Skip to main content
Antimicrobial Agents and Chemotherapy logoLink to Antimicrobial Agents and Chemotherapy
. 2021 Sep 17;65(10):e00317-21. doi: 10.1128/AAC.00317-21

Population Pharmacokinetics of Diethylcarbamazine in Patients with Lymphatic Filariasis and Healthy Individuals

Veenu Bala a,h, Yashpal S Chhonker a, Abdullah Alshehri a, Constant Edi b, Catherine M Bjerum c, Benjamin G Koudou b,d,e, Christopher L King c,f, Daryl J Murry a,g,
PMCID: PMC8448115  PMID: 34310218

ABSTRACT

Diethylcarbamazine (DEC) is a drug of choice to treat lymphatic filariasis (LF) either used alone or in combination as mass drug administration (MDA) preventive strategies. The objective of this study was to develop a population pharmacokinetics (PK) model for DEC in subjects infected with lymphatic filariasis (LF) compared to healthy individuals, and to evaluate the effect of covariates on the volume of distribution (V/F) and oral clearance (CL/F) of DEC. This was an open-label cohort study of treatment-naive Wuchereria bancrofti-infected (n = 32) and uninfected (n = 24) adults residing in the Agboville District of Côte d’Ivoire. The population pharmacokinetics model for DEC was built using Phoenix NLME 8.0 software. The covariates included in the model-building process were age, gender, body weight, infection status, creatinine clearance (CLCR), and aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels. A total of 56 adults were enrolled in the study, and a total of 728 samples were obtained over 168 h. A one-compartment linear pharmacokinetics model with first-order absorption with an absorption lag time (Tlag) best described the data. After determining the pharmacokinetics (PK) parameters of DEC, body weight and gender were found to be the significant covariates for DEC V/F. The final population pharmacokinetics model adequately described the pharmacokinetics of DEC in the studied population. Model-based simulation indicated that the body weight significantly impacted the exposure in both the male and female populations. This analysis may further support the drug-drug interaction model development of DEC with different coadministered drugs or agents in disease control programs. (This study is registered at clinicaltrials.gov under identifier NCT02845713.)

KEYWORDS: lymphatic filariasis, diethylcarbamazine, population pharmacokinetics

INTRODUCTION

Lymphatic filariasis (LF), commonly known as elephantiasis, is a neglected tropical disease and caused by infection with nematode parasites of Wuchereria bancrofti, Brugia malayi, or Brugia timori (1). In 2000, 120 million people were estimated to be infected with LF, and now 880 million people in 52 countries remain at risk of acquiring LF (2). LF has therefore been targeted for elimination by the WHO as it is identified as one of the six potentially eradicable infectious diseases identified by the International Task Force for Disease Eradication (3). The WHO recommended mass drug administration (MDA) for LF elimination (4). Diethylcarbamazine citrate (DEC) combined with albendazole (ALB) is the primary drug of choice for MDA to treat LF infection outside sub-Saharan Africa (5). Because DEC plus albendazole fails to sustain microfilarial (Mf) clearance for prolonged periods of time, annual treatment is required for at least 5 years (the approximate life span of adult worms) with sufficient coverage to reduce Mf levels in the community below the threshold to sustain transmission. The addition of ivermectin (IVM) to DEC plus albendazole, or triple-drug therapy, is able to sustain Mf clearance for up to 5 years after a single coadministered regimen (68). Consequently, WHO has recommended triple-drug therapy for areas outside sub-Saharan Africa that have failed to eliminate LF with two-drug regimens or in areas that have not started MDA (9). Simulation modeling suggests that only two rounds of triple-drug therapy with sufficient coverage can achieve the LF elimination target in most areas (10).

DEC is thought to be the most potent antifiliarial drug in the triple-drug regimen. It kills both microfilariae and adult worms (11). DEC is cytotoxic to microfilariae within hours after taking the drug; however, significant disappearance of adult worms requires several months to become apparent. DEC is also less efficient at killing adults than microfilariae. A single dose of DEC at 6 mg/kg of body weight inactivates 50 to 80% of adult worms (1214). Exactly how DEC works to kill microfilariae and adult worms is currently unknown. DEC interferes with parasitic arachidonic acid metabolism via inhibiting prostaglandin H synthase (PGHS) (cyclooxygenase) in the parasites and is thought to make them more susceptible to immune attack (15). Because of the essential role of DEC in MDA for LF outside sub-Saharan Africa and significant pharmacokinetic (PK) variability (16), it is essential to evaluate dosing regimens that produce effective concentrations in diverse populations (i.e., pediatric, obese, etc.).

