not involve a change in drug levels. An example of a pharmacodynamic drug–drug interaction is the interference of the antiparkinsonian effects of a dopamine receptor agonist (such as pramipexole) by a dopamine receptor antagonist (such as risperidone). Mixed interactions are those that are believed to involve both pharmacological and pharmacodynamic effects. Symptoms of serotonin toxicity, such as agitation and confusion that have been observed in some individuals on the combination of paroxetine and dextromethorphan, for example, may reflect the shared pharmacodynamic effect of the two agents at serotonin receptor sites as well as the elevation of dextromethorphan levels due to inhibition of its cytochrome P450 metabolism by paroxetine. Finally, idiosyncratic interactions are those that occur sporadically in a small number of patients in ways that are not yet predicted by the known pharmacokinetic and pharmacodynamic properties of the drugs involved.

Box 11-1
Classification of Drug–Drug Interactions

Pharmacokinetic

  • Alteration in blood level or tissue concentration (or both) resulting from interactions involving drug absorption, distribution, metabolism, or excretion

Pharmacodynamic

  • Alteration in pharmacological effect resulting from interactions at the same or inter-related biologically-active (receptor) sites

Mixed

  • Alterations in blood levels and pharmacological effects due to pharmacokinetic and pharmacodynamic interactions

Idiosyncractic

  • Sporadic interactions among drugs not accounted for by their currently known pharmacokinetic or pharmacodynamic properties

Pharmacokinetics

As described earlier, pharmacokinetic processes refer to absorption, distribution, metabolism, and excretion, factors that determine plasma levels and tissue concentrations of a drug. Pharmacokinetics refers to the mathematical analysis of these processes. Advances in analytic chemistry and computer methods of pharmacokinetic modeling and a growing understanding of the molecular pharmacology of the liver enzymes responsible for metabolism of most psychotropic medications have furnished increasingly sophisticated insights into the disposition and interaction of administered drugs. Although pharmacokinetics refers to only one of the two broad mechanisms by which drugs interact, pharmacokinetic interactions involve all classes of psychotropic and non-psychotropic medications. An overview of pharmacokinetic processes is a helpful prelude to a discussion of drug–drug interactions by psychotropic drug class.

Absorption

Factors that influence drug absorption are generally of less importance to drug–drug interactions involving psychiatric medications than are factors that influence subsequent drug disposition, particularly drug metabolism. Factors relevant to absorption generally pertain to orally rather than parenterally administered drugs, for which alterations in gastrointestinal drug absorption may affect the rate (time to reach maximum concentration) or the extent of absorption or both. The extent or completeness of absorption, also known as the fractional absorption, is measured as the area under the curve (AUC) when plasma concentration is plotted against time. The bioavailability of an oral dose of drug refers, in turn, to the fractional absorption for orally compared with intravenously (IV) administered drug. If an agent is reported to have a 90% bioavailability (e.g., lorazepam), this would indicate that the extent of absorption of an orally administered dose is nearly that of an IV-administered dose, although the rate of absorption may well be slower for the oral dose.

Because the upper part of the small intestine is the primary site of drug absorption through passive membrane diffusion and filtration and both passive and active transport processes, factors that speed gastric emptying (e.g., metoclopramide) or diminish intestinal motility (e.g., opiates or marijuana) may facilitate greater contact with, and absorption from, the mucosal surface into the systemic circulation, potentially increasing plasma drug concentrations. Conversely, antacids, charcoal, kaolin-pectin, and cholestyramine may bind to drugs, forming complexes that pass unabsorbed through the gastrointestinal lumen. Changes in gastric pH associated with food or other drugs alter the non-polar, un-ionized fraction of drug available for absorption. In the case of drugs that are very weak acids or bases, however, the extent of ionization is relatively invariant under physiological conditions. Properties of the preparation administered (e.g., tablet, capsule, or liquid) may also influence the rate or extent of absorption, and, for an increasing number of medications (e.g., lithium, bupropion, valproate, and methylphenidate), preparations intended for slow release are available. Finally, the local action of enzymes in the gastrointestinal tract (e.g., monoamine oxidase [MAO] and cytochrome P450 3A4) may be responsible for metabolism of drug before absorption. As described later, this is of critical relevance to the emergence of hypertensive crises that occur when excessive quantities of the dietary pressor tyramine are systemically absorbed in the setting of irreversible inhibition of the MAO isoenzymes for which tyramine is a substrate.

Distribution

Drugs distribute to tissues through the systemic circulation. The amount of drug ultimately reaching receptor sites in tissues is determined by a variety of factors, including the concentration of free (unbound) drug in plasma, regional blood flow, and physiochemical properties of drug (e.g., lipophilicity or structural characteristics). For entrance into the central nervous system (CNS), penetration across the blood–brain barrier is required. Fat-soluble drugs (such as benzodiazepines, neuroleptics, and cyclic antidepressants) distribute more widely in the body than water-soluble drugs (such as lithium), which distribute through a smaller volume of distribution. Changes with age, typically including an increase in the ratio of body fat to lean body mass, therefore result in a net greater volume of lipophilic drug distribution and potentially greater accumulation of drug in adipose tissue in older than in younger patients.

In general, psychotropic drugs have relatively high affinities for plasma proteins (some to albumin but others, such as antidepressants, to α 1 -acid glycoproteins and lipoproteins). Most psychotropic drugs are more than 80% protein-bound. A drug is considered highly protein-bound if more than 90% exists in bound form in plasma. Fluoxetine, aripiprazole, and diazepam are examples of the many psychotropic drugs that are highly protein-bound. In contrast, venlafaxine, lithium, topiramate, zonisamide, gabapentin, pregabalin, milnacipran, and memantine are examples of drugs with minimal protein-binding and therefore minimal risk of participating in drug–drug interactions related to protein-binding. A reversible equilibrium exists between bound and unbound drug. Only the unbound fraction exerts pharmacological effects. Competition by two or more drugs for protein-binding sites often results in displacement of a previously bound drug, which in the free state becomes pharmacologically active. Similarly, reduced concentrations of plasma proteins in a severely malnourished patient or a patient with a disease that is associated with severely lowered serum proteins (such as liver disease or nephrotic syndrome) may be associated with an increase in the fraction of unbound drug potentially available for activity at relevant receptor sites. Under most circumstances, the net changes in plasma concentration of active drug are, in fact, quite small because the unbound drug is available for redistribution to other tissues and for metabolism and excretion, thereby off-setting the initial rise in plasma levels. Nevertheless, clinically significant consequences can develop when protein-binding-interactions alter the unbound fraction of previously highly protein-bound drugs that have a low therapeutic index (e.g., warfarin). For these drugs, relatively small variations in plasma level may be associated with serious untoward effects.

An emerging understanding of the drug transport proteins, of which P-glycoproteins are the best characterized, indicates a crucial role in regulating permeability of intestinal epithelia, lymphocytes, renal tubules, the biliary tract, and the blood–brain barrier. These transport proteins are thought to account for the development of certain forms of drug resistance and tolerance, but are increasingly seen as likely also to mediate clinically-important drug interactions. Little is known yet about their relevance to drug interactions involving psychiatric medications; the capacity of St. John's wort to lower blood levels of several critical medications (including cyclosporine and indinavir) is hypothesized to be related, at least in part, to an effect of the botanical agent on this transport system.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here