1
|
Yin W, Mitra P, Copalu V, Marbury TC, Rondon JC, Lawitz EJ, Lloyd V, Baratta M, Asgharnejad M, Hui T, Khan Y. Phase 1 pharmacokinetic and safety study of soticlestat in participants with mild or moderate hepatic impairment or normal hepatic function. Pharmacol Res Perspect 2024; 12:e1213. [PMID: 38993008 PMCID: PMC11239955 DOI: 10.1002/prp2.1213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 07/13/2024] Open
Abstract
This phase 1, open-label, three-arm study (NCT05098054) compared the pharmacokinetics and safety of soticlestat (TAK-935) in participants with hepatic impairment. Participants aged ≥18 to <75 years had moderate (Child-Pugh B) or mild (Child-Pugh A) hepatic impairment or normal hepatic function (matched to hepatic-impaired participants by sex, age, and body mass index). Soticlestat was administered as a single oral 300 mg dose. Pharmacokinetic parameters of soticlestat and its metabolites TAK-935-G (M3) and M-I were assessed and compared by group. The incidence of treatment-emergent adverse events (TEAEs) and other safety parameters were also monitored. The pharmacokinetic analyses comprised 35 participants. Participants with moderate hepatic impairment had lower proportions of bound and higher proportions of unbound soticlestat than participants with mild hepatic impairment and normal hepatic function. Total plasma soticlestat pharmacokinetic parameters (maximum observed concentration [Cmax], area under the concentration-time curve from time 0 to time of last quantifiable concentration [AUClast], and AUC from time 0 to infinity [AUC∞]) were approximately 115%, 216%, and 199% higher with moderate and approximately 45%, 35%, and 30% higher with mild hepatic impairment, respectively, than healthy matched participants. Moderate hepatic impairment decreased the liver's ability to metabolize soticlestat to M-I; glucuronidation to M3 was also affected. Mild hepatic impairment resulted in a lower total plasma M-I exposure, but glucuronidation was unaffected. TEAEs were similar across study arms, mild, and no new safety findings were observed. A soticlestat dose reduction is required for individuals with moderate but not mild hepatic impairment.
Collapse
Affiliation(s)
- Wei Yin
- Takeda Development Center Americas, Inc.CambridgeMassachusettsUSA
| | - Pranab Mitra
- Takeda Development Center Americas, Inc.CambridgeMassachusettsUSA
| | - Veronique Copalu
- Takeda Development Center Americas, Inc.CambridgeMassachusettsUSA
| | | | | | - Eric J. Lawitz
- The Texas Liver InstituteUniversity of Texas Health San AntonioSan AntonioTexasUSA
| | - Valerie Lloyd
- Takeda Development Center Americas, Inc.CambridgeMassachusettsUSA
| | - Mike Baratta
- Takeda Development Center Americas, Inc.CambridgeMassachusettsUSA
| | | | - Tom Hui
- Takeda Development Center Americas, Inc.CambridgeMassachusettsUSA
| | - Yasir Khan
- Takeda Development Center Americas, Inc.CambridgeMassachusettsUSA
| |
Collapse
|
2
|
Basharat Z, Meshal A. Pan-genome mediated therapeutic target mining in Kingella kingae and inhibition assessment using traditional Chinese medicinal compounds: an informatics approach. J Biomol Struct Dyn 2024; 42:2872-2885. [PMID: 37144759 DOI: 10.1080/07391102.2023.2208221] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/23/2023] [Indexed: 05/06/2023]
Abstract
Kingella kingae causes bacteremia, endocarditis, osteomyelitis, septic arthritis, meningitis, spondylodiscitis, and lower respiratory tract infections in pediatric patients. Usually it demonstrates disease after inflammation of mouth, lips or infections of the upper respiratory tract. To date, therapeutic targets in this bacterium remain unexplored. We have utilized a battery of bioinformatics tools to mine these targets in this study. Core genes were initially inferred from 55 genomes of K. kingae and 39 therapeutic targets were mined using an in-house pipeline. We selected aroG product (KDPG aldolase) involved in chorismate pathway, for inhibition analysis of this bacterium using lead-like metabolites from traditional Chinese medicinal plants. Pharmacophore generation was done using control ZINC36444158 (1,16-bis[(dihydroxyphosphinyl)oxy]hexadecane), followed by molecular docking of top hits from a library of 36,000 compounds. Top prioritized compounds were ZINC95914016, ZINC33833283 and ZINC95914219. ADME profiling and simulation of compound dosing (100 mg tablet) was done to infer compartmental pharmacokinetics in a population of 300 individuals in fasting state. PkCSM based toxicity analysis revealed the compounds ZINC95914016 and ZINC95914219 as safe and with almost similar bioavailability. However, ZINC95914016 takes less time to reach maximum concentration in the plasma and shows several optimal parameters compared to other leads. In light of obtained data, we recommend this compound for further testing and induction in experimental drug design pipeline.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Alotaibi Meshal
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Albatin, Saudi Arabia
| |
Collapse
|
3
|
Hindi J, Fréchette-Le Bel M, Rouleau JL, de Denus S. Influence of Weight and Body Size on the Pharmacokinetics of Heart Failure Pharmacotherapy: A Systematic Review. Ann Pharmacother 2024; 58:255-272. [PMID: 37338205 DOI: 10.1177/10600280231179484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2023] Open
Abstract
OBJECTIVE To conduct a review of studies evaluating the influence of body size and weight (WT) on the pharmacokinetics (PK) of drugs recommended for heart failure (HF) treatment. DATA SOURCES A systematic search of the MEDLINE (1946 to April 2023) and EMBASE (1974 to April 2023) databases was conducted for articles that focused on the impact of WT or body size on the PK of drugs of interest used in HF patients. STUDY SELECTION AND DATA EXTRACTION Articles written in English or French related to the aim of our study were retained for analysis. DATA SYNTHESIS Of 6493 articles, 20 were retained for analysis. Weight was associated with the clearance of digoxin, carvedilol, enalapril, and candesartan as well as the volume of distribution of eplerenone and bisoprolol. There was no documented direct impact of WT on the PK of furosemide, valsartan, and metoprolol, although these studies were limited or confounded by the small sample size, adjustment of PK factors by WT, or the use of the Cockroff-Gault equation for the evaluation of creatinine clearance, which includes WT. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE This review highlights and summarizes the available data on the importance of WT on the PK of HF treatment. CONCLUSION Considering the significant impact of WT on most HF drugs in this review, it may be important to further investigate it in the context of personalized therapy, particularly in patients presenting extreme WTs.
Collapse
Affiliation(s)
- Jessica Hindi
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
- Montreal Heart Institute, Montreal, QC, Canada
- Beaulieu-Saucier Pharmacogenomics Center, Université de Montréal, Montreal, QC, Canada
| | | | - Jean Lucien Rouleau
- Montreal Heart Institute, Montreal, QC, Canada
- Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Simon de Denus
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
- Montreal Heart Institute, Montreal, QC, Canada
- Beaulieu-Saucier Pharmacogenomics Center, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
4
|
Bashiardes S, Christodoulou C. Orally Administered Drugs and Their Complicated Relationship with Our Gastrointestinal Tract. Microorganisms 2024; 12:242. [PMID: 38399646 PMCID: PMC10893523 DOI: 10.3390/microorganisms12020242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Orally administered compounds represent the great majority of all pharmaceutical compounds produced for human use and are the most popular among patients since they are practical and easy to self-administer. Following ingestion, orally administered drugs begin a "perilous" journey down the gastrointestinal tract and their bioavailability is modulated by numerous factors. The gastrointestinal (GI) tract anatomy can modulate drug bioavailability and accounts for interpatient drug response heterogeneity. Furthermore, host genetics is a contributor to drug bioavailability modulation. Importantly, a component of the GI tract that has been gaining notoriety with regard to drug treatment interactions is the gut microbiota, which shares a two-way interaction with pharmaceutical compounds in that they can be influenced by and are able to influence administered drugs. Overall, orally administered drugs are a patient-friendly treatment option. However, during their journey down the GI tract, there are numerous host factors that can modulate drug bioavailability in a patient-specific manner.
Collapse
Affiliation(s)
- Stavros Bashiardes
- Molecular Virology Department, Cyprus Institute of Neurology and Genetics, Iroon Avenue 6, Nicosia 2371, Cyprus;
| | | |
Collapse
|
5
|
Karsten Dafonte K, Weber L, Chmielewski F, Böhmer AM, Lutz P, Hartmann G, Jaehde U, Coenen M. Dose Recommendations for Common Drugs in Patients with Liver Cirrhosis: A Systematic Literature Review. Clin Drug Investig 2023; 43:475-502. [PMID: 37460783 PMCID: PMC10374753 DOI: 10.1007/s40261-023-01289-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND AND OBJECTIVES The presence of liver cirrhosis affects the selection and dosing of drugs metabolised by the liver as doses have to be adjusted to the remaining liver function. This is a major challenge in clinical practice as specific guidelines are lacking. The aim of this study was to identify drugs for which recommendations on selection and dose adjustments for patients with cirrhosis exist by assessing the literature according to certain quality standards, paying particular attention to the suitability of these recommendations for clinical practice. METHODS A systematic literature review included peer-reviewed publications that were published by October 2020 in PubMed in the English language and aimed to generate recommendations on dose adjustment in patients with liver cirrhosis. Subsequently, the identified publications were checked for reporting quality against the relevant reporting guidelines and the Oxford Centre for Evidence-Based Medicine Levels of Evidence. Finally, all specific dose recommendations were extracted, compared with the specifications of the Summaries of Product Characteristics and mapped according to the Anatomical Therapeutic Chemical/Defined Daily Dose Index. RESULTS Eighteen publications covering a total of 1145 dose recommendations for 481 active substances were identified. There were 706 recommendations for 316 substances sufficiently specific for application in clinical practice. For 22 active substances, the specific recommendations were consistent across multiple publications, of which only six were also consistent with the respective Summaries of Product Characteristics. CONCLUSIONS As the majority of dose recommendations were not sufficiently specific or even contradictory, there is an urgent need for the definition of standard parameters for a uniform assessment of drugs in liver cirrhosis. In addition, dose recommendations should be aligned by suitable methods.
Collapse
Affiliation(s)
- Katharina Karsten Dafonte
- Institute of Clinical Chemistry and Clinical Pharmacology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| | - Lisa Weber
- Pharmacy of the University Hospital Bonn, Bonn, Germany
| | | | - Anna Maria Böhmer
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Bonn, An der Immenburg 4, Bonn, 53121, Germany
| | - Philipp Lutz
- Department of Internal Medicine I, University Hospital Bonn, Bonn, 53127, Germany
| | - Gunther Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Ulrich Jaehde
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Bonn, An der Immenburg 4, Bonn, 53121, Germany
| | - Martin Coenen
- Institute of Clinical Chemistry and Clinical Pharmacology, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| |
Collapse
|
6
|
Hanley MJ, Kerstein D, Tugnait M, Narasimhan N, Marbury TC, Venkatakrishnan K, Gupta N. Brigatinib pharmacokinetics in patients with chronic hepatic impairment. Invest New Drugs 2023:10.1007/s10637-023-01339-6. [PMID: 37052729 DOI: 10.1007/s10637-023-01339-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/08/2023] [Indexed: 04/14/2023]
Abstract
Brigatinib is an anaplastic lymphoma kinase (ALK) inhibitor approved for the treatment of ALK-positive non-small cell lung cancer. This open-label, parallel-group study investigated the effect of chronic hepatic impairment on the pharmacokinetics (PK) of brigatinib to inform dosing recommendations for these patients. Participants with hepatic impairment classified according to Child-Pugh categories of mild (A), moderate (B), or severe (C) and matched-healthy participants with normal hepatic function received a single oral dose of 90-mg brigatinib. Plasma samples were collected for the determination of brigatinib plasma protein binding and estimation of plasma PK parameters. Twenty-seven participants were enrolled (Child-Pugh A-C, n = 6 each; matched-healthy participants, n = 9). The mean fraction of free plasma brigatinib was comparable for the Child-Pugh A (11.1%), Child-Pugh B (10.8%), and healthy participant groups (8.5%); free brigatinib was higher in the Child-Pugh C group (23.1%). There were no clinically meaningful effects of mild or moderate hepatic impairment on unbound systemic exposures (area under the plasma concentration-time curve [AUC]) of brigatinib (geometric least-squares mean ratios [90% CI] of 89.32% [69.79%-114.31%] and 99.55% [77.78%-127.41%], respectively). In the severe hepatic impairment group, brigatinib unbound AUC was approximately 37% higher (geometric least-squares mean ratio [90% CI] of 137.41% [107.37%-175.86%]) compared with healthy participants with normal hepatic function. Brigatinib was well tolerated in healthy participants and in participants with hepatic impairment. No dose adjustment is required for patients with mild or moderate hepatic impairment. The brigatinib dose should be reduced by approximately 40% for patients with severe hepatic impairment.