The study of population pharmacokinetics is an essential aspect of drug development and plays a vital role in finding the optimal and effective treatment dose of a drug of interest (17, 18). Population pharmacokinetics models are efficient in unfolding the pharmacokinetic behavior of investigated drug and to identify the possible source of variation (covariates) that may influence pharmacokinetics (19, 20). The WHO age-based dosing for DEC ranges from 100 to 300 mg across three different age groupings (2 to 5 years, 6 to 15 years, and >15 years) (21). Although age-based dosing is recommended by the WHO for LF MDA programs, the maximum recommended doses using these methods are lower than those indicated by weight-based dosing (6 mg/kg for DEC) and do not consider covariates that could affect drug levels. Using this dosing regimen for DEC may result in significant underdosing of DEC and reduced efficacy in some individuals. There are currently no studies describing the population pharmacokinetics and important covariates of DEC that would help to address this important limitation. The aim of this work is to develop a validated population pharmacokinetics model of DEC and explore the relation between drug levels and patient characteristics (age, sex, and weight) along with infection state and clinical determinates of organ function, including creatinine clearance (CLCR) and aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels.

RESULTS

Subject selection, sampling, and noncompartmental pharmacokinetics analysis.

A total of 56 subjects with a Wuchereria bancrofti-infected population (n = 32) and uninfected population (n = 24) were included in the data set. Table 1 summarizes the patient demographics. A total of 728 samples were collected in the study out to 168 h. The plasma DEC concentrations continuously declined after 8 h of administration and were detected only up to 72 h (as DEC was measurable only up to 72 h in all included volunteers) following oral administration of 6 mg/kg (DEC citrate). Cmax (1,550 ng/ml), apparent volume of distribution during terminal phase (Vz)/Fobs (114 liters), and CL/Fobs (8.36 liters/h) determined from noncompartmental analysis (NCA) were of further use as initial estimates in development of the population pharmacokinetics model of DEC.

TABLE 1.

Demographics of the study volunteers (n = 56)

Variable Result for:
Infected (n = 32) Uninfected (n = 24)
Age (yr), median (range) 43.50 (22–66) 36.5 (18–54)
Gender, no. (%)
    Male 20 (63) 12 (50)
    Female 12 (37) 12 (50)
Body wt (kg), median (range) 61.25 (55–135) 63.5 (51–89)
CLCR (ml/min), median (range) 79.80 (41.50–149.0) 79.25 (38.10–137.80)
AST (U/liter), median (range) 29.0 (21–53) 27.0 (15–52)
ALT (U/liter), median (range) 24.50 (14–46) 25.0 (16–67)

Population pharmacokinetics modeling.

On the basis of the goodness-of-fit criteria, DEC concentrations as a function of time were best described by a one-compartment model with an absorption lag time (22). Thus, pharmacokinetic models were parameterized in terms of absorption rate constant (Ka), lag time (Tlag), apparent clearance (CL/F) and apparent volume of distribution (V/F). A one-compartment additive plus proportional error model with Tlag for the given data was developed and compared on the basis of the maximum likelihood criterion (objective function value [OFV] of 2× the negative log likelihood value [−2LL]) and visual inspection of the fittings. The combined additive plus proportional error model with Tlag showed improvement in the OFV (−2LL, 7,635) in comparison to the additive model (−2LL, 8,475) or proportional error model (2LL, 7,765) alone. After adding a Tlag function with combined additive plus proportional error model (base model), OFV values further improved by 304 points (2LL, 7,331). Diagnostic plots for the base model showed a reasonable fit with no apparent trends of residuals over time or model predictions (see Fig. S1 in the supplemental material). The visual covariate screening procedure (scatter plots for continuous and box plots for discrete variables, shown in Fig. S2 and S3 in the supplemental material) indicated the effect of all covariates (age, body weight, gender, infection status, AST, ALT, and CLCR profile) with PK parameters. To check whether the covariates significantly altered the PK parameters, a model with all the included covariates was run using stepwise forward addition (P ≤ 0.01) followed by a backward elimination (P ≤ 0.001) with the likelihood ratio (−2LL) test. In the forward addition process, the effects of gender, body weight over V/F, and weight over CL/F were included in the model, while in the subtraction process, the effect of weight over CL/F was removed. The covariate analysis clearly indicated the significant effect of weight and gender over apparent volume of distribution (ΔOFV = −40) on DEC pharmacokinetics in this patient population. Eta covariate plots between population parameters and included covariates are shown in Fig. S2 and S3. The final selected population pharmacokinetics model incorporates the effects of body weight and gender covariate on volume (−2LL = 7,291). The final population values for Ka, V/F, CL/F, and Tlag were 1.37 h−1, 103.33 liters, 8.63 liters/h, and 0.74 h, respectively. The percentages of interindividual variability (IIV) calculated from final population model for Ka, V/F, CL/F, and Tlag are 77.45, 7.07, 18.59, and 15.91%, respectively, in comparison to the base model: i.e., 81.51% for Ka, 13.84% for V/F, 19.14% for CL/F, and 15.74% for Tlag. Goodness-of-fit plots for the final DEC population pharmacokinetics model are shown in Fig. 1. The plots of observed plasma concentrations (dependent variable [DV]) of DEC versus individual prediction (IPRED) and population predictions (PRED) were symmetrically distributed, which suggests the good prediction value of the final model. The values of weighted residuals (CWRES) ranged between 2 and −2 and were randomly scattered, indicating the minor differences in the prediction models and the suitability of this error model for this population. The population pharmacokinetics parameters for the final model are shown in Table 2. A summary of the full model development process is shown in Table S1 in the supplemental material in decreasing order of the OFVs depicting model improvement.

