scholarly journals Population Pharmacokinetics of Oxycodone and Metabolites in Patients with Cancer-Related Pain

Cancers ◽  
2021 ◽  
Vol 13 (11) ◽  
pp. 2768
Author(s):  
Bram C. Agema ◽  
Astrid W. Oosten ◽  
Sebastiaan D. T. Sassen ◽  
Wim J. R. Rietdijk ◽  
Carin C. D. van der Rijt ◽  
...  

Oxycodone is frequently used for treating cancer-related pain, while not much is known about the factors that influence treatment outcomes in these patients. We aim to unravel these factors by developing a population-pharmacokinetic model to assess the pharmacokinetics of oxycodone and its metabolites in cancer patients, and to associate this with pain scores, and adverse events. Hospitalized patients with cancer-related pain, who were treated with oral oxycodone, could participate. Pharmacokinetic samples and patient-reported pain scores and occurrence and severity of nine adverse events were taken every 12 h. In 28 patients, 302 pharmacokinetic samples were collected. A one-compartment model for oxycodone and each metabolite best described oxycodone, nor-oxycodone, and nor-oxymorphone pharmacokinetics. Furthermore, oxycodone exposure was not associated with average and maximal pain scores, and oxycodone, nor-oxycodone, and nor-oxymorphone exposure were not associated with adverse events (all p > 0.05). This is the first model to describe the pharmacokinetics of oxycodone including the metabolites nor-oxycodone and nor-oxymorphone in hospitalized patients with cancer pain. Additional research, including more patients and a more timely collection of pharmacodynamic data, is needed to further elucidate oxycodone (metabolite) pharmacokinetic/pharmacodynamic relationships. This model is an important starting point for further studies to optimize oxycodone dosing regiments in patients with cancer-related pain.

Pharmaceutics ◽  
2020 ◽  
Vol 12 (1) ◽  
pp. 54 ◽  
Author(s):  
Amaia Soraluce ◽  
Helena Barrasa ◽  
Eduardo Asín-Prieto ◽  
Jose Ángel Sánchez-Izquierdo ◽  
Javier Maynar ◽  
...  

Antimicrobial treatment in critically ill patients remains challenging. The aim of this study was to develop a population pharmacokinetic model for linezolid in critically ill patients and to evaluate the adequacy of current dosing recommendation (600 mg/12 h). Forty inpatients were included, 23 of whom were subjected to continuous renal replacement therapies (CRRT). Blood and effluent samples were drawn after linezolid administration at defined time points, and linezolid levels were measured. A population pharmacokinetic model was developed, using NONMEM 7.3. The percentage of patients that achieved the pharmacokinetic/pharmacodynamic (PK/PD) targets was calculated (AUC24/MIC > 80 and 100% T>MIC). A two-compartment model best described the pharmacokinetics of linezolid. Elimination was conditioned by the creatinine clearance and by the extra-corporeal clearance if the patient was subjected to CRRT. For most patients, the standard dose of linezolid did not cover infections caused by pathogens with MIC ≥ 2 mg/L. Continuous infusion may be an alternative, especially when renal function is preserved.


2006 ◽  
Vol 50 (7) ◽  
pp. 2281-2285 ◽  
Author(s):  
Elizabeth A. Ashley ◽  
Kasia Stepniewska ◽  
Niklas Lindegårdh ◽  
Rose McGready ◽  
Robert Hutagalung ◽  
...  

