Drug Interactions Clinical Pharmacology Spring Course 2006 M. E. Blair Holbein, Ph.D. Clinical Pharmacologist Presbyterian Hospital Why study drug interactions? Clinical Significance of Drug Interactions Over 2 MILLION serious ADRs and 100,000 deaths yearly ADRs 4th leading cause of death ahead of pulmonary disease, diabetes, AIDS, pneumonia, accidents and automobile deaths Greater than total costs of cardiovascular or diabetic care ADRs cause 1 out of 5 injuries or deaths per year to hospitalized patients Mean length of stay, cost and mortality for ADR patients are DOUBLE that for control patients Account for 6.5% hospital admissions Nursing home patients ADR rate—50,000 yearly Ambulatory patients ADR rate—unknown Many clinical implications Libby Zion case Clinical Trials, OPI International Intrigue? Ref: Institute of Medicine, National Academy Press, 2000, Lazarou J et al. JAMA 1998;279(15):1200–1205, Gurwitz JH et al. Am J Med 2000;109(2):87–94. Johnson JA et al. Arch Intern Med 1995;155(18):1949–1956, Leape LL et al. N Engl J Med 1991;324(6):377–384, Classen DC et al. JAMA 1997;277(4):301–306 er Preventable drug interactions 1/3 of adverse drug events and 1/2 cost. Definition A drug interaction is defined as a measurable modification (in magnitude or duration) of the action of one drug by prior or concomitant administration of another substance (including prescription and nonprescription drugs, food, or alcohol) May be harmful: toxicity, reduced efficacy May be beneficial: synergistic combinations, pharmacokinetic boosting, increased convenience, reduced toxicity, cost reduction . Wright JM. 2000. Drug Interactions. In: Carruthers SG, Hoffman BB, et al. , ed. Melmon and Morrelli’s Clinical Pharmacology: Basic Principles in Therapeutics, 4th ed. New York:McGraw-Hill. Characterizing Drug Interactions Mechanism Interacting agents Pharmacodynamic Receptor inhibition Additive effects Drug - Disease Drug-drug Pharmacokinetic Altered absorption, distribution, metabolism, or elimination Prescription Non-prescription Illicit, recreational Food, supplements, herbal products Clinical Significance Major Substantial morbidity and mortality Therapy altering Manageable Little or no change in therapy Optimize therapy Intentional Additive or synergistic effects Enhanced pharmacokinetics Mechanisms of Interactions Pharmacodynamic Pharmacokinetic Receptor Absorption Non-receptor Distribution Metabolism Excretion Mechanisms of Interactions Pharmacodynamic Receptor Non-receptor Pharmacodynamic: Pharmacological Interaction at the drug receptor Activity is function of intrinsic activity and affinity for receptor Agonist and antagonists Effect also function of concentration at receptor Effect can be additive Several agents that act via the same receptor Example, several agents with anticholinergic activity or side effects can result in serious anticholinergic toxicity especially in elderly patients. Pharmacodynamic: Physiological Agents that can act in concert or in opposition via different cellular mechanisms. theophylline and b-receptor agonists can cause bronchiolar muscle relaxation Sensitization of myocardium to arrhythmogenic action of catecholamines by general anesthetics. Combinations of antihypertensive (can be intentional) Both Pharmacodynamic: Altered physiology Altered Aging cellular environment effects Blunted sympathetic nervous system; blunted responses Agents that change the state of the host Ex. Hypokalemia caused by diuretics increases toxicity of digoxin. Pharmacodynamic: Neutralization Neutralization systemically in the host (as opposed to prior to absorption) Protamine used to neutralize heparin Purified antidigoxin Fab fragments used to treat digoxin toxicity Mechanisms of Interactions Pharmacodynamic Pharmacokinetic Receptor Absorption Non-receptor Distribution Metabolism Excretion Mechanisms of Interactions Pharmacokinetic Absorption Distribution Metabolism Excretion Mechanisms of Interactions Pharmacokinetic Absorption Distribution Metabolism Excretion Pharmacokinetic: Absorption Alters rate that drug enters the system with altered level or time to peak Mechanisms: Physical interaction, chelation, binding. e.g. tetracyclines and cations Altered GI function: changes in pH (ketoconazole), motility, mucosal function, metabolism, absorption sites, perfusion Absorption: in the gut Sucralfate, some milk products, antacids, and oral iron preparations Block absorption of quinolones, tetracycline, and azithromycin Omeprazole, lansoprazole, H2-antagonists Reduce absorption of ketoconazole, delavirdine Didanosine (given as a buffered tablet) Reduces ketoconazole absorption Cholestyramine Binds raloxifene,thyroid hormone, and digoxin Interactions: Presystemic Elimination Gut transit and metabolism Intestinal wall CYP3A4 metabolizes a number of drugs Inhibition/induction results in altered bioavailability Ex: grapefruit juice inhibits intestinal CYP3A4 Results in increased bioavailability of calcium channel blockers (dihydropyridine), cyclosporin, saquinavir (HIV-1 protease inhibitors), carbamazepine, lovastatin, terazosin, triazolam and midazolam. High intrinsic hepatic clearance dependent upon hepatic blood flow Inhibition results in increased bioavailabilty. Propranolol, metoprolol, labetalol, verapamil, hydralazine, felodipine, clhlorpromazine, imipramine, amitriptyline, morphine First-Pass Metabolism after Oral Administration of a Drug, as Exemplified by Felodipine and Its Interaction with Grapefruit Juice Wilkinson, G. R. N Engl J Med 2005;352:2211-2221 Some Common Drugs with Low Oral Bioavailability and Susceptibility to First-Pass Drug Interactions Wilkinson, G. R. N Engl J Med 2005;352:2211-2221 Consequences of the Inhibition of First-Pass Metabolism, as Exemplified by the Interaction between Felodipine and Grapefruit Juice Wilkinson, G. R. N Engl J Med 2005;352:2211-2221 Induction of P-glycoprotein and Intestinal CYP450 Intestinal epithelium with CYP450 Sufficient amout to result in presystemic clearance of some drugs Highly variable Enterocytes have transporter proteins Organic anion-transporting polypeptide (OATP) Organic cation transporters (OCTs) P-glycoprotein (P-gp) Product of human multidrug resistance gene (mdr1) Contributesto resistance to a variety of chenotherapeutic agents Decreases the intracellular accumulation of anticancer drugs Efflux transporter in Gi epithelium, liver, kidney, edothelial cells of blood-brain barrier Complements CYP450 interactions Intestinal Transporter - P-glycoprotein P-glycoprotein Substrates and Inhibitors Substrates Actinomycin Amprenavir Colchicines Cortisol Cyclosporine Daunorubicin Dexamethasone Digoxin Diltiazem Docetaxel Doxorubicin Erythromycin Etoposide Fexofenadine Hydrocortisone Indinavir Ivermectin Loperamide Mitomycin C Inhibitors Mitoxantrone Morphine Nelfinavir Nicardipine Nifedipine Paclitaxel Progesterone Rifampin Ritonavir Saquinavir Tacrolimus Taxol Teniposide Topotecan Verapamil Vinblastine Vincristine Amiodarone Bepridil Cefoperzone Ceftriaxone Clarithromycin Cortisol Cyclosporine Diltiazem Dipyridamole Erythromycin Itraconazole Felodipine Fluperazine Hydrocortisone Ketoconazole Lidocaine Mefloquine Nicardipine Nifedipine Nitrendipine Progesterone Propranolol Quercetin Quinine Quinidine Reserpine Tacrolimus Tamoxifen Testosterone Trifluoperaine Verapamil Intestinal Monoamine Oxidase Intestinal MAO inhibited by nonselective irreversible agents and inhibit metabolism of dietary tyramine resulting in increased release of norepi from sympathetic postganglionic neurons Less problematic for selective MAO B inhibitor selegiline and reversible agent moclobemide Mechanisms of Interactions Pharmacokinetic Absorption Distribution Metabolism Excretion Pharmacokinetic: Distribution Protein-binding Relative displacement to : Concentration - a high concentration of one drug relative to another will shift the binding equilibrium Relative binding affinity - only relatively highly bound drugs will be effected Volume of distribution - small Vd allows for greater proportional effect Therapeutic index - mostly drugs with a narrow TI are clinically significant Alterations in protein-binding capacity hypoalbuminemia (acidic drugs) a1-acid glycoprotein (basic drugs) acute phase reactants Pharmacokinetic: Distribution Protein-binding displacement Effect is rapid and transient and usually compensated by increased elimination May result in transient pharmacologic effect Overall result is unpredictable New steady-state attained Pharmacokinetic: Distribution Cellular distribution interactions Cellular transport systems “Promiscuous” and affect several agents requiring active transport Best studied example is P-glycoprotein (PGP) an organic anion transporter system. Cyclosporin A, quinidine, verapamil, itraconazole and clarithromycin inhibit PGP Some correlation with CYP3A4 affinities May be significant for some anticancer drugs Mechanisms of Interactions Pharmacokinetic Absorption Distribution Metabolism Excretion Drug Metabolism Phase I Oxidation Cytochrome P450 monooxygenase system Flavin-containing monooxygenase system Alcohol dehydrogenase and aldehyde dehyddrogenase Monoamine oxidase (Co-oxidation by peroxidases) Reduction NADPH-cytochrome P450 reductase Reduced (ferrous) cytochrome P450 Hydroloysis Esterases amd amidases Epoxide hydrolase Phase II Glutathione S-transferases UDP-Glucoron(os)yltranasferases N-Acetyltransferases Amino acid N-acyl transferases Sulfotransferases Interactions in the Phases of Drug Metabolism Drug interactions due to metabolic effects nearly always due to interaction at Phase I enzymes, rather than Phase II CYP450 system responsible for the majority of oxidative reactions and subsequent interactions Significant polymorphism in many. CYP2C9, CYP2C19, and CYP2D6—can be even be genetically absent! Drugs may be metabolized by a single isoenzyme Desipramine/CYP2D6; indinavir/CYP3A4; midazolam/CYP3A; caffeine/CYP1A2; omeprazole/CYP2C19 Drugs may be metabolized by multiple isoenzymes Most drugs metabolized by more than one isozyme Imipramine: CYP2D6, CYP1A2, CYP3A4, CYP2C19 If co-administered with CYP450 inhibitor, some isozymes may “pick up slack” for inhibited isozyme. Drugs may be metabolized by a combination of enzymatic systems. Pharmacokinetic: Elimination - Metabolism Interactions can result from increased as well as decreased metabolism Clinical relevance is dependent upon timing of interaction, therapeutic index of affected drug, duration of therapy, metabolic fate of affected drug, metabolic capacity of host. Host factors include age, genetic makeup (acetylation, CYP2D6), nutritional state, disease state, hormonal milieu, environmental and exogenous chemical exposure. P450 isoenzymes are variously affected. Isoenzymes characterized Substrates Inhibiting agents Inducing agents No consistent correlation of substrate versus inhibitor or inducer Good reference: http://medicine.iupui.edu/flockhart/ (alias: www.druginteractions.com) Pharmacokinetic: Elimination - Metabolism Characteristics of interactions with DECREASED metabolism Inhibition of metabolizing enzymes Timeframe is rapid Duration and extent of effect is dependent upon concentration of agents and enzyme affinities. Maximum effect seen in 4-5 half-lifes Mostly in hepatic microsomal enzymes (mixed-function oxidases of cytochrome P450 system) Other systems affected; less well characterized Conjugation, acetylation, etc. P450 isoenzymes are variously affected. Most important with drugs with narrow TI, brittle hosts, agents with few alternate metabolic pathways Ex: theophylline, antihypertensive agents, hypoglycemic agents, chemotherapeutic agents, some hormonal agents, HAART agents Pharmacokinetic: Elimination - Metabolism Characteristics of interactions due to INCREASED metabolism Induction of metabolizing enzymes Timeframe is slow “Recovery” to basal state is also slow Mostly in hepatic microsomal enzymes but also in other tissues Clinical relevance is dependent upon timing of interaction, therapeutic index of affected drug, duration of therapy. Most frequently encountered inducing agents: Phenobarbital, phenytoin, carbamazepine Rifampin > rifabutin Cigarettes and charred or smoked foods Prolonged and substantial ethyl alcohol ingestion Isoniazid Mechanism of Induction of CYP3A4-Mediated Metabolism of Drug Substrates (Panel A) Wilkinson, G. R. N Engl J Med 2005;352:2211-2221 The Resulting Reduced Plasma Drug Concentration (Panel B) Common CYP2D6 Drug Substrates and Clinically Important Inhibitors of Biotransformations Phase I Oxidation Cytochrome P450 monooxygenase system Flavin-containing monooxygenase system Alcohol dehydrogenase and aldehyde dehddrogenase Monoamine oxidase (Co-oxidation by peroxidases) Reduction NADPH-cytochrome P450 reductase Reduced (ferrous) cytochrome P450 Hydroloysis Esterases amd amidases Epoxide hydrolase Phase II Glutathione S-transferases Mercapturic acid biosynthesis UDP-Glucoron(os)yltranasferases N-Acetyltransferases Amino acid N-acyl transferases Sulfotransferases Proportion of Drugs Metabolized by CYP450 Enzymes CYP2D6 20% CYP3A4 38% CYP2C19 8% CYP1A2 11% CYP2C9 16% CYP2E1 4% CYP2A6 3% Cytochrome P450 3A4,5,7 Largest number of drugs metabolized Present in the largest amount in the liver. Present Not in GI tract polymorphic Inherent activity varies widely, e.g. 1,000 fold Activity has been shown to predominate in the gut. Responsible Most for metabolism of: calcium channel blockers Most benzodiazepines Most HIV protease inhibitors Most HMG-CoA-reductase inhibitors Cyclosporine Most non-sedating antihistamines Cisapride Cytochrome P450 3A4,5,7 -continued Substrates: antibiotics – clarithromycin, erythromycin; benzodiazeines- diazepam, midazolam; cyclosporine, tacrolimus,; HIV Protease Inhibitors – indinavir, ritonavir; chlorpheniramine; Calcium Channel Blockers – nifedipine, amlodipine; HMG Co A Reductase Inhibitors – atorvastatin, lovastatin; haloperidol, buspirone; sildenafil, tamoxifen, trazodone, vincristine macrolide Inhibited by: HIV Protease Inhibitors, cimetidine, clarithromycin, fluoxetine, fluvoxamine, grapefruit juice, itraconazole, ketoconazole, verapamil Induced by: carbamazepine, phenobarbital, phenytoin, rifampin, St. John’s wort, troglitazone Cytochrome P450 2D6 Second largest number of substrates. Polymorphic distribution Majority of the population is characterized as an extensive or even ultra-extensive metabolizer. Approximately 7% of the U.S. Caucasian population and 1-2% of African or Asian inheritance have a genetic defect in CYP2D6 that results in a poor metabolizer phenotype. include: many b-blockers – metoprolol, timolol, amitriptylline, imipramine, paroxetine, haloperidol, risperidone, thioridazine, codeine, dextromethorphan, ondansetron, tamoxifen, tramadol Inhibited by: amiodarone, chlorpheniramine, cimetidine, fluoxetine, ritonavir Substrates Pharmacogenetics of Nortriptyline Variability of CYP2D6 Expression Pharmacog enetics of Nortriptylin e Weinshilboum, R. N Engl J Med 2003;348:529-537 Pharmacogenetics of CYP2D6 Pharmacogenetics of CYP2D6 Weinshilboum, R. N Engl J Med 2003;348:529-537 Cytochrome P450 2C9 Note: Absent in 1% of Caucasian and African- Americans. Substrates include: many NSAIDs – ibuprofen, tolbutamide, glipizide, irbesartan, losartan, celecoxib, fluvastatin, phenytoin, sulfamethoxazole, tamoxifen, tolbutamide, warfarin Inhibited by: fluconazole, isoniazid, ticlopidine Induced by: rifampin Cytochrome P450 1A2 Substrates include: caffeine, theophylline, imipramine, clozapine Inhibited by: many fluoroquinolone antibiotics, fluvoxamine, cimetidine Induced by: smoking tobacco Cytochrome P450 2C19 Note: Absent in 20-30% of Asians, 3-5% of Caucasians Substrates include: omeprazole, diazepam, phenytoin, phenobarbitone, amitriptylline, clomipramine, cyclophosphamide, progesterone Inhibited by: fluoxetine, fluvoxamine, ketoconazole, lansoprazole, omeprazole, ticlopidine Cytochrome P450 2B6 Substrates include: bupropion, cyclophosphamide, efavirenz, methadone Inhibited by: thiotepa Induced by: phenobarbital, rifampin Cytochrome P450 2E1 Substrates include: acetaminophen Cytochrome P450 2C8 