FIG 1.

FIG 1

Goodness-of-fit plots of DEC for the final population pharmacokinetics model.

TABLE 2.

Population pharmacokinetics parameter estimates of the final model and bootstrap analysis

Parametera Final model
Bootstrap
Estimate % CV 95% CI Median % CV 95% CI
Primary
    tvKa (1/h) 1.38 12.96 1.02–1.73 1.37 12.26 1.09–1.77
    tvV (liters) 103.33 3.00 97.2–109.4 103.27 2.70 97.69–109.17
    tvCL (liters/h) 8.63 2.72 8.17–9.09 8.64 2.57 8.19–9.09
    tvTlag (h) 0.74 6.09 0.65–0.83 0.74 6.25 0.62–0.81
    tvCmultiSD 0.152 12.61 0.11–0.19 0.151 12.46 0.11–0.19
    δVGender1 0.159 23.42 0.08–0.23 0.160 16.53 0.10–0.21
    δVwt 0.415 21.99 0.23–0.59 0.405 19.49 0.21–0.53
    SD, 0 2.91 26.09 1.42–4.40 3.04 26.70 1.78–5.10
Random effect (% IIV)
    ω2Ka 0.47 77.45 0.47 77.45
    ω2V 0.005 7.07 0.005 7.07
    ω2CL 0.034 18.59 0.033 18.31
    ω2Tlag 0.025 15.91 0.03 17.45
a

CV, coefficient of variation; CI, confidence interval; tvKa, typical value of rate of absorption; tvV, typical value of apparent volume of distribution; tvCL, typical value of apparent clearance; tvTlag, typical value of absorption lag time; tvCmultiSD, proportional error on percent scale 0.152 (15.2%); δVGender1, exponent for gender as a covariate for volume of distribution; δVwt, exponent for weight as a covariate for volume of distribution; SD, 0 (square root of CEps variance), additive error on concentration scale (2.91 ng/ml); ω2Ka, variance of interindividual variability for Ka; ω2V, variance of interindividual variability for V; ω2CL, variance of interindividual variability for CL; ω2Tlag, variance of interindividual variability for Tlag.

Model evaluation.

The stability, precision, and robustness of the final model were assessed using nonparametric bootstrap analysis. Table 2 provides comparison of the final model parameter estimates and bootstrap replicates (n = 1,000) for DEC. The parameter estimates of the final population pharmacokinetics model were close to the median values obtained from bootstrap replications, showing the robustness, accuracy, and stability of the final population PK model. The low percentage of coefficient of variation (CV) for the bootstrap results suggests parameter uncertainty was small. The bootstrap calculated percentage of IIV was similar to the final model estimated for the patient‐to‐patient variability. Visual predictive check (VPC) plots were constructed to evaluate the model predictability. Concentration-time data were graphically superimposed on the median values and the 5th and 95th percentiles of the simulated concentration-time profiles. In addition, The VPC (n = 1,000) results for the final model presented in Fig. 2 showed that the most of observed DEC data fell within the boundaries of 5th and 95th percentiles, suggesting the adequacy of the final model to explain the observed data. Normalized prediction distribution error (NPDE) was also evaluated. The VPC and bootstrap parameter values both showed that the model was a good description of the data (Table 2 and Fig. 2).

FIG 2.

FIG 2

The visual predictive check of the final population pharmacokinetics model over the time (independent variable [IVAR]) from 0 and 72 h following DEC administration (n = 1,000) is shown. The inset shows the first 20 h after dosing.

Using the final model, simulations of plasma-concentration profiles were performed for DEC administered at a dose of 6 mg/kg. Descriptive statistics of AUClast (area under the plasma-concentration curve from dosing to 72 h postdose) and Cmax are provided in Table 3.

