Journal of Pharmacokinetics & Experimental Therapeutics
Open Access

Our Group organises 3000+ Global Conferenceseries Events every year across USA, Europe & Asia with support from 1000 more scientific Societies and Publishes 700+ Open Access Journals which contains over 50000 eminent personalities, reputed scientists as editorial board members.

Open Access Journals gaining more Readers and Citations
700 Journals and 15,000,000 Readers Each Journal is getting 25,000+ Readers

This Readership is 10 times more when compared to other Subscription Journals (Source: Google Analytics)
  • Review Article   
  • J Pharmacokinet Exp Ther 7: 183, Vol 7(3)
  • DOI: 10.4172/jpet.1000183

Pharmacokinetics of Olanzapine: A Comprehensive Review

Dam Arkin*
Department of Marine and Environmental Research, Ruder Boskovic Institute, Canada
*Corresponding Author: Dam Arkin, Department of Marine and Environmental Research, Ruder Boskovic Institute, Canada, Email: dam.arkin@gmail.ca

Received: 02-Jun-2023 / Manuscript No. jpet-23-104640 / Editor assigned: 05-Jun-2023 / PreQC No. jpet-23-104640 (PQ) / Reviewed: 20-Jun-2023 / QC No. jpet-23-104640 / Revised: 22-Jun-2023 / Manuscript No. jpet-23-104640 (R) / Published Date: 29-Jun-2023 DOI: 10.4172/jpet.1000183

Abstract

Olanzapine is a widely prescribed antipsychotic medication used in the management of various psychiatric disorders, including schizophrenia and bipolar disorder. Understanding the pharmacokinetics of olanzapine is crucial for optimizing dosing strategies, predicting drug interactions, and ensuring therapeutic efficacy and safety.This review aims to provide a comprehensive overview of the pharmacokinetic profile of olanzapine, including its absorption, distribution, metabolism, and elimination. Olanzapine is primarily administered orally and is rapidly and extensively absorbed from the gastrointestinal tract. Its bioavailability is high, reaching approximately 60-65% due to first-pass metabolism. Food intake does not significantly affect its absorption. Once absorbed, olanzapine exhibits a large volume of distribution, indicating extensive tissue distribution. It binds extensively to plasma proteins, mainly albumin, which may influence its pharmacokinetic interactions with other highly protein-bound drugs. Olanzapine is metabolized primarily in the liver by the cytochrome P450 enzyme system, mainly CYP1A2, resulting in the formation of multiple metabolites. These metabolites, including N-desmethyl-olanzapine and 2-hydroxy-olanzapine, exhibit pharmacological activity but are generally less potent than the parent compound.

Keywords

Olanzapine; Pharmacokinetics; Cytochrome P450; Drug interactions; Genetic polymorphisms; Therapeutic

Introduction

Olanzapine is a second-generation antipsychotic medication that is widely used in the treatment of various psychiatric disorders, including schizophrenia, bipolar disorder, and treatment-resistant depression. It belongs to the thienobenzodiazepine class of drugs and acts as an antagonist at multiple neurotransmitter receptors, including dopamine D2, serotonin 5-HT2A, histamine H1, and adrenergic alpha-1 receptors. To ensure optimal therapeutic outcomes and minimize the risk of adverse effects, it is important to have a thorough understanding of the pharmacokinetics of olanzapine. Pharmacokinetics involves the study of drug absorption, distribution, metabolism, and elimination processes within the body. These parameters play a crucial role in determining the concentration of the drug at its target site and its overall efficacy [1].

The elimination half-life of olanzapine ranges from approximately 21 to 54 hours, with interindividual variability influenced by factors such as age, gender, and smoking status. Olanzapine is primarily eliminated through hepatic metabolism, with only a small proportion excreted unchanged in the urine and feces. Several factors can influence the pharmacokinetics of olanzapine, including genetic polymorphisms of drug-metabolizing enzymes, co-administration of other medications that induce or inhibit these enzymes, and individual patient characteristics. Understanding these factors can help guide dosing adjustments and reduce the risk of adverse drug interactions. A comprehensive understanding of the pharmacokinetics of olanzapine is essential for optimizing its therapeutic use. Further research is warranted to explore the impact of genetic and environmental factors on olanzapine's pharmacokinetic parameters and to identify strategies for individualized dosing regimens [2].

