1
|
Chen L, Zhang Y, Zhang YX, Wang WL, Sun DM, Li PY, Feng XS, Tan Y. Pretreatment and analysis techniques development of TKIs in biological samples for pharmacokinetic studies and therapeutic drug monitoring. J Pharm Anal 2024; 14:100899. [PMID: 38634061 PMCID: PMC11022103 DOI: 10.1016/j.jpha.2023.11.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 10/26/2023] [Accepted: 11/15/2023] [Indexed: 04/19/2024] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have emerged as the first-line small molecule drugs in many cancer therapies, exerting their effects by impeding aberrant cell growth and proliferation through the modulation of tyrosine kinase-mediated signaling pathways. However, there exists a substantial inter-individual variability in the concentrations of certain TKIs and their metabolites, which may render patients with compromised immune function susceptible to diverse infections despite receiving theoretically efficacious anticancer treatments, alongside other potential side effects or adverse reactions. Therefore, an urgent need exists for an up-to-date review concerning the biological matrices relevant to bioanalysis and the sampling methods, clinical pharmacokinetics, and therapeutic drug monitoring of different TKIs. This paper provides a comprehensive overview of the advancements in pretreatment methods, such as protein precipitation (PPT), liquid-liquid extraction (LLE), solid-phase extraction (SPE), micro-SPE (μ-SPE), magnetic SPE (MSPE), and vortex-assisted dispersive SPE (VA-DSPE) achieved since 2017. It also highlights the latest analysis techniques such as newly developed high performance liquid chromatography (HPLC) and high-resolution mass spectrometry (HRMS) methods, capillary electrophoresis (CE), gas chromatography (GC), supercritical fluid chromatography (SFC) procedures, surface plasmon resonance (SPR) assays as well as novel nanoprobes-based biosensing techniques. In addition, a comparison is made between the advantages and disadvantages of different approaches while presenting critical challenges and prospects in pharmacokinetic studies and therapeutic drug monitoring.
Collapse
Affiliation(s)
- Lan Chen
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Yuan Zhang
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Yi-Xin Zhang
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Wei-Lai Wang
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - De-Mei Sun
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Peng-Yun Li
- Institute of Pharmacology and Toxicology Institution, National Engineering Research Center for Strategic Drugs, Beijing, 100850, China
| | - Xue-Song Feng
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Yue Tan
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| |
Collapse
|
2
|
Martin JH, Galettis P, Flynn A, Schneider J. Phenotype versus genotype to optimize cancer dosing in the clinical setting-focus on 5-fluorouracil and tyrosine kinase inhibitors. Pharmacol Res Perspect 2024; 12:e1182. [PMID: 38429945 PMCID: PMC10907881 DOI: 10.1002/prp2.1182] [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: 09/11/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 03/03/2024] Open
Abstract
Cancer medicines often have narrow therapeutic windows; toxicity can be severe and sometimes fatal, but inadequate dose intensity reduces efficacy and survival. Determining the optimal dose for each patient is difficult, with body-surface area used most commonly for chemotherapy and flat dosing for tyrosine kinase inhibitors, despite accumulating evidence of a wide range of exposures in individual patients with many receiving a suboptimal dose with these strategies. Therapeutic drug monitoring (measuring the drug concentration in a biological fluid, usually plasma) (TDM) is an accepted and well validated method to guide dose adjustments for individual patients to improve this. However, implementing TDM in routine care has been difficult outside a research context. The development of genotyping of various proteins involved in drug elimination and activity has gained prominence, with several but not all Guideline groups recommending dose reductions for particular variant genotypes. However, there is increasing concern that dosing recommendations are based on limited data sets and may lead to unnecessary underdosing and increased cancer mortality. This Review discusses the evidence surrounding genotyping and TDM to guide decisions around best practice.
Collapse
Affiliation(s)
- Jennifer H. Martin
- Drug Repurposing and Medicines Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Peter Galettis
- Drug Repurposing and Medicines Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Alex Flynn
- Drug Repurposing and Medicines Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Jennifer Schneider
- Drug Repurposing and Medicines Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| |
Collapse
|
3
|
Almurshedi AS, Radwan MA, Al Quadeib B, Aldosari B, Alfagih IM, Almarshidy SS. Pharmacokinetics of Afatinib after Intravenous and Oral Administrations in Rats Using Validated UPLC MS/MS Assay. J Chromatogr Sci 2024; 62:249-256. [PMID: 36617945 DOI: 10.1093/chromsci/bmac110] [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: 06/14/2022] [Revised: 11/24/2022] [Accepted: 12/26/2022] [Indexed: 01/10/2023]
Abstract
Afatinib is designated as the first-line management therapy for patients with advanced non-small cell lung cancer, and metastatic head and neck cancer. LC coupled to MS/MS can be utilised in therapeutic drug monitoring to ensure optimal use of Afatinib with the reduction of its possible adverse reactions. The aim of this investigation was to determine the pharmacokinetics of Afatinib in rats after single IV (2 mg/kg) and oral (8 mg/kg) doses. Therefore, a selective, sensitive and precise UPLC MS/MS assay thru electrospray ionisation basis with positive ionisation approach was established to measure Afatinib concentrations in the rat. The precision and accuracy of the developed assay method in the concentration range of 10-1000 ng/ml show no significant difference among inter- and-intra-day analysis (P > 0.05). Linearity was detected over the studied range with correlation coefficient, r > 0.995 (n = 6/day). The pharmacokinetics of Afatinib in the rat after a single IV dose showed a mean terminal half-life of 4.6 ± 0.97 h, and a mean clearance 480 ± 80 ml/h/kg. After PO administration, a short absorption phase with a mean Tmax of 1.3 ± 0.6 h with the highest concentration of 513.9 ± 281.1 ng/ml, and the lowest concentration detected after 24 h was 18.8 ± 10.7 ng/ml.
Collapse
Affiliation(s)
- Alanood S Almurshedi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, 12331 Riyadh, Saudi Arabia
| | - Mahasen A Radwan
- Department of Pharmacy Practice/Clinical Pharmacy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, 11829 Cairo, Egypt
| | - Bushra Al Quadeib
- Department of Pharmaceutics, College of Pharmacy, King Saud University, 12331 Riyadh, Saudi Arabia
| | - Basmah Aldosari
- Department of Pharmaceutics, College of Pharmacy, King Saud University, 12331 Riyadh, Saudi Arabia
| | - Iman M Alfagih
- Department of Pharmaceutics, College of Pharmacy, King Saud University, 12331 Riyadh, Saudi Arabia
| | - Salma S Almarshidy
- Department of Pharmaceutics, College of Pharmacy, King Saud University, 12331 Riyadh, Saudi Arabia
| |
Collapse
|
4
|
Raish M, Ahmad A, Karim BA, Jardan YAB, Ahad A, Iqbal M, Alkharfy KM, Al-Jenoobi FI, Mohammed OM. Pharmacokinetics of Dasatinib in Rats: a Potential Food-Drug Interaction with Naringenin. Eur J Drug Metab Pharmacokinet 2024; 49:239-247. [PMID: 38376657 DOI: 10.1007/s13318-024-00881-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2024] [Indexed: 02/21/2024]
Abstract
BACKGROUND AND OBJECTIVES The novel tyrosine kinase inhibitor (TKI) dasatinib, a multitarget inhibitor of Bcr-Abl and Src family kinases, has been licensed for the treatment of Ph+ acute lymphoblastic leukemia and chronic myeloid leukemia. Many citrus-based foods include the flavonoid naringenin, which is commonly available. Dasatinib is a Cyp3a4, P-gp, and Bcrp1 substrate, which makes it sensitive to potential food-drug interactions. The concurrent use of naringenin may change the pharmacokinetics of dasatinib, which could result in adverse effects and toxicity. The present investigation examined the impact of naringenin on the pharmacokinetics interactions of DAS and proposes a possible interaction mechanism in Wistar rats. METHODS Rats were provided with a single oral dose of dasatinib (25 mg/kg) with or without naringenin pretreatment (150 mg/kg p.o. daily for 7 days, n = 6 in each group). Dasatinib was quantified in plasma by UHPLC MS/MS assay. Noncompartmental analysis was used to compute the pharmacokinetic parameters, and immunoblot was used to assess the protein expression in the hepatic and intestinal tissues. RESULTS Following 7 days of naringenin pretreatment, the plasma mean concentration of dasatinib was enhanced compared with without pretreatment. In rats that were pretreated with naringenin, the pharmacokinetics of the orally administered dasatinib (25 mg/kg) was shown to be significantly different from that of dasatinib given without pretreatment (p < 0.05). There was a significant enhancement in pharmacokinetic parameters elimination half-life (T1/2), time to maximum concentration ( Tmax), maximum concentration )Cmax), area under the concentration-time curve (AUC0-t), area under the moment curve (AUMC0-∞), and mean residence time (MRT) by 28.41%, 50%, 103.54%, 72.64%, 115.08%, and 15.19%, respectively (p < 0.05) and suppression in elimination rate constant (Kel), volume of distribution (Vd), and clearance (CL) by 21.09%, 31.13%, and 46.25%, respectively, in comparison with dasatinib alone group (p < 0.05). The enhancement in dasatinib bioavailability and systemic exposure resulted from the significant inhibition of Cyp3a2, Mdr1/P-gp, and Bcrp1 expression and suppression of the dasatinib hepatic and intestinal metabolism, which enhanced the rate of dasatinib absorption and decreased its elimination. CONCLUSION Concurrent use of naringenin-containing supplements, herbs, or foods with dasatinib may cause serious and potentially life-threatening drug interactions. Further studies are necessary to determine the clinical significance of these findings.
Collapse
Affiliation(s)
- Mohammad Raish
- Department of Pharmaceutics, College of Pharmacy, King Saud University, 11451, Riyadh, Saudi Arabia.
| | - Ajaz Ahmad
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Badr Abdul Karim
- Department of Pharmaceutics, College of Pharmacy, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Yousef A Bin Jardan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Abdul Ahad
- Department of Pharmaceutics, College of Pharmacy, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Muzaffar Iqbal
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Khalid M Alkharfy
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Fahad I Al-Jenoobi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Omer Mansour Mohammed
- Experimental Animal Care Facility, College of Pharmacy, King Saud University, 11451, Riyadh, Saudi Arabia
| |
Collapse
|
5
|
Jiang X, Qu A, Xu X, Kuang H, Liu L, Xu C. Ultrasensitive detection of imatinib in human serum using a gold-based paper sensor. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1234:124001. [PMID: 38281369 DOI: 10.1016/j.jchromb.2024.124001] [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] [Received: 11/18/2023] [Revised: 12/30/2023] [Accepted: 01/04/2024] [Indexed: 01/30/2024]
Abstract
Imatinib is the tyrosine kinase inhibitor of choice for the treatment of chronic myeloid leukemia and gastrointestinal stromal tumors. However, imatinib has drawbacks such as drug resistance and significant differences in pharmacokinetics within patients. Therefore, a colloidal gold-based immunochromatographic assay (CG-IA) was developed for measuring and monitoring imatinib in human serum. An imatinib derivative containing carboxyl groups was used for the synthesis of the immunogen, and 4-(4-methyl-1-piperazinylmethyl) benzoic acid was selected as the hapten for the heterologous coating antigen. Next, a highly sensitive and specific monoclonal antibody (mAb), 2F7 was screened for the construction of a CG-IA, with an IC50 value of 0.091 ng/mL. For the qualification of imatinib in human serum, the visual limit of detection (vLOD) and cut-off values of the CG-IA were 2 and 20 ng/mL, respectively. For quantitative detection, the calculated LOD value of the CG-IA was 0.068 ng/mL, with a linearity range of 1.004 and 23.087 ng/mL. The recovery rate of spiked serum samples was between 88.24 % and 104.75 %. In addition, the concentration of imatinib in the serum samples from 10 patients was detected by CG-IA and revealed a good correlation with those from LC-MS/MS. These results indicated that the developed gold-based paper sensor could become an effective tool for the rapid monitoring of imatinib in human serum samples.
