Article Type
Changed
Tue, 12/11/2018 - 15:06
Display Headline
Loss of enzyme induction: Ups and downs of a hidden drug-drug interaction

Mr. P, age 35 with schizophrenia and seizure disorder, has been maintained on risperidone, 6 mg qhs, and phenytoin, 300 mg qhs. For clinical reasons, the treating neurologist changes the anticonvulsant to divalproex. One week later, Mr. P presents to the emergency room complaining of jaw and neck stiffness.

Ms. K, age 43 with a history of schizoaffective disorder, bipolar type, and erratic medication adherence, is being treated with quetiapine, 600 mg at bedtime, and carbamazepine, 1,000 mg/d. Between appointments she stops taking carbamazepine, believing it is causing her to hear voices from her television. Two weeks later, the manager of Ms. K’s independent living facility tells the psychiatrist that the patient appears excessively sedated and has fallen twice in the past few days.

Mrs. T, a 39-year-old state hospital resident with schizoaffective disorder, bipolar type, has been treated with clozapine, 250 mg bid for 6 months; her most recent trough serum level was 492 ng/mL. She smokes 15 cigarettes/d. Two weeks after the hospital institutes a no-smoking policy, Mrs. T complains of excessive drooling and lightheadedness. Her trough clozapine level is now 875 ng/mL.

Discontinuing a medication that has enzyme-inducing effects presents a hidden problem for patients receiving antipsychotic pharmacotherapy. Certain hepatic enzymes responsible for antipsychotic metabolism—as well those involved in intercellular drug transport—are induced by medication or environmental exposures.1,2 Adding a medication that induces these enzymes to the regimen of a patient receiving antipsychotic therapy can result in markedly reduced serum antipsychotic levels, and discontinuing an inducing agent can result in increased antipsychotic levels.

Drug-drug interactions (DDIs) are a substantial contributor to adverse drug reactions (Box).3-7 Antipsychotic prescribing information highlights potential DDIs from the use of enzyme inhibitors and inducers but identifies only effects caused by adding a second agent. The prescriber remains the sole line of defense for monitoring for DDIs when discontinuing a medication that has inducing or inhibiting effects.

Most psychiatrists are aware that certain medications have clinically significant effects on cytochrome P450 (CYP) activity and of the potential for CYP inhibitors to generate DDIs. Clinicians often are aware of antidepressant medications’ CYP-inhibiting effects, know that levels of other medications will change when discontinuing a potent P450 inhibitor, and understand the need to increase dosages of medications influenced by such agents.8

However, few studies have evaluated the effects of enzyme induction on antipsychotic drug levels,9,10 and the literature rarely discusses changes in serum drug levels after loss of enzyme or drug transport induction.11 If unrecognized, these changes may have significant clinical consequences.

Box

Drug interactions: A common cause of nonadherence

Drug-drug interactions (DDIs) are a common and often preventable cause of morbidity and mortality. National surveillance data showed 700,000 emergency room visits related to adverse drug reactions (ADRs) in the 2 years from January 2004 through 2005.3 ADRs are particularly concerning for psychiatrists managing polypharmacy regimens for patients with severe mental disorders such as schizophrenia.

Literature on DDIs with antipsychotics focuses primarily on kinetic interactions that generate supratherapeutic drug levels.4,5 Because development of side effects is associated with reduced adherence, these kinetic interactions may increase the risk of adverse effects and lead to patients stopping the antipsychotic treatment.6,7

Two induction pathways

The primary mechanism underlying clinically significant DDIs occurs during CYP-mediated phase I metabolism. Molecules undergo oxidative conversion into metabolites that can be conjugated by phase II enzymes, generating more soluble forms that facilitate excretion.

The workhorse of human CYP metabolism is 3A4 (Table 1),12,13 which comprises 30% of hepatic activity and 70% of gut cytochrome activity.14 CYP 1A2 is responsible for 10% to 15% of CYP activity.

Both CYP 3A4 and 1A2 are inducible. A wide variety of medications induce 3A4 activity. The list of 1A2 inducers is shorter; the most common are aryl hydrocarbons from cigarette smoke and proton pump inhibitors.

CYP 2D6 accounts for 20% of hepatic cytochrome activity but is not inducible. CYP 2D6 is well known to psychiatrists because some selective serotonin reuptake inhibitors (SSRIs) and the non-SSRI antidepressant bupropion are potent inhibitors of this enzyme.15,16

P-glycoprotein (PGP) induction. Transmembrane shuttles such as P-glycoprotein (PGP) are an important component of drug disposition. PGP belongs to the family of ATP binding cassette (ABC) transporters that bring molecules across cellular barriers.17,18 It was first described in cancer cells that developed multiple drug resistance (MDR) and is often referred to as MDR1.19 PGP is encoded on human chromosome 7 and expressed in normal tissues, particularly in areas where cells seek to limit drug influx, such as those lining the luminal surface of the small and large intestine and those lining the blood-brain barrier and blood-testis barrier. The expression of PGP in hepatic cells promotes drug clearance by enhancing biliary drug excretion.

 

 

PGP is encoded on the same chromosome as CYP 3A4, and these 2 proteins frequently are expressed in the same cells, particularly in the intestinal lining and liver. Moreover, PGP is inducible, and there is substantial overlap between medications that are substrates for—or inducers of—PGP and CYP 3A4 activity. This makes it challenging to determine whether the kinetic effects of a second medication are the result of interference of 3A4, PGP, or both.

Polymorphisms in PGP activity may influence the penetration of psychotropic medications into the CNS. Studies indicate an association between certain PGP polymorphisms and treatment outcomes.17,18

Table 1

What induces CYP 1A2 and 3A4?

EnzymeDescriptionInducers*
CYP 1A2
  • Responsible for 10% to 15% of all CYP P450 activity
  • Located on chromosome 15
  • Low affinity/high capacity enzyme
  • Low affinity/high capacity enzyme
  • Prevalence of genetic polymorphisms conferring poor metabolizer status: 12% to 13%
Aryl hydrocarbons (smoking), protonpump inhibitors (omeprazole > lansoprazole > pantoprazole), modafinil, St. John’s wort, chargrilled meat, cruciferous vegetables such as broccoli and cabbage, flavones, protein supplements
CYP 3A4
  • Responsible for 30% of hepatic CYP 450 activity, 70% of gut cytochrome activity
  • Located on chromosome 7 (same as PGP)
  • Low affinity/high capacity enzyme
  • Little evidence for significant functional polymorphisms
Carbamazepine, phenytoin, phenobarbital, rifampin, oxcarbazepine, efavirenz, glucocorticoids, modafinil, nevirapine, pioglitazone, St. John’s wort
* Listed in order from strongest to weakest induction
CYP: cytochrome P450; PGP: P-glycoprotein
Source: References 12,13

Stopping an inducer

In general, inducers of CYP enzymes stimulate gene transcription within hours of exposure; maximum transcriptional activity occurs after 10 to 12 hours of exposure. As transcription increases, the concentration of the CYP mRNA transcript steadily accumulates, as does concentration of CYP protein.

After an inducer is discontinued, transcription returns to basal levels within 18 hours; however, the degradation of CYP proteins is a first-order process, with a half-life of 8 to 30 hours. As a result, the decrease in cellular CYP concentration—and the level of activity—lags behind the decreased synthesis from reduced mRNA levels.

