Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
J Clin Pharmacol ; 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38652112

ABSTRACT

BI 187004, a selective small-molecule inhibitor of 11ß-hydroxysteroid dehydrogenase-1 (11ß-HSD1), displayed complex nonlinear pharmacokinetics (PK) in humans. Following nine single oral doses, BI 187004 exhibited nonlinear PK at low doses and linear PK at higher doses. Notably, substantial hepatic 11ß-HSD1 inhibition (50%) was detected in a very low-dose group, achieving a consistent 70% hepatic enzyme inhibition in subsequent ascending doses without any dose-dependent effects. The unusual PK and PD profiles of BI 187004 suggest the presence of pharmacological target-mediated drug disposition (TMDD), arising from the saturable binding of BI 187004 compound to its high-affinity and low-capacity target 11ß-HSD1. The non-intuitive dose, exposure, and response relationship for BI 187004 pose a significant challenge in rational dose selection. This study aimed to construct a TMDD model to explain the complex nonlinear PK behavior and underscore the importance of recognizing TMDD in this small-molecule compound. Among the various models explored, the best model was a two-compartment TMDD model with three transit absorption components. The final model provides insights into 11ß-HSD1 binding-related parameters for BI 187004, including the total amount of 11ß-HSD1 in the liver (estimated to be 8000 nmol), the second order association rate constant (estimated to be 0.102 nM-1h-1), and the first-order dissociation rate constant (estimated to be 0.11 h-1). Our final population PK model successfully characterized the intricate nonlinear PK of BI 187004 across a wide dose range. This modeling work serves as a valuable reference for the rational selection of the dose regimens for BI 187004's future clinical trials.

2.
J Clin Pharmacol ; 64(1): 45-57, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37589246

ABSTRACT

Small interfering RNAs (siRNAs) represent a new class of drugs with tremendous potential for battling previously "undruggable" diseases. After nearly 2 decades of efforts in addressing the problems of the poor drug profile of naked unmodified siRNAs, this new modality has finally come to fruition, with 5 agents (patisiran, givosiran, lumasiran, inclisiran, and vutrisiran) being approved since 2018, and with many others in the different phases of clinical development. Unlike small-molecule drugs and protein therapeutics, siRNAs have different sizes, distinct mechanisms of action, differing physicochemical and pharmacological properties, and, accordingly, a unique pharmacokinetic/pharmacodynamic (PK/PD) relationship. To support the continuous development of siRNAs, it is important to have a thorough and deep understanding of the PK/PD and clinical pharmacology related features of siRNAs. As most of the current siRNA products are conjugated by N-acetylgalactosamine (GalNAc), this review focuses on the PK/PD relationships and clinical pharmacology of GalNAc-conjugated siRNAs, including their absorption, distribution, metabolism, excretion (ADME) properties, PK/PD models, drug-drug interactions, clinical pharmacology in special populations, and safety evaluation. In addition, necessary background information related to the development of siRNAs as a therapeutic modality, including the mechanisms of action, the advantages of siRNAs, the problems of naked siRNAs, as well as the strategies used to enhance the clinical utility of siRNAs, have also been covered. The goal of this review is to serve as a "primer" on siRNA PK/PD, and I hope the readers, especially those who have a limited background on siRNA therapeutics, will have a fundamental understanding of siRNA PK/PD and clinical pharmacology after reading this review.


Subject(s)
Pharmacology, Clinical , Humans , RNA, Small Interfering , Drug Interactions , Pharmacokinetics
3.
J Pharm Sci ; 113(1): 278-289, 2024 01.
Article in English | MEDLINE | ID: mdl-37716531