TABLE 3.

Simulated AUClast (0 to 72 h) and Cmax values after 6 mg/kg of DEC

Parameter Median Mean SD 95% CI
AUClast (0–72 h) (ng · h/ml) 22,755 23,109 5,012.2 13064.7–33154.3
Cmax (ng/ml) 1,570.2 1,597.4 241.6 1113.1–2081.7

Cmax and AUClast for the 6-mg/kg dose were simulated for a male subject and female subject (age of 40 years) with a weight of 51 kg (minimum-weight person as per the given population), an average-weight person (65 kg), and an overweight person (135 kg) to assess the impact of gender along with weight on drug exposure.

A lower-weight female (51 kg) had 26% lower Cmax and AUClast values than the average-weight female (65 kg [average weight as per the studied population]), while an overweight female (135 kg) had 10% lower Cmax and 50% higher AUClast values than the average-weight female. On the other hand, A lower-weight male (51 kg) had 7% lower Cmax and 8% lower AUClast values than the average-weight male (65 kg), while an overweight male (135 kg) had 10% lower Cmax and 37% higher AUClast values than an average-weight male.

In comparison of the female to male population, a lower-weight female had 5% higher Cmax and 27% lower AUClast values, an average-weight female had 33% higher Cmax and 3% lower AUClast values, and an overweight female had 8% higher Cmax and 6% lower AUClast values.

DISCUSSION

The objective of this study was to describe the population PK parameters of DEC administered orally at 6 mg/kg in clinical patients infected with LF caused by Wuchereria bancrofti and uninfected healthy adults. Blood samples for drug levels were obtained at 1, 2, 3, 4, 6, 8, 12, 24, 36, 48, 72, and 168 h after treatment. This is the first population-based assessment of the pharmacokinetics of DEC in patients and healthy volunteers. Population modeling offers an opportunity to assess drug disposition properties taking into account individual’s pharmacokinetic variability. The pharmacokinetics data of DEC are well described by a one-compartment linear model with first-order absorption. We have considered age, weight, infection status, gender, AST, ALT, and CLCR as covariates to be included in the model development process. We have not included effect of coadministration of IVM and ALB on DEC pharmacokinetics as the findings of the prior study (7, 23) show no interaction between these drugs. We have previously shown that the triple-drug combination is also safe in uninfected individuals and can therefore be used in MDA programs (16). The combined additive-proportional residual error model with Tlag function was found to best describe the data. In this study, the inclusion of age and health condition/infected state in the base model did not appear to improve the data fitting. Among the considered covariates, a correlation between body weight and gender over volume of distribution was observed (OFV = −40). A previous finding described that intersubject variations in the pharmacokinetics parameters of DEC could be due to nonadjustment of dose to body weight (24). However, our DEC dosing was dependent on body weight, but to a certain degree. The unavailability of ideal body weight (due to lack of data) serves as a limitation of the generated model to clearly defining the gender and body weight correlation. In addition, this study only included adult subjects (18 to 66 years), which makes extrapolation of dosing into pediatric and elderly patients difficult. Goss and coworkers developed dosing pole recommendations for DEC to compare the current WHO’s height- or age-based dosing recommendation as alternatives to weight-based dosing for mass drug administration lymphatic filariasis (LF) elimination programs. Their results supported weight-based DEC dosing along with other factors affecting height-weight relationships (e.g., age, sex, and geographic location) (25).

The CWRES distribution of the final model revealed uniform distribution, and the value ranged between 2 and −2, indicating the slight differences in prediction models. As part of a validation process, the bootstrap has been run for a 1,000-data set random sampling of the subjects from the original data set, and the results were in line with our final population PK model. VPC (n = 1,000) also explains the adequacy of our final model. Finally, our study confirms the significant relationship between V/F and body weight and gender for DEC. Further simulation suggests that the average-weight female population had higher Cmax values than average-weight male subjects, but as the gender covariate impacted volume of distribution, the AUC of exposure was not changed significantly. Body weight significantly impacted the exposure (increased body weight increased exposure) in both the male and female populations. This effect was more pronounced in the male population. However, this population PK study was performed in a specific population in West Africa, and there may be limitations applying these findings globally.

Conclusion.

We developed a population pharmacokinetic model of DEC in Wuchereria bancrofti-infected (n = 32) and uninfected (n = 24) populations. In this study, nonlinear mixed-effect modeling (NLME) was performed. The final model showed a significant influence of body weight and gender on volume of distribution of DEC. The stability, robustness, and the predictive performance of the final population PK model were established by using goodness-of-fit plots, bootstrapping, and VPC. The developed model along with described simulations may guide future study designs and be useful for predicting pharmacokinetic/pharmacodynamic analyses.