As with other first-order kinetic processes, the expected decrease in CYP activity will require 5 half-lives to reach the new steady state (ie, back to baseline CYP activity). This suggests that drug levels previously decreased by CYP induction will reach their peak on average 1 to 2 weeks after the inducer is discontinued.20

Interactions with antipsychotics

Effects on serum antipsychotic levels caused by discontinuing a CYP or PGP inducer can be predicted from data on decreases in antipsychotic levels following inducer exposure. Except for ziprasidone and paliperidone, most atypical antipsychotics are prone to substantial decreases during concomitant inducer use (Table 2).21

The effect of enzyme inducers on risperidone is particularly interesting. Conversion of risperidone to its active metabolite 9-OH risperidone (paliperidone) occurs primarily via 2D6,22 yet concurrent use of carbamazepine—a potent CYP 3A4 inducer—results in a 50% decrease in the concentration of the active moiety (risperidone plus 9-OH risperidone). This finding and other early investigations suggested that CYP 3A had a role in risperidone metabolism,23,24 but these early studies and case series often involved molecules that had activity at both 3A4 and PGP. Further research clarified that effects on PGP—and not 3A4—are responsible for the changes in risperidone metabolism observed with the use of carbamazepine and other medications.25,26

Induction in case patients: Follow-up. Regardless of whether induction is mediated by ≥1 metabolic pathways, the loss of the inducer will result in serum antipsychotic increases that are proportional to the initial decrease.20 For example, with risperidone, the expected decrease is 50%. Therefore, after Mr. P stopped taking phenytoin, his serum risperidone level would be expected to double, which resulted in extrapyramidal side effects.

Quetiapine clearance is increased 5-fold by inducer exposure, so a clinician treating Ms. K would expect a marked increase in somnolence—and possibly orthostasis—as serum quetiapine levels peak 1 to 2 weeks following carbamazepine discontinuation.

The effects of smoking cessation on serum clozapine levels have been well-documented.1,27 Clinicians should anticipate median increases in serum clozapine levels of 55% after a patient discontinues smoking (aryl hydrocarbon exposure), but changes vary substantially among individuals. Mrs. T’s serum clozapine increased approximately 78%.

Careful clinical monitoring and slow downward adjustment of antipsychotic doses could have prevented the adverse effects these 3 patients experienced after loss of CYP/PGP induction and the consequences those side effects present for future medication adherence. When loss of induction is unplanned—as when Ms. K stopped taking carbamazepine but continued quetiapine—clinicians need to be alert to the fact that the patient was prescribed an inducer and include the loss of induction as a hypothesis for the patient’s somnolence.

 

 

Table 2

Effects of CYP/PGP induction on atypical antipsychotics

AntipsychoticMetabolic pathwaysEffect of induction
Aripiprazole2D6 and 3A4 convert aripiprazole to active metabolite dehydro-aripiprazole3A4 induction decreases maximum concentration of aripiprazole and metabolite by 70%
ClozapineMultiple enzymes convert clozapine to N-desmethylclozapine; mean contributions of CYP 1A2, 2C19, 3A4, 2C9, and 2D6 are 30%, 24%, 22%, 12%, and 6%, respectively, with CYP 1A2 predominantly involved at low concentrationsLoss of smoking-related 1A2 induction results in 50% increase in serum levels
OlanzapineDirect glucuronidation or 1A2-mediated oxidation to N-desmethlyolanzapineCarbamazepine use increases clearance by 50%. Olanzapine concentration:dose ratio is about 5-fold lower in smokers (7.9 +/- 2.6) than in nonsmokers (1.56 +/- 1.1; P
Paliperidone59% excreted unchanged in urine; phase I metabolism accounts for ≤10% of drug clearanceUnlikely to significantly impact levels, but impact of PGP induction is unknown
Quetiapine3A4-mediated sulfoxidation to inactive metabolite is primary pathway, but numerous metabolites noted, with 1 active metabolite (norquetiapine)Phenytoin increases clearance 5-fold
Risperidone2D6 converts risperidone to active metabolite 9-OH risperidoneIn a drug interaction study of risperidone, 6 mg/d for 3 weeks, followed by 3 weeks of carbamazepine, active moiety concentration was decreased by about 50%
Ziprasidone3A4 (~1/3); aldehyde oxidase (~2/3)Approximately 35% decrease in ziprasidone exposure by carbamazepine
CYP: cytochrome P450; PGP: P-glycoprotein
Source: Reference 21

Clinical considerations

In the absence of detailed data on antipsychotic metabolism, clinicians can make intelligent decisions regarding potential DDIs by:

  • knowing the extent of induction by common offenders (such as carbamazepine or phenytoin) documented in the medication’s prescribing information or demonstrated through convincing case reports or case series
  • memorizing the list of CYP 1A2 and CYP 3A4/PGP inducers.

Although the list of CYP 1A2 and CYP 3A4/PGP inducers is short, it is essential for clinicians to consult a readily available source of this information that is periodically updated to account for newer medications, such as the online table maintained by Flockhart (see Related Resources).28

Patients who may be susceptible to effects from loss of enzyme induction (including smokers receiving olanzapine or clozapine or others taking 3A4/PGP inducers) must be identified, and plans made for dosage adjustments if inducing agents are discontinued for a sufficient time (≥1 week) to result in downregulation of CYP or PGP activity. A slow taper of the antipsychotic over 1 to 2 weeks to the new target dose should compensate for loss of enzyme or PGP induction.

For newer antipsychotic medications with limited data, the proposed discontinuation of an inducer should, at the minimum, prompt a discussion between the psychiatrist and patient regarding the expected increase in serum antipsychotic levels and potential adverse effects that may result. Clinicians also must make every attempt to stay apprised of a patient’s current medications, bearing in mind that another provider may prescribe an inducer. Patients with schizophrenia always should be educated to contact the psychiatrist following any change in medication regimen, placing particular emphasis on the 1 or 2 medications that are known to be implicated in DDIs with the patient’s current antipsychotic.

Related Resources

  • Flockhart DA. Drug interactions: Cytochrome P450 drug interaction table. Indiana University School of Medicine. 2007. http://medicine.iupui.edu/flockhart/table.htm.
  • Cozza KL, Armstrong SC, Oesterheld JR. Concise guide to drug interaction principles for medical practice: Cytochrome P450s, UGTS, p-glycoproteins. Washington, DC: American Psychiatric Press, Inc; 2003.
Drug brand names

  • Aripiprazole • Abilify
  • Bupropion • Wellbutrin
  • Carbamazepine • Carbatrol, Tegretol
  • Clozapine • Clozaril
  • Divalproex • Depakote
  • Efavirenz • Sustiva
  • Lansoprazole • Prevacid
  • Modafinil • Provigil
  • Nevirapine • Viramune
  • Olanzapine • Zyprexa
  • Omeprazole • Prilosec
  • Oxcarbazepine • Trileptal
  • Paliperidone • Invega
  • Pantoprazole • Protonix
  • Phenobarbital • Barbita, Luminal, others
  • Phenytoin • Dilantin
  • Pioglitazone • Actos
  • Quetiapine • Seroquel
  • Rifampin • Rifadin, Rimactane
  • Risperidone • Risperdal
  • Ziprasidone • Geodon
Disclosure

Dr. Meyer receives grant/research support from the National Institute of Mental Health, Pfizer Inc., and the University of California. He is a consultant to Bristol-Myers Squibb, Organon, Vanda Pharmaceuticals, and Wyeth, and a speaker for AstraZeneca, Bristol-Myers Squibb, Dainippon Sumitomo Pharma, and Pfizer Inc.

Ms. Leckband reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. Meyer JM. Individual changes in clozapine levels after smoking cessation: results and a predictive model. J Clin Psychopharmacol. 2001;21:569-574.

2. Wong YW, Yeh C, Thyrum PT. The effects of concomitant phenytoin administration on the steady-state pharmacokinetics of quetiapine. J Clin Psychopharmacol. 2001;21:89-93.

3. Budnitz DS, Pollock DA, Weidenbach KN, et al. National surveillance of emergency department visits for outpatient adverse drug events. JAMA. 2006;296:1858-1866.

4. Prior TI, Baker GB. Interactions between the cytochrome P450 system and the second-generation antipsychotics. J Psychiatry Neurosci. 2003;28:99-112.