Collapse
Affiliation(s)
- Xiaoqian Jiang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, People's Republic of China; International Joint Research Laboratory for Biointerface and Biodetection, and School of Food Science and Technology, Jiangnan University, Wuxi, People's Republic of China
| | - Aihua Qu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, People's Republic of China; International Joint Research Laboratory for Biointerface and Biodetection, and School of Food Science and Technology, Jiangnan University, Wuxi, People's Republic of China
| | - Xinxin Xu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, People's Republic of China; International Joint Research Laboratory for Biointerface and Biodetection, and School of Food Science and Technology, Jiangnan University, Wuxi, People's Republic of China
| | - Hua Kuang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, People's Republic of China; International Joint Research Laboratory for Biointerface and Biodetection, and School of Food Science and Technology, Jiangnan University, Wuxi, People's Republic of China
| | - Liqiang Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, People's Republic of China; International Joint Research Laboratory for Biointerface and Biodetection, and School of Food Science and Technology, Jiangnan University, Wuxi, People's Republic of China.
| | - Chuanlai Xu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, People's Republic of China; International Joint Research Laboratory for Biointerface and Biodetection, and School of Food Science and Technology, Jiangnan University, Wuxi, People's Republic of China
| |
Collapse
|
6
|
Cafaro A, Baiardi G, Pigliasco F, Barco S, Mattioli F, Volpi S, Caorsi R, Gattorno M, Cangemi G. A Novel LC-MS/MS Method for Therapeutic Drug Monitoring of Baricitinib in Plasma of Pediatric Patients. Ther Drug Monit 2024; 46:67-72. [PMID: 37752637 DOI: 10.1097/ftd.0000000000001128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/13/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND Janus kinase inhibitors are antirheumatic immunosuppressive drugs that target intracellular Janus kinases (JAKs). Baricitinib is a selective and reversible orally administered JAK1/JAK2 inhibitor approved for treating rheumatoid arthritis, atopic dermatitis, and alopecia areata in adult patients. Expanded access to baricitinib has been approved for treating pediatric patients affected by rare Mendelian autoinflammatory diseases with type I interferon-mediated damage. Knowledge of the pharmacokinetic properties and target plasma levels of baricitinib in pediatric patients is limited. In this study, a novel LC-MS/MS method for measuring baricitinib in plasma, validated according to the ICH M10 guidelines, is presented. METHODS Sample preparation was performed by adding 10 µL of IS working solution (150 ng/mL) and 200 µL of MeOH to each plasma sample. Chromatographic separation was conducted using a Thermo Scientific Accucore Polar Premium column (50 mm × 2.1 mm, i.d. 2.6 m). This method was applied to 7 real anonymous plasma samples obtained from pediatric patients treated with baricitinib at IRCCS Istituto Giannina Gaslini (Genoa, Italy). Patients of both sexes had a median age of 14 years (range, 10-17 years). RESULTS The LC-MS/MS method resulted linear over wide concentration ranges (1.024-100 ng/mL) and was accurate and reproducible in the absence of matrix effects, allowing for robust, specific, and rapid quantification of baricitinib from a low amount of plasma (50 µL). The plasma concentration of baricitinib in the samples of the patients, expressed as mean ± SD, was 11.25 ± 10.86 ng/mL. CONCLUSIONS This novel LC-MS/MS method is suitable for the therapeutic drug monitoring of baricitinib and can help guide therapy optimization in pediatric patients.
Collapse
Affiliation(s)
- Alessia Cafaro
- Chromatography and Mass Spectrometry Section, Central Laboratory of Analysis, IRCCS Istituto Giannina Gaslini
| | - Giammarco Baiardi
- Department of Internal Medicine, Pharmacology, and Toxicology Unit, University of Genoa, Viale Benedetto XV
- Clinical Pharmacology Unit, EO Ospedali Galliera, Mura delle Cappuccine; and
| | - Federica Pigliasco
- Chromatography and Mass Spectrometry Section, Central Laboratory of Analysis, IRCCS Istituto Giannina Gaslini
| | - Sebastiano Barco
- Chromatography and Mass Spectrometry Section, Central Laboratory of Analysis, IRCCS Istituto Giannina Gaslini
| | - Francesca Mattioli
- Department of Internal Medicine, Pharmacology, and Toxicology Unit, University of Genoa, Viale Benedetto XV
- Clinical Pharmacology Unit, EO Ospedali Galliera, Mura delle Cappuccine; and
| | - Stefano Volpi
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Roberta Caorsi
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Marco Gattorno
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Giuliana Cangemi
- Chromatography and Mass Spectrometry Section, Central Laboratory of Analysis, IRCCS Istituto Giannina Gaslini
| |
Collapse
|
7
|
Tanabe KK, Zahrieh D, Strand CA, Hoshida Y, Flotte TJ, Della’Zanna G, Umar A, Chavin KD, Cleary S, Kubota N, Llovet JM, Patel T, Siegel C, Limburg PJ. Epidermal Growth Factor Receptor Inhibition With Erlotinib in Liver: Dose De-Escalation Pilot Trial as an Initial Step in a Chemoprevention Strategy. GASTRO HEP ADVANCES 2024; 3:426-439. [PMID: 39131140 PMCID: PMC11307768 DOI: 10.1016/j.gastha.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 01/19/2024] [Indexed: 08/13/2024]
Abstract
Background and Aims Effective approaches for prevention of hepatocellular carcinoma (HCC) will have a significant impact on HCC-related mortality. There are strong preclinical data and rationale to support targeting epidermal growth factor receptor (EGFR) for HCC chemoprevention. Small molecule inhibitors of EGFR have been Food and Drug Administration-approved for cancer therapy, which provides an opportunity to repurpose one of these drugs for chemoprevention of HCC. Unfortunately, the frequency of side effects associated with administration of these drugs at oncology doses renders them ineffective for chemoprevention. This clinical trial assesses whether lower doses of one of these inhibitors, erlotinib, still engages EGFR in the liver to block signaling (eg, EGFR phosphorylation). The objective of this clinical trial was determination of a safe and minimum effective dose of erlorinib for which ≥ 50% reduction phospho-EGFR immunohistochemical staining in the liver was observed. Methods Forty six participants were preregistered and 25 participants were registered in this multicenter trial. By dose de-escalation trial design, cohorts of participants received a 7-day course of erlotinib 75 mg/day, 50 mg/day or 25 mg/day with liver tissue acquisition prior to and after erlotinib. Results A ≥50% reduction phospho-EGFR immunohistochemical staining in the liver was observed in a minimum of 40% of participants (predetermined threshhold) at each of the dose levels. Erlotinib was very well tolerated with few side effects observed, particularly at the dose of 25 mg/day. Favorable modulation of the Prognostic Liver Signature was observed in participants who received erlotinib. Conclusion These data support the selection of erlotinib doses as low as 25 mg/day of for a longer intervention to assess for evidence of efficacy as an HCC chemoprevention drug (ClinicalTrials.govNCT02273362).
Collapse
Affiliation(s)
- Kenneth K. Tanabe
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - David Zahrieh
- Division of Clinical Trial and Biostatistics, Mayo Clinic, Rochester, New York
| | - Carrie A. Strand
- Division of Clinical Trial and Biostatistics, Mayo Clinic, Rochester, New York
| | - Yujin Hoshida
- Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Thomas J. Flotte
- Mayo Clinic Pathology Research Core, Mayo Clinic, Rochester, New York
| | - Gary Della’Zanna
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Asad Umar
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Kenneth D. Chavin
- Department of Surgery, UH Cleveland Medical Center and Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Sean Cleary
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, New York
| | - Naoto Kubota
- Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Josep M. Llovet
- Mount Sinai Liver Cancer Program, Division of Liver Diseases, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Liver Unit, Translational Research in Hepatic Oncology, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Tushar Patel
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| | | | - Paul J. Limburg
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, New York
| |
Collapse
|
8
|
Adawi DH, Fredj NB, Al-Barghouthi A, Dridi I, Lubada M, Manasra M, Aouam K. Pharmacokinetics of Imatinib Mesylate and Development of Limited Sampling Strategies for Estimating the Area under the Concentration-Time Curve of Imatinib Mesylate in Palestinian Patients with Chronic Myeloid Leukemia. Eur J Drug Metab Pharmacokinet 2024; 49:43-55. [PMID: 38006575 DOI: 10.1007/s13318-023-00868-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUND AND OBJECTIVE Imatinib is a tyrosine kinase inhibitor used in the treatment of chronic myeloid leukemia (CML). The area under the concentration-time curve (AUC) is a pharmacokinetic parameter that symbolizes overall exposure to a drug, which is correlated with complete cytogenetic and treatment responses to imatinib, as well as its side effects in patients with CML. The limited sampling strategy (LSS) is considered a sufficiently precise and practical method that can be used to estimate pharmacokinetic parameters such as AUC, without the need for frequent, costly, and inconvenient blood sampling. This study aims to investigate the pharmacokinetic parameters of imatinib, develop and validate a reliable and practical LSS for estimating imatinib AUC0-24, and determine the optimum sampling points for predicting the imatinib AUC after the administration of once-daily imatinib in Palestinian patients with CML. METHOD Pharmacokinetic profiles, involving six blood samples collected during a 24-h dosing interval, were obtained from 25 Palestinian patients diagnosed with CML who had been receiving imatinib for at least 7 days and had reached a steady-state level. Imatinib AUC0-24 was calculated using the trapezoidal rule, and linear regression analysis was performed to assess the relationship between measured AUC0-24 and concentrations at each sampling time. All developed models were analyzed to determine their effectiveness in predicting AUC0-24 and to identify the optimal sampling time. To evaluate predictive performance, two error indices were employed: the percentage of root mean squared error (% RMSE) and the mean predictive error (% MPE). Bland and Altman plots, along with mountain plots, were utilized to assess the agreement between measured and predicted AUC. RESULTS Among the one-timepoint estimations, predicted AUC0-24 based on concentration of imatinib at the eighth hour after administration (C8-predicted AUC0-24) demonstrated the highest correlation with the measured AUC (r2 = 0.97, % RMSE = 6.3). In two-timepoint estimations, the model consisting of C0 and C8 yielded the highest correlation between predicted and measured imatinib AUC (r2 = 0.993 and % RMSE = 3.0). In three-timepoint estimations, the combination of C0, C1, and C8 provided the most robust multilinear regression for predicting imatinib AUC0-24 (r2 = 0.996, % RMSE = 2.2). This combination also outperformed all other models in predicting AUC. The use of a two-timepoint limited sampling strategy (LSS) for predicting AUC was found to be reliable and practical. While C0/C8 exhibited the highest correlation, the use of C0/C4 could be a more practical and equally accurate choice. Therapeutic drug monitoring of imatinib based on C0 can also be employed in routine clinical practice owing to its reliability and practicality. CONCLUSION The LSS using one timepoint, especially C0, can effectively predict imatinib AUC. This approach offers practical benefits in optimizing dose regimens and improving adherence. However, for more precise estimation of imatinib AUC, utilizing two- or three-timepoint concentrations is recommended over relying on a single point.
Collapse
Affiliation(s)
- Deema Hilmi Adawi
- Department of Pharmacology, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia.
- Department of Pharmacology, Palestinian Ministry of Health, Ramallah, Palestine.
| | - Nadia Ben Fredj
- Department of Pharmacology, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Ahmad Al-Barghouthi
- Department of Pharmacology, Palestinian Ministry of Health, Ramallah, Palestine
| | - Ichrack Dridi
- Department of Pharmacology, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Mustafa Lubada
- Department of Pharmacology, Palestinian Ministry of Health, Ramallah, Palestine
| | - Mohammad Manasra
- Department of Pharmacology, Palestinian Ministry of Health, Ramallah, Palestine
| | - Karim Aouam
- Department of Pharmacology, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| |
Collapse
|
9
|
Bellouard M, Donadieu J, Thiebot P, Giroux Leprieur E, Saiag P, Etting I, Dugues P, Abe E, Alvarez JC, Larabi IA. Validation of Liquid Chromatography Coupled with Tandem Mass Spectrometry for the Determination of 12 Tyrosine Kinase Inhibitors (TKIs) and Their Application to Therapeutic Drug Monitoring in Adult and Pediatric Populations. Pharmaceutics 2023; 16:5. [PMID: 38276485 PMCID: PMC10818921 DOI: 10.3390/pharmaceutics16010005] [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: 11/15/2023] [Revised: 12/12/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are used as targeted cancer therapies in adults and have an off-label pediatric application for the treatment of Langerhans cell histiocytosis. A multitarget LC-MS/MS method was developed and validated for the determination of alectinib, alectinib-M4, binimetinib, cobimetinib, crizotinib, dabrafenib, encorafenib, imatinib, lorlatinib, osimertinib, AZ5104, and trametinib. A total of 150 µL of internal standard methanolic solution was added to 50 µL of plasma sample to precipitate proteins. After centrifugation, 10 µL of the supernatant was injected into the chromatographic system. The chromatographic separation was conducted on a Kinetex C18 Polar column with a gradient of 2 mM ammonium formate in 0.1% formic acid and acetonitrile over 5 min. Limits of detection and quantification, linearity, accuracy, precision, selectivity, carryover, matrix effect, recovery, and stability were evaluated and satisfied EMA guidelines on bioanalytical methods. This method has been successfully applied to the therapeutic drug monitoring (TDM) of adults with melanoma and lung cancer, as well as children with histiocytosis, to improve the pharmacokinetic data for these drugs, with the aim of enhancing the therapeutic management and follow-up of patients. Blood concentrations of trametinib and binimetinib were different in the two groups, highlighting the age-related inter-individual variability of these molecules and the need for TDM.
Collapse
Affiliation(s)
- Marie Bellouard
- Toxicology Laboratory, Raymond Poincaré Hospital, AP-HP, 92380 Garches, France; (I.E.); (P.D.); (E.A.); (I.-A.L.); (J.-C.A.)
- Pediatric Hemato-Oncology Department, Trousseau Hospital, AP-HP, 75012 Paris, France;
| | - Jean Donadieu
- Pediatric Hemato-Oncology Department, Trousseau Hospital, AP-HP, 75012 Paris, France;
| | - Pauline Thiebot
- Toxicology Laboratory, Lariboisière Hospital, AP-HP, 75010 Paris, France;
| | | | - Philippe Saiag
- Dermatology Department, Ambroise Paré Hospital, AP-HP, 92100 Boulogne-Billancourt, France;
| | - Isabelle Etting
- Toxicology Laboratory, Raymond Poincaré Hospital, AP-HP, 92380 Garches, France; (I.E.); (P.D.); (E.A.); (I.-A.L.); (J.-C.A.)
| | - Pamela Dugues
- Toxicology Laboratory, Raymond Poincaré Hospital, AP-HP, 92380 Garches, France; (I.E.); (P.D.); (E.A.); (I.-A.L.); (J.-C.A.)
- Inserm U-1018, CESP, Team MOODS, Plateform MasSpecLab, Paris-Saclay/Versailles University, 78180 Montigny-le-Bretonneux, France
| | - Emuri Abe
- Toxicology Laboratory, Raymond Poincaré Hospital, AP-HP, 92380 Garches, France; (I.E.); (P.D.); (E.A.); (I.-A.L.); (J.-C.A.)
| | - Jean-Claude Alvarez
- Toxicology Laboratory, Raymond Poincaré Hospital, AP-HP, 92380 Garches, France; (I.E.); (P.D.); (E.A.); (I.-A.L.); (J.-C.A.)