Collapse
Affiliation(s)
- Michael J Hanley
- Takeda Development Center Americas, Inc., 95 Hayden Avenue, Lexington, MA, 02421, USA
| | - David Kerstein
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
- Theseus Pharmaceuticals, Cambridge, MA, USA
| | - Meera Tugnait
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
- Cerevel Therapeutics, Cambridge, MA, USA
| | - Narayana Narasimhan
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
- Theseus Pharmaceuticals, Cambridge, MA, USA
| | | | - Karthik Venkatakrishnan
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
- EMD Serono Research and Development Institute, Inc., Billerica, MA, USA
| | - Neeraj Gupta
- Takeda Development Center Americas, Inc., 95 Hayden Avenue, Lexington, MA, 02421, USA.
| |
Collapse
|
7
|
Morales Castro D, Dresser L, Granton J, Fan E. Pharmacokinetic Alterations Associated with Critical Illness. Clin Pharmacokinet 2023; 62:209-220. [PMID: 36732476 PMCID: PMC9894673 DOI: 10.1007/s40262-023-01213-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2023] [Indexed: 02/04/2023]
Abstract
Haemodynamic, metabolic, and biochemical derangements in critically ill patients affect drug pharmacokinetics and pharmacodynamics making dose optimisation particularly challenging. Appropriate therapeutic dosing depends on the knowledge of the physiologic changes caused by the patient's comorbidities, underlying disease, resuscitation strategies, and polypharmacy. Critical illness will result in altered drug protein binding, ionisation, and volume of distribution; it will also decrease oral drug absorption, intestinal and hepatic metabolism, and renal clearance. In contrast, the resuscitation strategies and the use of vasoactive drugs may oppose these effects by leading to a hyperdynamic state that will increase blood flow towards the major organs including the brain, heart, kidneys, and liver, with the subsequent increase of drug hepatic metabolism and renal excretion. Metabolism is the main mechanism for drug clearance and is one of the main pharmacokinetic processes affected; it is influenced by patient-specific factors, such as comorbidities and genetics; therapeutic-specific factors, including drug characteristics and interactions; and disease-specific factors, like organ dysfunction. Moreover, organ support such as mechanical ventilation, renal replacement therapy, and extracorporeal membrane oxygenation may contribute to both inter- and intra-patient variability of drug pharmacokinetics. The combination of these competing factors makes it difficult to predict drug response in critically ill patients. Pharmacotherapy targeted to therapeutic goals and therapeutic drug monitoring is currently the best option for the safe care of the critically ill. The aim of this paper is to review the alterations in drug pharmacokinetics associated with critical illness and to summarise the available evidence.
Collapse
Affiliation(s)
- Diana Morales Castro
- Interdepartmental Division of Critical Care Medicine, Toronto General Hospital, University of Toronto, 585 University Avenue, 9-MaRS, Toronto, ON, M5G 2N2, Canada. .,Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada.
| | - Linda Dresser
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - John Granton
- Interdepartmental Division of Critical Care Medicine, Toronto General Hospital, University of Toronto, 585 University Avenue, 9-MaRS, Toronto, ON, M5G 2N2, Canada.,Department of Medicine, Toronto General Hospital, University of Toronto, Toronto, ON, Canada
| | - Eddy Fan
- Interdepartmental Division of Critical Care Medicine, Toronto General Hospital, University of Toronto, 585 University Avenue, 9-MaRS, Toronto, ON, M5G 2N2, Canada.,Department of Medicine, Toronto General Hospital, University of Toronto, Toronto, ON, Canada.,Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
8
|
Song I, Chen G, Wu J, Ilic K. Maribavir Pharmacokinetics and Safety in Participants With Moderate Hepatic Impairment: A Phase 1, Open-Label, Single-Dose, Parallel Group Study. J Clin Pharmacol 2023; 63:250-258. [PMID: 36089648 PMCID: PMC10091977 DOI: 10.1002/jcph.2155] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/08/2022] [Indexed: 01/18/2023]
Abstract
Maribavir, an orally bioavailable antiviral, has shown superior activity against posttransplant cytomegalovirus infection compared with conventional antivirals. It is primarily metabolized in the liver. This open-label, single-center study evaluated the effect of hepatic impairment on the pharmacokinetics of maribavir in nontransplant participants. A single 200-mg dose of maribavir was administered orally under fasting conditions to participants with moderate hepatic impairment (Child-Pugh class B) (n = 10) and healthy controls (n = 10) matched for age, weight, sex, and smoking status. Compared with participants with normal hepatic function, maximum plasma concentration (Cmax ) and area under the plasma concentration-time curve (AUC) from time 0 to infinity values for maribavir in participants with moderate hepatic impairment were 1.346-fold (90%CI of geometric mean ratio, 1.091-1.660) and 1.261-fold (0.889-1.787) higher, respectively. However, Cmax and AUC values for unbound maribavir were comparable. For VP 44469, the main metabolite of maribavir, the Cmax and AUC from time 0 to infinity values were 1.190-fold (0.836-1.693) and 1.309-fold (1.007-1.702) higher, respectively, in participants with moderate hepatic impairment. In total, 7 mild treatment-emergent adverse events were reported, all in the moderate hepatic impairment group. Dysgeusia was the most frequently reported treatment-emergent adverse event, at a frequency of 50%. These results indicated that total maribavir concentrations were mildly increased in participants with moderate hepatic impairment, while unbound concentrations were unaffected. Similar maribavir pharmacokinetics in participants with moderate hepatic impairment and normal hepatic function suggest that dose adjustment may not be required for patients with moderate hepatic impairment.
Collapse
Affiliation(s)
- Ivy Song
- Takeda Development Center Americas, Inc., Lexington, Massachusetts, USA
| | - Grace Chen
- Takeda Development Center Americas, Inc., Lexington, Massachusetts, USA
| | - Jingyang Wu
- Takeda Development Center Americas, Inc., Lexington, Massachusetts, USA
| | - Katarina Ilic
- Takeda Development Center Americas, Inc., Lexington, Massachusetts, USA
| |
Collapse
|
9
|
Torsemide Pharmacometrics in Healthy Adult Populations Including CYP2C9 Genetic Polymorphisms and Various Patient Groups through Physiologically Based Pharmacokinetic-Pharmacodynamic Modeling. Pharmaceutics 2022; 14:pharmaceutics14122720. [PMID: 36559213 PMCID: PMC9784843 DOI: 10.3390/pharmaceutics14122720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/28/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Torsemide is a widely used diuretic in clinical practice. In this study, pharmacokinetic (PK) and pharmacodynamic (PD) simulations of torsemide for various population groups and exposure scenarios were performed through human-scale physiologically-based PK-PD (PBPK-PD) modeling of torsemide. For PBPK-PD modeling of torsemide, invitro and clinical data of torsemide reported previously were used. After exposure to clinical doses of torsemide, observed plasma (or serum) concentration and urine torsemide excretion profiles were used as PK-data, and observed urinary sodium excretion rate was used as PD-data. The model was then extended to take into account physiological and biochemical factors according to different CYP2C9 phenotypes or patient populations. The established model captured various torsemide clinical results well. Differences in torsemide PKs and PDs between patient groups or CYP2C9 genetic polymorphisms were modelologically identified. It was confirmed that degrees of differences in torsemide PKs and PDs by disease groups were greater than those according to different CYP2C9 phenotypes. According to torsemide administration frequency or dose change, it was confirmed that although the difference in plasma PKs between groups (healthy adult and patient groups) could increase to 14.80 times, the difference in PDs was reduced to 1.01 times. Results of this study suggested that it is very important to consider disease groups in the setting of torsemide clinical therapy and that it is difficult to predict PD proportionally with only differences in PKs of torsemide between population groups. The PBPK-PD model established in this study is expected to be utilized for various clinical cases involving torsemide application in the future, enabling optimal drug therapy.
Collapse
|
10
|
Hu Y, Li X, Liu J, Chen H, Zheng W, Zhang H, Wu M, Li C, Zhu X, Lou J, Yan P, Wu N, Liu X, Ma S, Wang X, Ding Y, Xuan C. Safety, pharmacokinetics and pharmacodynamics of a novel γ-aminobutyric acid (GABA) receptor potentiator, HSK3486, in Chinese patients with hepatic impairment. Ann Med 2022; 54:2769-2780. [PMID: 36217101 PMCID: PMC9559057 DOI: 10.1080/07853890.2022.2129433] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND The primary objective of this study was to investigate if hepatic impairment alters the safety, pharmacokinetics, and pharmacodynamics of HSK3486. RESEARCH DESIGN AND METHODS This was a clinical trial of HSK3486 in subjects with normal hepatic function (n = 8), and mild (Child-Pugh A; n = 8), or moderate (Child-Pugh B; n = 8) hepatic impairment. Each subject received an IV bolus dose of 0.4 mg/kg HSK3486 for 1 min, immediately followed by a maintenance infusion of 0.4 mg/kg/h HSK3486 for 30 min. RESULTS In total, 24 subjects were enrolled and completed the study. HSK3486 was generally well tolerated by all subjects. There were no serious AEs and no deaths reported during the study. The incidence of AEs was numerically highest in subjects with moderate hepatic impairment. The exposure (AUC) of HSK3486 increased gradually with the decrease in hepatic function; however, degree of hepatic impairment had little effect on HSK3486 PD (MOAA/S and BIS). CONCLUSIONS Overall, there were no clinically relevant differences in HSK3486 exposure or PD in subjects with mild or moderate hepatic impairment compared to normal control. These data imply that HSK3486 dose adjustment is not warranted in subjects with mild or moderate hepatic impairment. TRIAL REGISTRATION The trial is registered at ClinicalTrials.gov (CT.gov identifier: NCT04145596).Key MessageHSK3486 at an IV bolus dose of 0.4 mg/kg and a maintenance infusion of 0.4 mg/kg/h was safe and well tolerated by all mild or moderate hepatic impairment subjects and normal hepatic function subjects.There were no clinically relevant differences in HSK3486 exposure or PD in subjects with mild or moderate hepatic impairment compared to subjects with normal hepatic function.HSK3486 dose adjustment is not required in subjects with mild or moderate hepatic impairment.
Collapse
Affiliation(s)
- Yue Hu
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Jilin, China
| | - Xiaojiao Li
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Jilin, China
| | - Jingrui Liu
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Jilin, China
| | - Hong Chen
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Jilin, China
| | - Wenbo Zheng
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Jilin, China
| | - Hong Zhang
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Jilin, China
| | - Min Wu
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Jilin, China
| | - Cuiyun Li
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Jilin, China
| | - Xiaoxue Zhu
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Jilin, China
| | - Jinfeng Lou
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Jilin, China
| | - Pangke Yan
- Haisco Pharmaceutical Group, Sichuan, China
| | - Nan Wu
- Haisco Pharmaceutical Group, Sichuan, China
| | - Xiao Liu
- Haisco Pharmaceutical Group, Sichuan, China
| | - Shiping Ma
- Haisco Pharmaceutical Group, Sichuan, China
| | - Xu Wang
- Haisco Pharmaceutical Group, Sichuan, China
| | - Yanhua Ding
- Phase I Clinical Trial Unit, First Hospital, Jilin University, Jilin, China
| | - Chengluan Xuan
- Department of Anesthesiology, First Hospital, Jilin University, Jilin, China
| |
Collapse
|
11
|
Topletz-Erickson AR, Lee AJ, Mayor JG, Sun H, Abdulrasool LI, Rustia EL, Walker LN, Endres CJ. The Pharmacokinetics and Safety of Tucatinib in Volunteers with Hepatic Impairment. Clin Pharmacokinet 2022; 61:1761-1770. [PMID: 36471222 PMCID: PMC9734226 DOI: 10.1007/s40262-022-01183-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND OBJECTIVE Tucatinib, a highly selective tyrosine kinase inhibitor of the human epidermal growth factor receptor 2 (HER2) approved for HER2-positive metastatic breast cancer, is cleared by hepatic metabolism and subsequent biliary excretion. Liver disease can alter drug disposition and pharmacokinetics (PK). The objective of this study is to characterize PK and safety of tucatinib in volunteers with hepatic impairment. METHODS This Phase 1 study compared the PK and safety of a single 300-mg oral dose of tucatinib in volunteers with mild, moderate, and severe hepatic impairment (Child-Pugh A/B/C) to healthy volunteers matched for sex, age, and body mass index. Pharmacokinetic parameters were determined for tucatinib and its predominant metabolite ONT-993. RESULTS Compared with healthy volunteers, tucatinib exposure was similar in volunteers with mild impairment and increased in those with moderate or severe impairment without reaching statistical significance. Respective fold increases in geometric mean ratios for AUC0-t and AUC0-∞ were 1.13 and 1.15 in moderate impairment, and 1.43 and 1.61 in severe impairment compared with healthy volunteers. Three treatment-emergent adverse events (nausea, dermatitis, and increased transaminases) were reported in three volunteers and showed no obvious association with hepatic impairment status. CONCLUSION The 1.61-fold geometric mean ratio AUC0-∞ increase in volunteers with severe hepatic impairment supports the recommendation in the tucatinib prescribing information to reduce the dose from 300 mg twice daily to 200 mg twice daily in patients with severe impairment; no dose adjustment is recommended for patients with mild or moderate hepatic impairment. This trial (NCT03722823) was registered on October 29, 2018.