ABSTRACT

In the current study, we established a comprehensive quantitative systems pharmacology (QSP) model using linagliptin as the model drug, where drug disposition, drug intervention on dipeptidyl peptidase-4 (DPP-4), glucose-dependent insulinotropic peptide (GIP), Glucagon-like peptide-1 (GLP-1), glucagon, glucose, and insulin are integrated together with the cross talk and feedback loops incorporated among the whole glycemic control system. In the final linagliptin QSP model, the complicated disposition of linagliptin was characterized by a 2-compartment pharmacokinetic (PK) model with an enterohepatic cycling (EHC) component as well as target-mediated drug disposition (TMDD) processes occurring in both tissues and plasma, and the inhibitory effect of linagliptin on DPP-4 was determined by the linagliptin-DPP-4 complex in the central compartment based on target occupancy principle. The integrated GIP-GLP1-glucagon-glucose-insulin system contains five indirect response models as the "skeleton" structure with 12 feedback loops incorporated within the glucose control system. Our model adequately characterized the substantial nonlinear PK of linagliptin, time course of DPP-4 inhibition, as well as the kinetics of GIP, GLP-1, glucagon, and glucose simultaneously in humans. Our model provided valuable insights on linagliptin pharmacokinetics/pharmacodynamics and complicated glucose homeostasis. Since the glucose regulation modeling framework within the QSP model is "drug-independent", our model can be easily adopted by others to evaluate the effect of other DPP-4 inhibitors on the glucose control system. In addition, our QSP model, which contains more components than other reported glucose regulation models, can potentially be used to evaluate the effect of combination antidiabetic therapy targeting different components of glucose control system.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors , Humans , Blood Glucose , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Gastric Inhibitory Polypeptide/therapeutic use , Glucagon/therapeutic use , Glucagon-Like Peptide 1 , Glucose , Hypoglycemic Agents/pharmacokinetics , Incretins , Insulin/therapeutic use , Linagliptin/pharmacology , Linagliptin/therapeutic use , Network Pharmacology
4.
Clin Pharmacokinet ; 62(9): 1275-1288, 2023 09.
Article in English | MEDLINE | ID: mdl-37452998

ABSTRACT

INTRODUCTION: SPI-62 is a small-molecule 11ß-hydroxysteroid dehydrogenase type 1 (HSD-1) inhibitor exhibiting complicated nonlinear pharmacokinetics (PK) in human. Previously, we developed a target-mediated drug disposition (TMDD) model to characterize the substantial nonlinear PK of SPI-62. OBJECTIVE: The aim of the current analysis was to perform population PK/PD analysis to further link SPI-62 exposure (i.e., PK) with its response (i.e., inhibition of hepatic HSD-1 activity) to gain a quantitative understanding of the SPI-62 dose-exposure-response relationship. METHODS: PK and PD data from the first-in-human (FIH) clinical trials, including single ascending dose (SAD) and multiple ascending dose (MAD) studies, were used for model development. During the model development process, the final model selection was based on biological and physiological plausibility, goodness-of-fit plots, stability of parameter estimates, and objective function value. The nonlinear-mixed effect modeling (NONMEM) software was used for both the implementation of the PK/PD model and model simulation. SPI-62 plasma levels and hepatic HSD-1 inhibition over time following various dose regimens were simulated. RESULTS: The final model was a two-compartment TMDD model component for SPI-62 and an inhibitory Imax model component for hepatic HSD-1 activity. The TMDD-hepatic PD model that we established adequately characterized all remarkable PK and PD behaviors of SPI-62, such as extremely low plasma exposures following the first low doses, nonlinear PK turned into linear PK after repeated low doses, and substantial and long-lasting hepatic HSD-1 inhibition following low doses. SPI-62 was estimated to bind to the target with a second-order association rate constant (Kon) of 8.43 nM-1 h-1 and first-order dissociation rate constant (Koff) value of 0.229 h-1, indicating that SPI-62 binds rapidly to, and dissociates slowly from, its pharmacological target. The estimated target capacity (Rtot) of 5460 nmol corresponds to approximately 2.2 mg of SPI-62, which comports well with the dose range in which PK nonlinearity is prominent. Model simulation results reveal that a 6 mg once-daily regimen can lead to long-lasting and substantial hepatic HSD-1 inhibition. CONCLUSIONS: A population TMDD-PD model that explains SPI-62 nonlinear PK and hepatic HSD-1 inhibition following different dose regimens in healthy adults was successfully established. Our simulation results provide a solid foundation for model-informed development of SPI-62.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1 , Software , Humans , Adult , Computer Simulation , Dose-Response Relationship, Drug , Drug Delivery Systems/methods , Models, Biological
5.
J Antimicrob Chemother ; 78(6): 1460-1470, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37071586