- Inserm U-1018, CESP, Team MOODS, Plateform MasSpecLab, Paris-Saclay/Versailles University, 78180 Montigny-le-Bretonneux, France
| | - Islam-Amine Larabi
- Toxicology Laboratory, Raymond Poincaré Hospital, AP-HP, 92380 Garches, France; (I.E.); (P.D.); (E.A.); (I.-A.L.); (J.-C.A.)
- Inserm U-1018, CESP, Team MOODS, Plateform MasSpecLab, Paris-Saclay/Versailles University, 78180 Montigny-le-Bretonneux, France
| |
Collapse
|
10
|
Shahid M, Ahmad A, Raish M, Bin Jardan YA, Alkharfy KM, Ahad A, Abul Kalam M, Ahmad Ansari M, Iqbal M, Ali N, Al-Jenoobi FI. Herb-drug interaction: Effect of sinapic acid on the pharmacokinetics of dasatinib in rats. Saudi Pharm J 2023; 31:101819. [PMID: 37860687 PMCID: PMC10582055 DOI: 10.1016/j.jsps.2023.101819] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/02/2023] [Indexed: 10/21/2023] Open
Abstract
Dasatinib (DAS) is a narrow therapeutic index drug and novel oral multitarget inhibitor of tyrosine kinase and approved for the first-line therapy for chronic myelogenous leukemia (CML) and Philadelphia chromosome (Ph + ) acute lymphoblastic leukemia (ALL). DAS, a known potent substrate of cytochrome (CYP) 3A, P-glycoprotein (Pgp) and breast cancer resistance protein (BCRP) and is subject to auto-induction. The dietary supplementation of sinapic acid (SA) or concomitant use of SA containing herbs/foods may alter the pharmacokinetics as well as pharmacodynamics of DAS, that may probably lead to potential interactions. Protein expression in rat hepatic and intestinal tissues, as well as the in vivo pharmacokinetics of DAS and the roles of CYP3 A2 and drug transporters Pgp-MDR1 and BCPR/ABCG2, suggested a likely interaction mechanism. The single dose of DAS (25 mg/kg) was given orally to rats with or without SA pretreatment (20 mg/kg p.o. per day for 7 days, n = 6). The plasma concentration of DAS was estimated by using Ultra-High-Performance Liquid Chromatography Mass spectrometry (UHPLC-MS/MS). The in vivo pharmacokinetics and protein expression study demonstrate that SA pretreatment has potential to alter the DAS pharmacokinetics. The increase in Cmax, AUC and AUMC proposes increase in bioavailability and rate of absorption via modulation of CYP3 A2, PgP-MDR1 and BCPR/ABCG2 protein expression. Thus, the concomitant use of SA alone or with DAS may cause serious life-threatening drug interactions.
Collapse
Affiliation(s)
- Mudassar Shahid
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ajaz Ahmad
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad Raish
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Yousef A Bin Jardan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Khalid M. Alkharfy
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdul Ahad
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohd Abul Kalam
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mushtaq Ahmad Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Muzaffer Iqbal
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Naushad Ali
- Quality Assurance Unit, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fahad I. Al-Jenoobi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
11
|
Cafaro A, Conti M, Pigliasco F, Barco S, Bandettini R, Cangemi G. Biological Fluid Microsampling for Therapeutic Drug Monitoring: A Narrative Review. Biomedicines 2023; 11:1962. [PMID: 37509602 PMCID: PMC10377272 DOI: 10.3390/biomedicines11071962] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/04/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Therapeutic drug monitoring (TDM) is a specialized area of laboratory medicine which involves the measurement of drug concentrations in biological fluids with the aim of optimizing efficacy and reducing side effects, possibly modifying the drug dose to keep the plasma concentration within the therapeutic range. Plasma and/or whole blood, usually obtained by venipuncture, are the "gold standard" matrices for TDM. Microsampling, commonly used for newborn screening, could also be a convenient alternative to traditional sampling techniques for pharmacokinetics (PK) studies and TDM, helping to overcome practical problems and offering less invasive options to patients. Although technical limitations have hampered the use of microsampling in these fields, innovative techniques such as 3-D dried blood spheroids, volumetric absorptive microsampling (VAMS), dried plasma spots (DPS), and various microfluidic devices (MDS) can now offer reliable alternatives to traditional samples. The application of microsampling in routine clinical pharmacology is also hampered by the need for instrumentation capable of quantifying analytes in small volumes with sufficient sensitivity. The combination of microsampling with high-sensitivity analytical techniques, such as liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS), is particularly effective in ensuring high accuracy and sensitivity from very small sample volumes. This manuscript provides a critical review of the currently available microsampling devices for both whole blood and other biological fluids, such as plasma, urine, breast milk, and saliva. The purpose is to provide useful information in the scientific community to laboratory personnel, clinicians, and researchers interested in implementing the use of microsampling in their routine clinical practice.
Collapse
Affiliation(s)
- Alessia Cafaro
- Chromatography and Mass Spectrometry Section, Central Laboratory of Analysis, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Matteo Conti
- Public Health Department, Imola Local Unit, Regione Emilia-Romagna Healthcare Service, 40026 Imola, Italy
| | - Federica Pigliasco
- Chromatography and Mass Spectrometry Section, Central Laboratory of Analysis, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Sebastiano Barco
- Chromatography and Mass Spectrometry Section, Central Laboratory of Analysis, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Roberto Bandettini
- Chromatography and Mass Spectrometry Section, Central Laboratory of Analysis, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Giuliana Cangemi
- Chromatography and Mass Spectrometry Section, Central Laboratory of Analysis, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| |
Collapse
|
12
|
Raish M, Ahmad A, Shahid M, Jardan YAB, Ahad A, Kalam MA, Ansari MA, Iqbal M, Ali N, Alkharfy KM, Al-Jenoobi FI. Effects of Apigenin on Pharmacokinetics of Dasatinib and Probable Interaction Mechanism. Molecules 2023; 28:molecules28041602. [PMID: 36838589 PMCID: PMC9964503 DOI: 10.3390/molecules28041602] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/19/2023] [Accepted: 01/27/2023] [Indexed: 02/10/2023] Open
Abstract
Dasatinib (DAS), a narrow-therapeutic index drug, Bcr-Abl, and Src family kinases multitarget inhibitor have been approved for chronic myelogenous leukemia (CML) and Ph-positive acute lymphocytic leukemia (Ph+ ALL). Apigenin (APG) has a long history of human usage in food, herbs, health supplements, and traditional medicine, and it poses low risk of damage. The concomitant use of APG containing herbs/foods and traditional medicine may alter the pharmacokinetics of DAS, that probably lead to possible herb-drug interactions. The pharmacokinetic interaction of APG pretreatment with DAS in rat plasma following single and co-oral dosing was successfully deliberated using the UPLC-MS/MS method. The in vivo pharmacokinetics and protein expression of CYP3A2, Pgp-MDR1, and BCPR/ABCG2 demonstrate that APG pretreatment has potential to drastically changed the DAS pharmacokinetics where escalation in the Cmax, AUC(0-t), AUMC(0-inf_obs), T1/2, Tmax, and MRT and reduction in Kel, Vd, and Cl significantly in rats pretreated with APG 40 mg/kg, thus escalating systemic bioavailability and increasing the rate of absorption via modulation of CYP3A2, Pgp-MDR1, and BCPR/ABCG2 protein expression. Therefore, the concomitant consumption of APG containing food or traditional herb with DAS may cause serious life-threatening drug interactions and more systematic clinical study on herb-drug interactions is required, as well as adequate regulation in herbal safety and efficacy.
Collapse
Affiliation(s)
- Mohammad Raish
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
- Correspondence:
| | - Ajaz Ahmad
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mudassar Shahid
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Yousef A. Bin Jardan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdul Ahad
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohd Abul Kalam
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mushtaq Ahmad Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Muzaffar Iqbal
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Naushad Ali
- Quality Assurance Unit, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Khalid M. Alkharfy
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fahad I. Al-Jenoobi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
13
|
Guo ZX, Wu YE, Shi HY, van den Anker J, Liang P, Zheng Y, Zhao XW, Feng R, Zhao W. A liquid chromatography-tandem mass spectrometry method for simultaneous quantification of thirty-nine tyrosine kinase inhibitors in human plasma. J Pharm Biomed Anal 2023; 224:115159. [PMID: 36442459 DOI: 10.1016/j.jpba.2022.115159] [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: 09/16/2022] [Revised: 10/26/2022] [Accepted: 11/09/2022] [Indexed: 11/23/2022]
Abstract
Currently, the use of targeted drugs such as tyrosine kinase inhibitors (TKIs) plays an important role in clinical therapy. As the number of approved TKIs continues to increase, existing analysis methods will not be able to meet the growing needs, and will hamper the development of therapeutic drug monitoring (TDM) of TKIs. Based on LC-MS/MS technology, this study tends to develop and validate a multi-component analysis method for simultaneous determination of the concentrations of 39 TKIs in plasma. Spiked plasma was blended with isotope labelled internal standards, and injected into the LC-MS/MS system after protein precipitation by acetonitrile. Chromatographic separation was achieved using an ODS-4 column with gradient elution of formic acid/water (1:1000; v/v) and acetonitrile. Analytes detection was conducted in positive ionisation mode using MRM. The total run time was 8 min. The method validation was conducted by assessing the following parameters: selectivity, linearity and the lower limit of qualification, accuracy and precision, stability, matrix effect and recovery. The concentrations of 39 TKIs showed good linearity within the range of their respective standard curves in plasma, the accuracy of all quality control samples ranged from 85.9% to 114.1%, and the precision was lower than 13.3%. The extraction recovery ranged from 92.6% to 114.7%, and the matrix effect of plasma was lower than 11.3%. This new method was successfully developed, can be used for the determination of drug concentrations in multiple patients with different kinds of TKIs, and will therefore be suitable for TDM of 39 TKIs.
Collapse
Affiliation(s)
- Zi-Xuan Guo
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China; Department of Pharmacy, China-Japan Friendship Hospital, Beijing, China
| | - Yue-E Wu
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hai-Yan Shi
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
| | - John van den Anker
- Division of Clinical Pharmacology, Children's National Hospital, Washington DC, USA; Departments of Pediatrics, Pharmacology & Physiology, Genomics & Precision Medicine, the George Washington University School of Medicine and Health Sciences, Washington DC, USA; Department of Paediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland
| | - Ping Liang
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ying Zheng
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xue-Wei Zhao
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Rui Feng
- Department of Pharmacy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Wei Zhao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China; NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Shandong University, Jinan, China.
| |
Collapse
|
14
|
Neuwirt E, Magnani G, Ćiković T, Wöhrle S, Fischer L, Kostina A, Flemming S, Fischenich NJ, Saller BS, Gorka O, Renner S, Agarinis C, Parker CN, Boettcher A, Farady CJ, Kesselring R, Berlin C, Backofen R, Rodriguez-Franco M, Kreutz C, Prinz M, Tholen M, Reinheckel T, Ott T, Groß CJ, Jost PJ, Groß O. Tyrosine kinase inhibitors can activate the NLRP3 inflammasome in myeloid cells through lysosomal damage and cell lysis. Sci Signal 2023; 16:eabh1083. [PMID: 36649377 DOI: 10.1126/scisignal.abh1083] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Inflammasomes are intracellular protein complexes that promote an inflammatory host defense in response to pathogens and damaged or neoplastic tissues and are implicated in inflammatory disorders and therapeutic-induced toxicity. We investigated the mechanisms of activation for inflammasomes nucleated by NOD-like receptor (NLR) protiens. A screen of a small-molecule library revealed that several tyrosine kinase inhibitors (TKIs)-including those that are clinically approved (such as imatinib and crizotinib) or are in clinical trials (such as masitinib)-activated the NLRP3 inflammasome. Furthermore, imatinib and masitinib caused lysosomal swelling and damage independently of their kinase target, leading to cathepsin-mediated destabilization of myeloid cell membranes and, ultimately, cell lysis that was accompanied by potassium (K+) efflux, which activated NLRP3. This effect was specific to primary myeloid cells (such as peripheral blood mononuclear cells and mouse bone marrow-derived dendritic cells) and did not occur in other primary cell types or various cell lines. TKI-induced lytic cell death and NLRP3 activation, but not lysosomal damage, were prevented by stabilizing cell membranes. Our findings reveal a potential immunological off-target of some TKIs that may contribute to their clinical efficacy or to their adverse effects.