Collapse
Affiliation(s)
- Ariel R Topletz-Erickson
- Clinical Pharmacology and Pharmacometrics, Seagen Inc., 21823 30th Drive SE, Bothell, WA, 98021, USA
| | - Anthony J Lee
- Translational ADME and PKPD, Seagen Inc., Bothell, WA, USA
| | - JoAl G Mayor
- Clinical Development, Seagen Inc., Bothell, WA, USA
| | - Hao Sun
- Translational ADME and PKPD, Seagen Inc., Bothell, WA, USA
| | | | - Evelyn L Rustia
- Clinical Development, Seagen Inc., Bothell, WA, USA
- Gilead Sciences, Seattle, WA, USA
| | - Luke N Walker
- Clinical Development, Seagen Inc., Bothell, WA, USA
- Harpoon Therapeutics, South San Francisco, CA, USA
| | - Christopher J Endres
- Clinical Pharmacology and Pharmacometrics, Seagen Inc., 21823 30th Drive SE, Bothell, WA, 98021, USA.
| |
Collapse
|
12
|
A review of the safety of buprenorphine in special populations. Am J Med Sci 2022; 364:675-684. [PMID: 35843298 DOI: 10.1016/j.amjms.2022.06.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/17/2022] [Accepted: 06/29/2022] [Indexed: 01/25/2023]
Abstract
Rates of opioid misuse and opioid use disorder have been increasing in recent years. Buprenorphine has emerged as an appealing medication for its use not only as treatment for opioid use disorder, but also as an opioid for chronic pain that has a ceiling effect on risks associated with opioid therapy. As other opioid prescribing decreases, buprenorphine prescribing continues to increase. As a result, it is imperative to understand the safety and efficacy of its use in special populations. This review article will explore the safety and efficacy of buprenorphine when used in subjects with hepatic and renal impairment, the elderly, and pregnant women. While manufacturer labeling for buprenorphine products may caution against their use in these populations, further examination of available data indicates that buprenorphine can be used safely and effectively for both chronic pain and/or opioid use disorder in all four of these populations.
Collapse
|
13
|
A Comprehensive Review on the Use of Herbal Dietary Supplements in the USA, Reasons for Their Use, and Review of Potential Hepatotoxicity. LIVERS 2022. [DOI: 10.3390/livers2030011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Herbal and dietary supplement (HDS) use has grown exponentially in the United States. Unfortunately, the incidence of HDS-related liver injury has proportionally increased. Despite the potential for certain HDSs to cause clinically significant liver injury, they are not regulated by the Food and Drug Administration. Recent efforts have been made to regulate HDSs but are far removed from the scrutiny of prescription medications. Scant literature exists on HDSs and their risks of causing liver injury. In this comprehensive review, we examine trends of HDS use in the United States and the pathophysiologic mechanisms of drug-induced liver injury (DILI) of certain HDSs. Finally, we review usage rates; benefits, if any; purported pathophysiology of DILI; and propensity for progression to fulminant hepatic failure of nine HDSs linked to clinically significant DILI.
Collapse
|
14
|
Park J, Kim CO, Oh ES, Lee JI, Kim JK, Ahn SH, Kim DY, Kim SU, Kim BK, Chung YE, Kim SM, Park MS. Effects of Hepatic Impairment on the Pharmacokinetic Profile and Safety of Lobeglitazone. Clin Pharmacol Drug Dev 2022; 11:576-584. [PMID: 35255191 DOI: 10.1002/cpdd.1045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/10/2021] [Indexed: 11/09/2022]
Abstract
In this open-label, single-dose, parallel-group study, we compared the pharmacokinetic profile and safety of lobeglitazone, a thiazolidinedione acting as an agonist for peroxisome proliferator-activated receptors, in patients with hepatic impairment (HI) and healthy matched controls for age, sex, and body weight. After a single oral dose of lobeglitazone (0.5 mg), the lobeglitazone (parent drug) and M7 (major metabolite) plasma concentrations and pharmacokinetic parameters were analyzed and compared between the HI patient groups and healthy matched control groups. The geometric mean ratio (GMR; 90% confidence interval [CI]) for maximum concentration (Cmax ) and area under the plasma concentration-time curve from time 0 extrapolated to infinity (AUCinf ) of lobeglitazone was 1.06 (0.90-1.24) and 1.07 (0.82-1.40), respectively, for mild HI vs control A. The GMR (90%CI) of Cmax and AUCinf was 0.70 (0.56-0.88) and 1.00 (0.72-1.37), respectively, for moderate HI vs control B. For M7, the GMR (90%CI) of Cmax and AUCinf was 1.09 (0.75-1.57) and 1.18 (0.71-1.97), respectively, for mild HI vs control A and 1.50 (0.95-2.38) and 1.79 (1.06-3.04), respectively, for moderate HI vs control B. Notable adverse events or tolerability issues were not observed. Lobeglitazone may be safely used in patients with mild or moderate HI without dose adjustment.
Collapse
Affiliation(s)
- Jungsin Park
- Department of Pharmaceutical Medicine and Regulatory Science, Colleges of Medicine and Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Choon Ok Kim
- Department of Clinical Pharmacology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Sil Oh
- Department of Pharmaceutical Medicine and Regulatory Science, Colleges of Medicine and Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Jung Il Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ja Kyung Kim
- Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Gyeonggi-do, Republic of Korea
| | - Sang Hoon Ahn
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Do Young Kim
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Up Kim
- Yonsei Liver Centre, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Beom Kyung Kim
- Yonsei Liver Centre, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong Eun Chung
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Se-Mi Kim
- Department of Nonclinical Development, Chong Kun Dang Research Institute, Chong Kun Dang Pharmaceutical Corp., Ltd, Seoul, Republic of Korea
| | - Min Soo Park
- Department of Pharmaceutical Medicine and Regulatory Science, Colleges of Medicine and Pharmacy, Yonsei University, Incheon, Republic of Korea
- Department of Clinical Pharmacology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
15
|
Duong JK, Nand RA, Patel A, Della Pasqua O, Gross AS. A physiologically based pharmacokinetic model of clopidogrel in populations of European and Japanese ancestry: An evaluation of CYP2C19 activity. Pharmacol Res Perspect 2022; 10:e00946. [PMID: 35307978 PMCID: PMC8934724 DOI: 10.1002/prp2.946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 11/21/2022] Open
Abstract
Treatment response to clopidogrel is associated with CYP2C19 activity through the formation of the active H4 metabolite. The aims of this study were to develop a physiologically based pharmacokinetic (PBPK) model of clopidogrel and its metabolites for populations of European ancestry, to predict the pharmacokinetics in the Japanese population by CYP2C19 phenotype, and to investigate the effect of clinical and demographic factors. A PBPK model was developed and verified to describe the two metabolic pathways of clopidogrel (H4 metabolite, acyl glucuronide metabolite) for a population of European ancestry using plasma data from published studies. Subsequently, model predictions in the Japanese population were evaluated. The effects of CYP2C19 activity, fluvoxamine coadministration (CYP2C19 inhibitor), and population-specific factors (age, sex, BMI, body weight, cancer, hepatic, and renal dysfunction) on the pharmacokinetics of clopidogrel and its metabolites were then characterized. The predicted/observed ratios for clopidogrel and metabolite exposure parameters were acceptable (twofold acceptance criteria). For all CYP2C19 phenotypes, steady-state AUC0-τ of the H4 metabolite was lower for the Japanese (e.g., EM, 7.69 [6.26-9.45] ng·h/ml; geometric mean [95% CI]) than European (EM, 24.8 [20.4-30.1] ng·h/ml, p < .001) population. In addition to CYP2C19-poor metabolizer phenotype, fluvoxamine coadministration, hepatic, and renal dysfunction were found to reduce H4 metabolite but not acyl glucuronide metabolite concentrations. This is the first PBPK model describing the two major metabolic pathways of clopidogrel, which can be applied to populations of European and Japanese ancestry by CYP2C19 phenotype. The differences between the two populations appear to be determined primarily by the effect of varying CYP2C19 liver activity.
Collapse
Affiliation(s)
- Janna K. Duong
- Clinical Pharmacology Modelling and SimulationGlaxoSmithKline R&DErmingtonAustralia
| | - Romina A. Nand
- Clinical Pharmacology Modelling and SimulationGlaxoSmithKline R&DErmingtonAustralia
| | - Aarti Patel
- Drug Metabolism and PharmacokineticsGlaxoSmithKline R&DStevenageUK
| | - Oscar Della Pasqua
- Clinical Pharmacology Modelling and SimulationGlaxoSmithKline R&DBrentfordUK
| | - Annette S. Gross
- Clinical Pharmacology Modelling and SimulationGlaxoSmithKline R&DErmingtonAustralia
| |
Collapse
|
16
|
Monroy-Jaramillo N, Martínez-Magaña JJ, Pérez-Aldana BE, Ortega-Vázquez A, Montalvo-Ortiz J, López-López M. The role of alcohol intake in the pharmacogenetics of treatment with clozapine. Pharmacogenomics 2022; 23:371-392. [PMID: 35311547 DOI: 10.2217/pgs-2022-0006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Clozapine (CLZ) is an atypical antipsychotic reserved for patients with refractory psychosis, but it is associated with a significant risk of severe adverse reactions (ADRs) that are potentiated with the concomitant use of alcohol. Additionally, pharmacogenetic studies have explored the influence of several genetic variants in CYP450, receptors and transporters involved in the interindividual response to CLZ. Herein, we systematically review the current multiomics knowledge behind the interaction between CLZ and alcohol intake, and how its concomitant use might modulate the pharmacogenetics. CYP1A2*1F, *1C and other alleles not yet discovered could support a precision medicine approach for better therapeutic effects and fewer CLZ ADRs. CLZ monitoring systems should be amended and include alcohol intake to protect patients from severe CLZ ADRs.
Collapse
Affiliation(s)
- Nancy Monroy-Jaramillo
- Department of Genetics, National Institute of Neurology & Neurosurgery, Manuel Velasco Suárez, La Fama, Tlalpan, Mexico City, 14269, Mexico
| | - José Jaime Martínez-Magaña
- Department of Psychiatry, Division of Human Genetics, Yale University School of Medicine, Orange, West Haven, CT 06477, USA
| | - Blanca Estela Pérez-Aldana
- Doctorado en Ciencias Biológicas y de la Salud, Metropolitan Autonomous University, Campus Xochimilco, Villa Quietud, Coyoacán, Mexico City, 04960, Mexico
| | - Alberto Ortega-Vázquez
- Metropolitan Autonomous University, Campus Xochimilco, Villa Quietud, Coyoacán, Mexico City, 04960, Mexico
| | - Janitza Montalvo-Ortiz
- Department of Psychiatry, Division of Human Genetics, Yale University School of Medicine, Orange, West Haven, CT 06477, USA
| | - Marisol López-López
- Metropolitan Autonomous University, Campus Xochimilco, Villa Quietud, Coyoacán, Mexico City, 04960, Mexico
| |
Collapse
|
17
|
Abstract
Background and Objective Dacomitinib is a kinase inhibitor indicated for the first-line treatment of patients with metastatic non-small cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR)-activating mutations. To evaluate the effect of hepatic impairment on the pharmacokinetics of dacomitinib, two dedicated studies were conducted to inform optimal dosing. Methods Study 1 (NCT01571388) evaluated the effect of mild and moderate hepatic impairment on the plasma pharmacokinetics, safety, and tolerability after a single oral dose of dacomitinib 30 mg, and Study 2 (NCT03865446) evaluated the same endpoints in a severe hepatic impairment population. Both studies were phase I, open-label, parallel-group studies. A one-way analysis of variance (ANOVA) with unequal variance assumption and hepatic impairment group as a fixed effect was used to compare the natural log of area under the plasma concentration-time curve extrapolated to infinite time (AUCinf), AUC from time zero to the last quantifiable concentration (AUClast), and maximum plasma concentration (Cmax) for each hepatic impairment group to the respective normal hepatic function group. Since dacomitinib is a cytochrome P450 (CYP) 2D6 substrate, only participants with extensive or intermediate CYP2D6 phenotypes were included in the primary analysis. Results The AUCinf for participants with mild, moderate, or severe hepatic impairment decreased by 6%, decreased by 23%, and increased by 4%, respectively, compared with normal hepatic function, while the Cmax for participants with mild, moderate, or severe hepatic impairment increased by 3%, decreased by 20%, and increased by 31%, respectively, compared with normal hepatic function. A single oral dose of dacomitinib 30 mg was well tolerated in all participants. Conclusion Based on these pharmacokinetic results, dacomitinib pharmacokinetics of participants with mild, moderate, or severe hepatic impairment were not statistically different relative to participants with normal hepatic function based on the ANOVA analysis. No dacomitinib dose adjustments for patients with hepatic impairment are recommended. Clinical Trial Registration ClinicalTrials.gov NCT01571388, registered 5 April 2012; ClinicalTrials.gov NCT03865446, registered 6 March 2019. Supplementary Information The online version contains supplementary material available at 10.1007/s40261-022-01125-x.
Collapse
|
18
|
Montanha MC, Cottura N, Booth M, Hodge D, Bunglawala F, Kinvig H, Grañana-Castillo S, Lloyd A, Khoo S, Siccardi M. PBPK Modelling of Dexamethasone in Patients With COVID-19 and Liver Disease. Front Pharmacol 2022; 13:814134. [PMID: 35153785 PMCID: PMC8832977 DOI: 10.3389/fphar.2022.814134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/11/2022] [Indexed: 11/13/2022] Open
Abstract
The aim of the study was to apply Physiologically-Based Pharmacokinetic (PBPK) modelling to predict the effect of liver disease (LD) on the pharmacokinetics (PK) of dexamethasone (DEX) in the treatment of COVID-19. A whole-body PBPK model was created to simulate 100 adult individuals aged 18–60 years. Physiological changes (e.g., plasma protein concentration, liver size, CP450 expression, hepatic blood flow) and portal vein shunt were incorporated into the LD model. The changes were implemented by using the Child-Pugh (CP) classification system. DEX was qualified using clinical data in healthy adults for both oral (PO) and intravenous (IV) administrations and similarly propranolol (PRO) and midazolam (MDZ) were qualified with PO and IV clinical data in healthy and LD adults. The qualified model was subsequently used to simulate a 6 mg PO and 20 mg IV dose of DEX in patients with varying degrees of LD, with and without shunting. The PBPK model was successfully qualified across DEX, MDZ and PRO. In contrast to healthy adults, the simulated systemic clearance of DEX decreased (35%–60%) and the plasma concentrations increased (170%–400%) in patients with LD. Moreover, at higher doses of DEX, the AUC ratio between healthy/LD individuals remained comparable to lower doses. The exposure of DEX in different stages of LD was predicted through PBPK modelling, providing a rational framework to predict PK in complex clinical scenarios related to COVID-19. Model simulations suggest dose adjustments of DEX in LD patients are not necessary considering the low dose administered in the COVID-19 protocol.