ABSTRACT A fixed artesunate-mefloquine combination, comprising three daily doses of 8 mg of mefloquine/kg of body weight and 4 mg of artesunate/kg, has been developed recently. This study was designed to construct a population pharmacokinetic model describing this new dosage regimen of mefloquine given as loose tablets together with artesunate. In two randomized trials in Thailand which evaluated the efficacy, safety, and tolerability of this new regimen, the members of a subgroup of 50 patients were randomized to have capillary blood sampling before treatment and at five randomly assigned time points during the 63-day follow-up period. Mefloquine levels in capillary whole blood were assayed by liquid chromatography with UV detection. A pharmacokinetic model for mefloquine was constructed using mixed-effects modeling. A one-compartment model with first-order absorption and elimination was selected to describe the kinetic properties of mefloquine. For capillary whole-blood mefloquine, the area under the concentration curve (AUC) was 40% higher than previous estimates for patients given the equivalent conventional-dose regimen (mefloquine given as 15 mg/kg and then 10 mg/kg on the second and third days of treatment). The half-life (t 1/2) of the carboxylic acid metabolite was estimated as 26 days, and the metabolite was eliminated more slowly than the parent drug (population t 1/2 estimate, 10.5 days). Splitting the 25 mg/kg dose of mefloquine into three doses of 8 mg/kg each resulted in improved oral bioavailability compared to the conventional split-dose regimen results. This new regimen is well tolerated and results in an equivalent therapeutic response.


2022 ◽  
Vol 12 ◽  
Author(s):  
SiChan Li ◽  
SanLan Wu ◽  
WeiJing Gong ◽  
Peng Cao ◽  
Xin Chen ◽  
...  

Purpose: The aims of this study were to establish a joint population pharmacokinetic model for voriconazole and its N-oxide metabolite in immunocompromised patients, to determine the extent to which the CYP2C19 genetic polymorphisms influenced the pharmacokinetic parameters, and to evaluate and optimize the dosing regimens using a simulating approach.Methods: A population pharmacokinetic analysis was conducted using the Phoenix NLME software based on 427 plasma concentrations from 78 patients receiving multiple oral doses of voriconazole (200 mg twice daily). The final model was assessed by goodness of fit plots, non-parametric bootstrap method, and visual predictive check. Monte Carlo simulations were carried out to evaluate and optimize the dosing regimens.Results: A one-compartment model with first-order absorption and mixed linear and concentration-dependent-nonlinear elimination fitted well to concentration-time profile of voriconazole, while one-compartment model with first-order elimination well described the disposition of voriconazole N-oxide. Covariate analysis indicated that voriconazole pharmacokinetics was substantially influenced by the CYP2C19 genetic variations. Simulations showed that the recommended maintenance dose regimen would lead to subtherapeutic levels in patients with different CYP2C19 genotypes, and elevated daily doses of voriconazole might be required to attain the therapeutic range.Conclusions: The joint population pharmacokinetic model successfully characterized the pharmacokinetics of voriconazole and its N-oxide metabolite in immunocompromised patients. The proposed maintenance dose regimens could provide a rationale for dosage individualization to improve clinical outcomes and minimize drug-related toxicities.


2018 ◽  
Vol 62 (4) ◽  
Author(s):  
Max Taubert ◽  
Mark Lückermann ◽  
Andreas Vente ◽  
Axel Dalhoff ◽  
Uwe Fuhr

ABSTRACTFinafloxacin is a novel fluoroquinolone with increased antibacterial activity at acidic pH and reduced susceptibility to several resistance mechanisms. A phase II study revealed a good efficacy/safety profile in patients with complicated urinary tract infections (cUTIs), while the pharmacokinetics was characterized by highly variable concentration-versus-time profiles, suggesting the need for an elaborated pharmacokinetic model. Data from three clinical trials were evaluated: 127 healthy volunteers were dosed orally (n= 77) or intravenously (n= 50), and 139 patients with cUTI received finafloxacin intravenously. Plasma (2,824 samples from volunteers and 414 samples from patients) and urine (496 samples from volunteers and 135 samples patients) concentrations were quantified by liquid chromatography-tandem mass spectrometry (LC-MS/MS). NONMEM was used to build a population pharmacokinetic model, and pharmacokinetic/pharmacodynamic relationships were investigated via simulations and logistic regression. A two-compartment model with first-order elimination described the data best (central volume of distribution [Vc] and peripheral volume of distribution [Vp] of 47 liters [20%] and 43 liters [67%], respectively, and elimination clearance and intercompartmental clearance of 21 liters/h [54%] and 2.8 liters/h [57%], respectively [median bootstrap estimates {coefficients of variation}]).Vcincreased with body surface area, and clearance was reduced in patients (−29%). Oral absorption was described best by parallel first- and zero-order processes (bioavailability of 75%). No pharmacodynamic surrogate parameter of clinical/microbiological outcome could be identified, which depended exclusively on the MIC of the causative pathogens. Despite the interindividual variability, the present data set does not support covariate-based dose adjustments. Based on the favorable safety and efficacy data, the clinical relevance of the observed variability appears to be limited. (This study has been registered at ClinicalTrials.gov under identifier NCT01928433.)