Collapse
Affiliation(s)
- Emilia Neuwirt
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Giovanni Magnani
- Institute of Clinical Chemistry and Pathobiochemistry, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Tamara Ćiković
- Institute of Clinical Chemistry and Pathobiochemistry, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany.,Center for Translational Cancer Research (TranslaTUM), Technical University of Munich, 81675 Munich, Germany
| | - Svenja Wöhrle
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Larissa Fischer
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Anna Kostina
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Stephan Flemming
- Bioinformatics Group, Faculty of Engineering, University of Freiburg, 79110 Freiburg, Germany
| | - Nora J Fischenich
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Benedikt S Saller
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Oliver Gorka
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Steffen Renner
- Novartis Institutes for BioMedical Research, 4056 Basel, Switzerland
| | - Claudia Agarinis
- Novartis Institutes for BioMedical Research, 4056 Basel, Switzerland
| | | | - Andreas Boettcher
- Novartis Institutes for BioMedical Research, 4056 Basel, Switzerland
| | | | - Rebecca Kesselring
- Department for General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) 69120 Heidelberg, Germany
| | - Christopher Berlin
- Department for General and Visceral Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) 69120 Heidelberg, Germany
| | - Rolf Backofen
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany.,Bioinformatics Group, Faculty of Engineering, University of Freiburg, 79110 Freiburg, Germany
| | | | - Clemens Kreutz
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany.,Institute of Medical Biometry and Statistics (IMBI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Martina Tholen
- Institute for Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Thomas Reinheckel
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) 69120 Heidelberg, Germany.,Institute for Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Thomas Ott
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany.,Faculty of Biology, Cell Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Christina J Groß
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Philipp J Jost
- Division of Clinical Oncology, Department of Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Olaf Groß
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
15
|
Gu EM, Liu YN, Pan L, Hu Y, Ye X, Luo P. A high throughput method for Monitoring of Sorafenib, regorafenib, cabozantinib and their metabolites with UPLC-MS/MS in rat plasma. Front Pharmacol 2022; 13:955263. [PMID: 36160432 PMCID: PMC9493307 DOI: 10.3389/fphar.2022.955263] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 08/04/2022] [Indexed: 11/18/2022] Open
Abstract
As multi-targeted tyrosine kinase inhibitors, sorafenib, regorafenib and cabozantinib are widely used in hepatocellular carcinoma (HCC) for systemic therapies with anti-proliferative and anti-angiogenic effects. Nevertheless, adverse effects or insufficient efficacy appear frequently due to the plasma concentration with individual variability of these drugs. To ensure the curative effect and safety by therapeutic drug monitoring (TDM), this study developed a high throughput method to quantify sorafenib, regorafenib, cabozantinib and their active metabolites in plasma simultaneously. The chromatographic separation analysis achievement was performed on a Waters-ACQUITY UPLC BEH C18 column by UPLC-MS/MS system using a gradient elution of solvent A (acetonitrile) and solvent B (water with 0.1% formic acid) in 3.0 min. This method presented satisfactory results of specificity, precision (the intra-day coefficient of variation was between 2.5% and 6.6%, and the inter-day coefficient of variation was between 4.0% and 11.1%) and accuracy (within ±15% for intra-day and inter-day), as well as the stability under certain conditions, the matrix effect in plasma, and extraction recovery (75.6%–94.4%). The linearity of each analyte in the proper concentration scope indicated excellent. This study strictly complied with the performance rules of assay validation in biological medium proposed by FDA and was successfully applied to the pharmacokinetic study in rats. Thus, it would be an advantageous option to research the relationship between concentration-efficacy and concentration-toxic in HCC patients who were supposed to take these medications.
Collapse
Affiliation(s)
- Er-Min Gu
- The First People’s Hospital of Jiashan, Jiaxing, Zhejiang, China
| | - Ya-Nan Liu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lvjun Pan
- The First People’s Hospital of Jiashan, Jiaxing, Zhejiang, China
| | - Yingying Hu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuemei Ye
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- *Correspondence: Pingping Luo, ; Xuemei Ye,
| | - Pingping Luo
- The People’s Hospital of Lishui, Lishui, Zhejiang, China
- *Correspondence: Pingping Luo, ; Xuemei Ye,
| |
Collapse
|
16
|
Zhang M, Tajima S, Suetsugu K, Hirota T, Tsuchiya Y, Yamauchi T, Yoshimoto G, Miyamoto T, Egashira N, Akashi K, Ieiri I. Development and Validation of an LC-MS/MS Method to Quantify Gilteritinib and Its Clinical Application in Patients With FLT3 Mutation-Positive Acute Myelogenous Leukemia. Ther Drug Monit 2022; 44:592-596. [PMID: 35149666 DOI: 10.1097/ftd.0000000000000971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/21/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Gilteritinib, a novel oral tyrosine kinase inhibitor, is used to treat acute myeloid leukemia (AML) with FMS-like tyrosine kinase-3 (FLT3) mutations. Therapeutic drug monitoring (TDM) of gilteritinib is important for improving clinical outcomes and ensuring safety. Therefore, this study aimed to develop a simplified method for quantifying gilteritinib in human plasma using liquid chromatography-tandem mass spectrometry. METHODS Liquid chromatography was performed by using an Acquity BEH C18 column (50 mm × 2.1 mm, 1.7 μm) and a gradient elution with 0.1% formic acid in water (A) and acetonitrile (B). Detection was performed by using a Shimadzu tandem mass spectrometer through multiple reaction monitoring in the positive-ion mode. RESULTS The developed method enabled quantification of gilteritinib in 4 minutes and was validated by evaluating selectivity, calibration curve (10-1000 ng/mL, r 2 > 0.99), a lower limit of quantification (LLOQ), accuracy (overall bias -4.2% to 1.9%), precision (intraday CV ≤ 7.9%; interday CV ≤ 13.6%), carryover, recovery, matrix effect, dilution integrity, and stability according to the US Food and Drug Administration (FDA) guidelines. This method was successfully applied to the TDM of gilteritinib trough concentrations in 3 patients with AML. CONCLUSIONS The developed method fulfilled the FDA guideline criteria and can easily be implemented to facilitate TDM in patients receiving gilteritinib in a clinical setting.
Collapse
Affiliation(s)
- Mengyu Zhang
- Department of Clinical Pharmacology and Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Soichiro Tajima
- Department of Pharmacy, Kyushu University Hospital, Fukuoka, Japan
| | | | - Takeshi Hirota
- Department of Pharmacy, Kyushu University Hospital, Fukuoka, Japan
| | - Yuichi Tsuchiya
- Department of Pharmacy, Kyushu University Hospital, Fukuoka, Japan
| | - Takuji Yamauchi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Goichi Yoshimoto
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Hematology, Saga-Ken Medical Centre Koseikan, Saga, Japan ; and
| | - Toshihiro Miyamoto
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Hematology, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan
| | - Nobuaki Egashira
- Department of Clinical Pharmacology and Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
- Department of Pharmacy, Kyushu University Hospital, Fukuoka, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ichiro Ieiri
- Department of Clinical Pharmacology and Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
- Department of Pharmacy, Kyushu University Hospital, Fukuoka, Japan
| |
Collapse
|
17
|
Determination of Osimertinib, Aumolertinib, and Furmonertinib in Human Plasma for Therapeutic Drug Monitoring by UPLC-MS/MS. Molecules 2022; 27:molecules27144474. [PMID: 35889345 PMCID: PMC9325192 DOI: 10.3390/molecules27144474] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 02/06/2023] Open
Abstract
The third-generation epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs), osimertinib, aumolertinib, and furmonertinib represent a new treatment option for patients with EGFR p.Thr790 Met (T790 M)-mutated non-small cell lung cancer (NSCLC). Currently, there are no studies reporting the simultaneous quantification of these three drugs. A simple ultra-performance liquid chromatography–tandem mass spectrometry (UPLC-MS/MS) method was developed and validated for the simultaneous quantitative determination of osimertinib, aumolertinib, and furmonertinib concentrations in human plasma, and it was applied for therapeutic drug monitoring (TDM). Plasma samples were processed using the protein precipitation method (acetonitrile). A positive ion monitoring mode was used for detecting analytes. D3-Sorafenib was utilized as the internal standard (IS), and the mobile phases were acetonitrile (containing 0.1% formic acid) and water with gradient elution on an XSelect HSS XP column (2.1 mm × 100.0 mm, 2.5 µm, Waters, Milford, MA, USA) at a flow rate of 0.5 mL·min−1. The method’s selectivity, precision (coefficient of variation of intra-day and inter-day ≤ 6.1%), accuracy (95.8–105.2%), matrix effect (92.3–106.0%), extraction recovery, and stability results were acceptable according to the guidelines. The linear ranges were 5–500 ng·mL−1, 2–500 ng·mL−1, and 0.5–200 ng·mL−1 for osimertinib, aumolertinib, and furmonertinib, respectively. The results show that the method was sensitive, reliable, and simple and that it could be successfully applied to simultaneously determine the osimertinib, aumolertinib, and furmonertinib blood concentrations in patients. These findings support using the method for TDM, potentially reducing the incidence of dosing blindness and adverse effects due to empirical dosing and inter-patient differences.
Collapse
|
18
|
Verougstraete N, Stove V, Verstraete AG, Stove CP. Therapeutic Drug Monitoring of Tyrosine Kinase Inhibitors Using Dried Blood Microsamples. Front Oncol 2022; 12:821807. [PMID: 35392223 PMCID: PMC8980857 DOI: 10.3389/fonc.2022.821807] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/01/2022] [Indexed: 12/14/2022] Open
Abstract
Therapeutic drug monitoring (TDM) of tyrosine kinase inhibitors (TKIs) is not yet performed routinely in the standard care of oncology patients, although it offers a high potential to improve treatment outcome and minimize toxicity. TKIs are perfect candidates for TDM as they show a relatively small therapeutic window, a wide inter-patient variability in pharmacokinetics and a correlation between drug concentration and effect. Moreover, most of the available TKIs are susceptible to various drug-drug interactions and medication adherence can be checked by performing TDM. Plasma, obtained via traditional venous blood sampling, is the standard matrix for TDM of TKIs. However, the use of plasma poses some challenges related to sampling and stability. The use of dried blood microsamples can overcome these limitations. Collection of samples via finger-prick is minimally invasive and considered convenient and simple, enabling sampling by the patients themselves in their home-setting. The collection of small sample volumes is especially relevant for use in pediatric populations or in pharmacokinetic studies. Additionally, working with dried matrices improves compound stability, resulting in convenient and cost-effective transport and storage of the samples. In this review we focus on the different dried blood microsample-based methods that were used for the quantification of TKIs. Despite the many advantages associated with dried blood microsampling, quantitative analyses are also associated with some specific difficulties. Different methodological aspects of microsampling-based methods are discussed and applied to TDM of TKIs. We focus on sample preparation, analytics, internal standards, dilution of samples, external quality controls, dried blood spot specific validation parameters, stability and blood-to-plasma conversion methods. The various impacts of deviating hematocrit values on quantitative results are discussed in a separate section as this is a key issue and undoubtedly the most widely discussed issue in the analysis of dried blood microsamples. Lastly, the applicability and feasibility of performing TDM using microsamples in a real-life home-sampling context is discussed.
Collapse
Affiliation(s)
- Nick Verougstraete
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium.,Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Veronique Stove
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium.,Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Alain G Verstraete
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium.,Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Christophe P Stove
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
19
|
Simultaneous Determination of Two Tyrosine Kinase Inhibitors in Tablets by HPLC-MS analysis. CURRENT HEALTH SCIENCES JOURNAL 2022; 48:75-80. [PMID: 35911934 PMCID: PMC9289587 DOI: 10.12865/chsj.48.01.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 02/08/2022] [Indexed: 11/18/2022]
Abstract
The class of tyrosine kinase inhibitors (TKIs) is represented by a group of compounds which are currently used in the treatment of different types of cancer. These oral medicines present a narrow therapeutic index and a large inter-and intra-individual variability. Within this work, a simple, accurate and rapid reversed phase ultra-high-performance liquid chromatographic (RP-UHPLC) method with mass spectrometric (MS) detection for simultaneous analysis of two TKIs, ibrutinib and ruxolitinib, using pentoxifylline as internal standard (IS) in tablet dosage forms is presented. The separation was carried out on a Waters (Milford, Massachusetts, USA) Arc System coupled with a Waters QDa mass detector. The column used was a Waters CORTECS C18 (4.6×50mm, 2.7μm); a gradient elution was carried out using a mixture of ammonium formate 10 mM aqueous solution and acetonitrile. The flow rate of the mobile phase was set to 0.5mL/min. The column temperature was equilibrated to 40°C. The injected volume was 5μL. All samples were kept at 20°C during the entire analysis. Mass spectra were recorded in positive ionization mode in the range of m/z 100-400 for ruxolitinib and m/z 100-500 for ibrutinib. Quantification was established in single ion recording (SIR) mode for each compound, using pentoxifylline as internal standard. The method was validated according to International Guidelines in terms of stability, limit of detection, limit of quantitation, linearity, precision and accuracy. The validated method can be successfully applied for simultaneous determination of TKIs in tablet dosage forms.
Collapse
|
20
|
Roušarová J, Šíma M, Slanař O. Therapeutic Drug Monitoring of Protein Kinase Inhibitors in Breast Cancer Patients. Prague Med Rep 2021; 122:243-256. [PMID: 34924102 DOI: 10.14712/23362936.2021.22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Protein kinase inhibitors (PKIs) represent up-to-date therapeutic approach in breast cancer treatment. Although cancer is a rapidly progressive disease, many substances, including PKIs, are usually used at fixed doses without regard to each patient's individuality. Therapeutic drug monitoring (TDM) is a tool that allows individualization of therapy based on drug plasma levels. For TDM conduct, exposure-response relationships of drug substances are required. The pharmacokinetic data and exposure-response evidence supporting the use of TDM for 6 PKIs used in breast cancer treatment, one of the most common female tumour diseases, are discussed in this review.
