Abstract

Clofazimine (CFZ) is a weakly basic, small-molecule antibiotic used for the treatment of mycobacterial infections including leprosy and multidrug-resistant tuberculosis. Upon prolonged oral administration, CFZ precipitates and accumulates within macrophages throughout the host. To model the pharmacokinetics of CFZ, the volume of distribution (Vd) was considered as a varying parameter that increases with continuous drug loading. Fitting the time-dependent change in drug mass and concentration data obtained from CFZ-treated mice, we performed a quantitative analysis of the systemic disposition of the drug over a 20-week treatment period. The pharmacokinetics data were fitted using various classical compartmental models sampling serum and spleen concentration data into separate matrices. The models were constructed in NONMEM together with linear and nonlinear sigmoidal expansion functions to the spleen compartment to capture the phase transition in Vd. The different modeling approaches were compared by Akaike information criteria, observed and predicted concentration correlations, and graphically. Using the composite analysis of the modeling predictions, adaptive fractional CFZ sequestration, Vd and half-life were evaluated. When compared to standard compartmental models, an adaptive Vd model yielded a more accurate data fit of the drug concentrations in both the serum and spleen. Including a nonlinear sigmoidal equation into compartmental models captures the phase transition of drugs such as CFZ, greatly improving the prediction of population pharmacokinetics and yielding further insight into the mechanisms of drug disposition.

Highlights

  • Clofazimine (CFZ) is an antimycobacterial agent used to treat leprosy and multidrugresistant tuberculosis alongside some non-tuberculosis mycobacterial infections

  • The results reported are the first time that volume of distribution changes resulting from soluble-to-insoluble phase transitions of drug molecules are directly modeled and analyzed, using a population pharmacokinetics modeling approach that should be applicable to the study of CFZ pharmacokinetics in the human population

  • To account for the nonlinearity expected from crystal-like drug inclusions (CLDIs) formation, we added an adaptive function (V1) to the volume of the spleen compartment, the intercompartmental rate constant from compartment two to compartment one (K21), and the elimination rate constant (K)

Read more

Summary

Introduction

Clofazimine (CFZ) is an antimycobacterial agent used to treat leprosy and multidrugresistant tuberculosis alongside some non-tuberculosis mycobacterial infections. CFZ has been found to inhibit SARS-CoV-2 infection in vitro and in animal models [1,2], and its efficacy is currently being tested in a phase II clinical trial [3]. The accumulation of CFZ in macrophage lysosomes has been shown to occur by thermodynamically favorable conditions that lead to the precipitation of drugs by pH-dependent ion trapping. The hydrochloride crystals are found within membrane-associated complexes referred to as crystal-like drug inclusions (CLDIs). This phenomenon has been observed throughout the body, but it occurs primarily in macrophages of the spleen, liver, gut, and lungs. The large amount of CFZ that accumulates in macrophages over time directly leads to a pronounced expansion of the volume of distribution (Vd) of the drug [9]

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call