Collapse
|
19
|
Zoratti C, Moretti R, Rebuzzi L, Albergati IV, Di Somma A, Decorti G, Di Bella S, Crocè LS, Giuffrè M. Antibiotics and Liver Cirrhosis: What the Physicians Need to Know. Antibiotics (Basel) 2021; 11:31. [PMID: 35052907 PMCID: PMC8772826 DOI: 10.3390/antibiotics11010031] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 12/13/2022] Open
Abstract
The liver is the primary site of drug metabolism, which can be altered by a variety of diseases affecting the liver parenchyma, especially in patients with liver cirrhosis. The use of antibiotics in patients with cirrhosis is usually a matter of concern for physicians, given the lack of practical knowledge for drug choice and eventual dose adjustments in several clinical scenarios. The aim of the current narrative review is to report, as broadly as possible, basic, and practical knowledge that any physician should have when approaching a patient with liver cirrhosis and an ongoing infection to efficiently choose the best antibiotic therapy.
Collapse
Affiliation(s)
- Caterina Zoratti
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy; (C.Z.); (R.M.); (L.R.); (I.V.A.); (A.D.S.); (S.D.B.); (L.S.C.)
| | - Rita Moretti
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy; (C.Z.); (R.M.); (L.R.); (I.V.A.); (A.D.S.); (S.D.B.); (L.S.C.)
| | - Lisa Rebuzzi
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy; (C.Z.); (R.M.); (L.R.); (I.V.A.); (A.D.S.); (S.D.B.); (L.S.C.)
| | - Irma Valeria Albergati
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy; (C.Z.); (R.M.); (L.R.); (I.V.A.); (A.D.S.); (S.D.B.); (L.S.C.)
| | - Antonietta Di Somma
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy; (C.Z.); (R.M.); (L.R.); (I.V.A.); (A.D.S.); (S.D.B.); (L.S.C.)
| | - Giuliana Decorti
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, 34137 Trieste, Italy;
| | - Stefano Di Bella
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy; (C.Z.); (R.M.); (L.R.); (I.V.A.); (A.D.S.); (S.D.B.); (L.S.C.)
| | - Lory Saveria Crocè
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy; (C.Z.); (R.M.); (L.R.); (I.V.A.); (A.D.S.); (S.D.B.); (L.S.C.)
- Italian Liver Foundation, 34149 Trieste, Italy
| | - Mauro Giuffrè
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy; (C.Z.); (R.M.); (L.R.); (I.V.A.); (A.D.S.); (S.D.B.); (L.S.C.)
- Italian Liver Foundation, 34149 Trieste, Italy
| |
Collapse
|
20
|
Abstract
Antifungals are effective antimicrobial agents broadly used in medical practice. Severe acute liver failure from oral or IV administration of antifungals is a rare but long-standing clinical challenge. We aimed to approximate the risk of clinical acute liver injury among users of oral antifungals in the general population. This review was completed based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Six articles were included, comprising case reports and cohort studies, after eliminating duplicate publications. No randomized control studies were found. In all studies, the duration of antifungal use was associated with significantly increased liver enzyme levels. Although it is not very common for patients on antifungals to develop acute liver failure, the prognosis is often good with swift discontinuation of the drug and proper treatment. Liver function evaluation before treatment and periodic monitoring every three to six weeks after commencement of treatment is suggested.
Collapse
Affiliation(s)
- Eyad Gadour
- Gastroenterology and Hepatology, University Hospitals of Morecambe Bay National Health Service (NHS) Foundation Trust, Lancaster, GBR
| | - Ahmed Kotb
- Hepatobiliary Surgery, Glan Clwyd Hospital, Wales, GBR
| |
Collapse
|
21
|
Wu X, Zhang X, Xu R, Shaik IH, Venkataramanan R. Physiologically based pharmacokinetic modelling of treprostinil after intravenous injection and extended-release oral tablet administration in healthy volunteers: An extrapolation to other patient populations including patients with hepatic impairment. Br J Clin Pharmacol 2021; 88:587-599. [PMID: 34190364 PMCID: PMC9290939 DOI: 10.1111/bcp.14966] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 11/30/2022] Open
Abstract
AIMS Pulmonary arterial hypertension (PAH) is characterized by increased pulmonary arterial pressure, resulting in right ventricular overload, right heart failure and eventually death. Treprostinil is a prostacyclin analogue that is used in the treatment of PAH. As an orphan drug, limited information is available regarding its disposition and its use in special populations such as elderly, paediatric and pregnant patients. The objective of the current study was to develop a robust physiologically based pharmacokinetic (PBPK) model for treprostinil intravenous injection and extended-release tablet as the first step to optimize treprostinil pharmacotherapy in patients. METHODS PBPK model was built using Simcyp simulator which integrated physicochemical properties, observed or predicted parameters for drug absorption, distribution and elimination for treprostinil, and population specific physiological characteristics. Three clinical trials after intravenous infusion and nine studies after oral administration of treprostinil extended-release tablet in healthy volunteers were used to develop and validate the model. The simulated PK profiles were compared with the observed data. Extrapolation of the model to patient populations including patients with hepatic impairment was conducted to validate the predictions. RESULTS Most of the observed data were within the 5th and 95th percentile interval of the prediction. Most of the percentage error in the PK parameters were within ±50% of the corresponding observed parameters. The developed model predicted the lung exposure of treprostinil to be approximately 0.17 times of concentration in plasma. CONCLUSION Predicted absorption, distribution, and metabolic enzyme kinetics gave an insight into the disposition of treprostinil in humans. Extrapolation of the established model to patient populations with hepatic impairment successfully documented the model reliability. The developed model has the potential to be used in the PK predictions in other special patient populations with different demographic, physiological and pathological characteristics.
Collapse
Affiliation(s)
- Xuemei Wu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Xiaohan Zhang
- College of Arts and Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - Ruichao Xu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Imam Hussain Shaik
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Raman Venkataramanan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA.,Thomas Starzl Transplantation Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
22
|
Trivedi A, Oberoi RK, Mackowski M, Jafarinasabian P, Zhang H, Flach S, Hutton S, Abbasi S, Dutta S, Lee E. Effect of Varying Degrees of Hepatic Impairment on the Pharmacokinetics of Omecamtiv Mecarbil. Clin Pharmacol Drug Dev 2021; 10:1442-1451. [PMID: 34107172 DOI: 10.1002/cpdd.969] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/03/2021] [Indexed: 12/13/2022]
Abstract
Omecamtiv mecarbil (OM) is a novel selective cardiac myosin activator under investigation for the treatment of heart failure with reduced ejection fraction. OM is primarily eliminated via metabolism mediated by multiple cytochrome P450 enzymes. This phase 1 single-dose, multicenter, open-label, nonrandomized study evaluated the pharmacokinetics (PK) of OM and major metabolites M3 and M4, safety, and tolerability following oral administration of a single dose of 25-mg MR tablet in subjects with mild (n = 6) or moderate (n = 6) hepatic impairment (according to Child-Pugh classification) versus subjects with normal hepatic function (n = 6). Relative to subjects with normal hepatic function, for subjects with mild or moderate hepatic impairment, OM AUCinf was 103.2% (90%CI, 58.0%-183.6%) and 94.8% (90%CI, 54.7%-164.1%), respectively, and OM Cmax was 126.8% (90%CI, 85.7%-187.7%) and 117.3% (90%CI, 80.7%-170.5%), respectively. Exposures to M3 were similar across groups, whereas slightly lower exposures were observed for M4 with worsening hepatic function. The OM, M3, and M4 tmax and t1/2 values were similar between groups. There were no serious adverse events (AEs) or treatment-related treatment-emergent AEs. Overall, OM, M3, and M4 PK were not meaningfully affected by mild or moderate hepatic impairment, suggesting the same dosing strategy can be used in subjects with mild or moderate hepatic impairment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Edward Lee
- Amgen Inc., Thousand Oaks, California, USA
| |
Collapse
|
23
|
Miura M, Tanaka S, Uchida S, Kamiya C, Katayama N, Hakamata A, Odagiri K, Inui N, Kawakami J, Watanabe H, Namiki N. Prediction of the Area under the Curve Using Limited-Point Blood Sampling in a Cocktail Study to Assess Multiple CYP Activities. Biol Pharm Bull 2021; 44:762-770. [PMID: 34078808 DOI: 10.1248/bpb.b20-00691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A cocktail study is an in vivo evaluation method to assess multiple CYP activities via a single trial and single administration of a cocktail drug that is a combination of multiple CYP substrates. However, multiple blood samples are required to evaluate the pharmacokinetics of a CYP probe drug. A limited-point sampling method is generally beneficial in clinical studies because of the simplified protocol and reduced participant burden. The aim of this study was to evaluate whether a limited-point plasma concentration analysis of CYP substrates in a cocktail drug could predict their area under the curve (AUC). We created prediction models of five CYP substrates (caffeine, losartan, omeprazole, dextromethorphan, and midazolam) using multiple linear regressions from the data of two cocktail studies, and then performed predictability analysis of these models using data derived from data in the co-administration with inducer (rifampicin) and inhibitors (fluvoxamine and cimetidine). For the administration of inhibitors, the AUC prediction accuracy (mean absolute error (MAE)) were <39.5% in Model 1 and <26.2% in Model 2 which were created using 1- and 4-point sampling data. MAE shows larger values in the administration of inducer in compared with the administration of inhibitors. The accuracy of the prediction in Model 2 could be acceptable for screening of inhibitions. MAE for caffeine, dextromethorphan, and midazolam were acceptable in the model that used 4 sampling points from all data. The use of this method could reduce the burden on the subject and make it possible to evaluate each AUC in a minimally invasive manner.
Collapse
Affiliation(s)
- Motoyasu Miura
- Department of Pharmacy Practice & Science, School of Pharmaceutical Sciences, University of Shizuoka.,Hospital Pharmacy, Hamamatsu University School of Medicine
| | - Shimako Tanaka
- Department of Pharmacy Practice & Science, School of Pharmaceutical Sciences, University of Shizuoka
| | - Shinya Uchida
- Department of Pharmacy Practice & Science, School of Pharmaceutical Sciences, University of Shizuoka
| | - Chiaki Kamiya
- Department of Clinical Pharmacology & Therapeutics, Hamamatsu University School of Medicine
| | - Naoki Katayama
- Department of Clinical Pharmacology & Therapeutics, Hamamatsu University School of Medicine
| | - Akio Hakamata
- Department of Clinical Pharmacology & Therapeutics, Hamamatsu University School of Medicine
| | - Keiichi Odagiri
- Department of Clinical Pharmacology & Therapeutics, Hamamatsu University School of Medicine
| | - Naoki Inui
- Department of Clinical Pharmacology & Therapeutics, Hamamatsu University School of Medicine
| | | | - Hiroshi Watanabe
- Department of Clinical Pharmacology & Therapeutics, Hamamatsu University School of Medicine
| | - Noriyuki Namiki
- Department of Pharmacy Practice & Science, School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
24
|
CFTR Modulators: Does One Dose Fit All? J Pers Med 2021; 11:jpm11060458. [PMID: 34073663 PMCID: PMC8224731 DOI: 10.3390/jpm11060458] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 12/11/2022] Open
Abstract
For many people with cystic fibrosis (pwCF), CFTR modulators will be the cornerstone of their treatment. These modulators show robust treatment effects at group level in pwCF with specific mutations. The individual effect however, is variable. In this review we will explain reasons for reconsideration of dosing regimens of CFTR modulating therapy in order to improve treatment response and prevent side effects. Since the effect of a drug depends on pharmacodynamics and pharmacokinetics, pharmacodynamics and pharmacokinetic properties of CFTR modulators will be discussed. Pharmacokinetic-pharmacodynamic relationships will be used to gain insight in dosage response and exposure response relationships. To understand the cause of variation in drug exposure, pharmacokinetic properties that may change due to CF disease will be explained. We show that with current insight, there are conceivable situations that give reason for reconsideration of dosing regimens, however many questions need to be unravelled.
Collapse
|
25
|
Trifu S, Țîbîrnă A, Costea RV, Popescu A. A multidisciplinary approach to the management of liver disease and alcohol disorders in psychiatric settings (Review). Exp Ther Med 2021; 21:271. [PMID: 33603878 PMCID: PMC7851668 DOI: 10.3892/etm.2021.9702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 11/24/2020] [Indexed: 11/19/2022] Open
Abstract
Society is burdened with the uncontrolled use of alcohol, an ongoing issue, with a substantial associated morbidity and a pressing economical reverberation. It is inevitable that a series of psychiatric patients who display alcohol disorders will be admitted to hospital while also suffering from health conditions, such as liver disease, due to the consumption of alcohol. Managing comorbid patients in a psychiatric facility is a delicate matter that requires a collaborative team. The aim of this systematic paper is to highlight the following: The possibility of treating alcohol use disorder (AUD) and alcohol withdrawal syndrome (AWS) overlapping alcohol liver disease (ALD) within a psychiatric institution, and the importance of a collaborative multidisciplinary team; correctly dosing psychoactive medication when metabolism is affected by ALD; deciding when is it necessary to seek a transfer to a general hospital. Prescribing medication in patients suffering from ALD is still a not a fully documented territory. Protein binding, metabolism, bioavailability, extraction ratios, excretion route, and half-life must be taken into consideration as well as frequently repeating liver panels. Studies suggest that short-acting benzodiazepines are preferred over their alternatives when treating AWS in ALD. All anticonvulsants can be used in patients with decompensated liver disease with caution, although newer generation antiepileptic agents should be first line. Propofol is favored to benzodiazepines or opioids in the case of decompensated cirrhosis. Patients with ALD are likely to be further compromised by the potential hepatocytotoxicity of some pharmacological agents. On that account, having an integrated perspective of the medical case while taking into consideration the underlying illness as well as possible drug interaction is crucial in treating AUD or AWS in a psychiatric institution.