MATERIALS AND METHODS

Subjects and study design.

This was an open-label cohort study of treatment-naive Wuchereria bancrofti-infected (n = 32) and uninfected (n = 24) adults residing in Agboville District of Côte d’Ivoire. LF is endemic in the Agboville District, and onchocerciasis is nonendemic. Informed consent was obtained from all participants. Individuals (18 to 70 years) included in this study were prescreened for W. bancrofti infection with fingerstick blood using a filariasis test strip (FTS; Alere, Inc., Waltham, MA, USA), to detect circulating filarial antigenemia. Exclusion criteria for the study were a positive pregnancy test, chronic kidney or liver disease, or a serum alanine transaminase, aspartate transaminase, or creatinine level >1.5 times the normal limits or severe anemia (blood hemoglobin, <7 g/dl), taking medication that could interfere with test drug metabolism within 1 week of study onset, evidence of urinary tract infection (as indicated by an active urinary sediment of >68 pus/neutrophil cells per field or 3+ nitrate on dipstick), or lactose and/or gluten intolerance. All individuals were tested for onchocerciasis by microscopic examination of two skin snips taken from the iliac crests and antibodies to a recombinant Onchocerca volvulus antigen (Ov16 rapid test; Standard Diagnostics, Inc., Youngin, South Korea) since DEC can cause serious adverse events in persons with onchocerciasis (26). Participants were given a single coadministered dose of IVM (200 μg/kg), DEC citrate (6 mg/kg), and ALB (400 mg) as directly observed therapy. A peripheral intravenous catheter was placed for the first 12 h due to frequent blood draws. Blood samples for drug concentrations were obtained prior to and at 1, 2, 3, 4, 6, 8, 12, 24, 36, 48, 72, and 168 h after treatment, and plasma was separated by centrifugation and stored at –20°C. Institutional review boards in Cleveland, OH (University Hospitals Cleveland Medical Center IRB no. 03-16-09), and in Côte d’Ivoire approved the study protocol (Comité National d’Ethique et de la Recherche, CNER, N/Ref: 022/MSLS/CNER-kp). This trial is registered at clinicaltrials.gov (identifier NCT02845713).

Bioanalytical assays.

Plasma concentrations of DEC were determined using a validated liquid chromatography-tandem mass spectrometry (LC-MS/MS) method as previously described (22). The assay was linear from 1 to 2,000 ng/ml. Control concentrations of 5, 500, and 1,500 ng/ml were within 15% of nominal values for all data.

Population pharmacokinetics modeling.

The concentration-time data of DEC was used to develop the population pharmacokinetics model by using a nonlinear mixed‐effects approach with Phoenix NLME software (version 8.0; Certara L.P., St. Louis, MO, USA). The first-order conditional estimation (FOCE) method was used throughout the model-building process. One- and two-compartment models with and without lag time were evaluated with different residual errors to identify the model that best described the set of data. Initially, the base structural model without covariates was generated by using the initial estimates (observed Cmax, Vz/Fobs, and CL/Fobs of noncompartmental analysis [NCA]). The best structural model was chosen on the basis of examination of objective function value (OFV; equal to the twice the negative log likelihood [−2LL] value), the Akaike information criterion (AIC), and the visual inspection of diagnostic standard goodness-of-fit plots (16, 17). The structural pharmacokinetics base model comprised of three fundamental fixed‐effect parameters: apparent clearance (CL/F), apparent volume of distribution (V/F), and the absorption rate constant (Ka).

The interindividual variability of the PK parameters was modeled by the log normal distribution of the exponential model as shown in equation 1:

V=tv×exp(η) (1)

Here, V, CL, and Ka are the fixed effects for the pharmacokinetic parameter, tv represents the typical value of the population mean (tvV, tvCL, and tvKa), and η (ηV, ηCL, and ηKa) is a random variable following a Gaussian distribution with a mean of 0 and a variance of ω2. The interindividual variation, η, for between-subject variability and the interindividual variation for random variables with high shrinkage value (>0.5) were not included in the model (27). The additive, proportional, and combined additive plus proportional error models were tested, and the residual error model was best described using a combined additive-proportional error model with Tlag function as shown in equation 2:

Cobs=C+CEps×sqrt(1+(C2×(CmultiSDsigma())2)) (2)

Cobs and C are the observed and predicted concentrations of the individual, respectively, CEps is a normally distributed residual error with a mean of 0 and ω2 (additive part of variance), sqrt, is the square root, CmultiSD is C with multiple standard deviations, and C2 × (CmultiSD/sigma())2) is proportional part variance.

Population covariate analysis.