ABSTRACT

OBJECTIVES: We aimed to identify rational empirical dosing strategies for cefepime treatment in critically ill patients by utilizing population pharmacokinetics and target attainment analysis. PATIENTS AND METHODS: A prospective and opportunistic pharmacokinetic (PK) study was conducted in 130 critically ill patients in two ICU sites. The plasma concentrations of cefepime were determined using a validated LC-MS/MS method. All cefepime PK data were analysed simultaneously using the non-linear mixed-effects modelling approach. Monte Carlo simulations were performed to evaluate the PTA of cefepime at different MIC values following different dose regimens in subjects with different renal functions. RESULTS: The PK of cefepime in critically ill patients was best characterized by a two-compartment model with zero-order input and first-order elimination. Creatinine clearance and body weight were identified to be significant covariates. Our simulation results showed that prolonged 3 h infusion does not provide significant improvement on target attainment compared with the traditional intermittent 0.5 h infusion. In contrast, for a given daily dose continuous infusion provided much higher breakpoint coverage than either 0.5 h or 3 h intermittent infusions. To balance the target attainment and potential neurotoxicity, cefepime 3 g/day continuous infusion appears to be a better dosing regimen than 6 g/day continuous infusion. CONCLUSIONS: Continuous infusion may represent a promising strategy for cefepime treatment in critically ill patients. With the availability of institution- and/or unit-specific cefepime susceptibility patterns as well as individual patients' renal function, our PTA results may represent useful references for physicians to make dosing decisions.


Subject(s)
Anti-Bacterial Agents , Critical Illness , Humans , Cefepime , Anti-Bacterial Agents/therapeutic use , Chromatography, Liquid , Prospective Studies , Tandem Mass Spectrometry , Monte Carlo Method , Microbial Sensitivity Tests
6.
AAPS J ; 25(3): 41, 2023 04 13.
Article in English | MEDLINE | ID: mdl-37055588

ABSTRACT

In general, small-molecule target-mediated drug disposition (TMDD) is caused by the interaction of a drug with its high-affinity, low-capacity pharmacological target. In the current work, we developed a pharmacometrics model to characterize a new type of TMDD, where the nonlinear pharmacokinetics (PK) is mediated by a high-capacity pharmacological target with cooperative binding instead of target saturation. The model drug we used was PF-07059013, a noncovalent hemoglobin modulator that demonstrated promising preclinical efficacy to treat sickle cell disease (SCD), and showed complex nonlinear PK in mice with the fraction of unbound drug in blood (fub) decreased with an increase in PF-07059013 concentrations/doses due to the positive cooperative binding of PF-07059013 to hemoglobin. Among the various models we evaluated, the best one is a semi-mechanistic model where only drug molecules not bound to hemoglobin were allowed for elimination, with the nonlinear pharmacokinetics being captured by incorporating cooperative binding for drug molecules bound to hemoglobin. Our final model provided valuable insight on target binding-related parameters, such as the Hill coefficient γ (estimated to be 1.6), binding constant KH (estimated to be 1450 µM), and the amount of total hemoglobin Rtot (estimated to be 2.13 µmol). As the dose selection of a compound with positive cooperative binding is tricky and challenging due to the nonproportional and steep response, our model may be valuable in facilitating the rational dose regimen selection for future preclinical animal and clinical trials for PF-07059013 and other compounds whose nonlinear pharmacokinetics are caused by similar mechanisms.


Subject(s)
Drug Delivery Systems , Models, Biological , Mice , Animals , Computer Simulation
7.
Antimicrob Agents Chemother ; 67(4): e0124822, 2023 04 18.
Article in English | MEDLINE | ID: mdl-36920230

ABSTRACT

In the current study, population pharmacokinetic (PK) of ampicillin-sulbactam was performed based on the clinical pharmacokinetics data collected from a prospective study conducted in 40 surgical patients undergoing prolonged surgery where antibiotic redosing was implemented. A population PK model was successfully developed to characterize the disposition of ampicillin and sulbactam. The final models were two-compartment models for both drugs, with creatinine clearance and heart failure affecting clearance and body surface area having an impact on the central volume of distribution of both ampicillin and sulbactam. Comprehensive Monte Carlo simulations were performed to evaluate the probability of target attainment (PTA) of 24 different redosing scenarios. Simulation results indicated that the ampicillin-sulbactam 2-h redosing scheme recommended by ASHP guidelines is likely too conservative given that 3-g dose (2-g ampicillin/1-g sulbactam) with 4-h redosing interval can reach the breakpoint of 2 mg/L for ampicillin in all populations even with the aggressive pharmacokinetic/pharmacodynamic (PK/PD) target of 100% fT > MIC. With the target 50% fT > MIC, all redosing schemes evaluated, including the 8-h redosing scenario, are predicted to be able to reach the breakpoint of 64 mg/L in all patients. According to our findings, redosing of ampicillin-sulbactam should be every 4 h instead of the currently recommended 2-h redosing schedule. Our PTA results should inform future updates to existing general antibiotic redosing guidelines; and, when used in combination with the availability of institution- and/or unit-specific ampicillin susceptibility patterns, our PTA results may be used to customize SSI prophylaxis redosing recommendations for ampicillin-sulbactam at individual hospitals.