Substrates; paclitaxel, torsemide, amodiaquine, cerivastatin, repaglinide Inhibited by: trimethoprim, quercetin, glitazones, gemfibrozil, montelukast Induced by: rifampin The “Usual Suspects” - Inhibitors Amiodarone Ketoconazole Cimetidine Ciprofloxacin (1A2) Diltiazem Erythromycin (3A4) Ethanol (acute) Fluconazole (3A4) Fluoxetine (2C9, 2C19, 2D6) Fluvoxamine (1A2, 2C19, 3A4) Grapefruit (3A4) Isoniazid (2E1) Itraconazole (3A4) Ketaconazole (3A4) Metronidazole Miconazole (3A4) Nefazodone (3A4) Oral contraceptives Paroxetine (2D6) Phenylbutazone Quinidine (2D6) Sulfinpyrazone Valproate Verapamil The “Usual Suspects” - Inducers Barbiturates (2B) Carbamazepine (2C19, 3A4/5/7) Charcoal-broiled food (1A2) Dexamethasone Ethanol (chronic) (2E1) Griseofulvin Isoniazid (2E1) Primidone (2B) Rifabutin (3A4) Rifampin (2B6, 2CB, 2C19, 2C9, 2D6, 3A4/5/7) Tobacco smoke (1A2) Probe Substrates and Inhibitors for P450s P450 Subs trates Preferred Acceptable CYP1A2 Ethoxyresorufin, phenacet in CYP2A6 Coumarin CYP2B6 CYP2C8 CYP2C9 S-Mepheytoin (Ndesmethyl metabolite) CYP2C19 CYP2D6 CYP2E1 Caffeine (low t urnover), theophylline (low turnover), acetanilide (most ly applied in hepatocytes), methoxyresorufin Ephenyt oin (N-desmethyl metabolite) Paclitxel (?) S-Warfarin, diclofenac Inhibitors Preferred Acceptable Furafylline a-Naphthoflavone (but coan also activate and inhibit CYP3A4) 8-Methoxypsoralen Coumarin (but high turnover), Sertraline (but also inhibits CYP 2D6) Bupropion (metabolite standards?) Glitazones (?) Tobut amine (low turnover) Sulphaphenazole T iclopidine (but also inhibits CYP2D6), noot kat one (also inhibits CYP2A6) S-Mephytoin (4hydroxy met abolite), omeprazole Bufuralol dextromet horphan Metoprolol, debrisoquine, codeine Quinidine Chlorzoxazone 4-Nit rophenol, lauric acid Clomethiazole 4-Methyl pyrazole Midazolam, Nifedipine, felodipine, Ketoconazole (not test osterone (test at cyclosporin A, t erfenadine, specific, slo inhibits Cyclosporin A CYP3A least 2) erythromycin, simvast atin CYP2C8) Adapted from Bjornsson TD, Callaghan JT , Einolf HJ, et al. Drug Met Disp 2003; 31:815-832; See also Tucker GT, Houst on JB and Hyang SM. Pharm Res 2001; 18: 1071-1080. Probe Substrates and Inhibitors for P450s Substrates P450 Preferred CYP1A2 Ethoxyresorufin, phenacetin CYP2A6 Coumarin CYP2B6 S-Mephytoin (4-hydroxy metabolite) Inhibitors Acceptable Caffeine (low turnover), theophylline (low turnover), acetanilide (mostly applied in hepatocytes), methoxyresorufin Ephenytoin (N-desmethyl metabolite) Preferred Acceptable Furafylline a-Naphthoflavone (but coan also activate and inhibit CYP3A4) Methoxypsoralen Coumarin (but high turnover), Sertraline (but also inhibits CYP2D6) Bupropion (metabolite standards?) Glitazones (?) CYP2C8 CYP2C9 CYP2C19 S-Mephytoin (4-hydroxy metabolite), omeprazole Ticlopidine (but also inhibits CYP2D6), nootkatone (also inhibits CYP2A6) CYP2D2 Bufuralol dextromethorphan Metoprolol, debrisoquine, codeine Quinidine CYP2E1 Chlorzoxazone 4-Nitrophenol, lauric acid Clomethiazole 4-Methyl pyrazole CYP3A Midazolam, testosterone (test at least 2) Nifedipine, felodipine, cyclosporin A, terfenadine, erythromycin, simvastatin Ketoconazole (not specific, also inhibits CYP2C8) Cyclosporin A Adapted from Bjornsson TD, Callaghan JT , Einolf HJ, etal. Drug Met Disp 2003; 31:815-832; See also Tucker GT, Houston JB and Hyang SM. Pharm Res 2001; 18: 1071-1080. Drug Metabolism Phase I Oxidation Cytochrome P450 monooxygenase system Flavin-containing monooxygenase system Alcohol dehydrogenase and aldehyde dehddrogenase Monoamine oxidase (Co-oxidation by peroxidases) Reduction NADPH-cytochrome P450 reductase Reduced (ferrous) cytochrome P450 Hydroloysis Esterases amd amidases Epoxide hydrolase Phase II Glutathione S-transferases UDP-Glucoron(os)yltranasferases N-Acetyltransferases Amino acid N-acyl transferases Sulfotransferases Monoamine Oxidase Many 112 May interactions listed for Selegiline! be very significant Used less frequently due to safer agents Relative Contribution to Drug