In the second step of model building, demographic variables were tested as potential covariates for the PK parameters. This study included sex and infection state as categorical covariates and age, weight, CLCR, and AST and ALT levels as continuous covariates. CLCR was calculated from serum creatinine data using the following equation:

CLCR=(140age [yr] × wt [kg])/(72 × serum CR [mg/dl]) (3)

(Multiply the result by 0.85 for the female.)

A visual covariate screening procedure was performed before modeling, which includes scatter plots for continuous covariates and box plots for discrete variables. After visual inspection, all the covariates were added, and the model with covariates was run using the stepwise forward addition procedure (P ≤ 0.01) followed by a backward elimination procedure (P ≤ 0.001) with the likelihood ratio test. All of the selected covariates were evaluated according to the difference in the OFV (−2LL), AIC, and further change in interindividual variability (percentage of IIV) among individual PK parameters and covariate plots. A covariate was considered significant when the addition of the covariate resulted in a decrease in the OFV greater than 6.63 (P < 0.01), and the elimination of the covariate resulted in an increase in the OFV greater than 10.82 (P < 0.001) (28). The percentage of IIV was calculated by using the following equation:

% IIV=e(ω2)1×100 (4)

After a full covariate regressive model was established, the analysis procedure was continued until only significant covariates remained in the model. At the end of the model‐building process, all significant covariates were then incorporated into the base model, which was designated the “final” population model.

Model validation and model-based simulations.

The suitability of the final full population model was evaluated using the nonparametric bootstrap analysis and visual predictive check (VPC) to evaluate the precision and robustness of the final model (29). A bootstrap (n = 1,000) was performed by random sampling of the subjects from the original data set. The final parameter estimates obtained from the original data set were compared with the median values and 95% confidence intervals (CIs) of the bootstrap estimates. The final population model of DEC was evaluated on the basis of a visual predictive check using a 1,000-data set simulation to determine whether the fitted model provides an adequate description of the data. The lower (5%), median (50%), and upper (95%) percentiles of the simulated concentrations were calculated and then visually checked to evaluate how the simulated results are related to the observed DEC concentrations.

Using the parameter estimates from the final model, plasma drug concentration profiles were simulated for the given doses 6 mg/kg, using the same population included in the population pharmacokinetics modeling regarding covariate distributions. One thousand plasma drug concentration profiles per dose were simulated. From the plasma drugconcentration profiles and estimated CL/F values, the pharmacokinetics parameters AUClast (area under the plasma-concentration curve from dosing to 72 h postdose) was estimated as dose/(CL/F) and Cmax (maximal plasma drug concentration) was read directly from the plasma drug concentration profiles. The arithmetic mean, median, standard deviation (SD), and the 2.5% and 97.5% percentiles for AUClast and Cmax were estimated.

ACKNOWLEDGMENTS

The project was funded by Bill & Melinda Gates Foundation grant OPPGH 5342. The sponsor of the study had no role in study design, data collection, data analysis, data interpretation, or writing of the report.

The corresponding author has had full access to all the data in the study and takes final responsibility for the decision to submit the paper for publication.

We declare that there are no conflicts of interest.

Footnotes

Supplemental material is available online only.

Supplemental file 1
Supplemental Figures S1 to S3 and Table S1. Download AAC.00317-21-s0001.pdf, PDF file, 1.0 MB (1,011.6KB, pdf)