Subject(s)
Ampicillin , Sulbactam , Humans , Sulbactam/pharmacology , Prospective Studies , Ampicillin/therapeutic use , Anti-Bacterial Agents/pharmacokinetics
8.
Clin Pharmacokinet ; 62(1): 127-139, 2023 01.
Article in English | MEDLINE | ID: mdl-36633812

ABSTRACT

BACKGROUND AND OBJECTIVE: Piperacillin/tazobactam is one of the most frequently used antimicrobials in older adults. Using an opportunistic study design, we evaluated the pharmacokinetics of piperacillin/tazobactam as a probe drug to evaluate changes in antibacterial drug exposure and dosing requirements, including in older adults. METHODS: A total of 121 adult patients were included. The population pharmacokinetic models that best characterized the observed plasma concentrations of piperacillin and tazobactam were one-compartment structural models with zero-order input and linear elimination. RESULTS: Among all potential covariates, estimated creatinine clearance had the most substantial impact on the elimination clearance for both piperacillin and tazobactam. After accounting for renal function and body size, there was no remaining impact of frailty on the pharmacokinetics of piperacillin and tazobactam. Monte Carlo simulations indicated that renal function had a greater impact on the therapeutic target attainment than age, although these covariates were highly correlated. Frailty, using the Canadian Study of Health and Aging Clinical Frailty Scale, was assessed in 60 patients who were ≥ 65 years of age. CONCLUSIONS: The simulations suggested that adults ≤ 50 years of age infected with organisms with higher minimum inhibitory concentrations may benefit from continuous piperacillin/tazobactam infusions (12 g/day of piperacillin component) or extended infusions of 4 g every 8 hours. However, for a target of 50% fT + minimum inhibitory concentration, dosing based on renal function is generally preferable to dosing by age, and simulations suggested that patients with creatinine clearance ≥ 120 mL/min may benefit from infusions of 4 g every 8 hours for organisms with higher minimum inhibitory concentrations.


Subject(s)
Frailty , Longevity , Humans , Aged , Creatinine , Penicillanic Acid/pharmacokinetics , Canada , Piperacillin, Tazobactam Drug Combination , Anti-Bacterial Agents/pharmacokinetics , Piperacillin/pharmacokinetics , Tazobactam , Microbial Sensitivity Tests
9.
Antimicrob Agents Chemother ; 67(1): e0131222, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36622154

ABSTRACT

In the present study, population pharmacokinetic (PK) analysis was performed based on meropenem data from a prospective study conducted in 114 critically ill patients with a wide range of renal functions and various disease conditions. The final model was a one-compartment model with linear elimination, with creatinine clearance and continuous renal replacement therapy affecting clearance, and total bodyweight impacting the volume of distribution. Our model is a valuable addition to the existing meropenem population PK models, and it could be particularly useful during implementation of a therapeutic drug monitoring program combined with Bayesian forecasting. Based on the final model developed, comprehensive Monte Carlo simulations were performed to evaluate the probability of target attainment (PTA) of 16 different dosing regimens. Simulation results showed that 2 g administered every 8 h with 3-h prolonged infusion (PI) and 4 g/day by continuous infusion (CI) appear to be two empirical dosing regimens that are superior to many other regimens when both target attainment and potential toxicity are considered and renal function information is not available. Following a daily CI dose of 6 g or higher, more than 30% of the population with a creatinine clearance of <60 mL/min is predicted to have neurotoxicity. With the availability of institution- and/or unit-specific meropenem susceptibility patterns, as well as an individual patient's renal function, our PTA results may represent useful references for physicians to make dosing decisions.