Collapse
Affiliation(s)
- Simona Trifu
- Department of Clinical Neurosciences, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Andrian Țîbîrnă
- Department of Psychiatry, 'Alex. Obregia' Clinical Hospital for Psychiatry, 041914 Bucharest, Romania
| | - Radu-Virgil Costea
- Department of General Surgery, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Alexandra Popescu
- Department of Psychiatry, 'Alex. Obregia' Clinical Hospital for Psychiatry, 041914 Bucharest, Romania
| |
Collapse
|
26
|
Hong T, Jin BH, Kim CO, Yoo BW, Kim D, Lee JI, Kim BK, Ahn SH, Kim DY, Park JY, Park MS. Pharmacokinetics and safety of evogliptin in hepatically impaired patients. Br J Clin Pharmacol 2020; 87:2757-2766. [PMID: 33245796 DOI: 10.1111/bcp.14680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 12/17/2022] Open
Abstract
AIMS Evogliptin is a potent and selective dipeptidyl peptidase-4 inhibitor for glycaemic control in patients with type 2 diabetes mellitus. Since evogliptin is mainly eliminated through hepatic metabolism, we investigated the pharmacokinetics (PKs) and safety characteristics of evogliptin in Korean patients with mild or moderate hepatic impairment. METHODS An open-label, parallel-group study was conducted in patients with mild or moderate hepatic impairment and healthy control subjects matched to each patient for sex, age and body mass index. A single dose (5 mg) of evogliptin was administered orally, and serial blood samples were collected over 120 h to assess the PK profile of evogliptin and its main metabolites (M7 and M8). RESULTS Patients with mild hepatic impairment and their matched healthy controls showed similar maximum concentration (Cmax ) and area under the concentration-time curve values from 0 to 120 h (AUClast ); the geometric mean ratio (GMR) and 90% confidence interval (CI) were 1.04 (0.80, 1.35) and 1.01 (0.90, 1.14), respectively. Exposure to evogliptin (Cmax and AUClast ) was increased by about 40% in patients with moderate hepatic impairment-the GMR and 90% CI were 1.37 (1.09, 1.72) and 1.44 (1.18, 1.75), respectively. The metabolic ratios of M7 and M8 were lower in patients with moderate hepatic impairment than in matched healthy controls. Evogliptin was well tolerated by both patients and healthy subjects. CONCLUSION Although evogliptin exposure was increased in patients with moderate hepatic impairment, the increase is unlikely to affect safety and efficacy adversely, and no dose adjustment is warranted.
Collapse
Affiliation(s)
- Taegon Hong
- Department of Clinical Pharmacology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Byung Hak Jin
- Department of Clinical Pharmacology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea.,Department of Pharmaceutical Medicine and Regulatory Science, Colleges of Medicine and Pharmacy, Yonsei University, Incheon, South Korea
| | - Choon Ok Kim
- Department of Clinical Pharmacology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Byung Won Yoo
- Department of Clinical Pharmacology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Dasohm Kim
- Department of Clinical Pharmacology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea.,Department of Pharmaceutical Medicine and Regulatory Science, Colleges of Medicine and Pharmacy, Yonsei University, Incheon, South Korea
| | - Jung Il Lee
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Beom Kyung Kim
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Hoon Ahn
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Do Young Kim
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Jun Yong Park
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Min Soo Park
- Department of Clinical Pharmacology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea.,Department of Pharmaceutical Medicine and Regulatory Science, Colleges of Medicine and Pharmacy, Yonsei University, Incheon, South Korea.,Department of Pediatrics, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
27
|
Hayward KL, Weersink RA. Improving Medication-Related Outcomes in Chronic Liver Disease. Hepatol Commun 2020; 4:1562-1577. [PMID: 33163829 PMCID: PMC7603526 DOI: 10.1002/hep4.1612] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/20/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022] Open
Abstract
Patients with chronic liver disease (CLD) are becoming increasingly complex due to the rising prevalence of multimorbidity and polypharmacy. Medications are often essential to manage the underlying liver disease, complications of cirrhosis and portal hypertension, and comorbidities. However, medication-related problems (MRPs) have been associated with adverse patient outcomes, including hospitalization and mortality. Factors that can contribute to MRPs in people with CLD are variable and often entwined. This narrative literature review discusses key barriers and opportunities to modify risk factors and improve medication-related outcomes for people with CLD.
Collapse
Affiliation(s)
- Kelly L Hayward
- Centre for Liver Disease Research, Faculty of Medicine The University of Queensland, Translational Research Institute Brisbane QLD Australia.,Department of Gastroenterology and Hepatology Princess Alexandra Hospital Brisbane QLD Australia
| | - Rianne A Weersink
- Department of Clinical Pharmacy Deventer Hospital Deventer The Netherlands
| |
Collapse
|
28
|
De Sutter PJ, Gasthuys E, Van Braeckel E, Schelstraete P, Van Biervliet S, Van Bocxlaer J, Vermeulen A. Pharmacokinetics in Patients with Cystic Fibrosis: A Systematic Review of Data Published Between 1999 and 2019. Clin Pharmacokinet 2020; 59:1551-1573. [DOI: 10.1007/s40262-020-00932-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
29
|
Relationship Between Change Rate of Tacrolimus Clearance During Continuous Intravenous Infusion and Recipient Recovery at an Early Stage After Living Donor Liver Transplantation. Eur J Drug Metab Pharmacokinet 2020; 45:619-626. [PMID: 32514937 DOI: 10.1007/s13318-020-00628-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND AND OBJECTIVE Tacrolimus clearance (CL) is significantly altered according to recovery of liver function at an early stage after living donor liver transplantation (LDLT). In this study, we aimed to examine the impact of the change rate from postoperative day (POD) 1 in CL (ΔCL) of tacrolimus during continuous intravenous infusion (CIVI) on recipient recovery. METHODS A tacrolimus population pharmacokinetic model on POD1 after LDLT was developed using Phoenix NLME 1.3. The CLPOD1 was calculated using the final model. The CLPOD4-7 was calculated by dividing total daily tacrolimus dose by the area under the concentration-time curve from 0 to 24 h. RESULTS Data were obtained from 57 LDLT recipients, along with 540 points (177 points on POD1, 363 points on POD4-7) of tacrolimus whole blood concentrations at CIVI. The median tacrolimus CL decreased from POD1 to POD4 (from 2.73 to 1.40 L/h) and was then stable until POD7. Stepwise Cox proportional hazards regression analyses showed that the graft volume (GV)/standard liver volume (SLV) ratio (GV/SLV) and the tacrolimus ΔCLPOD6 were independent factors predicting early discharge (within 64 days median value) of recipients after LDLT [hazard ratio (HR) = 1.041, P = 0.001 and HR = 1.023, P = 0.004]. CONCLUSIONS The tacrolimus ΔCL during CIVI immediately after LDLT in each recipient was a useful indicator for evaluation of recovery at an early stage after LDLT.
Collapse
|
30
|
Alkharfy KM, Ali FA, Alkharfy MA, Jan BL, Raish M, Alqahtani S, Ahmad A. Effect of compromised liver function and acute kidney injury on the pharmacokinetics of thymoquinone in a rat model. Xenobiotica 2020; 50:858-862. [PMID: 32216504 DOI: 10.1080/00498254.2020.1745319] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The current research explored the effect of hepatic and renal dysfunctions on the pharmacokinetics of thymoquinone (TQ) in a rat model.An acute kidney injury was induced using gentamicin and a liver damage was elicited using a single dose of d-galactosamine. For the pharmacokinetic studies, TQ was administered as IV injection or and PO route to rats.The concentrations of TQ and pharmacokinetic parameters were calculated using a non-compartmental analysis. The systemic clearance (Cl) of TQ after IV dosing was slightly reduced in the liver dysfunction group compared to healthy controls (p = 0.0013). Similarly, the estimated volume of distribution at steady state (Vss) was marginally decreased (p = 0.001). However, in rats with acute kidney injury exhibited a larger Vss as opposed to normal renal function (511.28 ± 21.03 ml/kg vs. 442.25 ± 31.43 ml/kg; p = 0.0001). Whereas oral Cl and terminal volume of distribution (Vz) of TQ were reduced by ∼50% in the liver dysfunction group (p = 0.0001). These changes were associated with more systemic exposure as measured by AUC0-∞ in rats with compromised liver functions. The estimated plasma protein binding TQ was 99.84 ± 0.03% in healthy controls, 97.05 ± 0.57% with kidney injury rats, and 95.75 ± 0.64% in liver dysfunctionThe findings of the present study suggest that liver dysfunction could potentially modify the disposition of TQ administered orally, and therefore, a smaller maintenance dose is probably required to avoid accumulation.
Collapse
Affiliation(s)
- Khalid M Alkharfy
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Fahad A Ali
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | | | - Basit L Jan
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad Raish
- Department Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saeed Alqahtani
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ajaz Ahmad
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
31
|
Ruoß M, Rebholz S, Weimer M, Grom-Baumgarten C, Athanasopulu K, Kemkemer R, Käß H, Ehnert S, Nussler AK. Development of Scaffolds with Adjusted Stiffness for Mimicking Disease-Related Alterations of Liver Rigidity. J Funct Biomater 2020; 11:17. [PMID: 32183326 PMCID: PMC7151584 DOI: 10.3390/jfb11010017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 12/21/2022] Open
Abstract
Drug-induced liver toxicity is one of the most common reasons for the failure of drugs in clinical trials and frequent withdrawal from the market. Reasons for such failures include the low predictive power of in vivo studies, that is mainly caused by metabolic differences between humans and animals, and intraspecific variances. In addition to factors such as age and genetic background, changes in drug metabolism can also be caused by disease-related changes in the liver. Such metabolic changes have also been observed in clinical settings, for example, in association with a change in liver stiffness, a major characteristic of an altered fibrotic liver. For mimicking these changes in an in vitro model, this study aimed to develop scaffolds that represent the rigidity of healthy and fibrotic liver tissue. We observed that liver cells plated on scaffolds representing the stiffness of healthy livers showed a higher metabolic activity compared to cells plated on stiffer scaffolds. Additionally, we detected a positive effect of a scaffold pre-coated with fetal calf serum (FCS)-containing media. This pre-incubation resulted in increased cell adherence during cell seeding onto the scaffolds. In summary, we developed a scaffold-based 3D model that mimics liver stiffness-dependent changes in drug metabolism that may more easily predict drug interaction in diseased livers.
Collapse
Affiliation(s)
- Marc Ruoß
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University, 72076 Tübingen, Germany; (S.R.); (M.W.); (C.G.-B.); (S.E.); (A.K.N.)
| | - Silas Rebholz
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University, 72076 Tübingen, Germany; (S.R.); (M.W.); (C.G.-B.); (S.E.); (A.K.N.)
| | - Marina Weimer
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University, 72076 Tübingen, Germany; (S.R.); (M.W.); (C.G.-B.); (S.E.); (A.K.N.)