5. Spina E, de Leon J. Metabolic drug interactions with newer antipsychotics: a comparative review. Basic Clin Pharmacol Toxicol. 2007;100:4-22.

6. Preskorn SH. Drug-drug interactions: proof of relevance (part II): cause of tolerability problems or noncompliance. J Psychiatr Pract. 2005;11:397-401.

7. Weiden PJ, Mackell JA, McDonnell D. Obesity as a risk factor for antipsychotic noncompliance. Schizophr Res. 2004;66:51-7.

8. Preskorn SH, Flockhart D. 2006 guide to psychiatric drug interactions. Prim Psychiatry. 2006;13:35-64.

9. Spina E, Perucca E. Clinical significance of pharmacokinetic interactions between antiepileptic and psychotropic drugs. Epilepsia. 2002;43(suppl 2):37-44.

10. Meyer JM. Drug-drug interactions with antipsychotics. CNS Spectr. 2007;12:6-9.

11. Takahashi H, Yoshida K, Higuchi H, et al. Development of parkinsonian symptoms after discontinuation of carbamazepine in patients concurrently treated with risperidone: two case reports. Clin Neuropharmacol. 2001;24:358-360.

12. Rendic S. Summary of information on human CYP enzymes: human P450 metabolism data. Drug Metab Rev. 2002;34:83-448.

13. Hong CC, Tang BK, Hammond GL, et al. Cytochrome P450 1A2 (CYP1A2) activity and risk factors for breast cancer: a cross-sectional study. Breast Cancer Res. 2004;6:R352-365.

14. Cozza KL, Armstrong SC, Oesterheld JR. Concise guide to drug interaction principles for medical practice: cytochrome P450s, UGTS, p-glycoproteins. Washington, DC: American Psychiatric Press, Inc; 2003.

15. Kirchheiner J, Seeringer A. Clinical implications of pharmacogenetics of cytochrome P450 drug metabolizing enzymes. Biochim Biophys Acta. 2007;1770:489-494.

16. Kotlyar M, Brauer LH, Tracy TS, et al. Inhibition of CYP2D6 activity by bupropion. J Clin Psychopharmacol. 2005;25:226-229.

17. Uhr M, Tontsch A, Namendorf C, et al. Polymorphisms in the drug transporter gene ABCB1 predict antidepressant treatment response in depression. Neuron. 2008;57:203-209.

18. Bozina N, Kuzman MR, Medved V, et al. Associations between MDR1 gene polymorphisms and schizophrenia and therapeutic response to olanzapine in female schizophrenic patients. J Psychiatr Res. 2008;42:89-97.

19. Kim RB. Drugs as p-glycoprotein substrates, inhibitors, and inducers. Drug Metab Rev. 2002;34:47-54.

20. Hollenberg PF. Characteristics and common properties of inhibitors, inducers, and activators of CYP enzymes. Drug Metab Rev. 2002;34:17-35.

21. Physicians’ desk reference. 62nd ed. Montvale, NJ: Thomson Healthcare Inc.; 2007.

22. Heykants J, Huang ML, Mannens G, et al. The pharmacokinetics of risperidone in humans: a summary. J Clin Psychiatry. 1994;55 (suppl):13-7.

23. de Leon J, Bork J. Risperidone and cytochrome P450 3A. J Clin Psychiatry. 1997;58:450.-

24. Lane HY, Chang WH. Risperidone-carbamazepine interactions: is cytochrome P450 3A involved? J Clin Psychiatry. 1998;59:430-431.

25. Ejsing TB, Pedersen AD, Linnet K. P-glycoprotein interaction with risperidone and 9-OH-risperidone studied in vitro, in knock-out mice and in drug-drug interaction experiments. Hum Psychopharmacol. 2005;20:493-500.

26. Cousein E, Barthelemy C, Poullain S, et al. P-glycoprotein and cytochrome P450 3A4 involvement in risperidone transport using an in vitro Caco-2/TC7 model and an in vivo model. Prog Neuropsychopharmacol Biol Psychiatry. 2007;31:878-886.

27. Rostami-Hodjegan A, Amin AM, et al. Influence of dose, cigarette smoking, age, sex, and metabolic activity on plasma clozapine concentrations: a predictive model and nomograms to aid clozapine dose adjustment and to assess compliance in individual patients. J Clin Psychopharmacol. 2004;24:70-78.

28. Flockhart DA. Drug interactions: cytochrome P450 drug interaction table. Indiana University School of Medicine. 2007. Available at: http://medicine.iupui.edu/flockhart/table.htm. Accessed October 22, 2008.

Article PDF
Author and Disclosure Information

Jonathan M. Meyer, MD
Assistant professor, department of psychiatry, University of California, San Diego, staff psychiatrist, VA San Diego Healthcare System

Susan G. Leckband, RPh, BCPP
Assistant clinical professor, school of pharmacy, University of California, San Diego, psychiatric clinical pharmacist specialist, VA San Diego Healthcare System

Issue
Current Psychiatry - 08(01)
Publications
Page Number
47-53
Legacy Keywords
enzyme induction; antipsychotics; drug interactions; DDIs; p-glycoprotein; PGP; P450 CYP; CYP1A2; CYP 3A4; Jonathan Meyer; Susan Leckband
Sections
Author and Disclosure Information

Jonathan M. Meyer, MD
Assistant professor, department of psychiatry, University of California, San Diego, staff psychiatrist, VA San Diego Healthcare System

Susan G. Leckband, RPh, BCPP
Assistant clinical professor, school of pharmacy, University of California, San Diego, psychiatric clinical pharmacist specialist, VA San Diego Healthcare System

Author and Disclosure Information

Jonathan M. Meyer, MD
Assistant professor, department of psychiatry, University of California, San Diego, staff psychiatrist, VA San Diego Healthcare System

Susan G. Leckband, RPh, BCPP
Assistant clinical professor, school of pharmacy, University of California, San Diego, psychiatric clinical pharmacist specialist, VA San Diego Healthcare System

Article PDF
Article PDF

Mr. P, age 35 with schizophrenia and seizure disorder, has been maintained on risperidone, 6 mg qhs, and phenytoin, 300 mg qhs. For clinical reasons, the treating neurologist changes the anticonvulsant to divalproex. One week later, Mr. P presents to the emergency room complaining of jaw and neck stiffness.

Ms. K, age 43 with a history of schizoaffective disorder, bipolar type, and erratic medication adherence, is being treated with quetiapine, 600 mg at bedtime, and carbamazepine, 1,000 mg/d. Between appointments she stops taking carbamazepine, believing it is causing her to hear voices from her television. Two weeks later, the manager of Ms. K’s independent living facility tells the psychiatrist that the patient appears excessively sedated and has fallen twice in the past few days.

Mrs. T, a 39-year-old state hospital resident with schizoaffective disorder, bipolar type, has been treated with clozapine, 250 mg bid for 6 months; her most recent trough serum level was 492 ng/mL. She smokes 15 cigarettes/d. Two weeks after the hospital institutes a no-smoking policy, Mrs. T complains of excessive drooling and lightheadedness. Her trough clozapine level is now 875 ng/mL.

Discontinuing a medication that has enzyme-inducing effects presents a hidden problem for patients receiving antipsychotic pharmacotherapy. Certain hepatic enzymes responsible for antipsychotic metabolism—as well those involved in intercellular drug transport—are induced by medication or environmental exposures.1,2 Adding a medication that induces these enzymes to the regimen of a patient receiving antipsychotic therapy can result in markedly reduced serum antipsychotic levels, and discontinuing an inducing agent can result in increased antipsychotic levels.

Drug-drug interactions (DDIs) are a substantial contributor to adverse drug reactions (Box).3-7 Antipsychotic prescribing information highlights potential DDIs from the use of enzyme inhibitors and inducers but identifies only effects caused by adding a second agent. The prescriber remains the sole line of defense for monitoring for DDIs when discontinuing a medication that has inducing or inhibiting effects.