2010 ◽  
Vol 104 (08) ◽  
pp. 252-260 ◽  
Author(s):  
Paul Zufferey ◽  
Denis Baylot ◽  
Philippe Nguyen ◽  
Jeanne-Yvonne Borg ◽  
Michaela Fontenay ◽  
...  

SummaryFondaparinux is a synthetic antithrombotic agent with specific anti-factor Xa activity. A population pharmacokinetic model of fondaparinux, based on data obtained in patients included in phase II/III trials, has been described. However, the validity of this model in everyday practice needed to be confirmed. This study was a multicenter, prospective cohort study in consecutive orthopaedic patients treated with 2.5 mg of fondaparinux. Anti-Xa activities were recorded in 809 patients. Population parameters and inter-individual variability were estimated using NONMEM VI software. A two-compartment model with first-order absorption best described fondaparinux pharmacokinetics. Covariates partly explaining inter-individual variability were body weight, age and creatinine clearance estimated by the simplified Modification of Diet in Renal Disease formula (MDRD). A body weight less than 50 kg and moderate renal failure increased drug exposure. Although the population pharmacokinetic model of fondaparinux was described, this one requires to be validated in everyday practice.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Yoonhyuk Jang ◽  
Seonghae Yoon ◽  
Tae-Joon Kim ◽  
SeungHwan Lee ◽  
Kyung-Sang Yu ◽  
...  

AbstractThis study aimed to develop a pharmacokinetic (PK) model of oxcarbazepine (OXC) and analyse the relationship between monohydroxylated derivative (MHD), an active metabolite of OXC, and the adverse events of OXC. We obtained 711 OXC samples from 618 patients with epilepsy who were enrolled in the Epilepsy Registry Cohort of Seoul National University Hospital from February 2011 to January 2014. The plasma PK model was developed using a nonlinear mixed-effect modelling method with NONMEM (ver 7.3). A one-compartment model with a first-order absorption model and proportional residual error adequately described the MHD concentration–time profiles. The only covariate incorporated for CL/F and V/F was body weight. Of the 447 patients analysed, 28 (6.26%) had dose-related adverse events (DRAEs), which were dizziness, somnolence, headache, and diplopia. For DRAE occurrence, the cut-off values of the MHD trough and AUC were 12.27 mg/L (specificity 0.570, sensitivity 0.643) and 698.5 mg h/L (specificity, sensitivity 0.571), respectively. Multivariate analysis showed the sole dizziness symptom was significantly associated with both the MHD trough and the AUC (p = 0.013, p = 0.038, respectively). We newly developed a population PK model using sparse sampling data from patients with epilepsy, and the model better reflects the actual clinical situation.


Author(s):  
Thanh Bach ◽  
Daryl J. Murry ◽  
Larissa V. Stebounova ◽  
Gregory Deye ◽  
Patricia Winokur ◽  
...  