- Faculty of Applied Chemistry, Reutlingen University, 72762 Reutlingen, Germany; (K.A.); (R.K.)
| | - Carl Grom-Baumgarten
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University, 72076 Tübingen, Germany; (S.R.); (M.W.); (C.G.-B.); (S.E.); (A.K.N.)
| | - Kiriaki Athanasopulu
- Faculty of Applied Chemistry, Reutlingen University, 72762 Reutlingen, Germany; (K.A.); (R.K.)
| | - Ralf Kemkemer
- Faculty of Applied Chemistry, Reutlingen University, 72762 Reutlingen, Germany; (K.A.); (R.K.)
| | - Hanno Käß
- Faculty of Basic Science, University of Applied Sciences Esslingen, 73728 Esslingen am Neckar, Germany;
| | - Sabrina Ehnert
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University, 72076 Tübingen, Germany; (S.R.); (M.W.); (C.G.-B.); (S.E.); (A.K.N.)
| | - Andreas K. Nussler
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University, 72076 Tübingen, Germany; (S.R.); (M.W.); (C.G.-B.); (S.E.); (A.K.N.)
| |
Collapse
|
32
|
Weersink RA, Burger DM, Hayward KL, Taxis K, Drenth JP, Borgsteede SD. Safe use of medication in patients with cirrhosis: pharmacokinetic and pharmacodynamic considerations. Expert Opin Drug Metab Toxicol 2019; 16:45-57. [DOI: 10.1080/17425255.2020.1702022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Rianne A. Weersink
- Department of Pharmacy, Unit of Pharmacotherapy, -Epidemiology and -Economics, University of Groningen, Groningen, The Netherlands
- Department of Clinical Decision Support, Health Base Foundation, Houten, The Netherlands
| | - David M. Burger
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kelly L. Hayward
- Centre for Liver Disease Research, Translational Research Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Pharmacy Department, Princess Alexandra Hospital, Brisbane, Australia
| | - Katja Taxis
- Department of Pharmacy, Unit of Pharmacotherapy, -Epidemiology and -Economics, University of Groningen, Groningen, The Netherlands
| | - Joost P.H. Drenth
- Department of Gastroenterology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sander D. Borgsteede
- Department of Clinical Decision Support, Health Base Foundation, Houten, The Netherlands
| |
Collapse
|
33
|
Takebe N, Beumer JH, Kummar S, Kiesel BF, Dowlati A, O'Sullivan Coyne G, Piekarz R, Rubinstein L, Fogli LK, Vaishampayan U, Goel S, O'Bryant CL, El‐Rayes BF, Chung V, Lenz H, Kim R, Belani CP, Tuscano JM, Schelman W, Moore N, Doroshow JH, Chen AP. A phase I pharmacokinetic study of belinostat in patients with advanced cancers and varying degrees of liver dysfunction. Br J Clin Pharmacol 2019; 85:2499-2511. [PMID: 31271459 PMCID: PMC6848909 DOI: 10.1111/bcp.14054] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 06/17/2019] [Accepted: 06/25/2019] [Indexed: 12/31/2022] Open
Abstract
AIMS The histone deacetylase inhibitor belinostat has activity in various cancers. Because belinostat is metabolized by the liver, reduced hepatic clearance could lead to excessive drug accumulation and increased toxicity. Safety data in patients with liver dysfunction are needed for this drug to reach its full potential in the clinic. METHODS We performed a phase 1 trial to determine the safety, maximum tolerated dose (MTD) and pharmacokinetics of belinostat in patients with advanced cancer and varying degrees of liver dysfunction. RESULTS Seventy-two patients were enrolled and divided into cohorts based on liver function. In patients with mild dysfunction, the MTD was the same as the recommended phase 2 dose (1000 mg/m2 /day). Belinostat was well tolerated in patients with moderate and severe liver dysfunction, although the trial was closed before the MTD in these cohorts could be determined. The mean clearance of belinostat was 661 mL/min/m2 in patients with normal liver function, compared to 542, 505 and 444 mL/min/m2 in patients with mild, moderate and severe hepatic dysfunction. Although this trial was not designed to assess clinical activity, of the 47 patients evaluable for response, 13 patients (28%) experienced stable disease. CONCLUSION While a statistically significant difference in clearance indicates increased belinostat exposure with worsening liver function, no relationship was observed between belinostat exposure and toxicity. An assessment of belinostat metabolites revealed significant differences in metabolic pathway capability in patients with differing levels of liver dysfunction. Further studies are needed to establish formal dosing guidelines in this patient population.
Collapse
Affiliation(s)
- Naoko Takebe
- Early Clinical Trials Development Program, Developmental Therapeutics Clinic, Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMDUSA
| | - Jan H. Beumer
- Cancer Therapeutics ProgramUPMC Hillman Cancer CenterPittsburghPAUSA
- Department of Pharmaceutical SciencesUniversity of Pittsburgh School of PharmacyPittsburghPAUSA
- Division of Hematology‐Oncology, Department of MedicineUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Shivaani Kummar
- Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMDUSA
| | - Brian F. Kiesel
- Cancer Therapeutics ProgramUPMC Hillman Cancer CenterPittsburghPAUSA
- Department of Pharmaceutical SciencesUniversity of Pittsburgh School of PharmacyPittsburghPAUSA
| | - Afshin Dowlati
- University Hospitals Seidman Cancer Center and Case Western Reserve UniversityClevelandOHUSA
| | - Geraldine O'Sullivan Coyne
- Early Clinical Trials Development Program, Developmental Therapeutics Clinic, Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMDUSA
| | - Richard Piekarz
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMDUSA
| | - Lawrence Rubinstein
- Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMDUSA
| | - Laura K. Fogli
- Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMDUSA
| | | | - Sanjay Goel
- Montefiore Medical CenterAlbert Einstein College of MedicineNew YorkNYUSA
| | | | | | | | - Heinz‐Josef Lenz
- Norris Comprehensive Cancer CenterUniversity of Southern CaliforniaLos AngelesCAUSA
| | - Richard Kim
- Department of Gastrointestinal OncologyMoffitt Cancer Center and Research InstituteTampaFLUSA
| | - Chandra P. Belani
- Penn State Cancer InstitutePenn State Health Milton S. Hershey Medical CenterHersheyPAUSA
| | - Joseph M. Tuscano
- Comprehensive Cancer CenterUniversity of California Davis Medical CenterSacramentoCAUSA
| | | | - Nancy Moore
- Early Clinical Trials Development Program, Developmental Therapeutics Clinic, Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMDUSA
| | - James H. Doroshow
- Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMDUSA
- Center for Cancer ResearchNational Cancer InstituteBethesdaMDUSA
| | - Alice P. Chen
- Early Clinical Trials Development Program, Developmental Therapeutics Clinic, Division of Cancer Treatment and DiagnosisNational Cancer InstituteBethesdaMDUSA
| |
Collapse
|
34
|
Miura M, Uchida S, Tanaka S, Inui N, Kawakami J, Watanabe H, Namiki N. The Prediction of the Area under the Curve and Clearance of Midazolam from Single-Point Plasma Concentration and Urinary Excretion in Healthy Volunteers. Biol Pharm Bull 2019; 42:1590-1595. [DOI: 10.1248/bpb.b19-00122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Motoyasu Miura
- Departments of Pharmacy Practice & Science, School of Pharmaceutical Sciences, University of Shizuoka
- Hospital Pharmacy, Hamamatsu University School of Medicine
| | - Shinya Uchida
- Departments of Pharmacy Practice & Science, School of Pharmaceutical Sciences, University of Shizuoka
| | - Shimako Tanaka
- Departments of Pharmacy Practice & Science, School of Pharmaceutical Sciences, University of Shizuoka
| | - Naoki Inui
- Department of Clinical Pharmacology & Therapeutics, Hamamatsu University School of Medicine
| | | | - Hiroshi Watanabe
- Department of Clinical Pharmacology & Therapeutics, Hamamatsu University School of Medicine
| | - Noriyuki Namiki
- Departments of Pharmacy Practice & Science, School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
35
|
Papazafiropoulou A, Melidonis A. Antidiabetic agents in patients with hepatic impairment. World J Meta-Anal 2019; 7:380-388. [DOI: 10.13105/wjma.v7.i8.380] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/07/2019] [Accepted: 08/20/2019] [Indexed: 02/06/2023] Open
|
36
|
Pharmacokinetics of Fentanyl and Its Derivatives in Children: A Comprehensive Review. Clin Pharmacokinet 2019; 57:125-149. [PMID: 28688027 DOI: 10.1007/s40262-017-0569-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Fentanyl and its derivatives sufentanil, alfentanil, and remifentanil are potent opioids. A comprehensive review of the use of fentanyl and its derivatives in the pediatric population was performed using the National Library of Medicine PubMed. Studies were included if they contained original pharmacokinetic parameters or models using established routes of administration in patients younger than 18 years of age. Of 372 retrieved articles, 44 eligible pharmacokinetic studies contained data of 821 patients younger than 18 years of age, including more than 46 preterm infants, 64 full-term neonates, 115 infants/toddlers, 188 children, and 28 adolescents. Underlying diagnoses included congenital heart and pulmonary disease and abdominal disorders. Routes of drug administration were intravenous, epidural, oral-transmucosal, intranasal, and transdermal. Despite extensive use in daily clinical practice, few studies have been performed. Preterm and term infants have lower clearance and protein binding. Pharmacokinetics was not altered by chronic renal or hepatic disease. Analyses of the pooled individual patients' data revealed that clearance maturation relating to body weight could be best described by the Hill function for sufentanil (R 2 = 0.71, B max 876 mL/min, K 50 16.3 kg) and alfentanil (R 2 = 0.70, B max (fixed) 420 mL/min, K 50 28 kg). The allometric exponent for estimation of clearance of sufentanil was 0.99 and 0.75 for alfentanil clearance. Maturation of remifentanil clearance was described by linear regression to bodyweight (R 2 = 0.69). The allometric exponent for estimation of remifentanil clearance was 0.76. For fentanyl, linear regression showed only a weak correlation between clearance and bodyweight in preterm and term neonates (R 2 = 0.22) owing to a lack of data in older age groups. A large heterogeneity regarding study design, clinical setting, drug administration, laboratory assays, and pharmacokinetic estimation was observed between studies introducing bias into the analyses performed in this review. A limitation of this review is that pharmacokinetic data, based on different modes of administration, dosing schemes, and parameter estimation methods, were combined.
Collapse
|
37
|
Wuensch T, Heucke N, Wizenty J, Quint J, Sinn B, Arsenic R, Jara M, Kaffarnik M, Pratschke J, Stockmann M. Hepatic CYP1A2 activity in liver tumors and the implications for preoperative volume-function analysis. Am J Physiol Gastrointest Liver Physiol 2019; 316:G608-G614. [PMID: 30869529 DOI: 10.1152/ajpgi.00335.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dynamic liver function assessment by the [13C]methacetin maximal liver function capacity (LiMAx) test reflects the overall hepatic cytochrome P-450 (CYP) 1A2 activity. One proven strategy for preoperative risk assessment in liver surgery includes the combined assessment of the dynamic liver function by the LiMAx test, the volumetric analysis of the liver, and calculation of future liver remnant function. This so-called volume-function analysis assumes that the remaining CYP1A2 activity in any tumor lesion is zero. The here presented study aims to assess the remaining CYP1A2 activities in different hepatic tumor lesions and its consequences for the preoperative volume-function analysis in patients undergoing liver surgery. The CYP1A2 activity analysis of neoplastic lesions and adjacent nontumor liver tissue from resected tumor specimens revealed a significantly higher CYP1A2 activity (median, interquartile range) in nontumor tissues (35.5, 15.9-54.4 µU/mg) compared with hepatocellular adenomas (7.35, 1.2-32.5 µU/mg), hepatocellular carcinomas (0.18, 0.0-2.0 µU/mg), or colorectal liver metastasis (0.17, 0.0-2.1 µU/mg). In nontumor liver tissue, a gradual decline in CYP1A2 activity with exacerbating fibrosis was observed. The CYP1A2 activity differences were also reflected in CYP1A2 protein signals in the assessed hepatic tissues. Volume-function analysis showed a minimal deviation compared with the current standard calculation for hepatocellular carcinomas or colorectal liver metastasis (<1% difference), whereas a difference of 11.9% was observed for hepatocellular adenomas. These findings are important for a refined preoperative volume-function analysis and improved surgical risk assessment in hepatocellular adenoma cases with low LiMAx values. NEW & NOTEWORTHY The cytochrome P-450 (CYP) 1A2-dependent maximal liver function capacity test reflects the overall functional capacity of the liver. To which extent hepatocellular tumors harbor CYP1A2 activity and thus contribute to the maximal liver function capacity test outcome is unknown. We here show that hepatocellular adenomas but not hepatocellular carcinomas or colorectal liver metastasis contain significant residual CYP1A2 activity. These findings are important for an improved preoperative volume-function analysis and an accurate surgical risk assessment in hepatocellular adenoma cases.
Collapse
Affiliation(s)
- Tilo Wuensch
- Department of Surgery, Campus Charité Mitte and Campus Virchow-Klinikum, Charité- Universitätsmedizin Berlin, Berlin , Germany
| | - Niklas Heucke
- Department of Surgery, Campus Charité Mitte and Campus Virchow-Klinikum, Charité- Universitätsmedizin Berlin, Berlin , Germany
| | - Jonas Wizenty
- Department of Surgery, Campus Charité Mitte and Campus Virchow-Klinikum, Charité- Universitätsmedizin Berlin, Berlin , Germany
| | - Janina Quint
- Department of Surgery, Campus Charité Mitte and Campus Virchow-Klinikum, Charité- Universitätsmedizin Berlin, Berlin , Germany
| | - Bruno Sinn
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin , Germany
| | - Ruza Arsenic
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin , Germany
| | - Maximilian Jara
- Department of Surgery, Campus Charité Mitte and Campus Virchow-Klinikum, Charité- Universitätsmedizin Berlin, Berlin , Germany
| | - Magnus Kaffarnik
- Department of Surgery, Campus Charité Mitte and Campus Virchow-Klinikum, Charité- Universitätsmedizin Berlin, Berlin , Germany
| | - Johann Pratschke
- Department of Surgery, Campus Charité Mitte and Campus Virchow-Klinikum, Charité- Universitätsmedizin Berlin, Berlin , Germany
| | - Martin Stockmann
- Department of Surgery, Campus Charité Mitte and Campus Virchow-Klinikum, Charité- Universitätsmedizin Berlin, Berlin , Germany
| |
Collapse
|
38
|
Zhou X, Lockhart AC, Fu S, Nemunaitis J, Sarantopoulos J, Muehler A, Rangachari L, Bargfrede M, Venkatakrishnan K. Pharmacokinetics of the Investigational Aurora A Kinase Inhibitor Alisertib in Adult Patients With Advanced Solid Tumors or Relapsed/Refractory Lymphoma With Varying Degrees of Hepatic Dysfunction. J Clin Pharmacol 2019; 59:1204-1215. [PMID: 30985952 DOI: 10.1002/jcph.1416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/16/2019] [Indexed: 11/11/2022]
Abstract
This clinical trial was designed to evaluate the effect of moderate or severe hepatic impairment on the single-dose pharmacokinetics (PK) of the investigational anticancer agent, alisertib, in adult patients with advanced solid tumors or lymphoma. Patients with normal hepatic function (total bilirubin and alanine transaminase [ALT] ≤ upper limit of normal [ULN]), moderate hepatic impairment (1.5 × ULN < total bilirubin ≤ 3 × ULN, with any ALT) or severe hepatic impairment (total bilirubin > 3 × ULN, with any ALT), received a single 50-mg oral dose of alisertib. Blood samples for PK were collected up to 168 hours postdose. Predose samples were also used to assess alisertib plasma protein binding. Patients could continue to receive alisertib for 7 days in 21-day cycles (50, 30, or 10 mg twice daily for normal hepatic function, moderate hepatic impairment, and severe hepatic impairment, respectively). Alisertib was approximately 99% protein bound in all hepatic function groups. Alisertib exposure was similar in moderate and severe hepatic impairment groups, but higher than the normal hepatic function group. The geometric least-squares mean ratios (90% confidence intervals) for unbound alisertib area under the curve extrapolated to infinity for moderate/severe impairment groups versus the normal hepatic function group was 254% (184%, 353%). Patients with moderate or severe hepatic impairment have approximately 150% higher unbound alisertib exposures compared with patients with normal hepatic function. An approximately 60% reduction of the starting dose of alisertib in patients with moderate/severe hepatic impairment is recommended based on pharmacokinetic considerations.