Most psychiatrists are aware that certain medications have clinically significant effects on cytochrome P450 (CYP) activity and of the potential for CYP inhibitors to generate DDIs. Clinicians often are aware of antidepressant medications’ CYP-inhibiting effects, know that levels of other medications will change when discontinuing a potent P450 inhibitor, and understand the need to increase dosages of medications influenced by such agents.8

However, few studies have evaluated the effects of enzyme induction on antipsychotic drug levels,9,10 and the literature rarely discusses changes in serum drug levels after loss of enzyme or drug transport induction.11 If unrecognized, these changes may have significant clinical consequences.

Box

Drug interactions: A common cause of nonadherence

Drug-drug interactions (DDIs) are a common and often preventable cause of morbidity and mortality. National surveillance data showed 700,000 emergency room visits related to adverse drug reactions (ADRs) in the 2 years from January 2004 through 2005.3 ADRs are particularly concerning for psychiatrists managing polypharmacy regimens for patients with severe mental disorders such as schizophrenia.

Literature on DDIs with antipsychotics focuses primarily on kinetic interactions that generate supratherapeutic drug levels.4,5 Because development of side effects is associated with reduced adherence, these kinetic interactions may increase the risk of adverse effects and lead to patients stopping the antipsychotic treatment.6,7

Two induction pathways

The primary mechanism underlying clinically significant DDIs occurs during CYP-mediated phase I metabolism. Molecules undergo oxidative conversion into metabolites that can be conjugated by phase II enzymes, generating more soluble forms that facilitate excretion.

The workhorse of human CYP metabolism is 3A4 (Table 1),12,13 which comprises 30% of hepatic activity and 70% of gut cytochrome activity.14 CYP 1A2 is responsible for 10% to 15% of CYP activity.

Both CYP 3A4 and 1A2 are inducible. A wide variety of medications induce 3A4 activity. The list of 1A2 inducers is shorter; the most common are aryl hydrocarbons from cigarette smoke and proton pump inhibitors.

CYP 2D6 accounts for 20% of hepatic cytochrome activity but is not inducible. CYP 2D6 is well known to psychiatrists because some selective serotonin reuptake inhibitors (SSRIs) and the non-SSRI antidepressant bupropion are potent inhibitors of this enzyme.15,16

P-glycoprotein (PGP) induction. Transmembrane shuttles such as P-glycoprotein (PGP) are an important component of drug disposition. PGP belongs to the family of ATP binding cassette (ABC) transporters that bring molecules across cellular barriers.17,18 It was first described in cancer cells that developed multiple drug resistance (MDR) and is often referred to as MDR1.19 PGP is encoded on human chromosome 7 and expressed in normal tissues, particularly in areas where cells seek to limit drug influx, such as those lining the luminal surface of the small and large intestine and those lining the blood-brain barrier and blood-testis barrier. The expression of PGP in hepatic cells promotes drug clearance by enhancing biliary drug excretion.

 

 

PGP is encoded on the same chromosome as CYP 3A4, and these 2 proteins frequently are expressed in the same cells, particularly in the intestinal lining and liver. Moreover, PGP is inducible, and there is substantial overlap between medications that are substrates for—or inducers of—PGP and CYP 3A4 activity. This makes it challenging to determine whether the kinetic effects of a second medication are the result of interference of 3A4, PGP, or both.

Polymorphisms in PGP activity may influence the penetration of psychotropic medications into the CNS. Studies indicate an association between certain PGP polymorphisms and treatment outcomes.17,18

Table 1

What induces CYP 1A2 and 3A4?

EnzymeDescriptionInducers*
CYP 1A2
  • Responsible for 10% to 15% of all CYP P450 activity
  • Located on chromosome 15
  • Low affinity/high capacity enzyme
  • Low affinity/high capacity enzyme
  • Prevalence of genetic polymorphisms conferring poor metabolizer status: 12% to 13%
Aryl hydrocarbons (smoking), protonpump inhibitors (omeprazole > lansoprazole > pantoprazole), modafinil, St. John’s wort, chargrilled meat, cruciferous vegetables such as broccoli and cabbage, flavones, protein supplements
CYP 3A4
  • Responsible for 30% of hepatic CYP 450 activity, 70% of gut cytochrome activity
  • Located on chromosome 7 (same as PGP)
  • Low affinity/high capacity enzyme
  • Little evidence for significant functional polymorphisms
Carbamazepine, phenytoin, phenobarbital, rifampin, oxcarbazepine, efavirenz, glucocorticoids, modafinil, nevirapine, pioglitazone, St. John’s wort
* Listed in order from strongest to weakest induction
CYP: cytochrome P450; PGP: P-glycoprotein
Source: References 12,13

Stopping an inducer

In general, inducers of CYP enzymes stimulate gene transcription within hours of exposure; maximum transcriptional activity occurs after 10 to 12 hours of exposure. As transcription increases, the concentration of the CYP mRNA transcript steadily accumulates, as does concentration of CYP protein.

After an inducer is discontinued, transcription returns to basal levels within 18 hours; however, the degradation of CYP proteins is a first-order process, with a half-life of 8 to 30 hours. As a result, the decrease in cellular CYP concentration—and the level of activity—lags behind the decreased synthesis from reduced mRNA levels.

As with other first-order kinetic processes, the expected decrease in CYP activity will require 5 half-lives to reach the new steady state (ie, back to baseline CYP activity). This suggests that drug levels previously decreased by CYP induction will reach their peak on average 1 to 2 weeks after the inducer is discontinued.20

Interactions with antipsychotics

Effects on serum antipsychotic levels caused by discontinuing a CYP or PGP inducer can be predicted from data on decreases in antipsychotic levels following inducer exposure. Except for ziprasidone and paliperidone, most atypical antipsychotics are prone to substantial decreases during concomitant inducer use (Table 2).21

The effect of enzyme inducers on risperidone is particularly interesting. Conversion of risperidone to its active metabolite 9-OH risperidone (paliperidone) occurs primarily via 2D6,22 yet concurrent use of carbamazepine—a potent CYP 3A4 inducer—results in a 50% decrease in the concentration of the active moiety (risperidone plus 9-OH risperidone). This finding and other early investigations suggested that CYP 3A had a role in risperidone metabolism,23,24 but these early studies and case series often involved molecules that had activity at both 3A4 and PGP. Further research clarified that effects on PGP—and not 3A4—are responsible for the changes in risperidone metabolism observed with the use of carbamazepine and other medications.25,26

Induction in case patients: Follow-up. Regardless of whether induction is mediated by ≥1 metabolic pathways, the loss of the inducer will result in serum antipsychotic increases that are proportional to the initial decrease.20 For example, with risperidone, the expected decrease is 50%. Therefore, after Mr. P stopped taking phenytoin, his serum risperidone level would be expected to double, which resulted in extrapyramidal side effects.

Quetiapine clearance is increased 5-fold by inducer exposure, so a clinician treating Ms. K would expect a marked increase in somnolence—and possibly orthostasis—as serum quetiapine levels peak 1 to 2 weeks following carbamazepine discontinuation.

The effects of smoking cessation on serum clozapine levels have been well-documented.1,27 Clinicians should anticipate median increases in serum clozapine levels of 55% after a patient discontinues smoking (aryl hydrocarbon exposure), but changes vary substantially among individuals. Mrs. T’s serum clozapine increased approximately 78%.

Careful clinical monitoring and slow downward adjustment of antipsychotic doses could have prevented the adverse effects these 3 patients experienced after loss of CYP/PGP induction and the consequences those side effects present for future medication adherence. When loss of induction is unplanned—as when Ms. K stopped taking carbamazepine but continued quetiapine—clinicians need to be alert to the fact that the patient was prescribed an inducer and include the loss of induction as a hypothesis for the patient’s somnolence.