Oxfendazole is a potent veterinary benzimidazole anthelmintic under transition to human for the treatment of multiple parasitic infectious diseases. The first-in-human study evaluating the disposition of oxfendazole and its metabolites in healthy adults following single ascending oral doses from 0.5 to 60 mg/kg shows that oxfendazole pharmacokinetics is substantially nonlinear, which complicates correlating oxfendazole dose to exposure. To quantitatively capture the relation between oxfendazole dose and exposure, a population pharmacokinetic model for oxfendazole and its metabolites, oxfendazole sulfone and fenbendazole, in humans was developed using nonlinear mixed-effect modeling approach. Our final model incorporated mechanistic characterization of dose limited bioavailability as well as different oxfendazole metabolic processes and provided insight to the significance of pre-systemic metabolism in oxfendazole and metabolites disposition. Oxfendazole clinical pharmacokinetics was best described by a one-compartment model with nonlinear absorption and linear elimination. Oxfendazole apparent clearance and apparent volume of distribution were estimated to be 2.57 L/h and 35.2 L, respectively, at the lowest dose (0.5 mg/kg), indicating that oxfendazole is a low extraction drug with moderate distribution. The disposition of both metabolites was adequately characterized by one-compartment model with formation-rate limited elimination. Fenbendazole formation from oxfendazole was primarily through systemic metabolism while both pre-systemic and systemic metabolism were critical to the formation of oxfendazole sulfone. Our model adequately captured the concentration-time profiles of both oxfendazole and its two metabolites in healthy adults over a wide dose range. The model can be used to predict oxfendazole disposition under new dosing regimens to support dose optimization in humans.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5084-5084
Author(s):  
Li Zhang ◽  
Xin-mei Yang ◽  
Jing Chen ◽  
Lei Hu ◽  
Fan Yang ◽  
...  

# The first two authors contributed equally * The last two authors contributed equally Background: Realgar-Indigo Naturalis Formula (RIF), the only commercially oral arsenic agent, was launched in China from 2009. It has been recommended as consolidation or maintenance therapy in adults with acute promyelocytic leukemia (APL). Recently, RIF had been off-label used in pediatric APL in China. However, the knowledge of efficacy and dosage of RIF in pediatric was limited until now. Objective: The purpose of this pilot study was to evaluate effectiveness and pharmacokinetics of RIF as consolidation or maintenance therapy for newly diagnosed and relapsed APL in pediatric after induction therapy with intravenous arsenic trioxide. Meanwhile, modeling and simulation techniques were used to evaluate and optimize RIF dosing regimen in pediatric population. Patients and Methods: A total of 11 newly diagnosed APL and 1 relapsed patient (4-14years of age) were included from July 2016 to August 2017.The one relapsed and six newly diagnosed patients were treated with RIF (60 mg/kg/day TID) as maintenance therapy. Five newly diagnosed patients were treated with RIF (60 mg/kg/day TID) as consolidation and maintenance therapy. The treatment protocol and MRD results are shown in Figure 1. Event-free survival and overall survival were used to evaluate the efficacy. The plasma concentration of arsenic was quantified by inductively coupled plasma mass spectrometry (ICP-MS). Population pharmacokinetic analysis was carried out using the nonlinear mixed effects modeling program NONMEM V 7.2 (Icon Development Solutions, USA). Results: All newly diagnosed APL patients were alive as of July 1, 2019 and were in MCR with a median follow-up of 28 months (range, 23 to 37 months). Both the estimated 3-year EFS and OS rates were 100%. Of note, all the patients completed the postremission therapy containing RIF on an outpatient basis. A total of 107 arsenic concentrations were used for population pharmacokinetic model building. The arsenic concentrations of the blood samples ranged from 0.1 to 75.0 µg/L. The steady-state trough plasma arsenic concentration during the RIF treatment was 47.43μg/L (range, 25.74 to 62.97μg/L).A one-compartment model with first-order elimination fitted the data. Body weight was the only significant covariate of CL for the final model development. The developmental pharmacokinetic model was evaluated by goodness-of-fit plots and bootstrap analysis. The mean and variance of NPDE were -0.118 and 0.93, respectively. Conclusions: The effectiveness and pharmacokinetics were evaluated for the first time in pediatricAPL.The developmental population pharmacokinetic model for RIF in children revealed that one-compartment model with first-order elimination fitted the data.The current dosing regimen of 60 mg/kg/day TID resulted higher steady-state through concentration than that in adults (24.4μg/L, range:11.5 to 64μg/L). Disclosures:No relevant conflicts of interest to declare. This trial was conducted in accordance with the Declaration of Helsinki. OffLabel Disclosure: The combination of all-trans-retinoic acid (ATRA) and arsenic trioxide (ATO) has become the front-line treatment for patients with acute promyelocytic leukemia (APL). Realgar-Indigo Naturalis Formula (RIF), the only commercially oral arsenic agent, was launched in China from 2009. Since then, RIF had been used for over 5000 adults with acute promyelocytic leukemia (APL). One pill of RIF is 270 mg which contains 30 mg of Realgar, 125 mg of Indigo naturalis, 50 mg of Radix salviae miltiorrhizae, 45 mg of Radix pseudostellariae, and 20mg of garment film. However, the pharmacokinetics of RIF in children has not been studied.