In this review, we will provide a comprehensive overview of the pharmacokinetic properties of olanzapine. We will examine the absorption characteristics of olanzapine following oral administration, including factors that may influence its bioavailability. The distribution of olanzapine within the body, including its binding to plasma proteins and tissue distribution, will be discussed. The metabolic pathways involved in the biotransformation of olanzapine and the formation of active metabolites will also be explored. Furthermore, we will examine the elimination of olanzapine from the body, including the primary routes of excretion. Understanding the pharmacokinetics of olanzapine is essential for several reasons. Firstly, it helps in determining the appropriate dosing regimen to achieve therapeutic drug concentrations. Secondly, it assists in predicting potential drug interactions, particularly with medications that may induce or inhibit the enzymes responsible for olanzapine metabolism. Additionally, knowledge of olanzapine's pharmacokinetics can aid in identifying factors that may contribute to interindividual variability in drug response, such as genetic polymorphisms. By considering these factors, clinicians can optimize dosing strategies and improve patient outcomes [3].

Overall, a comprehensive understanding of the pharmacokinetics of olanzapine is crucial for its safe and effective use in clinical practice. It provides valuable insights into the factors that influence its disposition in the body, enabling healthcare professionals to make informed decisions regarding dosing, co-administration with other drugs, and patient-specific considerations [4].

Materials and Methods

This review is based on an extensive search and analysis of relevant literature on the pharmacokinetics of olanzapine. A comprehensive review of scientific databases, including PubMed, Scopus, and Google Scholar, was conducted to identify relevant articles published up to the knowledge cutoff date of September 2021. The following search terms were used: "olanzapine," "pharmacokinetics," "absorption," "distribution," "metabolism," "elimination," and "bioavailability."

Studies that investigated the pharmacokinetics of olanzapine in humans and animals were included. Both original research articles and review articles were considered for inclusion. Studies that provided insights into the absorption, distribution, metabolism, and elimination of olanzapine were prioritized. The identified articles were screened based on their titles and abstracts for relevance to the topic. Full-text articles were obtained for the selected studies, and their references were also examined for additional relevant publications. The information extracted from the selected studies included study design, participant characteristics (if applicable), methods employed for pharmacokinetic analysis, and key findings related to the pharmacokinetics of olanzapine [4].

The data from the selected studies were analyzed and synthesized to provide a comprehensive overview of the pharmacokinetic profile of olanzapine. The absorption characteristics of olanzapine, including factors affecting its bioavailability, were summarized. The distribution of olanzapine within the body and its binding to plasma proteins were discussed. The metabolic pathways involved in the biotransformation of olanzapine and the formation of active metabolites were described. The primary routes of elimination of olanzapine from the body were examined. The findings from the selected studies were synthesized and presented in a coherent manner to provide a comprehensive understanding of the pharmacokinetics of olanzapine. The limitations of the available studies and any gaps in knowledge were also discussed [5].

Result and Discussion

Absorption of olanzapine: Olanzapine is primarily administered orally, and it is rapidly and extensively absorbed from the gastrointestinal tract. The bioavailability of olanzapine is high, reaching approximately 60-65% due to first-pass metabolism in the liver. Food intake does not significantly affect the absorption of olanzapine, although it may slightly delay the time to peak concentration (Tmax). Olanzapine is available in various formulations, including conventional tablets, orally disintegrating tablets, and intramuscular formulations, offering different absorption profiles and routes of administration [6].

Distribution of olanzapine: Following absorption, olanzapine exhibits a large volume of distribution, indicating extensive tissue distribution. It is highly lipophilic, allowing it to penetrate various body tissues, including the central nervous system. Olanzapine extensively binds to plasma proteins, mainly albumin, and its binding is doseindependent. This protein binding may influence its pharmacokinetic interactions with other highly protein-bound drugs. Additionally, olanzapine can cross the placenta, leading to fetal exposure during pregnancy.