 

 

Table 2

Effects of CYP/PGP induction on atypical antipsychotics

AntipsychoticMetabolic pathwaysEffect of induction
Aripiprazole2D6 and 3A4 convert aripiprazole to active metabolite dehydro-aripiprazole3A4 induction decreases maximum concentration of aripiprazole and metabolite by 70%
ClozapineMultiple enzymes convert clozapine to N-desmethylclozapine; mean contributions of CYP 1A2, 2C19, 3A4, 2C9, and 2D6 are 30%, 24%, 22%, 12%, and 6%, respectively, with CYP 1A2 predominantly involved at low concentrationsLoss of smoking-related 1A2 induction results in 50% increase in serum levels
OlanzapineDirect glucuronidation or 1A2-mediated oxidation to N-desmethlyolanzapineCarbamazepine use increases clearance by 50%. Olanzapine concentration:dose ratio is about 5-fold lower in smokers (7.9 +/- 2.6) than in nonsmokers (1.56 +/- 1.1; P
Paliperidone59% excreted unchanged in urine; phase I metabolism accounts for ≤10% of drug clearanceUnlikely to significantly impact levels, but impact of PGP induction is unknown
Quetiapine3A4-mediated sulfoxidation to inactive metabolite is primary pathway, but numerous metabolites noted, with 1 active metabolite (norquetiapine)Phenytoin increases clearance 5-fold
Risperidone2D6 converts risperidone to active metabolite 9-OH risperidoneIn a drug interaction study of risperidone, 6 mg/d for 3 weeks, followed by 3 weeks of carbamazepine, active moiety concentration was decreased by about 50%
Ziprasidone3A4 (~1/3); aldehyde oxidase (~2/3)Approximately 35% decrease in ziprasidone exposure by carbamazepine
CYP: cytochrome P450; PGP: P-glycoprotein
Source: Reference 21

Clinical considerations

In the absence of detailed data on antipsychotic metabolism, clinicians can make intelligent decisions regarding potential DDIs by:

  • knowing the extent of induction by common offenders (such as carbamazepine or phenytoin) documented in the medication’s prescribing information or demonstrated through convincing case reports or case series
  • memorizing the list of CYP 1A2 and CYP 3A4/PGP inducers.

Although the list of CYP 1A2 and CYP 3A4/PGP inducers is short, it is essential for clinicians to consult a readily available source of this information that is periodically updated to account for newer medications, such as the online table maintained by Flockhart (see Related Resources).28

Patients who may be susceptible to effects from loss of enzyme induction (including smokers receiving olanzapine or clozapine or others taking 3A4/PGP inducers) must be identified, and plans made for dosage adjustments if inducing agents are discontinued for a sufficient time (≥1 week) to result in downregulation of CYP or PGP activity. A slow taper of the antipsychotic over 1 to 2 weeks to the new target dose should compensate for loss of enzyme or PGP induction.

For newer antipsychotic medications with limited data, the proposed discontinuation of an inducer should, at the minimum, prompt a discussion between the psychiatrist and patient regarding the expected increase in serum antipsychotic levels and potential adverse effects that may result. Clinicians also must make every attempt to stay apprised of a patient’s current medications, bearing in mind that another provider may prescribe an inducer. Patients with schizophrenia always should be educated to contact the psychiatrist following any change in medication regimen, placing particular emphasis on the 1 or 2 medications that are known to be implicated in DDIs with the patient’s current antipsychotic.

Related Resources

  • Flockhart DA. Drug interactions: Cytochrome P450 drug interaction table. Indiana University School of Medicine. 2007. http://medicine.iupui.edu/flockhart/table.htm.
  • Cozza KL, Armstrong SC, Oesterheld JR. Concise guide to drug interaction principles for medical practice: Cytochrome P450s, UGTS, p-glycoproteins. Washington, DC: American Psychiatric Press, Inc; 2003.
Drug brand names

  • Aripiprazole • Abilify
  • Bupropion • Wellbutrin
  • Carbamazepine • Carbatrol, Tegretol
  • Clozapine • Clozaril
  • Divalproex • Depakote
  • Efavirenz • Sustiva
  • Lansoprazole • Prevacid
  • Modafinil • Provigil
  • Nevirapine • Viramune
  • Olanzapine • Zyprexa
  • Omeprazole • Prilosec
  • Oxcarbazepine • Trileptal
  • Paliperidone • Invega
  • Pantoprazole • Protonix
  • Phenobarbital • Barbita, Luminal, others
  • Phenytoin • Dilantin
  • Pioglitazone • Actos
  • Quetiapine • Seroquel
  • Rifampin • Rifadin, Rimactane
  • Risperidone • Risperdal
  • Ziprasidone • Geodon
Disclosure

Dr. Meyer receives grant/research support from the National Institute of Mental Health, Pfizer Inc., and the University of California. He is a consultant to Bristol-Myers Squibb, Organon, Vanda Pharmaceuticals, and Wyeth, and a speaker for AstraZeneca, Bristol-Myers Squibb, Dainippon Sumitomo Pharma, and Pfizer Inc.

Ms. Leckband reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

Mr. P, age 35 with schizophrenia and seizure disorder, has been maintained on risperidone, 6 mg qhs, and phenytoin, 300 mg qhs. For clinical reasons, the treating neurologist changes the anticonvulsant to divalproex. One week later, Mr. P presents to the emergency room complaining of jaw and neck stiffness.

Ms. K, age 43 with a history of schizoaffective disorder, bipolar type, and erratic medication adherence, is being treated with quetiapine, 600 mg at bedtime, and carbamazepine, 1,000 mg/d. Between appointments she stops taking carbamazepine, believing it is causing her to hear voices from her television. Two weeks later, the manager of Ms. K’s independent living facility tells the psychiatrist that the patient appears excessively sedated and has fallen twice in the past few days.

Mrs. T, a 39-year-old state hospital resident with schizoaffective disorder, bipolar type, has been treated with clozapine, 250 mg bid for 6 months; her most recent trough serum level was 492 ng/mL. She smokes 15 cigarettes/d. Two weeks after the hospital institutes a no-smoking policy, Mrs. T complains of excessive drooling and lightheadedness. Her trough clozapine level is now 875 ng/mL.

Discontinuing a medication that has enzyme-inducing effects presents a hidden problem for patients receiving antipsychotic pharmacotherapy. Certain hepatic enzymes responsible for antipsychotic metabolism—as well those involved in intercellular drug transport—are induced by medication or environmental exposures.1,2 Adding a medication that induces these enzymes to the regimen of a patient receiving antipsychotic therapy can result in markedly reduced serum antipsychotic levels, and discontinuing an inducing agent can result in increased antipsychotic levels.

Drug-drug interactions (DDIs) are a substantial contributor to adverse drug reactions (Box).3-7 Antipsychotic prescribing information highlights potential DDIs from the use of enzyme inhibitors and inducers but identifies only effects caused by adding a second agent. The prescriber remains the sole line of defense for monitoring for DDIs when discontinuing a medication that has inducing or inhibiting effects.

Most psychiatrists are aware that certain medications have clinically significant effects on cytochrome P450 (CYP) activity and of the potential for CYP inhibitors to generate DDIs. Clinicians often are aware of antidepressant medications’ CYP-inhibiting effects, know that levels of other medications will change when discontinuing a potent P450 inhibitor, and understand the need to increase dosages of medications influenced by such agents.8

However, few studies have evaluated the effects of enzyme induction on antipsychotic drug levels,9,10 and the literature rarely discusses changes in serum drug levels after loss of enzyme or drug transport induction.11 If unrecognized, these changes may have significant clinical consequences.