Collapse
Affiliation(s)
- Jaroslava Roušarová
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.
| | - Martin Šíma
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Ondřej Slanař
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| |
Collapse
|
21
|
Abstract
BACKGROUND AND OBJECTIVE Gilteritinib is a novel, highly selective tyrosine kinase inhibitor approved in the USA, Canada, Europe, Brazil, Korea, and Japan for the treatment of FLT3 mutation-positive acute myeloid leukemia. This article describes the clinical pharmacokinetic profile of gilteritinib. METHODS The pharmacokinetic profile of gilteritinib was assessed from five clinical studies. RESULTS Dose-proportional pharmacokinetics was observed following once-daily gilteritinib administration (dose range 20-450 mg). Median maximum concentration was reached 2-6 h following single and repeat dosing of gilteritinib; mean elimination half-life was 113 h. Elimination was primarily via feces. Exposure to gilteritinib was comparable under fasted and fed conditions. Gilteritinib is primarily metabolized via cytochrome P450 (CYP) 3A4; coadministration of gilteritinib with itraconazole (a strong P-glycoprotein inhibitor and CYP3A4 inhibitor) or rifampicin (a strong P-glycoprotein inducer and CYP3A inducer) significantly affected the gilteritinib pharmacokinetic profile. No clinically relevant interactions were observed when gilteritinib was coadministered with midazolam (a CYP3A4 substrate) or cephalexin (a multidrug and toxin extrusion 1 substrate). Unbound gilteritinib exposure was similar between subjects with hepatic impairment and normal hepatic function. CONCLUSIONS Gilteritinib exhibits a dose-proportional pharmacokinetic profile in healthy subjects and in patients with relapsed/refractory acute myeloid leukemia. Gilteritinib exposure is not significantly affected by food. Moderate-to-strong CYP3A inhibitors demonstrated a significant effect on gilteritinib exposure. Coadministration of gilteritinib with CYP3A4 or multidrug and toxin extrusion 1 substrates did not impact substrate concentrations. Unbound gilteritinib was comparable between subjects with hepatic impairment and normal hepatic function; dose adjustment is not warranted for patients with hepatic impairment. CLINICAL TRIAL REGISTRATION NCT02014558, NCT02456883, NCT02571816.
Collapse
|
22
|
Panetta JC, Campagne O, Gartrell J, Furman W, Stewart CF. Pharmacokinetically guided dosing of oral sorafenib in pediatric hepatocellular carcinoma: A simulation study. Clin Transl Sci 2021; 14:2152-2160. [PMID: 34060723 PMCID: PMC8604221 DOI: 10.1111/cts.13069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/05/2021] [Accepted: 04/04/2021] [Indexed: 02/01/2023] Open
Abstract
Sorafenib improves outcomes in adult hepatocellular carcinoma; however, hand foot skin reaction (HFSR) is a dose limiting toxicity of sorafenib that limits its use. HFSR has been associated with sorafenib systemic exposure. The objective of this study was to use modeling and simulation to determine whether using pharmacokinetically guided dosing to achieve a predefined sorafenib target range could reduce the rate of HFSR. Sorafenib steady‐state exposures (area under the concentration curve from 0 to 12‐h [AUC0–>12 h]) were simulated using published sorafenib pharmacokinetics at either a fixed dosage (90 mg/m2/dose) or a pharmacokinetically guided dose targeting an AUC0–>12 h between 20 and 55 h µg/ml. Dosages were either rounded to the nearest quarter of a tablet (50 mg) or capsule (10 mg). A Cox proportional hazard model from a previously published study was used to quantify HFSR toxicity. Simulations showed that in‐target studies increased from 50% using fixed doses with tablets to 74% using pharmacokinetically guided dosing with capsules. The power to observe at least 4 of 6 patients in the target range increased from 33% using fixed dosing with tablets to 80% using pharmacokinetically guided with capsules. The expected HFSR toxicity rate decreased from 22% using fixed doses with tablets to 16% using pharmacokinetically guided dosing with capsules. The power to observe less than 6 of 24 studies with HFSR toxicity increased from 51% using fixed dosing with tablets to 88% using pharmacokinetically guided with capsules. Our simulations provide the rationale to use pharmacokinetically guided sorafenib dosing to maintain effective exposures that potentially improve tolerability in pediatric clinical trials.
Collapse
Affiliation(s)
- John C Panetta
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Olivia Campagne
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jessica Gartrell
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Wayne Furman
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Clinton F Stewart
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
23
|
Quantification of sorafenib, lenvatinib, and apatinib in human plasma for therapeutic drug monitoring by UPLC-MS/MS. J Pharm Biomed Anal 2021; 202:114161. [PMID: 34052550 DOI: 10.1016/j.jpba.2021.114161] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 05/08/2021] [Accepted: 05/19/2021] [Indexed: 12/24/2022]
Abstract
Sorafenib, lenvatinib, and apatinib, as multi-targeted tyrosine kinase inhibitors with anti-proliferative and anti-angiogenic effects, are widely used for systemic therapy in advanced hepatocellular carcinoma patients. Nevertheless, insufficient efficacy or adverse effects often appear due to the significant inter-individual variability of plasma concentration for these drugs. In order to carry out therapeutic drug monitoring of these drugs and then ensure the effectiveness and safety of the medical treatment, the first method allowing to quantify sorafenib, lenvatinib, and apatinib simultaneously in human plasma was developed in this study. The analysis was performed by UPLC-MS/MS system and the chromatographic separation was achieved on a C18 column using a gradient elution of water-acetonitrile in 3.5 min. This method presented satisfactory results in terms of specificity, precision (coefficient of variation of intra-day and inter-day:1.4-6.6 %), accuracy (92.6-105.4 %), matrix effects (96.9-107.2 %), extraction recovery (90.5-99.4 %), as well as stability in human plasma and even whole blood under certain conditions. This sensitive, rapid and simple method was successfully applied to the analysis of sorafenib, lenvatinib and apatinib for therapeutic drug monitoring in hepatocellular carcinoma patients, and it was expected to be applied to further study about clarifying the concentration- efficacy and concentration-toxic relationship of sorafenib, lenvatinib, and apatinib in hepatocellular carcinoma patients.
Collapse
|
24
|
Chang YY, Wu HL, Wang T, Fang H, Tong GY, Chen Y, Wang ZY, Chen W, Yu RQ. Three efficient chemometrics assisted fluorimetric detection methods for interference-free, rapid, and simultaneous determination of ibrutinib and pralatrexate in various complicated biological fluids. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 252:119419. [PMID: 33524816 DOI: 10.1016/j.saa.2020.119419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/27/2020] [Accepted: 12/30/2020] [Indexed: 06/12/2023]
Abstract
In this study, a series of green, interference-free fluorimetric detection methods of the excitation-emission matrix coupled with the second-order calibration methods were proposed for the determination of ibrutinib and pralatrexate in various complicated biological fluids. The second-order advantage of the proposed method can overcome the problem of poor selectivity caused by the wide spectra of the fluorescence method. Even in the presence of uncalibrated interferences and severe peak overlap, the signal of pure substance and accurate quantitative results were still obtained. The average recoveries of the three methods were 94.5-104.9% for Alternating Trilinear Decomposition (ATLD) algorithm, 95.5-105.8% for Alternating Normalization Weighted Error (ANWE) algorithm and 94.4-105.7% for Parallel Factor Analysis (PARAFAC) algorithm, respectively. For ATLD, ANWE and PARAFAC, the relative standard deviations (RSD) were lower than 9.2%, 6.8% and 9.2%, and the RMSEPs were less than 8.1, 8.4 and 8.6 ng mL-1, respectively. In addition, the elliptic joint confidence region (EJCR) was adopted to further prove the accuracy of the three methods. The results showed that the three methods can accurately be quantified without significant difference. Good figures of merit parameters were also obtained. Among them, the limit of detection (LOD) and limit of quantification (LOQ) of ibrutinib and pralatrexate were in the range of 0.11-0.76 ng mL-1 and 0.21-1.12 ng mL-1, respectively, which were lower than the corresponding blood concentrations. These results indicate that the proposed method provides a promising, alternative and universal analysis strategy for clinical drug monitoring.
Collapse
Affiliation(s)
- Yue-Yue Chang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Hai-Long Wu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China.
| | - Tong Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Huan Fang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Gao-Yan Tong
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Yue Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Zhao-Yang Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Wei Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Ru-Qin Yu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
25
|
Verougstraete N, Stove V, Verstraete AG, Stove C. Quantification of eight hematological tyrosine kinase inhibitors in both plasma and whole blood by a validated LC-MS/MS method. Talanta 2021; 226:122140. [PMID: 33676691 DOI: 10.1016/j.talanta.2021.122140] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/17/2021] [Accepted: 01/21/2021] [Indexed: 01/18/2023]
Abstract
Therapeutic drug monitoring (TDM) of tyrosine kinase inhibitors (TKIs) in cancer therapy offers the potential to improve treatment efficacy while minimizing toxicity. Therefore, a high-throughput, sensitive LC-MS/MS method was developed and validated, to be used for personalized treatment of hematologic malignancies. The assay allows the simultaneous quantification in plasma (EDTA and heparin) and whole blood of eight TKIs, including bosutinib, dasatinib, gilteritinib, ibrutinib, imatinib, midostaurin, nilotinib and ponatinib, which are used in the treatment of chronic and acute myeloid leukemia (CML, AML) and chronic lymphocytic leukemia (CLL). The procedure involves simple protein precipitation of 50 μL of sample, a 4-min chromatographic separation by applying gradient elution on a standard reverse phase column, and tandem mass spectrometric detection. The method was successfully validated based on international guidelines in terms of calibration curves, precision (within-run CV 0.74-16.4%; between-run CV 1.65-17.8%), accuracy (within-run bias 0.07-19.8%; between-run bias 0.05 to -17.6%), carry-over (max 19.4%, for ponatinib), selectivity, matrix-effects, recovery (ranging from 61 to 128%), stability (only issues observed for ibrutinib) and dilution integrity. Furthermore, the accuracy of the method was demonstrated by analyzing external quality controls, with a maximum bias of -11.3%. Assay applicability was demonstrated by analyzing authentic plasma and whole blood samples in order to derive blood-plasma ratios and the variation thereof. The latter are important to allow possible blood-plasma conversion when envisaging possible future implementation of TDM via dried blood microsampling. The presented method can be applied in clinical practice for performing TDM of TKIs in plasma and whole blood samples.
Collapse
Affiliation(s)
- Nick Verougstraete
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium; Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Veronique Stove
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium; Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Alain G Verstraete
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium; Department of Diagnostic Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Christophe Stove
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium.
| |
Collapse
|
26
|
Hirasawa T, Kikuchi M, Shigeta K, Takasaki S, Sato Y, Sato T, Ogura J, Onodera K, Fukuhara N, Onishi Y, Maekawa M, Mano N. High-throughput liquid chromatography/electrospray ionization-tandem mass spectrometry method using in-source collision-induced dissociation for simultaneous quantification of imatinib, dasatinib, bosutinib, nilotinib, and ibrutinib in human plasma. Biomed Chromatogr 2021; 35:e5124. [PMID: 33772839 DOI: 10.1002/bmc.5124] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 01/30/2023]
Abstract
Recent studies have shown that therapeutic drug monitoring of tyrosine kinase inhibitors (TKIs) could improve treatment efficacy and safety. A simple analytical method using high-performance LC/electrospray ionization-tandem mass spectrometry has been developed and validated for simultaneous quantification of BCR-ABL and Bruton's TKIs used for chronic leukemia (imatinib, dasatinib, bosutinib, nilotinib, and ibrutinib) in human plasma. Although these structures and physical properties are similar, owing to their different linear ranges, simultaneously determining the plasma levels of these five TKIs by applying optimal MS parameters remains difficult. A quantitative range exceeding 60,000-fold was required, and the linear dynamic ranges of imatinib, bosutinib, and nilotinib were limited because of the presence of a saturated detection signal. In this study, we applied the in-source collision-induced dissociation technique to control the ion amounts in mass spectrometry. This new method allowed rapid determination within 5 min with simple pretreatment. The method was validated according to the US Food and Drug Administration guidelines. Moreover, all samples of patients with chronic leukemia were successfully measured and their values were within the linear range of measurement. Therefore, our high-throughput analytical system is useful to measure the plasma concentrations of imatinib, dasatinib, bosutinib, nilotinib, and ibrutinib in clinical practice.