Subject(s)
Anti-Bacterial Agents , Intensive Care Units , Humans , Meropenem/pharmacokinetics , Anti-Bacterial Agents/pharmacokinetics , Prospective Studies , Creatinine , Bayes Theorem , Critical Illness/therapy , Monte Carlo Method , Microbial Sensitivity Tests
10.
J Clin Pharmacol ; 63(5): 526-538, 2023 05.
Article in English | MEDLINE | ID: mdl-36479709

ABSTRACT

With more potent drug candidates being developed, the incidence of target-mediated drug disposition (TMDD) in small-molecule compounds has significantly increased in the past decade. Moreover, TMDD appears to apply to some small-molecule compound classes. The main purpose of the current review is to increase the awareness of TMDD in a series of small-molecule inhibitors of 11ß-hydroxysteroid dehydrogenase type 1 (HSD-1) using ABT-384, SPI-62, MK-0916, BMS-823778, and BI-187004 as case examples. Although developed independently by different pharmaceutical companies, these HSD-1 inhibitors demonstrated strikingly similar nonlinear pharmacokinetic behaviors when wide dose ranges were evaluated in first-in-human (FIH) single ascending dose (SAD) and multiple ascending dose (MAD) studies. Recognizing TMDD in small-molecule compounds is important, as the information can be leveraged to select the appropriate dose regimen, improve clinical trial design, as well as predict pharmacological target occupancy. In this review, we summarize the general pharmacokinetic features that facilitate the recognition of small-molecule TMDD, provide case examples of specific HSD-1 inhibitors, highlight the importance of recognizing TMDD of small-molecule compounds during clinical development, and comment on the importance of utilizing pharmacometric modeling to facilitate the quantitative understanding of small-molecule compounds exhibiting TMDD.


Subject(s)
Drug Development , Models, Biological , Humans , Drug Delivery Systems , Enzyme Inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 1
11.
J Pharmacokinet Pharmacodyn ; 50(1): 63-74, 2023 02.
Article in English | MEDLINE | ID: mdl-36376611

ABSTRACT

Recently, a new mechanism of drug-drug interaction (DDI) was reported between agalsidase, a therapeutic protein, and migalastat, a small molecule, both of which are treatment options of Fabry disease. Migalastat is a pharmacological chaperone that stabilizes the native form of both endogenous and exogenous agalsidase. In Fabry patients co-administrated with agalsidase and migalastat, the increase in active agalsidase exposure is considered a pharmacokinetic effect of agalsidase infusion but a pharmacodynamic effect of migalastat administration, which makes this new DDI mechanism even more interesting. To quantitatively characterize the interaction between agalsidase and migalastat in human, a pharmacometric DDI model was developed using literature reported concentration-time data. The final model includes three components: a 1-compartment linear model component for migalastat; a 2-compartment linear model component for agalsidase; and a DDI component where the agalsidase-migalastat complex is formed via second order association constant kon, dissociated with first order dissociation constant koff, and distributed/eliminated with same rates as agalsidase alone, albeit the complex (i.e., bound agalsidase) has higher enzyme activity compared to free agalsidase. The final model adequately captured several key features of the unique interaction between agalsidase and migalastat, and successfully characterized the kinetics of migalastat as well as the kinetics and activities of agalsidase when both drugs were used alone or in combination following different doses. Most parameters were reasonably estimated with good precision. Because the model includes mechanistic basis of therapeutic protein and small molecule pharmacological chaperone interaction, it can potentially serve as a foundational work for DDIs with similar mechanism.


Subject(s)
1-Deoxynojirimycin , alpha-Galactosidase , Humans , alpha-Galactosidase/genetics , alpha-Galactosidase/metabolism , Mutation , 1-Deoxynojirimycin/pharmacology , 1-Deoxynojirimycin/therapeutic use , Drug Interactions
12.
J Pharm Sci ; 111(9): 2620-2629, 2022 09.
Article in English | MEDLINE | ID: mdl-35691608

ABSTRACT

Our aim was to build a mechanistic full target-mediated drug disposition (TMDD) model for rhEpo to better understand rhEpo disposition, Epo receptor (EpoR) synthesis, and degradation in hematopoietic transplant patients with four distinct bone marrow conditions. All PK data were analyzed simultaneously using the nonlinear mixed effect modeling approach with NONMEM. The final model was a two-compartmental full TMDD model, which adequately characterizes rhEpo PK in patients and provides insight into the dynamics of free EpoR, rhEpo-EpoR, and total EpoR. The model predicted association rate constant (kon), dissociation rate constant (koff), and internalization rate constant (kint) were 0.0276 pM-1h-1, 0.647 h-1, and 0.255h-1, respectively, which were supported by experimental data. Also, the EpoR degradation rate constant (kdeg) was estimated to be 0.461 h-1. EpoR production rate was estimated to be 37.5 pM/h for adults at pre-ablation baseline and 5.91 pM/h, and 4.19 pM/h in the early post-transplant post-engraftment, and late post-transplant full engraftment. Our model provides extensive information on the dynamics of free EpoR, total EpoR and rhEpo-EpoR, and proven to be more robust and can provide more physiologically relevant binding parameters than previous models.