REFERENCES

  • 1.Sheel M, Sheridan S, Gass K, Won K, Fuimaono S, Kirk M, Gonzales A, Hedtke SM, Graves PM, Lau CL. 2018. Identifying residual transmission of lymphatic filariasis after mass drug administration: comparing school-based versus community-based surveillance—American Samoa, 2016. PLoS Negl Trop Dis 12:e0006583. doi: 10.1371/journal.pntd.0006583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.WHO. 2017. Summary of global update on preventive chemotherapy implementation in 2016: crossing the billion. Wkly Epidemiol Rec 92:589–593. [PubMed] [Google Scholar]
  • 3.Gyapong JO, Owusu IO, da-Costa Vroom FB, Mensah EO, Gyapong M. 2018. Elimination of lymphatic filariasis: current perspectives on mass drug administration. Res Rep Trop Med 9:25–33. doi: 10.2147/RRTM.S125204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.WHO. 2016. Global programme to eliminate lymphatic filariasis: progress report, 2015. Wkly Epidemiol Rec 91:441–455. [PubMed] [Google Scholar]
  • 5.Rizzo JÂ, Belo C, Lins R, Dreyer G. 2007. Children and adolescents infected with Wuchereria bancrofti in Greater Recife, Brazil: a randomized, year-long clinical trial of single treatments with diethylcarbamazine or diethylcarbamazine-albendazole. Ann Trop Med Parasitol 101:423–433. doi: 10.1179/136485907X176517. [DOI] [PubMed] [Google Scholar]
  • 6.King CL, Suamani J, Sanuku N, Cheng Y-C, Satofan S, Mancuso B, Goss CW, Robinson LJ, Siba PM, Weil GJ, Kazura JW. 2018. A trial of a triple-drug treatment for lymphatic filariasis. N Engl J Med 379:1801–1810. doi: 10.1056/NEJMoa1706854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Thomsen EK, Sanuku N, Baea M, Satofan S, Maki E, Lombore B, Schmidt MS, Siba PM, Weil GJ, Kazura JW, Fleckenstein LL, King CL. 2016. Efficacy, safety, and pharmacokinetics of coadministered diethylcarbamazine, albendazole, and ivermectin for treatment of Bancroftian filariasis. Clin Infect Dis 62:334–341. doi: 10.1093/cid/civ882. [DOI] [PubMed] [Google Scholar]
  • 8.King CL, Weil GJ, Kazura JW. 2020. Single-dose triple-drug therapy for Wuchereria bancrofti—5-year follow-up. N Engl J Med 382:1956–1957. doi: 10.1056/NEJMc1914262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.WHO. 2017. Guideline: alternative mass drug administration regimens to eliminate lymphatic filariasis. Contract no. WHO/HTM/NTD/PCT/2017.0. WHO, Geneva, Switzerland. [PubMed] [Google Scholar]
  • 10.Irvine MA, Stolk WA, Smith ME, Subramanian S, Singh BK, Weil GJ, Michael E, Hollingsworth TD. 2017. Effectiveness of a triple-drug regimen for global elimination of lymphatic filariasis: a modelling study. Lancet Infect Dis 17:451–458. doi: 10.1016/S1473-3099(16)30467-4. [DOI] [PubMed] [Google Scholar]
  • 11.Ottesen EA. 1985. Efficacy of diethylcarbamazine in eradicating infection with lymphatic-dwelling filariae in humans. Rev Infect Dis 7:341–356. doi: 10.1093/clinids/7.3.341. [DOI] [PubMed] [Google Scholar]
  • 12.Ismail MM, Jayakody RL, Weil GJ, Nirmalan N, Jayasinghe KS, Abeyewickrema W, Rezvi Sheriff MH, Rajaratnam HN, Amarasekera N, de Silva DC, Michalski ML, Dissanaike AS. 1998. Efficacy of single dose combinations of albendazole, ivermectin and diethylcarbamazine for the treatment of Bancroftian filariasis. Trans R Soc Trop Med Hyg 92:94–97. doi: 10.1016/S0035-9203(98)90972-5. [DOI] [PubMed] [Google Scholar]
  • 13.Ismail MM, Weil GJ, Jayasinghe KS, Premaratne UN, Abeyewickreme W, Rajaratnam HN, Sheriff MH, Perera CS, Dissanaike AS. 1996. Prolonged clearance of microfilaraemia in patients with bancroftian filariasis after multiple high doses of ivermectin or diethylcarbamazine. Trans R Soc Trop Med Hyg 90:684–688. doi: 10.1016/s0035-9203(96)90437-x. [DOI] [PubMed] [Google Scholar]
  • 14.Noroes J, Dreyer G, Santos A, Mendes VG, Medeiros Z, Addiss D. 1997. Assessment of the efficacy of diethylcarbamazine on adult Wuchereria bancrofti in vivo. Trans R Soc Trop Med Hyg 91:78–81. doi: 10.1016/s0035-9203(97)90405-3. [DOI] [PubMed] [Google Scholar]
  • 15.Peixoto CA, Silva BS. 2014. Anti-inflammatory effects of diethylcarbamazine: a review. Eur J Pharmacol 734:35–41. doi: 10.1016/j.ejphar.2014.03.046. [DOI] [PubMed] [Google Scholar]