Collapse
Affiliation(s)
- Tensei Hirasawa
- Faculty of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Masafumi Kikuchi
- Faculty of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan.,Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Kensuke Shigeta
- Faculty of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Shinya Takasaki
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Yu Sato
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Toshihiro Sato
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Jiro Ogura
- Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Miyagi, Japan.,Yamagata University Graduate School of Medical Science/Department of Pharmacy, Yamagata University Hospital, Yamagata, Japan
| | - Koichi Onodera
- Department of Hematology and Rheumatology, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Noriko Fukuhara
- Department of Hematology and Rheumatology, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Yasushi Onishi
- Department of Hematology and Rheumatology, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Masamitsu Maekawa
- Faculty of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan.,Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Nariyasu Mano
- Faculty of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan.,Department of Pharmaceutical Sciences, Tohoku University Hospital, Sendai, Miyagi, Japan
| |
Collapse
|
27
|
Validation and Comparison of Two Analytical Methods for Imatinib Therapeutic Drug Monitoring. Chromatographia 2021. [DOI: 10.1007/s10337-021-04041-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
28
|
Sharma JB, Krishnamurthy MN, Awase A, Joshi A, Patil V, Noronha V, Prabhash K, Gota V. Validation of a novel causality assessment scale for adverse events in non-small cell lung carcinoma patients treated with platinum and pemetrexed doublet chemotherapy. Ther Adv Drug Saf 2021; 12:2042098621991280. [PMID: 33628419 PMCID: PMC7882752 DOI: 10.1177/2042098621991280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/07/2021] [Indexed: 01/25/2023] Open
Abstract
Aim: Accurate causality assessment (CA) of adverse events (AEs) is important in clinical research and routine clinical practice. The Naranjo scale (NS) used for CA lacks specificity, leading to a high rate of false positive causal associations. NS is a simple scale for CA; however, its limitations have reduced its popularity in favour of other scales. We therefore attempted to improvise the algorithm by addressing specific lacunae in NS. Methods: We attempted to modify the existing NS by (a) changing the weightage given to certain responses, (b) achieving higher resolution to certain responses for delineating drug related and unrelated AEs and (c) modifying the slabs for classification of association as ‘likely’ and ‘unlikely’. The new scale, named as the Sharma-Nookala-Gota (SNG) algorithm, was evaluated in a training set of 19 AEs in a tertiary care cancer hospital in western India, and further validated in a set of 104 AEs. Consensus of four physician opinion was taken as gold standard for comparison. Results: Of the 19 AEs in the training set, 6 were described by the treating physician as ‘not related’ and 13 as related to the drug. The SNG algorithm had 100% concordance with physician opinion, whereas the NS had only 73.7% concordance. NS showed a tendency to misclassify AEs as ‘related’ when they were indeed ‘not related’. In the validation set of 104 AEs, NS and SNG algorithms misclassified 30 and 2 AEs, respectively, leading to a concordance of 70.2% and 98.1%, respectively, with physician opinion. Conclusion: Decisive modifications of the NS resulted in the SNG scale, with superior specificity while retaining sensitivity against the gold standard. Plain Language Summary SNG algorithm – A novel tool for causality assessment of adverse drug reactions Adverse events (AEs) can cause increased morbidity, hospitalisation, and even death. Hence it is essential to recognise AEs and to establish their correct causal relationship to a drug. Many causality assessment methods, scales and algorithms are available to assess the relationship between an AE and a drug. The Naranjo algorithm is most commonly employed in spite of its many drawbacks as it is simple to use. Concerns have been raised regarding the performance of the scale, and researchers have tried to answer them, but none of them could address all issues satisfactorily. We too experienced many problems while using it in our routine clinical practice and in clinical trials. For instance, the Naranjo scale is non-specific and shows a bias toward implicating the drug as the causal factor for AEs. This improper assessment has often led to drug discontinuation, thereby compromising the efficacy of treatment. Hence, we modified the existing Naranjo scale to a new one (the Sharma-Nookala-Gota – SNG algorithm) to address these shortcomings. We piloted the SNG causality assessment algorithm in patients suffering from AEs due to various drugs. The SNG algorithm was found to have good concordance with the physicians’ assessment of causality. As a next step, we validated the SNG algorithm in patients receiving a standard drug combination of pemetrexed and carboplatin for lung cancer combination. Out of the 104 AEs observed in 65 patients, the SNG causality assessment algorithm showed good concordance (except in two cases) with the physicians’ decision of causality assessment, while the Naranjo algorithm was not so successful. Hence, the SNG algorithm can be a better guide for causality assessment of AEs.
Collapse
Affiliation(s)
- Jyoti Bhagatram Sharma
- Department of Clinical Pharmacology, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India
| | - Manjunath Nookala Krishnamurthy
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Sector 22, Kharghar, Maharashtra, Navi Mumbai 410210, India; Homi Bhabha National Institute, Mumbai, India
| | - Ankita Awase
- Department of Clinical Pharmacology, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India
| | - Amit Joshi
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Dr. E. Borge's Road, Parel, Mumbai, India; Homi Bhabha National Institute, Mumbai, India
| | - Vijay Patil
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Dr. E. Borge's Road, Parel, Mumbai, India; Homi Bhabha National Institute, Mumbai, India
| | - Vanita Noronha
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Dr. E. Borge's Road, Parel, Mumbai, India; Homi Bhabha National Institute, Mumbai, India
| | - Kumar Prabhash
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Dr. E. Borge's Road, Parel, Mumbai, India; Homi Bhabha National Institute, Mumbai, India
| | - Vikram Gota
- Department of Clinical Pharmacology, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
29
|
Tartaggia S, Meneghello A, Bellotto O, Poetto AS, Zanchetta M, Posocco B, Bunka D, Polo F, Toffoli G. An SPR investigation into the therapeutic drug monitoring of the anticancer drug imatinib with selective aptamers operating in human plasma. Analyst 2021; 146:1714-1724. [DOI: 10.1039/d0an01860k] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
An ss-DNA aptamer-based biosensor was devised to detect the anticancer drug imatinib by means of surface plasmon resonance.
Collapse
Affiliation(s)
- Stefano Tartaggia
- Clinical and Experimental Pharmacology
- Centro di Riferimento Oncologico di Aviano (CRO) IRCCS
- 33081, Aviano
- Italy
| | - Anna Meneghello
- Clinical and Experimental Pharmacology
- Centro di Riferimento Oncologico di Aviano (CRO) IRCCS
- 33081, Aviano
- Italy
| | - Ottavia Bellotto
- Clinical and Experimental Pharmacology
- Centro di Riferimento Oncologico di Aviano (CRO) IRCCS
- 33081, Aviano
- Italy
- Department of Chemical and Pharmaceutical Sciences
| | - Ariana Soledad Poetto
- Clinical and Experimental Pharmacology
- Centro di Riferimento Oncologico di Aviano (CRO) IRCCS
- 33081, Aviano
- Italy
- Department of Pharmacological and Pharmaceutical Sciences
| | - Martina Zanchetta
- Clinical and Experimental Pharmacology
- Centro di Riferimento Oncologico di Aviano (CRO) IRCCS
- 33081, Aviano
- Italy
- Department of Chemical and Pharmaceutical Sciences
| | - Bianca Posocco
- Clinical and Experimental Pharmacology
- Centro di Riferimento Oncologico di Aviano (CRO) IRCCS
- 33081, Aviano
- Italy
| | | | - Federico Polo
- Clinical and Experimental Pharmacology
- Centro di Riferimento Oncologico di Aviano (CRO) IRCCS
- 33081, Aviano
- Italy
| | - Giuseppe Toffoli
- Clinical and Experimental Pharmacology
- Centro di Riferimento Oncologico di Aviano (CRO) IRCCS
- 33081, Aviano
- Italy
| |
Collapse
|
30
|
Niessen WMA, Hillebrand MJX, Rosing H, Beijnen JH. Tandem mass spectrometry of small-molecule signal transduction inhibitors: Accurate-m/z data to adapt structure proposals of product ions. J Pharm Biomed Anal 2020; 195:113864. [PMID: 33387839 DOI: 10.1016/j.jpba.2020.113864] [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] [Received: 07/14/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 01/22/2023]
Abstract
Protein kinases inhibitors or, more generally, signal transduction inhibitors (STIs) can be used to treat diseases in which deregulation of the protein kinase activity plays a role, such as in cancer. A wide variety of drugs has been developed and/or is under investigation to act as protein kinase inhibitors, especially in tyrosine kinase inhibition. The bioanalysis of STIs has received considerable attention in the past 20 years. Liquid chromatography-tandem mass spectrometry (LC-MS-MS) in selected-reaction monitoring (SRM) mode is the method-of-choice in such studies. In several of these studies from us and others, structures are proposed for the product ions applied in SRM. A critical review of these proposed structures is presented using accurate-m/z data, which we have now generated with a linear-ion-trap-Orbitrap hybrid mass spectrometer. This led to adaptation and new structural proposals of 18 product ions for 13 STIs. Our investigation endorses the power of accurate-m/z analysis in structure elucidation of product ions in bioanalytical LC-MS-MS studies and for which the SRM mode in tandem-quadrupole instruments is apparently less suitable.
Collapse
Affiliation(s)
- W M A Niessen
- hyphen MassSpec, Margrietstraat 34, 2215 HJ, Voorhout, the Netherlands.
| | - M J X Hillebrand
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - H Rosing
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - J H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| |
Collapse
|
31
|
Garrison DA, Talebi Z, Eisenmann ED, Sparreboom A, Baker SD. Role of OATP1B1 and OATP1B3 in Drug-Drug Interactions Mediated by Tyrosine Kinase Inhibitors. Pharmaceutics 2020; 12:E856. [PMID: 32916864 PMCID: PMC7559291 DOI: 10.3390/pharmaceutics12090856] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/02/2020] [Accepted: 09/02/2020] [Indexed: 12/20/2022] Open
Abstract
Failure to recognize important features of a drug's pharmacokinetic characteristics is a key cause of inappropriate dose and schedule selection, and can lead to reduced efficacy and increased rate of adverse drug reactions requiring medical intervention. As oral chemotherapeutic agents, tyrosine kinase inhibitors (TKIs) are particularly prone to cause drug-drug interactions as many drugs in this class are known or suspected to potently inhibit the hepatic uptake transporters OATP1B1 and OATP1B3. In this article, we provide a comprehensive overview of the published literature and publicly-available regulatory documents in this rapidly emerging field. Our findings indicate that, while many TKIs can potentially inhibit the function of OATP1B1 and/or OATP1B3 and cause clinically-relevant drug-drug interactions, there are many inconsistencies between regulatory documents and the published literature. Potential explanations for these discrepant observations are provided in order to assist prescribing clinicians in designing safe and effective polypharmacy regimens, and to provide researchers with insights into refining experimental strategies to further predict and define the translational significance of TKI-mediated drug-drug interactions.
Collapse
Affiliation(s)
| | | | | | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (D.A.G.); (Z.T.); (E.D.E.)
| | - Sharyn D. Baker
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (D.A.G.); (Z.T.); (E.D.E.)
| |
Collapse
|
32
|
Hopkins AM, Menz BD, Wiese MD, Kichenadasse G, Gurney H, McKinnon RA, Rowland A, Sorich MJ. Nuances to precision dosing strategies of targeted cancer medicines. Pharmacol Res Perspect 2020; 8:e00625. [PMID: 32662214 PMCID: PMC7358594 DOI: 10.1002/prp2.625] [Citation(s) in RCA: 8] [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: 03/05/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 01/05/2023] Open
Abstract
Selecting the dose of a targeted cancer medicine that is most appropriate for a specific individual is a rational approach to maximize therapeutic outcomes and minimize toxicity. There are many different options for optimizing the dose of targeted cancer medicines and the purpose of this review is to provide a comprehensive comparison of the main options explored in prospective studies. Precision initial dose selection of targeted cancer therapies has been minimally explored to date; however, concentration, toxicity, and therapeutic outcome markers are used to guide on-therapy dose adaption of targeted cancer therapies across several medicines and cancers. While a specific concentration, toxicity, or therapeutic outcome marker commonly dominates an investigated precision on-therapy dose adaption strategy, greater attention to simultaneously account for exposure, toxicity, therapeutic outcomes, disease status, time since treatment initiation and patient preferences are required for optimal patient outcomes. To enable successful implementation of precision dosing strategies for targeted cancer medicines into clinical practice, future prospective studies aiming to develop strategies should consider these elements in their design.
Collapse
Affiliation(s)
- Ashley M. Hopkins
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Bradley D. Menz
- Division of PharmacySouthern Adelaide Local Health Network, Flinders Medical CentreAdelaideSouth AustraliaAustralia
| | - Michael D. Wiese
- School of Pharmacy and Medical SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Ganessan Kichenadasse
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Howard Gurney
- Department of Medical OncologyWestmead HospitalSydneyNew South WalesAustralia
| | - Ross A. McKinnon
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Andrew Rowland
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Michael J. Sorich
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| |
Collapse
|
33
|
Leenhardt F, Gracia M, Perrin C, Muracciole-Bich C, Marion B, Roques C, Alexandre M, Firmin N, Pouderoux S, Mbatchi L, Gongora C, Jacot W, Evrard A. Liquid chromatography-tandem mass spectrometric assay for the quantification of CDK4/6 inhibitors in human plasma in a clinical context of drug-drug interaction. J Pharm Biomed Anal 2020; 188:113438. [PMID: 32623316 DOI: 10.1016/j.jpba.2020.113438] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/16/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022]
Abstract
The CDK4/6 inhibitors palbociclib and ribociclib are kinase inhibitors used in association with hormonal therapy for the management of patients with metastatic breast cancer. Like most kinase inhibitors, therapeutic drug monitoring may be used for personalize their dosage. To this aim, we developed and validated a sensitive and specific HPLC-MS/MS method for palbociclib and ribociclib quantification in blood samples. We then quantified exposure to palbociclib (plasma trough concentration; Ctrough) in a real-life cohort of patients with locally invasive or metastatic breast cancer (n = 18) at day 15 of the first cycle of palbociclib treatment to characterize palbociclib concentration at steady state (Clinicaltrials.gov identifier NCT04025541, IdRCB n° 2018-A00064-51, 03/07/2018). The geometric mean (± standard deviation [min-max]) of palbociclib plasma Ctrough was 88.58 ng/mL (± 26.4 [46.5 ng/mL - 133 ng/mL]) at day 15. Some covariates, such as drug-drug interactions, could explain the concentration variations observed in our Caucasian cohort. These first results in real-life settings obtained with our HPLC-MS/MS method give important information on palbociclib monitoring and pharmacokinetic variability.