Subject(s)
Erythropoietin , Hematopoietic Stem Cell Transplantation , Bone Marrow/metabolism , Erythropoietin/pharmacokinetics , Humans , Receptors, Erythropoietin/metabolism , Recombinant Proteins
13.
J Pharm Biomed Anal ; 210: 114521, 2022 Feb 20.
Article in English | MEDLINE | ID: mdl-34979494

ABSTRACT

Cefazolin is widely used during surgery to prevent surgical site infections (SSIs). Although cefazolin redosing is often needed due to its short half-life, the appropriate redosing schedule remains controversial and there is limited information on cefazolin disposition following repeated doses during surgery. In parallel with an ongoing cefazolin redosing clinical study, we have developed and fully validated a simple and robust liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for the quantification of cefazolin in human plasma. A simple protein precipitation was used for sample preparation. MS/MS analysis was performed using multiple reaction monitoring (MRM) under a positive ionization mode. The lower limit of quantification (LLOQ) for cefazolin was evaluated at 0.25 µg/mL and a linearity ranging from 0.25 to 300 µg/mL. Accuracy was ≤ 114.3% for quality controls and ≤ 118.2% for LLOQ; intra-day and inter-day precision ranging from 1.9% to 14.2% for all quality controls and LLOQ. Matrix effect, extraction recovery, stability testing, dilution integrity, hemolysis effects and whole blood stability have all been investigated. A total of 17 parameters were validated and passed their validation criteria. The method was applied in the quantification of cefazolin in clinical plasma samples and was able to successfully determine the concentrations in patients undergoing various surgeries. In comparison with other prior published methods, our method has a simple sample preparation combined with a short analysis run time, a wide dynamic range and low limit of quantification, and is a fully validated assay that abides by FDA guidance.


Subject(s)
Cefazolin , Tandem Mass Spectrometry , Chromatography, Liquid , Humans , Plasma , Reproducibility of Results
14.
Antimicrob Agents Chemother ; 66(1): e0143221, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34606333

ABSTRACT

Oxfendazole is a potent veterinary antiparasitic drug undergoing development for human use to treat multiple parasitic infections. Results from two recently completed phase I clinical trials conducted in healthy adults showed that the pharmacokinetics of oxfendazole is nonlinear, affected by food, and, after the administration of repeated doses, appeared to mildly affect hemoglobin concentrations. To facilitate oxfendazole dose optimization for its use in patient populations, the relationship among oxfendazole dose, pharmacokinetics, and hemoglobin concentration was quantitatively characterized using population pharmacokinetic-pharmacodynamic modeling. In fasting subjects, oxfendazole pharmacokinetics was well described by a one-compartment model with first-order absorption and elimination. The change in oxfendazole pharmacokinetics when administered following a fatty meal was captured by an absorption model with one transit compartment and increased bioavailability. The effect of oxfendazole exposure on hemoglobin concentration in healthy adults was characterized by a life span indirect response model in which oxfendazole has positive but minor inhibitory effect on red blood cell synthesis. Further simulation indicated that oxfendazole has a low risk of posing a safety concern regarding hemoglobin concentration, even at a high oxfendazole dose of 60 mg/kg of body weight once daily. The final model was further used to perform comprehensive target attainment simulations for whipworm infection and filariasis at various dose regimens and target attainment criteria. The results of our modeling work, when adopted appropriately, have the potential to greatly facilitate oxfendazole dose regimen optimization in patient populations with different types of parasitic infections.