Collapse
Affiliation(s)
- Xiaofei Zhou
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | - A Craig Lockhart
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Siqing Fu
- University of Texas - MD Anderson Cancer Center - Houston, TX, USA
| | | | - John Sarantopoulos
- Institute for Drug Development, Mays Cancer Center at University of Texas Health San Antonio MD Anderson Cancer Center, San Antonio, TX, USA
| | - Andreas Muehler
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | - Lakshmi Rangachari
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | | | - Karthik Venkatakrishnan
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| |
Collapse
|
39
|
Choi GW, Lee YB, Cho HY. Interpretation of Non-Clinical Data for Prediction of Human Pharmacokinetic Parameters: In Vitro-In Vivo Extrapolation and Allometric Scaling. Pharmaceutics 2019; 11:E168. [PMID: 30959827 PMCID: PMC6523982 DOI: 10.3390/pharmaceutics11040168] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/22/2019] [Accepted: 04/02/2019] [Indexed: 02/06/2023] Open
Abstract
Extrapolation of pharmacokinetic (PK) parameters from in vitro or in vivo animal to human is one of the main tasks in the drug development process. Translational approaches provide evidence for go or no-go decision-making during drug discovery and the development process, and the prediction of human PKs prior to the first-in-human clinical trials. In vitro-in vivo extrapolation and allometric scaling are the choice of method for projection to human situations. Although these methods are useful tools for the estimation of PK parameters, it is a challenge to apply these methods since underlying biochemical, mathematical, physiological, and background knowledge of PKs are required. In addition, it is difficult to select an appropriate methodology depending on the data available. Therefore, this review covers the principles of PK parameters pertaining to the clearance, volume of distribution, elimination half-life, absorption rate constant, and prediction method from the original idea to recently developed models in order to introduce optimal models for the prediction of PK parameters.
Collapse
Affiliation(s)
- Go-Wun Choi
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Korea.
| | - Yong-Bok Lee
- College of Pharmacy, Chonnam National University, 77 Yongbong-ro, Buk-Gu, Gwangju 61186, Korea.
| | - Hea-Young Cho
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Korea.
| |
Collapse
|
40
|
Zhang M, You X, Ke M, Jiao Z, Wu H, Huang P, Lin C. Prediction of Ticagrelor and its Active Metabolite in Liver Cirrhosis Populations Using a Physiologically Based Pharmacokinetic Model Involving Pharmacodynamics. J Pharm Sci 2019; 108:2781-2790. [PMID: 30928308 DOI: 10.1016/j.xphs.2019.03.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/17/2019] [Accepted: 03/19/2019] [Indexed: 12/14/2022]
Abstract
Ticagrelor, a P2Y12 receptor antagonist, has been highly recommended for use in acute coronary syndrome. The major active metabolite (AM) is similar to the parent drug, which exhibits antiplatelet activity. The inhibition of platelet aggregation (IPA) is used as an assay to demonstrate the anticoagulant efficacy of ticagrelor. In this study, we developed a physiologically based pharmacokinetic (PBPK) model to predict the pharmacokinetics of ticagrelor and its AM and combined this model with a pharmacodynamics model to reflect potential pharmacodynamic alterations in liver cirrhosis populations. The simulated results obtained using the PBPK model were validated by fold error values, which were all smaller than 2. Comparisons of exposure in different classifications of liver cirrhosis indicated that exposure to ticagrelor increased significantly with an increase in the degree of cirrhosis severity, whereas exposure to AM was decreased. The total concentration of ticagrelor and AM was related to the IPA included in the Sigmoid Emax model. The PBPK model of ticagrelor and AM could predict the pharmacokinetics of all populations, and a combination of PD models was used to extrapolate for predicting unknown scenarios. Liver cirrhosis may result in prolonged IPA, depending on the severity degree of this disease. The combined PBPK model including IPA can reveal changes in pharmacokinetics and pharmacodynamics in populations affected by liver cirrhosis and indicate the risk potential.
Collapse
Affiliation(s)
- Min Zhang
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, 20 Cha Zhong M. Rd, Fuzhou 350005, People's Republic of China
| | - Xiang You
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, 20 Cha Zhong M. Rd, Fuzhou 350005, People's Republic of China
| | - Meng Ke
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, 20 Cha Zhong M. Rd, Fuzhou 350005, People's Republic of China
| | - Zheng Jiao
- Department of Pharmacy, Huashan Hospital of Fudan University, 12 Wu Lu Mu Qi M. Rd, Shanghai 20040, People's Republic of China.
| | - Hongwei Wu
- Department of Antibiotics, Xiamen Institute for Food and Drug Quality Control, 33 Hai Shan. Rd, Xiamen 361012, People's Republic of China
| | - Pinfang Huang
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, 20 Cha Zhong M. Rd, Fuzhou 350005, People's Republic of China
| | - Cuihong Lin
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, 20 Cha Zhong M. Rd, Fuzhou 350005, People's Republic of China.
| |
Collapse
|
41
|
AbuHalimeh B, Krowka MJ, Tonelli AR. Treatment Barriers in Portopulmonary Hypertension. Hepatology 2019; 69:431-443. [PMID: 30063259 PMCID: PMC6460471 DOI: 10.1002/hep.30197] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 07/23/2018] [Indexed: 12/16/2022]
Abstract
Portopulmonary hypertension (PoPH) is a form of pulmonary arterial hypertension (PAH) that can develop as a complication of portal hypertension. Treatment of PoPH includes PAH-specific therapies, and in certain cases, such therapies are necessary to facilitate a successful liver transplantation. A significant number of barriers may limit the adequate treatment of patients with PoPH and explain the poorer survival of these patients when compared to patients with other types of PAH. Until recently, only one randomized controlled trial has included PoPH patients, and the majority of treatment data have been derived from relatively small observational studies. In the present article, we review some of the barriers in the treatment of patients with PoPH and implications for liver transplantation.
Collapse
Affiliation(s)
- Batool AbuHalimeh
- Pathobiology Division, Lerner Research Institute. Cleveland Clinic, OH, USA.
| | - Michael J Krowka
- Department of Gastroenterology and Hepatology and Department of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota, USA.
| | - Adriano R. Tonelli
- Department of Pulmonary, Allergy and Critical Care Medicine. Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
42
|
Dubovsky SL. Applications of calcium channel blockers in psychiatry: pharmacokinetic and pharmacodynamic aspects of treatment of bipolar disorder. Expert Opin Drug Metab Toxicol 2018; 15:35-47. [PMID: 30558453 DOI: 10.1080/17425255.2019.1558206] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Introduction: Calcium channel blockers (CCBs) comprise a heterogeneous group of medications that reduce calcium influx and attenuate cellular hyperactivity. Evidence of hyperactive intracellular calcium ion signaling in multiple peripheral cells of patients with bipolar disorder, calcium antagonist actions of established mood stabilizers, and a relative dearth of treatments have prompted research into potential uses of CCBs for this common and disabling condition. Areas covered: This review provides a comprehensive overview of intracellular calcium signaling in bipolar disorder, structure and function of calcium channels, pharmacology of CCBs, evidence of efficacy of CCBs in bipolar disorder, clinical applications, and directions for future research. Expert opinion: Despite mixed evidence of efficacy, CCBs are a promising novel approach to a demonstrated cellular abnormality in both poles of bipolar disorder. Potential advantages include low potential for sedation and weight gain, and possible usefulness for pregnant and neurologically impaired patients. Further research should focus on markers of a preferential response, studies in specific bipolar subtypes, development of CCBs acting preferentially in the central nervous system and on calcium channels that are primarily involved in neuronal signaling and plasticity.
Collapse
Affiliation(s)
- Steven L Dubovsky
- a Department of Psychiatry , State University of New York at Buffalo , Buffalo , NY , USA.,b Departments of Psychiatry and Medicine , University of Colorado , Denver , CO , USA
| |
Collapse
|
43
|
Pre-Existing Liver Disease and Toxicity of Antifungals. J Fungi (Basel) 2018; 4:jof4040133. [PMID: 30544724 PMCID: PMC6309049 DOI: 10.3390/jof4040133] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/05/2018] [Accepted: 12/07/2018] [Indexed: 12/17/2022] Open
Abstract
Pre-existing liver disease in patients with invasive fungal infections further complicates their management. Altered pharmacokinetics and tolerance issues of antifungal drugs are important concerns. Adjustment of the dosage of antifungal agents in these cases can be challenging given that current evidence to guide decision-making is limited. This comprehensive review aims to evaluate the existing evidence related to antifungal treatment in individuals with liver dysfunction. This article also provides suggestions for dosage adjustment of antifungal drugs in patients with varying degrees of hepatic impairment, after accounting for established or emerging pharmacokinetic–pharmacodynamic relationships with regard to antifungal drug efficacy in vivo.
Collapse
|
44
|
Pilla Reddy V, Bui K, Scarfe G, Zhou D, Learoyd M. Physiologically Based Pharmacokinetic Modeling for Olaparib Dosing Recommendations: Bridging Formulations, Drug Interactions, and Patient Populations. Clin Pharmacol Ther 2018; 105:229-241. [PMID: 29717476 PMCID: PMC6585620 DOI: 10.1002/cpt.1103] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/21/2018] [Indexed: 12/13/2022]
Abstract
We report physiologically based pharmacokinetic-modeling analyses to determine olaparib (tablet or capsule) drug-drug interactions (DDIs). Verified DDI simulations provided dose recommendations for olaparib coadministration with clinically relevant CYP3A4 modulators to eliminate potential risk to patient safety or olaparib efficacy. When olaparib is given with strong/moderate CYP3A inhibitors, the dose should be reduced to 100/150 mg b.i.d. (tablet), and 150/200 mg b.i.d. (capsule). Olaparib administration is not recommended with strong/moderate CYP3A inducers. No dose reductions are required with weak CYP3A inhibitors/inducers. Olaparib was shown to be a weak inhibitor of CYP3A (1.6-fold increase in exposure of a sensitive CYP3A probe) and to have no effect on P-glycoprotein or UGT1A1 substrates. Finally, this model was used to simulate exposure in scenarios where clinical data of olaparib are lacking, such as severe renal or hepatic impairment populations, and provided initial dosing recommendations in pediatric patients.
Collapse
Affiliation(s)
- Venkatesh Pilla Reddy
- Modelling and Simulation, Oncology DMPK, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Khanh Bui
- Quantitative Clinical Pharmacology, Oncology IMED Biotech Unit, AstraZeneca, Waltham, Massachusetts, USA
| | - Graeme Scarfe
- Modelling and Simulation, Oncology DMPK, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Diansong Zhou
- Quantitative Clinical Pharmacology, Oncology IMED Biotech Unit, AstraZeneca, Waltham, Massachusetts, USA
| | - Maria Learoyd
- Quantitative Clinical Pharmacology, Oncology IMED Biotech Unit, AstraZeneca, Cambridge, UK
| |
Collapse
|
45
|
Weersink RA, Bouma M, Burger DM, Drenth JPH, Harkes-Idzinga SF, Hunfeld NGM, Metselaar HJ, Monster-Simons MH, van Putten SAW, Taxis K, Borgsteede SD. Safe use of proton pump inhibitors in patients with cirrhosis. Br J Clin Pharmacol 2018; 84:1806-1820. [PMID: 29688583 PMCID: PMC6046475 DOI: 10.1111/bcp.13615] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/09/2018] [Accepted: 04/10/2018] [Indexed: 12/16/2022] Open
Abstract
Aims Proton pump inhibitors (PPIs) belong to the most frequently used drugs, also in patients with cirrhosis. PPIs are extensively metabolized by the liver, but practice guidance on prescribing in cirrhosis is lacking. We aim to develop practical guidance on the safe use of PPIs in patients with cirrhosis. Methods A systematic literature search identified studies on the safety (i.e. adverse events) and pharmacokinetics of PPIs in cirrhotic patients. This evidence and data from the product information was reviewed by an expert panel who classified drugs as safe; no additional risks known; additional risks known; unsafe; or unknown. Guidance was aimed at the oral use of PPIs and categorized by the severity of cirrhosis, using the Child–Turcotte–Pugh (CTP) classification. Results A total of 69 studies were included. Esomeprazole, omeprazole and rabeprazole were classified as having ‘no additional risks known’. A reduction in maximum dose of omeprazole and rabeprazole is recommended for CTP A and B patients. For patients with CTP C cirrhosis, the only PPI advised is esomeprazole at a maximum dosage of 20 mg per day. Pantoprazole and lansoprazole were classified as unsafe because of 4‐ to 8‐fold increased exposure. The use of PPIs in cirrhotic patients has been associated with the development of infections and hepatic encephalopathy and should be carefully considered. Conclusions We suggest using esomeprazole, omeprazole or rabeprazole in patients with CTP A or B cirrhosis and only esomeprazole in patients with CTP C. Pharmacokinetic changes are also important to consider when prescribing PPIs to vulnerable, cirrhotic patients.