Metabolism - Phase I Evans & Relling Science 1999 Pharmacogenetics of Phase I Drug Metabolism Weinshilboum, R. N Engl J Med 2003;348:529-537 Relative Contribution to Drug Metabolism - Phase II Evans & Relling Science 1999 Pharmacogenetics of Phase II Drug Metabolism Pharmacogen etics of Phase II Drug Metabolism Weinshilboum, R. N Engl J Med 2003;348:529-537 Pharmacogenetics of Acetylation Pharma cogene tics of Acetyla tion Weinshilboum, R. N Engl J Med 2003;348:529-537 Drug Interactions: Phase II Rarely rate-limiting step in either elimination or detoxification Phase I reactions increase polarity and excretion due to increased water solubility Assessing the Clinical Relevance of CYP450 Drug Interactions 1. 2. 3. 4. 5. 6. 7. 8. Therapeutic Index and toxic potential of the substrate Alternate pathways of metabolism Role of active metabolites Consequences of metabolic inhibition of metabolites Are multiple P450s inhibited by inhibitor Polymorphism of isoenzyme and patient’s metabolizer status Inhibitory potential of metabolites Is inhibition helpful or harmful Mechanisms of Interactions Pharmacokinetic Absorption Distribution Metabolism Excretion Pharmacokinetic: Excretion Filtration Renally cleared drugs affected notably digoxin and aminoglycoside antibiotics Metabolic products of parent drug Highly dependent upon GFR of host, elderly of great concern Active Two secretion non-specific active transport systems (pars recta) Organic acids Organic bases Also digoxin in distal tubule Reabsorption Distal tubule and collecting duct Dependent on flow, pH Useful for enhancing excretion of selected agents with inhibition Probenecid, drug ingestions Interactions Due to Altered Renal Excretion Drugs excreted by glomerular filtration unlikely to have significant interactions Drugs that are actively secreted into the tubular lumen can be inhibited by other drugs Sometimes useful: Probenecid decreases Cl of penicillin Sometimes toxic Methotrexate secretion inhibited by aspirin Lithium carbonate excretion affected by total body Na balance Altered sodium balance: thiazide and loop diuretics, some NSAIDs Characterizing Drug Interactions Interacting agents Mechanism Pharmacodynamic Receptor inhibition Additive effects Pharmacokinetic Altered absorption, distribution, metabolism, or elimination Drug - Disease Drug-drug Prescription Non-prescription Clinical Significance Major Manageable Substantial morbidity and mortality Therapy altering Little or no change in therapy Optimize therapy Intentional Additive or synergistic effects Enhanced pharmacokinetics Illicit, Food, recreational supplements, herbal products Drug-Disease Interactions Liver disease Renal disease Cardiac disease (hepatic blood flow) Acute myocardial infarction? Acute viral infection? Hypothyroidism or hyperthyroidism? SIRS ? Drug-Food Interactions Tetracycline and milk products Warfarin and vitamin K-containing foods Grapefruit juice Effects of grapefruit juice on felodipine pharmacokinetics and pharmacodynamics. Effects of grapefruit juice on felodipine pharmacokinetics and pharmacodynamics Dresser GK et al Clin Pharmacol Ther 2000;68(1):28–34 Drug-Herbal Interactions St. John’s wort with indinavir St. John’s wort with cyclosporin St. John’s wort with digoxin? Many others After St. John’s wort Prediction of Drug Interactions, In vitro In Vitro Screening Non-mammalian in vivo systems have very limited clinical utility In vitro systems to screen for CYP450-mediated drug interactions include microsomes, hepatocytes, liver slices, purified P450 systems, and recombinant human P450 enzymes. Most useful for screening inhibitory effects. Less useful for drugs with multiple metabolic pathways. Least useful for studying induction. Unknown appropriate concentration of inhibitor in vitro that would correlate with in vivo exposure. Utility in guiding subsequent clinical trials In Vivo Drug-Drug Interaction Studies Pharmacokinetic interactions must be evaluated relative to clinical relevance. Studies should be used for OPI Study design dictated