Subject(s)
Benzimidazoles , Adult , Benzimidazoles/pharmacokinetics , Biological Availability , Body Weight , Computer Simulation , Dose-Response Relationship, Drug , Humans
15.
J Pharm Sci ; 111(5): 1245-1249, 2022 05.
Article in English | MEDLINE | ID: mdl-34919967

ABSTRACT

Liquid chromatography-tandem mass spectrometry (LC-MS/MS) is a widely used quantitative method in small-molecule pharmacokinetic sample analysis. The linear dynamic range of mass analyzers, typically spanning 3 orders of magnitude, is usually insufficient for this purpose. Utilization of multiple isotopologues has been proposed as a compelling approach to expand the linear dynamic range of LC-MS/MS assays, particularly when the detector is saturated. Isotopologues are a statistical mixture of molecules of the same compound but of different exact masses due to the presence of natural chemical isotopes. While the concept of isotopologues is widely recognized in large-molecule bioanalysis and small-molecule metabolite profiling, it has not been commonly implemented in small-molecule targeted quantification. To increase the awareness of the value of isotopologues in small-molecule LC-MS/MS analysis, this minireview provides the basis of isotopologue distribution in MS/MS and summarizes published studies as well as our own experience in utilizing multiple isotopologues to expand the linear dynamic range of small-molecule LC-MS/MS assays. Considering that utilizing natural isotopologue transitions in the LC-MS/MS assays represents an easy, straightforward, and robust way to expand the linear dynamic range, we believe this method deserves wide application in small-molecule pharmacokinetic sample analysis and can particularly benefit people working in pharmacokinetic labs as well as the GLP bioanalytical labs in pharmaceutical industry.


Subject(s)
Isotopes , Tandem Mass Spectrometry , Biological Assay , Chromatography, Liquid/methods , Humans , Tandem Mass Spectrometry/methods
16.
J Clin Pharmacol ; 61(11): 1442-1453, 2021 11.
Article in English | MEDLINE | ID: mdl-34110620

ABSTRACT

SPI-62 is a selective and potent small-molecule inhibitor of 11ß-hydroxysteroid dehydrogenase type 1 (HSD-1). SPI-62 has demonstrated substantial and complex nonlinear pharmacokinetics (PK) in humans that is characterized by unusually low plasma exposure at low doses, dose-dependent volume of distribution, nonlinear PK following the first dose, and dose-proportional PK at steady state, as well as unusually high accumulation ratios at low doses. The most likely explanation for the observed nonlinearity of SPI-62 is the saturable binding of SPI-62 to its pharmacological target HSD-1, a phenomenon known as target-mediated drug disposition (TMDD). Because of the nonlinear and complex PK of SPI-62, the relationship among SPI-62 dose, exposure, and response is no longer intuitive and consequently dose selection can be challenging. To facilitate dose selection and clinical trial design, in the current study population PK analysis was performed to characterize SPI-62 dose-exposure relationship in humans quantitatively. SPI-62 PK was best characterized by a 2-compartment TMDD model with 3 transit absorption compartments. The model was successfully established to explain the substantial and unusual nonlinear PK of SPI-62 in humans, and it provided adequate fitting for both single- and multiple-dose data. Our modeling work has provided a strong foundation for dose selection in future SPI-62 clinical trials.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Benzamides/pharmacokinetics , Triazoles/pharmacokinetics , Dose-Response Relationship, Drug , Double-Blind Method , Humans , Models, Biological
17.
J Pharmacokinet Pharmacodyn ; 48(4): 581-595, 2021 08.
Article in English | MEDLINE | ID: mdl-33884580

ABSTRACT

First-order conditional estimation (FOCE) has been the most frequently used estimation method in NONMEM, a leading program for population pharmacokinetic/pharmacodynamic modeling. However, with growing data complexity, the performance of FOCE is challenged by long run time, convergence problem and model instability. In NONMEM 7, expectation-maximization (EM) estimation methods and FOCE with FAST option (FOCE FAST) were introduced. In this study, we compared the performance of FOCE, FOCE FAST, and two EM methods, namely importance sampling (IMP) and stochastic approximation expectation-maximization (SAEM), utilizing the rich pharmacokinetic data of oxfendazole and its two metabolites obtained from the first-in-human single ascending dose study in healthy adults. All methods yielded similar parameter estimates, but great differences were observed in parameter precision and modeling time. For simpler models (i.e., models of oxfendazole and/or oxfendazole sulfone), FOCE and FOCE FAST were more efficient than EM methods with shorter run time and comparable parameter precision. FOCE FAST was about two times faster than FOCE but it was prone to premature termination. For the most complex model (i.e., model of all three analytes, one of which having high level of data below quantification limit), FOCE failed to reliably assess parameter precision, while parameter precision obtained by IMP and SAEM was similar with SAEM being the faster method. IMP was more sensitive to model misspecification; without pre-systemic metabolism, IMP analysis failed to converge. With parallel computing introduced in NONMEM 7.2, modeling speed increased less than proportionally with the increase in the number of CPUs from 1 to 16.