Box

Drug interactions: A common cause of nonadherence

Drug-drug interactions (DDIs) are a common and often preventable cause of morbidity and mortality. National surveillance data showed 700,000 emergency room visits related to adverse drug reactions (ADRs) in the 2 years from January 2004 through 2005.3 ADRs are particularly concerning for psychiatrists managing polypharmacy regimens for patients with severe mental disorders such as schizophrenia.

Literature on DDIs with antipsychotics focuses primarily on kinetic interactions that generate supratherapeutic drug levels.4,5 Because development of side effects is associated with reduced adherence, these kinetic interactions may increase the risk of adverse effects and lead to patients stopping the antipsychotic treatment.6,7

Two induction pathways

The primary mechanism underlying clinically significant DDIs occurs during CYP-mediated phase I metabolism. Molecules undergo oxidative conversion into metabolites that can be conjugated by phase II enzymes, generating more soluble forms that facilitate excretion.

The workhorse of human CYP metabolism is 3A4 (Table 1),12,13 which comprises 30% of hepatic activity and 70% of gut cytochrome activity.14 CYP 1A2 is responsible for 10% to 15% of CYP activity.

Both CYP 3A4 and 1A2 are inducible. A wide variety of medications induce 3A4 activity. The list of 1A2 inducers is shorter; the most common are aryl hydrocarbons from cigarette smoke and proton pump inhibitors.

CYP 2D6 accounts for 20% of hepatic cytochrome activity but is not inducible. CYP 2D6 is well known to psychiatrists because some selective serotonin reuptake inhibitors (SSRIs) and the non-SSRI antidepressant bupropion are potent inhibitors of this enzyme.15,16

P-glycoprotein (PGP) induction. Transmembrane shuttles such as P-glycoprotein (PGP) are an important component of drug disposition. PGP belongs to the family of ATP binding cassette (ABC) transporters that bring molecules across cellular barriers.17,18 It was first described in cancer cells that developed multiple drug resistance (MDR) and is often referred to as MDR1.19 PGP is encoded on human chromosome 7 and expressed in normal tissues, particularly in areas where cells seek to limit drug influx, such as those lining the luminal surface of the small and large intestine and those lining the blood-brain barrier and blood-testis barrier. The expression of PGP in hepatic cells promotes drug clearance by enhancing biliary drug excretion.

 

 

PGP is encoded on the same chromosome as CYP 3A4, and these 2 proteins frequently are expressed in the same cells, particularly in the intestinal lining and liver. Moreover, PGP is inducible, and there is substantial overlap between medications that are substrates for—or inducers of—PGP and CYP 3A4 activity. This makes it challenging to determine whether the kinetic effects of a second medication are the result of interference of 3A4, PGP, or both.

Polymorphisms in PGP activity may influence the penetration of psychotropic medications into the CNS. Studies indicate an association between certain PGP polymorphisms and treatment outcomes.17,18

Table 1

What induces CYP 1A2 and 3A4?

EnzymeDescriptionInducers*
CYP 1A2
  • Responsible for 10% to 15% of all CYP P450 activity
  • Located on chromosome 15
  • Low affinity/high capacity enzyme
  • Low affinity/high capacity enzyme
  • Prevalence of genetic polymorphisms conferring poor metabolizer status: 12% to 13%
Aryl hydrocarbons (smoking), protonpump inhibitors (omeprazole > lansoprazole > pantoprazole), modafinil, St. John’s wort, chargrilled meat, cruciferous vegetables such as broccoli and cabbage, flavones, protein supplements
CYP 3A4
  • Responsible for 30% of hepatic CYP 450 activity, 70% of gut cytochrome activity
  • Located on chromosome 7 (same as PGP)
  • Low affinity/high capacity enzyme
  • Little evidence for significant functional polymorphisms
Carbamazepine, phenytoin, phenobarbital, rifampin, oxcarbazepine, efavirenz, glucocorticoids, modafinil, nevirapine, pioglitazone, St. John’s wort
* Listed in order from strongest to weakest induction
CYP: cytochrome P450; PGP: P-glycoprotein
Source: References 12,13

Stopping an inducer

In general, inducers of CYP enzymes stimulate gene transcription within hours of exposure; maximum transcriptional activity occurs after 10 to 12 hours of exposure. As transcription increases, the concentration of the CYP mRNA transcript steadily accumulates, as does concentration of CYP protein.

After an inducer is discontinued, transcription returns to basal levels within 18 hours; however, the degradation of CYP proteins is a first-order process, with a half-life of 8 to 30 hours. As a result, the decrease in cellular CYP concentration—and the level of activity—lags behind the decreased synthesis from reduced mRNA levels.

As with other first-order kinetic processes, the expected decrease in CYP activity will require 5 half-lives to reach the new steady state (ie, back to baseline CYP activity). This suggests that drug levels previously decreased by CYP induction will reach their peak on average 1 to 2 weeks after the inducer is discontinued.20

Interactions with antipsychotics

Effects on serum antipsychotic levels caused by discontinuing a CYP or PGP inducer can be predicted from data on decreases in antipsychotic levels following inducer exposure. Except for ziprasidone and paliperidone, most atypical antipsychotics are prone to substantial decreases during concomitant inducer use (Table 2).21

The effect of enzyme inducers on risperidone is particularly interesting. Conversion of risperidone to its active metabolite 9-OH risperidone (paliperidone) occurs primarily via 2D6,22 yet concurrent use of carbamazepine—a potent CYP 3A4 inducer—results in a 50% decrease in the concentration of the active moiety (risperidone plus 9-OH risperidone). This finding and other early investigations suggested that CYP 3A had a role in risperidone metabolism,23,24 but these early studies and case series often involved molecules that had activity at both 3A4 and PGP. Further research clarified that effects on PGP—and not 3A4—are responsible for the changes in risperidone metabolism observed with the use of carbamazepine and other medications.25,26

Induction in case patients: Follow-up. Regardless of whether induction is mediated by ≥1 metabolic pathways, the loss of the inducer will result in serum antipsychotic increases that are proportional to the initial decrease.20 For example, with risperidone, the expected decrease is 50%. Therefore, after Mr. P stopped taking phenytoin, his serum risperidone level would be expected to double, which resulted in extrapyramidal side effects.

Quetiapine clearance is increased 5-fold by inducer exposure, so a clinician treating Ms. K would expect a marked increase in somnolence—and possibly orthostasis—as serum quetiapine levels peak 1 to 2 weeks following carbamazepine discontinuation.

The effects of smoking cessation on serum clozapine levels have been well-documented.1,27 Clinicians should anticipate median increases in serum clozapine levels of 55% after a patient discontinues smoking (aryl hydrocarbon exposure), but changes vary substantially among individuals. Mrs. T’s serum clozapine increased approximately 78%.

Careful clinical monitoring and slow downward adjustment of antipsychotic doses could have prevented the adverse effects these 3 patients experienced after loss of CYP/PGP induction and the consequences those side effects present for future medication adherence. When loss of induction is unplanned—as when Ms. K stopped taking carbamazepine but continued quetiapine—clinicians need to be alert to the fact that the patient was prescribed an inducer and include the loss of induction as a hypothesis for the patient’s somnolence.