Collapse
Affiliation(s)
- Fanny Leenhardt
- Laboratoire de Pharmacocinétique, Université de Montpellier, Faculté de Pharmacie, France; Service Pharmacie, Institut du Cancer de Montpellier, Université de Montpellier, 208 rue des Apothicaires, 34298, Montpellier, France; Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Montpellier, France.
| | - Matthieu Gracia
- Laboratoire de Pharmacocinétique, Université de Montpellier, Faculté de Pharmacie, France; Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Montpellier, France
| | - Catherine Perrin
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247-CNRS-UM-ENSCM, Montpellier, France
| | | | - Bénédicte Marion
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247-CNRS-UM-ENSCM, Montpellier, France
| | - Celine Roques
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247-CNRS-UM-ENSCM, Montpellier, France
| | - Marie Alexandre
- Département d'Oncologie Médicale, Institut du Cancer de Montpellier, Université de Montpellier, 208 rue des Apothicaires, 34298, Montpellier, France
| | - Nelly Firmin
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Montpellier, France; Département d'Oncologie Médicale, Institut du Cancer de Montpellier, Université de Montpellier, 208 rue des Apothicaires, 34298, Montpellier, France
| | - Stephane Pouderoux
- Département d'Oncologie Médicale, Institut du Cancer de Montpellier, Université de Montpellier, 208 rue des Apothicaires, 34298, Montpellier, France
| | - Litaty Mbatchi
- Laboratoire de Pharmacocinétique, Université de Montpellier, Faculté de Pharmacie, France; Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Montpellier, France; Laboratoire de Biochimie et Biologie moléculaire, Centre Hospitalier Universitaire Nîmes, France
| | - Celine Gongora
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Montpellier, France
| | - William Jacot
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Montpellier, France; Département d'Oncologie Médicale, Institut du Cancer de Montpellier, Université de Montpellier, 208 rue des Apothicaires, 34298, Montpellier, France
| | - Alexandre Evrard
- Laboratoire de Pharmacocinétique, Université de Montpellier, Faculté de Pharmacie, France; Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Montpellier, France; Laboratoire de Biochimie et Biologie moléculaire, Centre Hospitalier Universitaire Nîmes, France
| |
Collapse
|
34
|
Menz BD, Stocker SL, Verougstraete N, Kocic D, Galettis P, Stove CP, Reuter SE. Barriers and opportunities for the clinical implementation of therapeutic drug monitoring in oncology. Br J Clin Pharmacol 2020; 87:227-236. [PMID: 32430968 DOI: 10.1111/bcp.14372] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 12/12/2022] Open
Abstract
There are few fields of medicine in which the individualisation of medicines is more important than in the area of oncology. Under-dosing can have significant ramifications due to the potential for therapeutic failure and cancer progression; by contrast, over-dosing may lead to severe treatment-limiting side effects, such as agranulocytosis and neutropenia. Both circumstances lead to poor patient prognosis and contribute to the high mortality rates still seen in oncology. The concept of dose individualisation tailors dosing for each individual patient to ensure optimal drug exposure and best clinical outcomes. While the value of this strategy is well recognised, it has seen little translation to clinical application. However, it is important to recognise that the clinical setting of oncology is unlike that for which therapeutic drug monitoring (TDM) is currently the cornerstone of therapy (e.g. antimicrobials). Whilst there is much to learn from these established TDM settings, the challenges presented in the treatment of cancer must be considered to ensure the implementation of TDM in clinical practice. Recent advancements in a range of scientific disciplines have the capacity to address the current system limitations and significantly enhance the use of anticancer medicines to improve patient health. This review examines opportunities presented by these innovative scientific methodologies, specifically sampling strategies, bioanalytics and dosing decision support, to enable optimal practice and facilitate the clinical implementation of TDM in oncology.
Collapse
Affiliation(s)
- Bradley D Menz
- SA Pharmacy, Flinders Medical Centre, Adelaide, SA, Australia
| | - Sophie L Stocker
- Department of Clinical Pharmacology & Toxicology, St Vincent's Hospital, Sydney, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Nick Verougstraete
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium.,Laboratory of Toxicology, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Danijela Kocic
- Department of Clinical Pharmacology & Toxicology, St Vincent's Hospital, Sydney, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Peter Galettis
- Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
| | - Christophe P Stove
- Laboratory of Toxicology, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Stephanie E Reuter
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
35
|
Ahmed OS, Ladner Y, Bousquet C, Montels J, Dubský P, Philibert L, Perrin C. Direct salting-out assisted liquid–liquid extraction (SALLE) from human blood: Application for the analysis of tyrosine kinase inhibitors. Microchem J 2020. [DOI: 10.1016/j.microc.2020.104791] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
36
|
Buclin T, Thoma Y, Widmer N, André P, Guidi M, Csajka C, Decosterd LA. The Steps to Therapeutic Drug Monitoring: A Structured Approach Illustrated With Imatinib. Front Pharmacol 2020; 11:177. [PMID: 32194413 PMCID: PMC7062864 DOI: 10.3389/fphar.2020.00177] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 02/07/2020] [Indexed: 01/07/2023] Open
Abstract
Pharmacometric methods have hugely benefited from progress in analytical and computer sciences during the past decades, and play nowadays a central role in the clinical development of new medicinal drugs. It is time that these methods translate into patient care through therapeutic drug monitoring (TDM), due to become a mainstay of precision medicine no less than genomic approaches to control variability in drug response and improve the efficacy and safety of treatments. In this review, we make the case for structuring TDM development along five generic questions: 1) Is the concerned drug a candidate to TDM? 2) What is the normal range for the drug's concentration? 3) What is the therapeutic target for the drug's concentration? 4) How to adjust the dosage of the drug to drive concentrations close to target? 5) Does evidence support the usefulness of TDM for this drug? We exemplify this approach through an overview of our development of the TDM of imatinib, the very first targeted anticancer agent. We express our position that a similar story shall apply to other drugs in this class, as well as to a wide range of treatments critical for the control of various life-threatening conditions. Despite hurdles that still jeopardize progress in TDM, there is no doubt that upcoming technological advances will shape and foster many innovative therapeutic monitoring methods.
Collapse
Affiliation(s)
- Thierry Buclin
- Service of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Yann Thoma
- School of Management and Engineering Vaud (HEIG-VD), University of Applied Science Western Switzerland (HES-SO), Yverdon-les-Bains, Switzerland
| | - Nicolas Widmer
- Service of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Pharmacy of Eastern Vaud Hospitals, Rennaz, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Pascal André
- Service of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Monia Guidi
- Service of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Chantal Csajka
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.,Center for Research and Innovation in Clinical Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - Laurent A Decosterd
- Service of Clinical Pharmacology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| |
Collapse
|
37
|
|
38
|
Effective quantification of 11 tyrosine kinase inhibitors and caffeine in human plasma by validated LC-MS/MS method with potent phospholipids clean-up procedure. Application to therapeutic drug monitoring. Talanta 2020; 208:120450. [DOI: 10.1016/j.talanta.2019.120450] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/30/2019] [Accepted: 10/04/2019] [Indexed: 12/11/2022]
|
39
|
Labots M, Pham TV, Honeywell RJ, Knol JC, Beekhof R, de Goeij-de Haas R, Dekker H, Neerincx M, Piersma SR, van der Mijn JC, van der Peet DL, Meijerink MR, Peters GJ, van Grieken NC, Jiménez CR, Verheul HM. Kinase Inhibitor Treatment of Patients with Advanced Cancer Results in High Tumor Drug Concentrations and in Specific Alterations of the Tumor Phosphoproteome. Cancers (Basel) 2020; 12:cancers12020330. [PMID: 32024067 PMCID: PMC7072422 DOI: 10.3390/cancers12020330] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/20/2020] [Accepted: 01/29/2020] [Indexed: 12/22/2022] Open
Abstract
Identification of predictive biomarkers for targeted therapies requires information on drug exposure at the target site as well as its effect on the signaling context of a tumor. To obtain more insight in the clinical mechanism of action of protein kinase inhibitors (PKIs), we studied tumor drug concentrations of protein kinase inhibitors (PKIs) and their effect on the tyrosine-(pTyr)-phosphoproteome in patients with advanced cancer. Tumor biopsies were obtained from 31 patients with advanced cancer before and after 2 weeks of treatment with sorafenib (SOR), erlotinib (ERL), dasatinib (DAS), vemurafenib (VEM), sunitinib (SUN) or everolimus (EVE). Tumor concentrations were determined by LC-MS/MS. pTyr-phosphoproteomics was performed by pTyr-immunoprecipitation followed by LC-MS/MS. Median tumor concentrations were 2–10 µM for SOR, ERL, DAS, SUN, EVE and >1 mM for VEM. These were 2–178 × higher than median plasma concentrations. Unsupervised hierarchical clustering of pTyr-phosphopeptide intensities revealed patient-specific clustering of pre- and on-treatment profiles. Drug-specific alterations of peptide phosphorylation was demonstrated by marginal overlap of robustly up- and downregulated phosphopeptides. These findings demonstrate that tumor drug concentrations are higher than anticipated and result in drug specific alterations of the phosphoproteome. Further development of phosphoproteomics-based personalized medicine is warranted.
Collapse
Affiliation(s)
- Mariette Labots
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Thang V. Pham
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Richard J. Honeywell
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Jaco C. Knol
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Robin Beekhof
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Richard de Goeij-de Haas
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Henk Dekker
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Maarten Neerincx
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Sander R. Piersma
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Johannes C. van der Mijn
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Donald L. van der Peet
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
| | - Martijn R. Meijerink
- Department of Radiology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
| | - Godefridus J. Peters
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
| | - Nicole C.T. van Grieken
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
| | - Connie R. Jiménez
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (M.L.); (T.V.P.); (R.J.H.); (J.C.K.); (R.B.); (R.d.G.-d.H.); (H.D.); (M.N.); (S.R.P.); (J.C.v.d.M.); (G.J.P.)
- Correspondence: or (C.R.J.); (H.M.W.V.)
| | - Henk M.W. Verheul
- Department of Medical Oncology, RadboudUMC, Radboud University, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, The Netherlands
- Correspondence: or (C.R.J.); (H.M.W.V.)
| |
Collapse
|
40
|
Zhang XW, Li QH, Xu ZD, Dou JJ. Mass spectrometry-based metabolomics in health and medical science: a systematic review. RSC Adv 2020; 10:3092-3104. [PMID: 35497733 PMCID: PMC9048967 DOI: 10.1039/c9ra08985c] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/14/2019] [Indexed: 01/15/2023] Open
Abstract
Metabolomics is the study of the investigation of small molecules derived from cellular and organism metabolism, which reflects the outcomes of the complex network of biochemical reactions in living systems. As the most recent member of the omics family, there has been notable progress in metabolomics in the last decade, mainly driven by the improvement in mass spectrometry (MS). MS-based metabolomic strategies in modern health and medical science studies provide innovative tools for novel diagnostic and prognostic approaches, as well as an augmented role in drug development, nutrition science, toxicology, and forensic science. In the present review, we not only introduce the application of MS-based metabolomics in the above fields, but also discuss the MS analysis technologies commonly used in metabolomics and the application of metabolomics in precision medicine, and further explore the challenges and perspectives of metabolomics in the field of health and medical science, which are expected to make a little contribution to the better development of metabolomics.
Collapse
Affiliation(s)
- Xi-Wu Zhang
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine Heping Road 24 Harbin 150040 China +86-451-87266827 +86-451-87266827
| | - Qiu-Han Li
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine Heping Road 24 Harbin 150040 China +86-451-87266827 +86-451-87266827
| | - Zuo-di Xu
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine Heping Road 24 Harbin 150040 China +86-451-87266827 +86-451-87266827
| | - Jin-Jin Dou
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine Heping Road 24 Harbin 150040 China +86-451-87266827 +86-451-87266827
| |
Collapse
|
41
|
Ezzeldin E, Iqbal M, Herqash RN, ElNahhas T. Simultaneous quantitative determination of seven novel tyrosine kinase inhibitors in plasma by a validated UPLC-MS/MS method and its application to human microsomal metabolic stability study. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1136:121851. [DOI: 10.1016/j.jchromb.2019.121851] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 10/02/2019] [Accepted: 10/26/2019] [Indexed: 12/17/2022]
|
42
|
Cost effectiveness of therapeutic drug monitoring for imatinib administration in chronic myeloid leukemia. PLoS One 2019; 14:e0226552. [PMID: 31869360 PMCID: PMC6927594 DOI: 10.1371/journal.pone.0226552] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 11/28/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Imatinib mesylate (IM) is a first-line treatment option for patients with chronic myeloid leukemia (CML). Patients who fail or are intolerant to IM therapy are treated with more expensive second and third-generation tyrosine kinase inhibitors. Patients show wide variation in trough concentrations in response to standard dosing. Thus, many patients receive subtherapeutic or supratherapeutic doses. Therapeutic drug monitoring (TDM) may improve dose management that, in turn, may reduce costs and improve outcomes. However, TDM also adds to the cost of patient care. The objective of this study was to determine the cost-effectiveness of TDM for generic IM therapy. METHODS We developed a microsimulation model for the trough plasma concentration of IM which is related to a cytogenetic or molecular response. We compared two cohorts: one with TDM and one without TDM (NTDM). The lifetime incremental cost-effectiveness ratio (ICER) was calculated using quality-adjusted life years (QALYs) as the effectiveness measure. One-way and probabilistic sensitivity analyses were performed. RESULTS The lifetime cost and QALY of treatment with TDM were $2,137K [95% Ci: 2,079K; 2,174K] and 12.37 [95% CI: 12.07; 12.55], respectively. The cost and QALY of NTDM were $2,132K [95% CI: 2,091K; 2,197K] and 12.23 [95% CI: 11.96; 12.50], respectively. The incremental cost and QALY for TDM relative to NTDM was $4,417 [95% CI: -52,582; 32,097]) and 0.15 [95% CI: -0.13; 0.28]. The ICER for TDM relative to NTDM was $30,450/QALY. Probabilistic sensitivity analysis showed that TDM was cost-effective relative to NTDM in 90% of the tested scenarios at a willingness-to-pay threshold of $100,000/QALY. CONCLUSIONS Although the impact of TDM is modest, the cost-effectiveness over a lifetime horizon (societal perspective, ($30,450/QALY) falls within the acceptable range (< $100k/QALY).