Subject(s)
Models, Statistical , Pharmacokinetics , Benzimidazoles/pharmacokinetics , Benzimidazoles/pharmacology , Fenbendazole/pharmacokinetics , Fenbendazole/pharmacology , Humans , Nonlinear Dynamics , Pharmacology
18.
Article in English | MEDLINE | ID: mdl-33526484

ABSTRACT

Oxfendazole is a potent veterinary benzimidazole anthelmintic under transition to humans 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 of body weight 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 a 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 into the significance of presystemic metabolism in oxfendazole and metabolite 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 liters/h and 35.2 liters, 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 a one-compartment model with formation rate-limited elimination. Fenbendazole formation from oxfendazole was primarily through systemic metabolism, while both presystemic 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.


Subject(s)
Anthelmintics , Benzimidazoles , Administration, Oral , Adult , Fenbendazole , Humans , Metabolic Clearance Rate
19.
J Pharm Biomed Anal ; 196: 113899, 2021 Mar 20.
Article in English | MEDLINE | ID: mdl-33508765

ABSTRACT

Ampicillin-sulbactam is a broad-spectrum combination antibiotic used for a variety of clinical applications, including as a prophylactic agent to reduce the risk of surgical site infection. The pharmacokinetics of ampicillin-sulbactam after redosing during prolonged surgeries remains incompletely understood. In anticipation of further studying the intra-operative pharmacokinetics of this drug, we have developed a novel liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for the quantification of ampicillin and sulbactam. The plasma samples were prepared using a simple protein precipitation method. Gradient chromatographic elution was used to separate analytes, and MS/MS analysis was performed in negative ionization mode for both analytes via multiple reaction monitoring (MRM). All validation parameters were evaluated under a good laboratory practice (GLP) environment. For both ampicillin and sulbactam, the lower limit of quantitation (LLOQ) was established as 0.25 µg/mL. The calibration curve ranged from 0.25 to 200 µg/mL for ampicillin and 0.25-100 µg/mL for sulbactam. Inter- and intra-day precisions for both analytes were ≤11.5 % for quality controls and ≤17.4 % for LLOQ; accuracies ranged from -11.5 to 12.5% for 3 quality control levels and -18.1-18.7% for LLOQ. In addition to sensitivity, accuracy and precision, 13 other parameters were also validated for both analytes, and the results met the acceptance criteria. Our method was successfully applied to quantify ampicillin and sulbactam concentrations in patients undergoing surgery.


Subject(s)
Pharmaceutical Preparations , Tandem Mass Spectrometry , Ampicillin , Chromatography, Liquid , Humans , Reproducibility of Results , Sulbactam
20.
J Clin Pharmacol ; 61(4): 531-537, 2021 04.
Article in English | MEDLINE | ID: mdl-33078430

ABSTRACT

Pharmacological target-mediated drug disposition (TMDD) represents a special source of nonlinear pharmacokinetics, and its occurrence in large-molecule compounds has been well recognized because numerous protein drugs have been reported to have TMDD due to specific binding to their pharmacological targets. Although TMDD can also happen in small-molecule compounds, it has been largely overlooked. In this mini-review, we summarize the occurrence of TMDD that we discovered recently in a series of small-molecule soluble epoxide hydrolase (sEH) inhibitors. Our journey started with an accidental discovery of target-mediated kinetics of 1-(1-propanoylpiperidin-4-yl)-3-[4-(trifluoromethoxy)phenyl]urea (TPPU), a potent sEH inhibitor, in a pilot clinical study. To confirm what we observed in humans, we conducted a series of mechanism experiments in animals, including pharmacokinetic experiments using sEH knockout mice as well as in vivo displacement experiments with co-administration of another potent sEH inhibitor. Our mechanism studies confirmed that the TMDD of TPPU is due to its pharmacological target sEH. We further expanded our evaluation to various other sEH inhibitors and found that TMDD is a class effect of this group of small-molecule sEH inhibitors. In addition to summarizing the occurrence of TMDD in sEH inhibitors, in this mini-review we also highlighted the importance of recognizing TMDD of small-molecule compounds and its impact in clinical development as well as using pharmacometric modeling in facilitating quantitative understanding of TMDD.


Subject(s)
Epoxide Hydrolases/antagonists & inhibitors , Phenylurea Compounds/pharmacokinetics , Piperidines/pharmacokinetics , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...