 

 

Table 2

Effects of CYP/PGP induction on atypical antipsychotics

AntipsychoticMetabolic pathwaysEffect of induction
Aripiprazole2D6 and 3A4 convert aripiprazole to active metabolite dehydro-aripiprazole3A4 induction decreases maximum concentration of aripiprazole and metabolite by 70%
ClozapineMultiple enzymes convert clozapine to N-desmethylclozapine; mean contributions of CYP 1A2, 2C19, 3A4, 2C9, and 2D6 are 30%, 24%, 22%, 12%, and 6%, respectively, with CYP 1A2 predominantly involved at low concentrationsLoss of smoking-related 1A2 induction results in 50% increase in serum levels
OlanzapineDirect glucuronidation or 1A2-mediated oxidation to N-desmethlyolanzapineCarbamazepine use increases clearance by 50%. Olanzapine concentration:dose ratio is about 5-fold lower in smokers (7.9 +/- 2.6) than in nonsmokers (1.56 +/- 1.1; P
Paliperidone59% excreted unchanged in urine; phase I metabolism accounts for ≤10% of drug clearanceUnlikely to significantly impact levels, but impact of PGP induction is unknown
Quetiapine3A4-mediated sulfoxidation to inactive metabolite is primary pathway, but numerous metabolites noted, with 1 active metabolite (norquetiapine)Phenytoin increases clearance 5-fold
Risperidone2D6 converts risperidone to active metabolite 9-OH risperidoneIn a drug interaction study of risperidone, 6 mg/d for 3 weeks, followed by 3 weeks of carbamazepine, active moiety concentration was decreased by about 50%
Ziprasidone3A4 (~1/3); aldehyde oxidase (~2/3)Approximately 35% decrease in ziprasidone exposure by carbamazepine
CYP: cytochrome P450; PGP: P-glycoprotein
Source: Reference 21

Clinical considerations

In the absence of detailed data on antipsychotic metabolism, clinicians can make intelligent decisions regarding potential DDIs by:

  • knowing the extent of induction by common offenders (such as carbamazepine or phenytoin) documented in the medication’s prescribing information or demonstrated through convincing case reports or case series
  • memorizing the list of CYP 1A2 and CYP 3A4/PGP inducers.

Although the list of CYP 1A2 and CYP 3A4/PGP inducers is short, it is essential for clinicians to consult a readily available source of this information that is periodically updated to account for newer medications, such as the online table maintained by Flockhart (see Related Resources).28

Patients who may be susceptible to effects from loss of enzyme induction (including smokers receiving olanzapine or clozapine or others taking 3A4/PGP inducers) must be identified, and plans made for dosage adjustments if inducing agents are discontinued for a sufficient time (≥1 week) to result in downregulation of CYP or PGP activity. A slow taper of the antipsychotic over 1 to 2 weeks to the new target dose should compensate for loss of enzyme or PGP induction.

For newer antipsychotic medications with limited data, the proposed discontinuation of an inducer should, at the minimum, prompt a discussion between the psychiatrist and patient regarding the expected increase in serum antipsychotic levels and potential adverse effects that may result. Clinicians also must make every attempt to stay apprised of a patient’s current medications, bearing in mind that another provider may prescribe an inducer. Patients with schizophrenia always should be educated to contact the psychiatrist following any change in medication regimen, placing particular emphasis on the 1 or 2 medications that are known to be implicated in DDIs with the patient’s current antipsychotic.

Related Resources

  • Flockhart DA. Drug interactions: Cytochrome P450 drug interaction table. Indiana University School of Medicine. 2007. http://medicine.iupui.edu/flockhart/table.htm.
  • Cozza KL, Armstrong SC, Oesterheld JR. Concise guide to drug interaction principles for medical practice: Cytochrome P450s, UGTS, p-glycoproteins. Washington, DC: American Psychiatric Press, Inc; 2003.
Drug brand names

  • Aripiprazole • Abilify
  • Bupropion • Wellbutrin
  • Carbamazepine • Carbatrol, Tegretol
  • Clozapine • Clozaril
  • Divalproex • Depakote
  • Efavirenz • Sustiva
  • Lansoprazole • Prevacid
  • Modafinil • Provigil
  • Nevirapine • Viramune
  • Olanzapine • Zyprexa
  • Omeprazole • Prilosec
  • Oxcarbazepine • Trileptal
  • Paliperidone • Invega
  • Pantoprazole • Protonix
  • Phenobarbital • Barbita, Luminal, others
  • Phenytoin • Dilantin
  • Pioglitazone • Actos
  • Quetiapine • Seroquel
  • Rifampin • Rifadin, Rimactane
  • Risperidone • Risperdal
  • Ziprasidone • Geodon
Disclosure

Dr. Meyer receives grant/research support from the National Institute of Mental Health, Pfizer Inc., and the University of California. He is a consultant to Bristol-Myers Squibb, Organon, Vanda Pharmaceuticals, and Wyeth, and a speaker for AstraZeneca, Bristol-Myers Squibb, Dainippon Sumitomo Pharma, and Pfizer Inc.

Ms. Leckband reports no financial relationship with any company whose products are mentioned in this article or with manufacturers of competing products.

References

1. Meyer JM. Individual changes in clozapine levels after smoking cessation: results and a predictive model. J Clin Psychopharmacol. 2001;21:569-574.

2. Wong YW, Yeh C, Thyrum PT. The effects of concomitant phenytoin administration on the steady-state pharmacokinetics of quetiapine. J Clin Psychopharmacol. 2001;21:89-93.

3. Budnitz DS, Pollock DA, Weidenbach KN, et al. National surveillance of emergency department visits for outpatient adverse drug events. JAMA. 2006;296:1858-1866.

4. Prior TI, Baker GB. Interactions between the cytochrome P450 system and the second-generation antipsychotics. J Psychiatry Neurosci. 2003;28:99-112.

5. Spina E, de Leon J. Metabolic drug interactions with newer antipsychotics: a comparative review. Basic Clin Pharmacol Toxicol. 2007;100:4-22.

6. Preskorn SH. Drug-drug interactions: proof of relevance (part II): cause of tolerability problems or noncompliance. J Psychiatr Pract. 2005;11:397-401.

7. Weiden PJ, Mackell JA, McDonnell D. Obesity as a risk factor for antipsychotic noncompliance. Schizophr Res. 2004;66:51-7.

8. Preskorn SH, Flockhart D. 2006 guide to psychiatric drug interactions. Prim Psychiatry. 2006;13:35-64.

9. Spina E, Perucca E. Clinical significance of pharmacokinetic interactions between antiepileptic and psychotropic drugs. Epilepsia. 2002;43(suppl 2):37-44.

10. Meyer JM. Drug-drug interactions with antipsychotics. CNS Spectr. 2007;12:6-9.

11. Takahashi H, Yoshida K, Higuchi H, et al. Development of parkinsonian symptoms after discontinuation of carbamazepine in patients concurrently treated with risperidone: two case reports. Clin Neuropharmacol. 2001;24:358-360.

12. Rendic S. Summary of information on human CYP enzymes: human P450 metabolism data. Drug Metab Rev. 2002;34:83-448.

13. Hong CC, Tang BK, Hammond GL, et al. Cytochrome P450 1A2 (CYP1A2) activity and risk factors for breast cancer: a cross-sectional study. Breast Cancer Res. 2004;6:R352-365.

14. Cozza KL, Armstrong SC, Oesterheld JR. Concise guide to drug interaction principles for medical practice: cytochrome P450s, UGTS, p-glycoproteins. Washington, DC: American Psychiatric Press, Inc; 2003.

15. Kirchheiner J, Seeringer A. Clinical implications of pharmacogenetics of cytochrome P450 drug metabolizing enzymes. Biochim Biophys Acta. 2007;1770:489-494.

16. Kotlyar M, Brauer LH, Tracy TS, et al. Inhibition of CYP2D6 activity by bupropion. J Clin Psychopharmacol. 2005;25:226-229.

17. Uhr M, Tontsch A, Namendorf C, et al. Polymorphisms in the drug transporter gene ABCB1 predict antidepressant treatment response in depression. Neuron. 2008;57:203-209.

18. Bozina N, Kuzman MR, Medved V, et al. Associations between MDR1 gene polymorphisms and schizophrenia and therapeutic response to olanzapine in female schizophrenic patients. J Psychiatr Res. 2008;42:89-97.

19. Kim RB. Drugs as p-glycoprotein substrates, inhibitors, and inducers. Drug Metab Rev. 2002;34:47-54.

20. Hollenberg PF. Characteristics and common properties of inhibitors, inducers, and activators of CYP enzymes. Drug Metab Rev. 2002;34:17-35.

21. Physicians’ desk reference. 62nd ed. Montvale, NJ: Thomson Healthcare Inc.; 2007.

22. Heykants J, Huang ML, Mannens G, et al. The pharmacokinetics of risperidone in humans: a summary. J Clin Psychiatry. 1994;55 (suppl):13-7.

23. de Leon J, Bork J. Risperidone and cytochrome P450 3A. J Clin Psychiatry. 1997;58:450.-

24. Lane HY, Chang WH. Risperidone-carbamazepine interactions: is cytochrome P450 3A involved? J Clin Psychiatry. 1998;59:430-431.

25. Ejsing TB, Pedersen AD, Linnet K. P-glycoprotein interaction with risperidone and 9-OH-risperidone studied in vitro, in knock-out mice and in drug-drug interaction experiments. Hum Psychopharmacol. 2005;20:493-500.

26. Cousein E, Barthelemy C, Poullain S, et al. P-glycoprotein and cytochrome P450 3A4 involvement in risperidone transport using an in vitro Caco-2/TC7 model and an in vivo model. Prog Neuropsychopharmacol Biol Psychiatry. 2007;31:878-886.

27. Rostami-Hodjegan A, Amin AM, et al. Influence of dose, cigarette smoking, age, sex, and metabolic activity on plasma clozapine concentrations: a predictive model and nomograms to aid clozapine dose adjustment and to assess compliance in individual patients. J Clin Psychopharmacol. 2004;24:70-78.

28. Flockhart DA. Drug interactions: cytochrome P450 drug interaction table. Indiana University School of Medicine. 2007. Available at: http://medicine.iupui.edu/flockhart/table.htm. Accessed October 22, 2008.

References

1. Meyer JM. Individual changes in clozapine levels after smoking cessation: results and a predictive model. J Clin Psychopharmacol. 2001;21:569-574.

2. Wong YW, Yeh C, Thyrum PT. The effects of concomitant phenytoin administration on the steady-state pharmacokinetics of quetiapine. J Clin Psychopharmacol. 2001;21:89-93.

3. Budnitz DS, Pollock DA, Weidenbach KN, et al. National surveillance of emergency department visits for outpatient adverse drug events. JAMA. 2006;296:1858-1866.

4. Prior TI, Baker GB. Interactions between the cytochrome P450 system and the second-generation antipsychotics. J Psychiatry Neurosci. 2003;28:99-112.

5. Spina E, de Leon J. Metabolic drug interactions with newer antipsychotics: a comparative review. Basic Clin Pharmacol Toxicol. 2007;100:4-22.

6. Preskorn SH. Drug-drug interactions: proof of relevance (part II): cause of tolerability problems or noncompliance. J Psychiatr Pract. 2005;11:397-401.

7. Weiden PJ, Mackell JA, McDonnell D. Obesity as a risk factor for antipsychotic noncompliance. Schizophr Res. 2004;66:51-7.

8. Preskorn SH, Flockhart D. 2006 guide to psychiatric drug interactions. Prim Psychiatry. 2006;13:35-64.

9. Spina E, Perucca E. Clinical significance of pharmacokinetic interactions between antiepileptic and psychotropic drugs. Epilepsia. 2002;43(suppl 2):37-44.

10. Meyer JM. Drug-drug interactions with antipsychotics. CNS Spectr. 2007;12:6-9.

11. Takahashi H, Yoshida K, Higuchi H, et al. Development of parkinsonian symptoms after discontinuation of carbamazepine in patients concurrently treated with risperidone: two case reports. Clin Neuropharmacol. 2001;24:358-360.

12. Rendic S. Summary of information on human CYP enzymes: human P450 metabolism data. Drug Metab Rev. 2002;34:83-448.

13. Hong CC, Tang BK, Hammond GL, et al. Cytochrome P450 1A2 (CYP1A2) activity and risk factors for breast cancer: a cross-sectional study. Breast Cancer Res. 2004;6:R352-365.

14. Cozza KL, Armstrong SC, Oesterheld JR. Concise guide to drug interaction principles for medical practice: cytochrome P450s, UGTS, p-glycoproteins. Washington, DC: American Psychiatric Press, Inc; 2003.

15. Kirchheiner J, Seeringer A. Clinical implications of pharmacogenetics of cytochrome P450 drug metabolizing enzymes. Biochim Biophys Acta. 2007;1770:489-494.

16. Kotlyar M, Brauer LH, Tracy TS, et al. Inhibition of CYP2D6 activity by bupropion. J Clin Psychopharmacol. 2005;25:226-229.

17. Uhr M, Tontsch A, Namendorf C, et al. Polymorphisms in the drug transporter gene ABCB1 predict antidepressant treatment response in depression. Neuron. 2008;57:203-209.

18. Bozina N, Kuzman MR, Medved V, et al. Associations between MDR1 gene polymorphisms and schizophrenia and therapeutic response to olanzapine in female schizophrenic patients. J Psychiatr Res. 2008;42:89-97.

19. Kim RB. Drugs as p-glycoprotein substrates, inhibitors, and inducers. Drug Metab Rev. 2002;34:47-54.

20. Hollenberg PF. Characteristics and common properties of inhibitors, inducers, and activators of CYP enzymes. Drug Metab Rev. 2002;34:17-35.

21. Physicians’ desk reference. 62nd ed. Montvale, NJ: Thomson Healthcare Inc.; 2007.

22. Heykants J, Huang ML, Mannens G, et al. The pharmacokinetics of risperidone in humans: a summary. J Clin Psychiatry. 1994;55 (suppl):13-7.

23. de Leon J, Bork J. Risperidone and cytochrome P450 3A. J Clin Psychiatry. 1997;58:450.-

24. Lane HY, Chang WH. Risperidone-carbamazepine interactions: is cytochrome P450 3A involved? J Clin Psychiatry. 1998;59:430-431.

25. Ejsing TB, Pedersen AD, Linnet K. P-glycoprotein interaction with risperidone and 9-OH-risperidone studied in vitro, in knock-out mice and in drug-drug interaction experiments. Hum Psychopharmacol. 2005;20:493-500.

26. Cousein E, Barthelemy C, Poullain S, et al. P-glycoprotein and cytochrome P450 3A4 involvement in risperidone transport using an in vitro Caco-2/TC7 model and an in vivo model. Prog Neuropsychopharmacol Biol Psychiatry. 2007;31:878-886.

27. Rostami-Hodjegan A, Amin AM, et al. Influence of dose, cigarette smoking, age, sex, and metabolic activity on plasma clozapine concentrations: a predictive model and nomograms to aid clozapine dose adjustment and to assess compliance in individual patients. J Clin Psychopharmacol. 2004;24:70-78.

28. Flockhart DA. Drug interactions: cytochrome P450 drug interaction table. Indiana University School of Medicine. 2007. Available at: http://medicine.iupui.edu/flockhart/table.htm. Accessed October 22, 2008.

Issue
Current Psychiatry - 08(01)
Issue
Current Psychiatry - 08(01)
Page Number
47-53
Page Number
47-53
Publications
Publications
Article Type
Display Headline
Loss of enzyme induction: Ups and downs of a hidden drug-drug interaction
Display Headline
Loss of enzyme induction: Ups and downs of a hidden drug-drug interaction
Legacy Keywords
enzyme induction; antipsychotics; drug interactions; DDIs; p-glycoprotein; PGP; P450 CYP; CYP1A2; CYP 3A4; Jonathan Meyer; Susan Leckband
Legacy Keywords
enzyme induction; antipsychotics; drug interactions; DDIs; p-glycoprotein; PGP; P450 CYP; CYP1A2; CYP 3A4; Jonathan Meyer; Susan Leckband
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media