by clinical objective (ex. cross-over versus parallel) Chronic versus acute dosing Sequence Relevant concentrations Steady-state versus acute short interval Endpoints (pharmacokinetic vs. pharmacodynamic) Sample size, statistical considerations Demonstration of “Lack of effect” vs. “Magnitude of effect” In Vivo Drug-Drug Interaction Studies, cont’d. Study populations Population pharmacokinetic approach In vitro characterization of likely targets Subgroups Safety concerns Clinical trials Concurrent Case pharmacokinetic studies Reports Prediction of Drug Interactions, Resources Clinical Trials CDER Guidance for Industry [http://0www.fda.gov.lilac.une.edu/cder/guidance/clin3.pdf] The Conduct of In Vitro and In Vivo Drug-Drug Interaction Studies: A Pharmaceutical Research and Manufacturers of America (PhRMA) Perspective. TD Bjornsson, and Others. Drug Met Disp 2003; 31: 815832. Case Reports: MedWatch @ FDA General Approach to Managing Drug Interactions Each contact with the patient includes a review of all medications prescribed and OTC. Information on medications prescribed by any and all health-care providers is reviewed Specifically query for problematic food and nutriceutical products Keep a high “Index of Suspicion” for all toxic events and therapeutic failures When possible, use agents which are the least problematic Sometimes, timing of doses may minimize interactions, especially with food Proactively instruct patients about avoiding interactions Usually, management of interactions requires minimal alterations in therapeutic plan Conclusions Drug-drug interactions are part of drug therapy May be beneficial or hazardous Polypharmacy (therapy with many agents) is often unavoidable Estimated that for 5 or more agents the probability of interaction approaches 100% Managing drug interactions is often more important than avoiding Be most cautious with narrow TI agents Make use of resources Some interactions are absolutely contraindicated Drug interactions are significant cause of adverse drug events and cost billions in additional health care costs. At-risk patients are most affected, e.g. the elderly, the very young, the critically ill. Summary: Drug Interactions Pharmacokinetic drug interactions are defined as those that alter drug absorption, distribution, metabolism, or excretion. Pharmacodynamic drug interactions result in an alteration of the biochemical or physiological effects of a drug. Interactions of this type are more difficult to characterize than pharmacokinetic interactions. Summary: Drug Interactions Drug interactions that alter the rate of absorption are usually of lesser concern that those that affect the extent. Overall outcomes of interactions of agonists and antagonists at the drug receptor are dependent on the varying affinities and activities of the different agents involved. Summary: Drug Interactions Alteration of metabolism of drugs in the liver, gut and other sites is an important but not singular source of significant drug interactions. In general, those drugs that are susceptible to the effects of induction of metabolism are also subject to inhibition. Drug interactions involving induction of metabolism develop more slowly than those involving inhibition. Summary: Drug Interactions A full profile of the interaction potential of any given drug generally takes an extended amount of time in the marketplace to be characterized. Many, but not all, important drug interactions are described in the official labeling. Summary Drug Metabolism Polymorphism of CYP gene(s) can result in a “poor metabolizer” phenotype, but occurs in less than 20% of the U.S. general population. Prototypic inhibiting agents include: Ciprofloxacin, Erythromycin, Fluconazole, Fluoxetine, Grapefruit juice, Itraconazole Prototypic inducing agents include: Carbamazepine (2C19, 3A4/5/7) Rifampin (2B6, 2CB, 2C19, 2C9, 2D6, 3A4/5/7) Questions? Blair Holbein, Ph.D. Presbyterian Hospital of Dallas Email: bholbein@hcin.net Website: http://phdres.caregate.net Annotated bibliography Slides