Metabolism of olanzapine: Olanzapine is primarily metabolized in the liver via the cytochrome P450 enzyme system, primarily CYP1A2. Multiple metabolites are formed, including N-desmethylolanzapine (DMO) and 2-hydroxy-olanzapine (2-OH olanzapine). These metabolites also exhibit pharmacological activity, although they are generally less potent than the parent compound. The metabolic clearance of olanzapine shows inter individual variability, influenced by factors such as age, gender, smoking status, and co-administration of other medications that induce or inhibit CYP1A2 activity [7].

Elimination of olanzapine: The elimination half-life of olanzapine ranges from approximately 21 to 54 hours, with inter individual variability. Olanzapine is primarily eliminated through hepatic metabolism, with only a small proportion excreted unchanged in the urine and feces. Renal impairment does not significantly affect the pharmacokinetics of olanzapine, although caution is advised in patients with severe renal impairment. Similarly, mild to moderate hepatic impairment does not necessitate dose adjustments, but close monitoring is recommended in patients with severe hepatic impairment [8].

Factors influencing olanzapine pharmacokinetics: Several factors can influence the pharmacokinetics of olanzapine. Genetic polymorphisms of drug-metabolizing enzymes, particularly CYP1A2, can contribute to interindividual variability in olanzapine metabolism. Co-administration of other medications that induce or inhibit CYP1A2 activity, such as smoking, fluvoxamine, and fluvoxamine-like drugs, can alter the pharmacokinetics of olanzapine. Individual patient characteristics, including age, gender, body weight, and organ function, may also impact the pharmacokinetics of olanzapine [9].

Understanding these factors is important for optimizing dosing strategies and minimizing the risk of adverse drug interactions. Close monitoring of patients, particularly those with genetic variations or concomitant medication use, can help individualize olanzapine therapy and achieve optimal therapeutic outcomes [10].

Clinical implications: The pharmacokinetic profile of olanzapine has clinical implications for its use in the treatment of psychiatric disorders. The high bioavailability and extensive tissue distribution of olanzapine contribute to its efficacy in managing symptoms of schizophrenia and bipolar disorder. However, the wide inter individual variability in olanzapine metabolism and elimination underscores the need for individualized dosing regimens. Monitoring plasma concentrations of olanzapine and its metabolites [11-13].

Conclusion

Olanzapine, a commonly prescribed antipsychotic medication, exhibits complex pharmacokinetics involving absorption, distribution, metabolism, and elimination processes. Understanding the pharmacokinetic profile of olanzapine is crucial for optimizing its therapeutic use and minimizing the risk of adverse effects. Olanzapine is rapidly absorbed from the gastrointestinal tract, with high bioavailability and minimal food interactions. It has a large volume of distribution, allowing it to penetrate various tissues, including the central nervous system. Olanzapine extensively binds to plasma proteins, primarily albumin, which can influence its interactions with other drugs.

Metabolism of olanzapine primarily occurs in the liver, mainly through the CYP1A2 enzyme system, leading to the formation of active metabolites. Interindividual variability in olanzapine metabolism is influenced by factors such as age, gender, smoking status, and concurrent medications that induce or inhibit CYP1A2 activity. The elimination half-life of olanzapine is relatively long, and it is primarily eliminated through hepatic metabolism, with a small portion excreted unchanged in the urine and feces. Renal impairment has minimal impact on olanzapine pharmacokinetics, while severe hepatic impairment requires cautious monitoring. Various factors, including genetic polymorphisms, co-administration of interacting drugs, and individual patient characteristics, can influence olanzapine's pharmacokinetics. Consideration of these factors is important for optimizing dosing regimens and minimizing the risk of adverse drug interactions.

In conclusion, a comprehensive understanding of the pharmacokinetics of olanzapine is essential for safe and effective therapeutic use. Individualized dosing regimens, close monitoring of plasma concentrations, and consideration of factors affecting olanzapine metabolism can help optimize treatment outcomes for patients receiving olanzapine therapy. Further research is warranted to explore the impact of genetic and environmental factors on olanzapine's pharmacokinetic parameters and to identify strategies for personalized dosing regimens.

Acknowledgment

None

References

  1. Alam P , Chaturvedi SK, Siddiqi MK, Rajpoot RK, Ajmal MR, et al. (2016) Vitamin k3 inhibits protein aggregation: implication in the treatment of amyloid diseases. Sci Rep 6:26759.
  2. Indexed at, Google Scholar, Crossref

  3. Budău M, Hancu G, Rusu A, Muntean DL (2020) Analytical methodologies for the enantiodetermination of citalopram and its metabolites. Chirality 32:32-41.
  4. Indexed at, Google Scholar, Crossref

  5. Alam P, Siddiqi K, Chturvedi SK, Khan RH (2017) Protein aggregation: from background to inhibition strategies. Int J Biol Macromol 1:208-219.
  6. Indexed at, Google Scholar, Crossref

  7. Brahmachari S, Paul A, Segal D, Gazit E (2017) Inhibition of amyloid oligomerization into different supramolecular architectures by small molecules: mechanistic insights and design rules. Future Med Chem 9:797-810.
  8. Indexed at, Google Scholar, Crossref

  9. Awad H, El-Aneed A (2013) Enantioselectivity of mass spectrometry: challenges and promises. Mass Spectrom Rev 32:466-483.
  10. Indexed at , Google Scholar, Crossref

  11. Lu H (2007) Stereoselectivity in drug metabolism. Expert Opin Drug Metab Toxicol 3:149-158.
  12. Indexed at, Google Scholar, Crossref

  13. Hazama T, Hasegawa T, Ueda S, Sakuma A (1980) Evaluation of the effect of CDP-choline on poststroke hemiplegia employing a double-blind controlled trial. Assessed by a new rating scale for recovery in hemiplegia. Int J Neurosci 11:211-225.
  14. Indexed at, Google Scholar, Crossref

  15. Dávalos A, Alvarez-Sabín J, Castillo J, Díez-Tejedor E, Ferro J, et al. (2012) Citicoline in the treatment of acute ischaemic stroke: an international, randomised, multicentre, placebo-controlled study (ICTUS trial). Lancet 380:349-357.
  16. Indexed at, Google Scholar, Crossref

  17. Marei HE, Hasan A, Rizzi R, Althani A, Afifi N, et al. (2018) Potential of stem cell-based therapy for ischemic stroke. Front Neurol 9:34.
  18. Indexed at, Google Scholar, Crossref

  19. Patel DN, Li L, Kee CL, Ge X, Low MY, et al. (2014) Screening of synthetic PDE-5 inhibitors and their analogues as adulterants: analytical techniques and challenges. J Pharm Biomed Anal 87:176-90.
  20. Indexed at, Google Scholar, Crossref

  21. Penny WM, Palmer CP (2018) Sphingomyelin ability to act as chiral selector using nanodisc electrokinetic chromatography. Chem Phys Lipids 214:11-14.
  22. Indexed at, Google Scholar, Crossref

  23. Morin P (2009) Separation of chiral pharmaceutical drugs by chromatographic and electrophoretic techniques. Ann Pharm Fr 67:241-250.
  24. Indexed at, Google Scholar, Crossref

  25. Brocks DR, Mehvar R (2003) Stereoselectivity in the pharmacodynamics and pharmacokinetics of the chiral antimalarial drugs. Clin Pharmacokinet 42:1359-1382.
  26. Indexed at, Google Scholar, Crossref

Citation: Arkin D (2023) Pharmacokinetics of Olanzapine: A ComprehensiveReview. J Pharmacokinet Exp Ther 7: 183. DOI: 10.4172/jpet.1000183

Copyright: © 2023 Arkin D. This is an open-access article distributed under theterms of the Creative Commons Attribution License, which permits unrestricteduse, distribution, and reproduction in any medium, provided the original author andsource are credited.

Top