  • 16.Edi C, Bjerum CM, Ouattara AF, Chhonker YS, Penali LK, Méité A, Koudou BG, Weil GJ, King CL, Murry DJ. 2019. Pharmacokinetics, safety, and efficacy of a single co-administered dose of diethylcarbamazine, albendazole and ivermectin in adults with and without Wuchereria bancrofti infection in Cote d'Ivoire. PLoS Negl Trop Dis 13:e0007325. doi: 10.1371/journal.pntd.0007325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Illamola SM, Sherwin CM, van Hasselt JGC. 2018. Clinical pharmacokinetics of amikacin in pediatric patients: a comprehensive review of population pharmacokinetic analyses. Clin Pharmacokinet 57:1217–1228. doi: 10.1007/s40262-018-0641-x. [DOI] [PubMed] [Google Scholar]
  • 18.Krekels EHJ, van Hasselt JGC, van den Anker JN, Allegaert K, Tibboel D, Knibbe CAJ. 2017. Evidence-based drug treatment for special patient populations through model-based approaches. Eur J Pharm Sci 109S:S22–S26. doi: 10.1016/j.ejps.2017.05.022. [DOI] [PubMed] [Google Scholar]
  • 19.Kodati D, Kotakonda HK, Yellu N. 2017. Population pharmacokinetic modeling of olmesartan, the active metabolite of olmesartan medoxomil, in patients with hypertension. Eur J Drug Metab Pharmacokinet 42:573–581. doi: 10.1007/s13318-016-0371-0. [DOI] [PubMed] [Google Scholar]
  • 20.Ali AM, Penny MA, Smith TA, Workman L, Sasi P, Adjei GO, Aweeka F, Kiechel J-R, Jullien V, Rijken MJ, McGready R, Mwesigwa J, Kristensen K, Stepniewska K, Tarning J, Barnes KI, Denti P, WWARN Amodiaquine PK Study Group. 2018. Population pharmacokinetics of the antimalarial amodiaquine: a pooled analysis to optimize dosing. Antimicrob Agents Chemother 62:e02193-17. doi: 10.1128/AAC.02193-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.WHO. 2006. Preventive chemotherapy in human helminthiasis: coordinated use of anthelminthic drugs in control interventions: a manual for health professionals and programme managers. https://apps.who.int/iris/bitstream/handle/10665/43545/9241547103_eng.pdf?sequence=1.
  • 22.Chhonker YS, Edi C, Murry DJ. 2018. LC-MS/MS method for simultaneous determination of diethylcarbamazine, albendazole and albendazole metabolites in human plasma: application to a clinical pharmacokinetic study. J Pharm Biomed Anal 151:84–90. doi: 10.1016/j.jpba.2017.12.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Shenoy RK, Suma TK, John A, Arun SR, Kumaraswami V, Fleckenstein LL, Na-Bangchang K. 2002. The pharmacokinetics, safety and tolerability of the co-administration of diethylcarbamazine and albendazole. Ann Trop Med Parasitol 96:603–614. doi: 10.1179/000349802125001663. [DOI] [PubMed] [Google Scholar]
  • 24.Bolla S, Boinpally RR, Poondru S, Devaraj R, Jasti BR. 2002. Pharmacokinetics of diethylcarbamazine after single oral dose at two different times of day in human subjects. J Clin Pharmacol 42:327–331. doi: 10.1177/00912700222011247. [DOI] [PubMed] [Google Scholar]
  • 25.Goss CW, O'Brian K, Dubray C, Fischer PU, Hardy M, Jambulingam P, King CL, Laman M, Lemoine JF, Robinson LJ, Samuela J, Subramanian S, Supali T, Weil GJ, Schechtman KB. 2019. Dosing pole recommendations for lymphatic filariasis elimination: a height-weight quantile regression modeling approach. PLoS Negl Trop Dis 13:e0007541. doi: 10.1371/journal.pntd.0007541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Fischer PU, King CL, Jacobson JA, Weil GJ. 2017. Potential value of triple drug therapy with ivermectin, diethylcarbamazine, and albendazole (IDA) to accelerate elimination of lymphatic filariasis and onchocerciasis in Africa. PLoS Negl Trop Dis 11:e0005163. doi: 10.1371/journal.pntd.0005163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Liu S, Chen P, Zhao Y, Dai G, Sun B, Wang Y, Ding A, Ju W. 2017. Pharmacokinetic and pharmacodynamic modeling of oral mitiglinide on glucose lowering in healthy Chinese volunteers. BMC Pharmacol Toxicol 18:54. doi: 10.1186/s40360-017-0161-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Song L, He CY, Yin NG, Liu F, Jia YT, Liu Y. 2017. A population pharmacokinetic model for individualised dosage regimens of vancomycin in Chinese neonates and young infants. Oncotarget 8:105211–105221. doi: 10.18632/oncotarget.22114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kodati D, Yellu N. 2017. Population pharmacokinetic modeling of furosemide in patients with hypertension and fluid overload conditions. Pharmacol Rep 69:492–496. doi: 10.1016/j.pharep.2017.01.006. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental file 1

Supplemental Figures S1 to S3 and Table S1. Download AAC.00317-21-s0001.pdf, PDF file, 1.0 MB (1,011.6KB, pdf)


Articles from Antimicrobial Agents and Chemotherapy are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES