ISSN: 2167-065X

Clinical Pharmacology & Biopharmaceutics
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)
  • Mini Review   
  • Clin Pharmacol Biopharm, Vol 13(6)

Translational Pharmacokinetics: Bridging Bench to Bedside

Adamu E Abechi*
Department of Chemistry, Ahmadu Bello University, Nigeria
*Corresponding Author: Adamu E Abechi, Department of Chemistry, Ahmadu Bello University, Nigeria, Email: adamu8989@gmail.com

Received: 04-Jun-2024 / Manuscript No. cpb-24-140298 / Editor assigned: 07-Jun-2024 / PreQC No. cpb-24-140298 / Reviewed: 20-Jun-2024 / QC No. cpb-24-140298 / Revised: 25-Jun-2024 / Manuscript No. cpb-24-140298 / Published Date: 28-Jun-2024

Abstract

Translational pharmacokinetics (PK) is crucial in bridging the gap between preclinical drug research and clinical application, providing essential insights into a drug's absorption, distribution, metabolism, and excretion (ADME). This article explores the importance of PK in drug development, highlighting the transition from laboratory studies to clinical practice. Key stages in this transition include preclinical PK studies, scaling and modeling, first-in-human studies, and extensive clinical trials. The case study of Infliximab, a monoclonal antibody used to treat chronic inflammatory diseases, illustrates the application of translational PK. Despite challenges such as inter-species differences and biological complexity, advancements in modeling techniques and personalized medicine hold promise for improving drug development and patient care.

Keywords

Translational pharmacokinetics; Drug development; Absorption; Distribution; Metabolism; Excretion Preclinical studies; Physiologically-based pharmacokinetic (PBPK) models; Infliximab; Personalized medicine; Dosing regimens; Therapeutic efficacy

Introduction

Translational pharmacokinetics (PK) is a field of study that aims to bridge the gap between preclinical drug research and clinical application. It involves understanding how a drug is absorbed, distributed, metabolized, and excreted in the body and translating this knowledge from laboratory settings to real-world clinical scenarios. This process is critical for developing safe and effective medications, optimizing dosing regimens, and ensuring therapeutic efficacy while minimizing adverse effects [1].

The importance of pharmacokinetics in drug development

Pharmacokinetics is a cornerstone of drug development and clinical pharmacology. It provides essential insights into a drug's behavior in the human body, influencing decisions at every stage of drug development, from initial discovery to post-marketing surveillance. Key parameters studied in PK include:

  • Absorption: How the drug enters the bloodstream.
  • Distribution: How the drug spreads throughout the body.
  • Metabolism: How the drug is chemically altered, usually in the liver.
  • Excretion: How the drug and its metabolites are eliminated from the body.

Understanding these processes helps in predicting drug concentrations at various sites of action, optimizing dosing intervals, and anticipating potential interactions with other medications [2].

Preclinical pharmacokinetics: the foundation

In the preclinical phase, PK studies are conducted using animal models and in vitro systems to predict human pharmacokinetics. These studies provide crucial data on:

  • Bioavailability: The proportion of the drug that reaches systemic circulation.
  • Half-life: The time it takes for the drug concentration to reduce by half.
  • Volume of distribution: The distribution of the drug within the body's compartments.

These parameters guide the selection of appropriate drug candidates for further development and inform the design of initial human trials [3].

Translational pharmacokinetics: from lab to clinic

The transition from preclinical to clinical PK involves several steps:

Scaling and Modeling: Mathematical models, such as physiologically-based pharmacokinetic (PBPK) models, are used to extrapolate preclinical data to humans. These models consider physiological differences between species to predict human responses.

First-in-Human (FIH) Studies: Initial clinical trials involve a small number of healthy volunteers to assess the drug's safety, tolerability, and PK profile in humans. These studies help to refine dosing regimens for subsequent trials.

Clinical Trials: As the drug progresses through Phase I, II, and III trials, PK data continue to play a pivotal role. They help in understanding dose-response relationships, identifying optimal dosing strategies, and assessing the impact of variables such as age, gender, and disease state on drug kinetics [4].

Case study: infliximab

Infliximab, a monoclonal antibody used to treat chronic inflammatory diseases such as Crohn's disease and rheumatoid arthritis, provides a compelling example of translational PK in action. Preclinical studies in animal models provided initial PK data, which were then scaled to predict human pharmacokinetics. Subsequent clinical trials refined these predictions, leading to the establishment of dosing regimens that balance efficacy and safety.

Challenges and future directions

Despite its successes, translational PK faces several challenges:

  • Inter-species Differences: Variability between animal models and humans can complicate the prediction of human pharmacokinetics.
  • Complexity of Biological Systems: Human bodies are complex and variable, making it challenging to predict drug behavior accurately.
  • Technological Limitations: While advanced modeling techniques exist, they require extensive data and computational resources.

Future directions in translational PK include the integration of more sophisticated models, such as machine learning algorithms, to predict PK parameters more accurately. Additionally, the use of biomarkers and personalized medicine approaches promises to tailor drug therapy to individual patients, enhancing therapeutic outcomes. [5].

Materials and methods

Materials

Preclinical models:

  • Animal Models: Rodents (mice, rats) and larger animals (dogs, primates) used for initial PK studies.
  • In vitro Systems: Human liver microsomes, hepatocytes, and other relevant cell lines for studying drug metabolism and transport.

Analytical techniques:

  • High-Performance Liquid Chromatography (HPLC) and Liquid Chromatography-Mass Spectrometry (LC-MS) for quantifying drug concentrations in biological samples.
  • Enzyme-linked Immunosorbent Assay (ELISA) for detecting and quantifying monoclonal antibodies like Infliximab [6].

Software and modeling tools:

  • Physiologically-based Pharmacokinetic (PBPK) modeling software such as Simcyp, GastroPlus, and PK-Sim.
  • Statistical software for data analysis, such as R or SAS.

Methods

Preclinical pharmacokinetic studies:

Animal studies:

  • Administer the drug to animals at varying doses.
  • Collect blood, tissue, and urine samples at predefined intervals.
  • Analyze samples using HPLC or LC-MS to determine drug concentrations.
  • Calculate PK parameters such as bioavailability, half-life, and volume of distribution using non-compartmental analysis [7].

In vitro studies:

  • Incubate the drug with human liver microsomes or hepatocytes.
  • Monitor metabolic stability and identify metabolites using LC-MS.
  • Assess drug transport and interaction with drug transporters in relevant cell lines.

Scaling and modeling:

  • Utilize PBPK models to extrapolate animal PK data to predict human pharmacokinetics.
  • Incorporate physiological differences between species into the models.
  • Validate models using available human data and refine as necessary [8].

First-in-human (FIH) studies:

  • Conduct Phase I clinical trials with a small cohort of healthy volunteers.
  • Administer single ascending doses of the drug to evaluate safety and tolerability.
  • Collect blood and urine samples at various time points.
  • Analyze samples for drug concentrations using HPLC or LC-MS.
  • Determine human PK parameters and compare with preclinical predictions.

Clinical trials:

  • Conduct Phase II and III clinical trials to further evaluate PK in patients with the target condition.
  • Collect extensive PK data at different dose levels and dosing regimens.
  • Analyze the impact of demographic factors (age, gender, weight) and disease state on drug pharmacokinetics.
  • Use population PK modeling to analyze data and optimize dosing regimens [9].

Case study: infliximab:

  • Review preclinical PK data from animal models and in vitro studies.
  • Analyze FIH and subsequent clinical trial data for PK parameters.
  • Evaluate the dosing regimens used in clinical practice based on PK data.

Assess the impact of factors such as immunogenicity and concomitant medications on Infliximab pharmacokinetics [10].

Discussion

Translational pharmacokinetics plays a pivotal role in the drug development process, ensuring that preclinical findings are effectively and safely translated into clinical applications. The comprehensive approach detailed in the Materials and Methods section demonstrates the systematic steps taken to bridge this gap, from preclinical models to patient care. This discussion will highlight key aspects and challenges of this translational journey, using the case study of Infliximab to illustrate practical applications.

One of the primary challenges in translational pharmacokinetics is the accurate extrapolation of animal data to humans. Physiological differences between species can lead to significant discrepancies in drug behavior. The use of PBPK models helps to address this issue by incorporating species-specific physiological parameters. These models, however, require extensive validation with human data to ensure their predictive accuracy.

First-in-human (FIH) studies are a critical milestone in the translational pathway. These studies provide the first insights into human pharmacokinetics and help to refine dosing regimens. The careful design of FIH studies, with single ascending doses and comprehensive safety monitoring, ensures that potential adverse effects are identified early. The data obtained from these studies are crucial for planning subsequent clinical trials.

The case of Infliximab exemplifies the translational process. Initial preclinical studies provided a foundation for understanding its pharmacokinetics, which were then extrapolated to humans using PBPK modeling. FIH and subsequent clinical trials refined these predictions, leading to the establishment of effective dosing regimens. Infliximab's PK profile, including its long half-life and limited bioavailability when administered orally, necessitated intravenous administration and specific dosing intervals to maintain therapeutic levels.

A significant challenge in the clinical application of pharmacokinetics is the variability among patients. Factors such as age, gender, weight, genetic polymorphisms, and comorbidities can influence drug metabolism and distribution. Population PK modeling addresses this variability by analyzing data from diverse patient populations, allowing for more personalized dosing strategies. For Infliximab, such modeling has been essential in optimizing doses for different patient groups, including those with varying degrees of inflammation and immune responses.

Another important consideration is the interaction of the drug with other medications. Drug-drug interactions can significantly alter pharmacokinetics, leading to reduced efficacy or increased toxicity. Continuous monitoring and updating of PK data throughout the drug's lifecycle are necessary to manage these interactions. For Infliximab, concomitant use with immunosuppressants like methotrexate has been shown to reduce immunogenicity and enhance therapeutic outcomes.

The integration of advanced technologies, such as machine learning and artificial intelligence, holds promise for the future of translational pharmacokinetics. These technologies can analyze vast datasets to identify patterns and predict PK behaviors more accurately, potentially reducing the reliance on animal models and streamlining the drug development process.