2021 ◽  
Author(s):  
Yoonhyuk Jang ◽  
Seonghae Yoon ◽  
Tae-Joon Kim ◽  
SeungHwan Lee ◽  
Kyung-Sang Yu ◽  
...  

Abstract This study aimed to develop a pharmacokinetic (PK) model of oxcarbazepine (OXC) and analyse the relationship between monohydroxylated derivative (MHD), an active metabolite of OXC, and the adverse events of OXC. We obtained 711 OXC samples from 618 patients with epilepsy who were enrolled in the Epilepsy Registry Cohort of Seoul National University Hospital from February 2011 to January 2014. The plasma PK model was developed using a nonlinear mixed-effect modelling method with NONMEM (ver 7.3). A one-compartment model with a first-order absorption model and proportional residual error adequately described the MHD concentration–time profiles. The only covariate incorporated for CL/F and V/F was body weight. Of the 447 patients analysed, 28 (6.26%) had dose-related adverse events (DRAEs), which were dizziness, somnolence, headache, and diplopia. For DRAE occurrence, the cut-off values of the MHD trough and AUC were 12.27 mg/L (specificity 0.570, sensitivity 0.643) and 698.5 mg⋅h/L (specificity, sensitivity 0.571), respectively. Multivariate analysis showed the sole dizziness symptom was significantly associated with both the MHD trough and the AUC (p = 0.013, p = 0.038, respectively). We newly developed a population PK model using sparse sampling data from patients with epilepsy, and the model better reflects the actual clinical situation.


2020 ◽  
Vol 64 (10) ◽  
Author(s):  
Yi Zheng ◽  
Déborah Hirt ◽  
Sandrine Delmas ◽  
Gabrielle Lui ◽  
Sihem Benaboud ◽  
...  

ABSTRACT A population pharmacokinetic model was developed to explore the pharmacokinetics modification of unbound raltegravir during pregnancy. The RalFe ANRS160 study was a nonrandomized, open-label, multicenter trial enrolling HIV-infected pregnant women receiving a combined antiretroviral regimen containing 400 mg raltegravir twice daily. Biological samples were collected during the third trimester of pregnancy (between 30 and 37 weeks of gestational age) and at postpartum (4 to 6 weeks after delivery). A population pharmacokinetic model was developed with Monolix software. A total of 360 plasma samples were collected from 43 women during pregnancy and postpartum. The unbound raltegravir was described by a one-compartment model with a transit compartment with first-order absorption, evolving to bound raltegravir (by a linear binding to albumin) or metabolism to RAL-glucuronide or to a first-order elimination, with a circadian rhythm. During pregnancy, the absorption was decreased and delayed and the raltegravir elimination clearance and glucuronidation increased by 37%. Median total and unbound area under the curve from 0 to 12 h significantly decreased by 36% and 27% during pregnancy. Median total trough concentration (Ctrough) decreased significantly in the evening (28%); however, the median total Ctrough in the morning, unbound Ctrough in the morning, and unbound Ctrough in the evening showed a nonsignificant decrease of 16%, 1%, and 15%, respectively, during pregnancy compared to the postpartum period. This is the first study reporting the pharmacokinetics of unbound raltegravir during pregnancy. As unbound Ctrough did not significantly decrease during the third trimester, the pregnancy effect on raltegravir unbound concentrations was not considered clinically relevant. (This study has been registered at ClinicalTrials.gov under identifier NCT02099474.)


Sign in / Sign up

Export Citation Format

Share Document