Collapse
Affiliation(s)
- Rianne A Weersink
- Health Base Foundation, Houten, The Netherlands.,Department of Pharmacy, Unit of Pharmacotherapy, -Epidemiology & -Economics, University of Groningen, Groningen, The Netherlands
| | - Margriet Bouma
- Department of Guideline Development, Dutch College of General Practice, Utrecht, The Netherlands
| | - David M Burger
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost P H Drenth
- Department of Gastroenterology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - S Froukje Harkes-Idzinga
- Center for Information on Medicines, Royal Dutch Pharmacists Association (KNMP), The Hague, The Netherlands
| | - Nicole G M Hunfeld
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Intensive Care, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Herold J Metselaar
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Margje H Monster-Simons
- Dutch Medicines Evaluation Board, Utrecht, The Netherlands.,Department of Clinical Pharmacy and Pharmacology, University of Groningen, Groningen, The Netherlands
| | | | - Katja Taxis
- Department of Pharmacy, Unit of Pharmacotherapy, -Epidemiology & -Economics, University of Groningen, Groningen, The Netherlands
| | - Sander D Borgsteede
- Health Base Foundation, Houten, The Netherlands.,Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
46
|
Weersink RA, Bouma M, Burger DM, Drenth JPH, Harkes-Idzinga SF, Hunfeld NGM, Metselaar HJ, Monster-Simons MH, Taxis K, Borgsteede SD. Evidence-Based Recommendations to Improve the Safe Use of Drugs in Patients with Liver Cirrhosis. Drug Saf 2018; 41:603-613. [PMID: 29330714 PMCID: PMC5966501 DOI: 10.1007/s40264-017-0635-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
INTRODUCTION The presence of liver cirrhosis can have a major impact on pharmacodynamics and pharmacokinetics, but guidance for prescribing is lacking. OBJECTIVE The aim of this study is to provide an overview of evidence-based recommendations developed for the safe use of drugs in liver cirrhosis. METHODS Recommendations were based on a systematic literature search combined with expert opinion from a panel of 10 experts. The safety of each drug was classified as safe, no additional risks known, additional risks known, unsafe, unknown or the safety class was dependent on the severity of liver cirrhosis (Child-Pugh classification). If applicable, drug-specific dosing advice was provided. All recommendations were implemented in clinical decision support systems and on a website. RESULTS We formulated 218 recommendations for a total of 209 drugs. For nine drugs, two recommendations were formulated for different administration routes or indications. Drugs were classified as 'safe' in 29 recommendations (13.3%), 'no additional risks known' in 60 (27.5%), 'additional risks known' in 3 (1.4%), and 'unsafe' in 30 (13.8%). In 57 (26.1%) of the recommendations, safety depended on the severity of liver cirrhosis and was 'unknown' in 39 (17.9%) recommendations. Large alterations in pharmacodynamics were the main reason for classifying a drug as 'unsafe'. For 67 drugs (31%), a dose adjustment was needed. CONCLUSIONS Over 200 recommendations were developed for the safe use of drugs in patients with liver cirrhosis. Implementing these recommendations into clinical practice can possibly enhance medication safety in this vulnerable patient group.
Collapse
Affiliation(s)
- Rianne A Weersink
- Department of Clinical Decision Support, Health Base Foundation, Houten, The Netherlands.
- Department of Pharmacy, Unit of Pharmacotherapy, Epidemiology and Economics, University of Groningen, Groningen, The Netherlands.
| | - Margriet Bouma
- Department of Guideline Development, Dutch College of General Practice, Utrecht, The Netherlands
| | - David M Burger
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost P H Drenth
- Department of Gastroenterology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - S Froukje Harkes-Idzinga
- Center for Information on Medicines, Royal Dutch Pharmacists Association (KNMP), The Hague, The Netherlands
| | - Nicole G M Hunfeld
- Department of Pharmacy and Department of Intensive Care, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Herold J Metselaar
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Margje H Monster-Simons
- Dutch Medicines Evaluation Board, Utrecht, The Netherlands
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, Groningen, The Netherlands
| | - Katja Taxis
- Department of Pharmacy, Unit of Pharmacotherapy, Epidemiology and Economics, University of Groningen, Groningen, The Netherlands
| | - Sander D Borgsteede
- Department of Clinical Decision Support, Health Base Foundation, Houten, The Netherlands
- SIR Institute for Pharmacy Practice and Policy, Leiden, The Netherlands
| |
Collapse
|
47
|
Ye J, Deng G, Gao F. Theoretical overview of clinical and pharmacological aspects of the use of etelcalcetide in diabetic patients undergoing hemodialysis. Drug Des Devel Ther 2018; 12:901-909. [PMID: 29719376 PMCID: PMC5914547 DOI: 10.2147/dddt.s160223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Etelcalcetide is the first intravenous calcimimetic agent authorized for the treatment of secondary hyperparathyroidism (sHPT) in patients undergoing hemodialysis in Europe, the US, and Japan. The relationship between sHPT and diabetes resides on complex, bidirectional effects and largely unknown homeostatic mechanisms. Although 30% or more patients with end-stage renal disease are diabetics and about the same percentage of those patients suffer from sHPT associated with hemodialysis, no data on the specificities of the use of etelcalcetide in such patients are available yet. Regarding pharmacokinetic interactions, etelcalcetide may compete with oral hypoglycemics recommended for use in patients undergoing hemodialysis and insulins detemir and degludec, causing unexpected hypocalcemia or hypoglycemia. More importantly, hypocalcemia, a common side effect of etelcalcetide, may cause decompensation of preexisting cardiac insufficiency in diabetic patients or worsen dialysis-related hypotension and lead to hypotension-related cardiac events, such as myocardial ischemia. In diabetic patients, hypocalcemia may lead to dangerous ventricular arrhythmias, as both insulin-related hypoglycemia and hemodialysis prolong QT interval. Patients with diabetes, therefore, should be strictly monitored for hypocalcemia and associated effects. Due to an altered parathormone activity in this patient group, plasma calcium should be the preferred indicator of etelcalcetide effects. Until more clinical experience with etelcalcetide is available, the clinicians should be cautious when using this calcimimetic in patients with diabetes.
Collapse
Affiliation(s)
- Jianzhen Ye
- Department of Endocrinology, Huangzhou District People’s Hospital, Huanggang, People’s Republic of China
| | - Guangrui Deng
- Department of Endocrinology, Huangzhou District People’s Hospital, Huanggang, People’s Republic of China
| | - Feng Gao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
48
|
Masman AD, Tibboel D, Baar FPM, van Dijk M, Mathot RAA, van Gelder T. Prevalence and Implications of Abnormal Laboratory Results in Patients in the Terminal Phase of Life. J Palliat Med 2018; 19:822-9. [PMID: 27494223 DOI: 10.1089/jpm.2015.0548] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Pathophysiological changes at the end of life may affect pharmacokinetics of drugs. However, caregivers typically do not extensively monitor patients' laboratory parameters at the end of life. OBJECTIVE Our aim was to describe laboratory parameters of hospice patients in the week before death. METHODS A cohort study was conducted on available laboratory results in the week before death, including clinical chemistry and hematology tests. RESULTS Laboratory data of 125 patients in a palliative care center were included, assessed at a median of 3 days before death. Eighty percent of patients had anemia and almost all had hypoalbuminemia (97%). Elevated levels of gamma-glutamyl transferase (gGT) were found in 75%, of alkaline phosphatase (ALP) in 60%, of aspartate aminotransferase (ASAT) in 60%, and of calcium (Ca) in 68%. Alanine aminotransferase (ALAT), bilirubin, sodium (Na), and potassium (K) were abnormal in from 8.8% to 36.0% of patients. A previous unknown poor kidney function was found in 60% of patients. Thirteen patients (22%) with a regular morphine prescription and one patient treated with a non-steroidal anti-inflammatory drug (NSAID) had severe kidney failure. CONCLUSIONS Abnormal laboratory results were expected due to the pathophysiological changes that occur during the last phase of life. Remarkably, however, electrolytes (Na and K) were balanced even shortly before death. Estimated glomerular filtration rate (eGFR), reflecting the kidney function, seems the most clinically relevant laboratory parameter, because it may guide drug choice and dosing.
Collapse
Affiliation(s)
- Anniek D Masman
- 1 Palliative Care Centre , Laurens Cadenza, Rotterdam, the Netherlands .,2 Pain Expertise Centre, Erasmus MC-Sophia Children's Hospital , Rotterdam, the Netherlands
| | - Dick Tibboel
- 2 Pain Expertise Centre, Erasmus MC-Sophia Children's Hospital , Rotterdam, the Netherlands .,3 Intensive Care, Department of Paediatric Surgery, Erasmus MC-Sophia Children's Hospital , Rotterdam, the Netherlands
| | - Frans P M Baar
- 1 Palliative Care Centre , Laurens Cadenza, Rotterdam, the Netherlands .,2 Pain Expertise Centre, Erasmus MC-Sophia Children's Hospital , Rotterdam, the Netherlands
| | - Monique van Dijk
- 2 Pain Expertise Centre, Erasmus MC-Sophia Children's Hospital , Rotterdam, the Netherlands .,3 Intensive Care, Department of Paediatric Surgery, Erasmus MC-Sophia Children's Hospital , Rotterdam, the Netherlands
| | - Ron A A Mathot
- 4 Hospital Pharmacy-Clinical Pharmacology , Academic Medical Centre, Amsterdam, the Netherlands
| | - Teun van Gelder
- 5 Department of Hospital Pharmacy, Erasmus Medical Centre , Rotterdam, the Netherlands
| |
Collapse
|
49
|
Bahar MA, Wang Y, Bos JHJ, Wilffert B, Hak E. Discontinuation and dose adjustment of metoprolol after metoprolol-paroxetine/fluoxetine co-prescription in Dutch elderly. Pharmacoepidemiol Drug Saf 2018; 27:621-629. [PMID: 29575226 PMCID: PMC6001522 DOI: 10.1002/pds.4422] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/12/2018] [Accepted: 02/16/2018] [Indexed: 12/19/2022]
Abstract
Purpose Co‐prescription of paroxetine/fluoxetine (a strong CYP2D6 inhibitor) in metoprolol (a CYP2D6 substrate) users is common, but data on the clinical consequences of this drug‐drug interaction are limited and inconclusive. Therefore, we assessed the effect of paroxetine/fluoxetine initiation on the existing treatment with metoprolol on the discontinuation and dose adjustment of metoprolol among elderly. Methods We performed a cohort study using the University of Groningen IADB.nl prescription database (www.IADB.nl). We selected all elderly (≥60 years) who had ever been prescribed metoprolol and had a first co‐prescription of paroxetine/fluoxetine, citalopram (weak CYP2D6 inhibitor), or mirtazapine (negative control) from 1994 to 2015. The exposure group was metoprolol and paroxetine/fluoxetine co‐prescription, and the other groups acted as controls. The outcomes were early discontinuation and dose adjustment of metoprolol. Logistic regression was applied to estimate adjusted odds ratios (OR) and 95% confidence intervals (CI). Results Combinations of metoprolol‐paroxetine/fluoxetine, metoprolol‐citalopram, and metoprolol‐mirtazapine were started in 528, 673, and 625 patients, respectively. Compared with metoprolol‐citalopram, metoprolol‐paroxetine/fluoxetine was not significantly associated with the early discontinuation and dose adjustment of metoprolol (OR = 1.07, 95% CI:0.77‐1.48; OR = 0.87, 95% CI:0.57‐1.33, respectively). In comparison with metoprolol‐mirtazapine, metoprolol‐paroxetine/fluoxetine was associated with a significant 43% relative increase in early discontinuation of metoprolol (OR = 1.43, 95% CI:1.01‐2.02) but no difference in the risk of dose adjustment. Stratified analysis by gender showed that women have a significantly high risk of metoprolol early discontinuation (OR = 1.62, 95% CI:1.03‐2.53). Conclusion Paroxetine/fluoxetine initiation in metoprolol prescriptions, especially for female older patients, is associated with the risk of early discontinuation of metoprolol.
Collapse
Affiliation(s)
- Muh Akbar Bahar
- Groningen Research Institute of Pharmacy, Department of PharmacoTherapy, -Epidemiology and -Economics, University of Groningen, Groningen, The Netherlands.,Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Yuanyuan Wang
- Groningen Research Institute of Pharmacy, Department of PharmacoTherapy, -Epidemiology and -Economics, University of Groningen, Groningen, The Netherlands
| | - Jens H J Bos
- Groningen Research Institute of Pharmacy, Department of PharmacoTherapy, -Epidemiology and -Economics, University of Groningen, Groningen, The Netherlands
| | - Bob Wilffert
- Groningen Research Institute of Pharmacy, Department of PharmacoTherapy, -Epidemiology and -Economics, University of Groningen, Groningen, The Netherlands.,Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, The Netherlands
| | - Eelko Hak
- Groningen Research Institute of Pharmacy, Department of PharmacoTherapy, -Epidemiology and -Economics, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
50
|
Pharmacokinetics and Safety of Velpatasvir and Sofosbuvir/Velpatasvir in Subjects with Hepatic Impairment. Clin Pharmacokinet 2018. [DOI: 10.1007/s40262-018-0645-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|