Conclusion

Translational pharmacokinetics is a vital component of the drug development process, serving as the bridge between laboratory research and clinical application. By thoroughly understanding and predicting how drugs behave within the human body, translational PK ensures that new therapies are safe, effective, and tailored to meet the needs of diverse patient populations.

The process begins with preclinical studies, where animal models and in vitro systems provide initial insights into a drug's absorption, distribution, metabolism, and excretion (ADME). These studies form the foundation upon which human pharmacokinetic predictions are built. However, the physiological differences between species present a significant challenge, necessitating the use of advanced PBPK models to extrapolate preclinical data to humans accurately.

First-in-human studies are a critical step in the translational pathway, offering the first real-world insights into a drug's behavior in humans. These studies help to refine dosing regimens and ensure safety before larger clinical trials. The subsequent phases of clinical trials further refine these regimens, incorporating data from diverse patient populations to optimize therapeutic outcomes.

The case study of Infliximab, a monoclonal antibody used to treat chronic inflammatory diseases, illustrates the practical application of translational pharmacokinetics. From its preclinical beginnings to its extensive clinical use, Infliximab's development highlights the importance of robust PK modeling, careful clinical trial design, and continuous monitoring to manage patient variability and drug-drug interactions effectively.

One of the primary challenges in translational pharmacokinetics is the variability among patients. Factors such as age, gender, genetic makeup, and comorbidities can significantly influence drug behavior. Population PK modeling addresses this challenge by analyzing data from a broad range of patients, allowing for more personalized and precise dosing strategies. For Infliximab, this approach has been crucial in optimizing treatment regimens and improving patient outcomes.

As technology advances, the field of translational pharmacokinetics is poised to become even more precise and efficient. Machine learning and artificial intelligence offer promising tools for analyzing complex datasets, predicting PK behaviors, and reducing reliance on animal models. These advancements will streamline the drug development process, making it faster and more cost-effective while maintaining a high standard of safety and efficacy.

References

  1. Carson PE, Flanagan CL, Ickes CE (1956) Enzymatic deficiency in primaquine-sensitive erythrocytes. Science 124: 484-485.
  2. Indexed at, Google Scholar, Crossref

  3. Chowbay B, Li H, David M (2005) Meta-analysis of the influence of MDR1 C3435T polymorphism on digoxin pharmacokinetics and MDR1 gene expression. Br J Clin Pharmacol 60: 159-171.
  4. Indexed at, Google Scholar, Crossref

  5. Beutler E, Gelbart T, Demina A (1998) Racial variability in the UDP-glucuronosyltransferase 1 (UGT1A1) promoter: a balanced polymorphism for regulation of bilirubin metabolism? Proc Natl Acad Sci U S A  95: 8170-8174.
  6. Indexed at, Google Scholar, Crossref

  7. Bosch TM, Meijerman I, Beijnen JH (2006) Genetic polymorphisms of drug-metabolising enzymes and drug transporters in the chemotherapeutic treatment of cancer. Clin Pharmacokinet 45: 253-285.
  8.         Indexed at, Google Scholar, Crossref

  9. Amado RG, Wolf M, Peeters M (2008) Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol 26: 1626-1634.
  10. Indexed at, Google Scholar, Crossref

  11. Apperley JF (2007) Part I: mechanisms of resistance to imatinib in chronic myeloid leukaemia. Lancet Oncol 8: 1018-1029.
  12. Indexed at, Google Scholar, Crossref

  13. Diasio RB, Beavers TL, Carpenter JT (1988) Familial deficiency of dihydropyrimidine dehydrogenase. Biochemical basis for familial pyrimidinemia and severe 5-fluorouracil-induced toxicity. J Clin Invest 81: 47-51.
  14. Indexed at, Google Scholar, Crossref

  15. Etienne MC, Lagrange JL, Dassonville O (1994) Population study of dihydropyrimidine dehydrogenase in cancer patients. J Clin Oncol 12: 2248-2253.
  16. Indexed at, Google Scholar, Crossref

  17. Iacopetta B, Grieu F, Joseph D (2001) A polymorphism in the enhancer region of the thymidylate synthase promoter influences the survival of colorectal cancer patients treated with 5-fluorouracil. Br J Cancer 85: 827-830.
  18. Indexed at, Google Scholar, Crossref

  19. Ingelman-Sundberg M, Sim SC, Gomez A (2007) Influence of cytochrome P450 polymorphisms on drug therapies: pharmacogenetic, pharmacoepigenetic and clinical aspects . Pharmacol Ther 116: 496-526.
  20. Indexed at, Google Scholar, Crossref

Citation: Adamu EA (2024) Translational Pharmacokinetics: Bridging Bench toBedside. Clin Pharmacol Biopharm, 13: 464.

Copyright: © 2024 Adamu EA. This is an open-access article distributed underthe terms of the Creative Commons Attribution License, which permits unrestricteduse, distribution, and reproduction in any medium, provided the original author andsource are credited.

Top