Collapse
|
43
|
A Pharmacokinetic Interaction Study of Sorafenib and Iced Teas in Rats Using UPLC-MS/MS: An Illustration of Beverage-Drug Interaction. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2410845. [PMID: 31871933 PMCID: PMC6907072 DOI: 10.1155/2019/2410845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 09/01/2019] [Accepted: 10/22/2019] [Indexed: 11/18/2022]
Abstract
Iced teas (ITs), also known as ready-to-drink teas, have gained much popularity among many nations. The modulatory effect of tea beverages on CYP3A4 increases the possibility of their potential interactions with many coadministered medications. Being a substrate of CYP3A4, sorafenib (SOR), the first-line therapy for the treatment of hepatocellular carcinoma, shows a great probability to exhibit pharmacokinetic (PK) interaction with ITs. For this purpose, different groups of Wistar rats were given oral doses of SOR (40 mg/kg), along with different types of ITs. The concentration of SOR in rat plasma was determined using UPLC-MS/MS. Chromatographic analysis was performed on a C18 analytical column, Acquity UPLC BEH™ (100 × 1.0 mm, i.d., 1.7 μm particle size), using erlotinib (ERL) as an internal standard. Isocratic elution was performed with a mobile phase consisting of two solvents: solvent A (water with 0.1% formic acid) and solvent B (acetonitrile with 0.1% formic acid), in a ratio of 30 : 70, v/v, respectively. Quantitation was performed using MRM of the transitions from protonated precursor ions [M+H]+ to product ions at m/z 465.12 > 252.02 (SOR) and m/z 394.29 > 278.19 (ERL). The method was fully validated as per the FDA guidance for bioanalytical method validation in the concentration range of 2.5–500 ng/mL. Different PK parameters were calculated for SOR in all rat groups and groups administered with ITs and SOR, compared with groups with simply water and SOR. Experimental data revealed that ITs caused a general reduction in SOR bioavailability; an approximate reduction of 30% was recorded for all types of tested ITs. These data indicate that ITs could affect the PK profile of SOR in rats.
Collapse
|
44
|
Ahmed OS, Malý M, Ladner Y, Philibert L, Dubský P, Perrin C. Influence of salt and acetonitrile on the capillary zone electrophoresis analysis of imatinib in plasma samples. Electrophoresis 2019; 40:2810-2819. [DOI: 10.1002/elps.201900188] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/06/2019] [Accepted: 08/09/2019] [Indexed: 01/28/2023]
Affiliation(s)
- Omar S. Ahmed
- Institut des Biomolécules Max Mousseron (IBMM)UMR 5247‐CNRS‐UM‐ENSCM Montpellier France
- Department of Analytical ChemistryFaculty of PharmacyMisr University of Science and Technology (MUST). 6th of October City Egypt
| | - Michal Malý
- Department of Physical and Macromolecular ChemistryFaculty of ScienceCharles University Prague Czech Republic
| | - Yoann Ladner
- Institut des Biomolécules Max Mousseron (IBMM)UMR 5247‐CNRS‐UM‐ENSCM Montpellier France
| | - Laurent Philibert
- Institut régional du Cancer de Montpellier (ICM)Département de Pharmacie et Pharmacologie Montpellier France
| | - Pavel Dubský
- Department of Physical and Macromolecular ChemistryFaculty of ScienceCharles University Prague Czech Republic
| | - Catherine Perrin
- Institut des Biomolécules Max Mousseron (IBMM)UMR 5247‐CNRS‐UM‐ENSCM Montpellier France
| |
Collapse
|
45
|
Onmaz DE, Abusoglu S, Unlu A, Basturk A, Dagli M, Bagci M, Tok O, Abusoglu G. Determination of serum imatinib and its' metabolite in patients chronic myeloid leukemia. Clin Chim Acta 2019; 497:120-124. [PMID: 31351054 DOI: 10.1016/j.cca.2019.07.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Imatinib has favorable pharmacokinetic properties, but primary and secondary resistance mechanisms may cause a decrease in clinical response over time. There is a positive correlation between serum imatinib concentrations and treatment response. Our aim was to develop a method for the measurement of imatinib and its' active metabolite N-desmethyl imatinib. METHODS Serum imatinib and N-desmethyl imatinib levels were determined by liquid chromatography-tandem mass spectrometry (LC-MS/MS) and validation studies were carried out according to CLSI (The Clinical & Laboratory Standards Institute) protocols. Serum samples were collected from 40 patients with chronic myeloid leukemia (CML) and analyzed with LC-MS/MS and ultra high-performance liquid chromatography (UHPLC) methods. RESULTS The linearity range and correlation coefficient were 12.2-12,500 ng/mL and 0.9987 for LC-MS/MS method, respectively. Limit of quantitation was determined as 24.4 ng/mL. The retention times of imatinib and N-desmethyl imatinib were 1.66 and 1.60 min, respectively. There was no statistically significant difference between the results of both methods. DISCUSSION This LC-MS/MS method is cost-effective and has adavantages such as using low serum volumes, requiring simple pretreatment steps (only protein precipitation) and reduced turnaround times for analysis.
Collapse
Affiliation(s)
- Duygu Eryavuz Onmaz
- Department of Biochemistry, Selcuk University Faculty of Medicine, Konya, Turkey.
| | - Sedat Abusoglu
- Department of Biochemistry, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Ali Unlu
- Department of Biochemistry, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Abdulkadir Basturk
- Department of Internal Medicine, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Mehmet Dagli
- Department of Internal Medicine, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Metin Bagci
- Department of Internal Medicine, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Oguzhan Tok
- Department of Biochemistry, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Gulsum Abusoglu
- Department of Medical Laboratory Techniques, Selcuk University Vocational School of Health, Konya, Turkey
| |
Collapse
|
46
|
Mould DR, Hutson PR. Critical Considerations in Anticancer Drug Development and Dosing Strategies: The Past, Present, and Future. J Clin Pharmacol 2019; 57 Suppl 10:S116-S128. [PMID: 28921645 DOI: 10.1002/jcph.983] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/19/2017] [Indexed: 01/26/2023]
|
47
|
Charlier B, Marino L, Dal Piaz F, Pingeon M, Coglianese A, Izzo B, Serio B, Selleri C, Filippelli A, Izzo V. Development and Validation of a Reverse-Phase High-Performance Liquid Chromatography with Fluorescence Detection (RP-HPLC-FL) Method to Quantify Ruxolitinib in Plasma Samples. ANAL LETT 2019. [DOI: 10.1080/00032719.2018.1537283] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Bruno Charlier
- Postgraduate School in Hospital Pharmacy, University of Salerno, Fisciano, Salerno, Italy
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, Salerno, Italy
| | - Luigi Marino
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Salerno, Italy
| | - Fabrizio Dal Piaz
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, Salerno, Italy
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Salerno, Italy
| | - Marine Pingeon
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Salerno, Italy
| | - Albino Coglianese
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Salerno, Italy
| | - Barbara Izzo
- Department of Clinical Medicine and Surgery, University Federico II of Naples, Naples, Italy
- CEINGE-Biotecnologie Avanzate, Naples, Italy
| | - Bianca Serio
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, Salerno, Italy
| | - Carmine Selleri
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, Salerno, Italy
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Salerno, Italy
| | - Amelia Filippelli
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, Salerno, Italy
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Salerno, Italy
| | - Viviana Izzo
- University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, Salerno, Italy
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Salerno, Italy
| |
Collapse
|
48
|
Sogawa R, Saita T, Yamamoto Y, Kimura S, Narisawa Y, Kimura S, Shin M. Development of a competitive enzyme-linked immunosorbent assay for therapeutic drug monitoring of afatinib. J Pharm Anal 2018; 9:49-54. [PMID: 30740257 PMCID: PMC6355467 DOI: 10.1016/j.jpha.2018.09.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/31/2018] [Accepted: 09/17/2018] [Indexed: 11/29/2022] Open
Abstract
Afatinib is an oral tyrosine kinase inhibitor (TKI) approved for treating advanced non-small cell lung cancer. It is necessary to develop a simple quantification method for TKIs in order to facilitate therapeutic drug monitoring (TDM) in clinical settings. This study sought to develop a simple and sensitive competitive enzyme-linked immunosorbent assay (ELISA) to quantify afatinib in plasma for routine pharmacokinetic applications. An anti-afatinib antibody was obtained using (S)-N-4-(3-chloro-4-fluorophenyl)-7-(tetrahydrofuran-3-yloxy)-quinazoline-4,6-diamine (CTQD), which has the same substructure as afatinib, as a hapten. Enzyme labeling of afatinib with horseradish peroxidase was similarly performed using CTQD. A simple competitive ELISA for afatinib was developed based on the principle of direct competition between afatinib and the enzyme marker for the anti-afatinib antibody, which had been immobilized on the plastic surface of a microtiter plate. Plasma afatinib concentrations below the limit of quantification of 30 pg/mL were reproducibly measurable. Also, the values of plasma afatinib levels measured from 20 patients were comparable with those measured by high-performance liquid chromatography, and there was a strong correlation between the values determined by both methods (Y = 0.976X – 0.207, r = 0.975). As indicated by its specificity and sensitivity, this newly developed ELISA for afatinib is an important tool for TDM and studies of the pharmacokinetics of afatinib.
Collapse
Affiliation(s)
- Rintaro Sogawa
- Department of Pharmacy, Saga University Hospital, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Tetsuya Saita
- Applied Life Science Department, Faculty of Biotechnology and Life Science, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | - Yuta Yamamoto
- Applied Life Science Department, Faculty of Biotechnology and Life Science, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | - Sakiko Kimura
- Department of Pharmacy, Saga University Hospital, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Yutaka Narisawa
- Department of Pharmacy, Saga University Hospital, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 849‑8501, Japan
| | - Masashi Shin
- Applied Life Science Department, Faculty of Biotechnology and Life Science, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| |
Collapse
|
49
|
Natarajan H, Kumar L, Bakhshi S, Sharma A, Velpandian T, Kabra M, Gogia A, Ranjan Biswas N, Gupta YK. Imatinib trough levels: a potential biomarker to predict cytogenetic and molecular response in newly diagnosed patients with chronic myeloid leukemia. Leuk Lymphoma 2018; 60:418-425. [PMID: 30124353 DOI: 10.1080/10428194.2018.1485907] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Therapeutic drug monitoring of imatinib in patients with chronic myeloid leukemia (CML) is an ongoing debate. We studied the influence of imatinib trough levels on therapeutic response in 206 newly diagnosed patients with CML. We also compared the drug levels in patients taking branded and generic imatinib. Imatinib levels were measured using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Marked inter-individual variability was seen in imatinib levels (coefficient of variation = 69%). Trough levels were significantly higher in patients who attained complete cytogenetic response than those who did not (2213.9 ± 1101 vs. 1648.6 ± 1403.4ng/mL; p < .001). Patients with major molecular response (MMR) had higher trough levels than those without MMR (2333.4 ± 1112 vs. 1643.4 ± 1383.9ng/mL; p = .001). Patients with trough levels ≤1000ng/mL were at high risk for failure of imatinib therapy [RR =1.926; 95%CI (1.562, 2.374); p < .001]. Trough levels emerged as an independent predictor of imatinib response in multivariate analysis. To conclude, imatinib trough levels significantly influence cytogenetic and molecular response and might emerge as a potential biomarker for therapeutic response in CML.
Collapse
Affiliation(s)
- Harivenkatesh Natarajan
- a Department of Pharmacology , Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER) , Puducherry , India
| | - Lalit Kumar
- b Department of Medical Oncology , All India Institute of Medical Sciences , New Delhi , India
| | - Sameer Bakhshi
- b Department of Medical Oncology , All India Institute of Medical Sciences , New Delhi , India
| | - Atul Sharma
- b Department of Medical Oncology , All India Institute of Medical Sciences , New Delhi , India
| | - Thirumurthy Velpandian
- d Department of Pharmacology , All India Institute of Medical Sciences , New Delhi , India
| | - Madhulika Kabra
- c Department of Pediatrics , All India Institute of Medical Sciences , New Delhi , India
| | - Ajay Gogia
- b Department of Medical Oncology , All India Institute of Medical Sciences , New Delhi , India
| | - Nihar Ranjan Biswas
- d Department of Pharmacology , All India Institute of Medical Sciences , New Delhi , India
| | - Yogendra Kumar Gupta
- d Department of Pharmacology , All India Institute of Medical Sciences , New Delhi , India
| |
Collapse
|
50
|
Ultra-performance LC–MS/MS study of the pharmacokinetic interaction of imatinib with selected vitamin preparations in rats. Bioanalysis 2018; 10:1099-1113. [DOI: 10.4155/bio-2018-0043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: The growing interest of cancerous patients in using vitamins, while on imatinib (IMA) therapy, increased the risk of their pharmacokinetic interactions. Methodology: Ultra-performance LC–MS/MS method was developed and validated for the determination of IMA following oral administration of selected vitamin preparations (vitamin A, E, D3 and C) in rat plasma using a hybrid sample preparation technique of protein precipitation followed by SPE. Results: The method showed good linear response for IMA over the concentration range 1–500 ng/ml. Co-administered vitamin preparations could affect IMA pharmacokinetic profiling through either an increase (vitamin A and E) or a decrease (vitamin C) in IMA bioavailability. Vitamin D3 produced no significant effect on IMA bioavailability. Conclusion: Particular concern should be paid when vitamin preparations are administered with IMA.
Collapse
|