1
|
Persaud AK, Bernier MC, Massey MA, Agrawal S, Kaur T, Nayak D, Xie Z, Weadick B, Raj R, Hill K, Abbott N, Joshi A, Anabtawi N, Bryant C, Somogyi A, Cruz-Monserrate Z, Amari F, Coppola V, Sparreboom A, Baker SD, Unadkat JD, Phelps MA, Govindarajan R. Increased renal elimination of endogenous and synthetic pyrimidine nucleosides in concentrative nucleoside transporter 1 deficient mice. Nat Commun 2023; 14:3175. [PMID: 37264059 PMCID: PMC10235067 DOI: 10.1038/s41467-023-38789-8] [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: 03/31/2022] [Accepted: 05/16/2023] [Indexed: 06/03/2023] Open
Abstract
Concentrative nucleoside transporters (CNTs) are active nucleoside influx systems, but their in vivo roles are poorly defined. By generating CNT1 knockout (KO) mice, here we identify a role of CNT1 in the renal reabsorption of nucleosides. Deletion of CNT1 in mice increases the urinary excretion of endogenous pyrimidine nucleosides with compensatory alterations in purine nucleoside metabolism. In addition, CNT1 KO mice exhibits high urinary excretion of the nucleoside analog gemcitabine (dFdC), which results in poor tumor growth control in CNT1 KO mice harboring syngeneic pancreatic tumors. Interestingly, increasing the dFdC dose to attain an area under the concentration-time curve level equivalent to that achieved by wild-type (WT) mice rescues antitumor efficacy. The findings provide new insights into how CNT1 regulates reabsorption of endogenous and synthetic nucleosides in murine kidneys and suggest that the functional status of CNTs may account for the optimal action of pyrimidine nucleoside analog therapeutics in humans.
Collapse
Affiliation(s)
- Avinash K Persaud
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Matthew C Bernier
- Campus Chemical Instrument Center Mass Spectrometry and Proteomics Facility, The Ohio State University, Columbus, OH, 43210, USA
| | - Michael A Massey
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
- The Center for Life Sciences Education, College of Arts and Sciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Shipra Agrawal
- Division of Nephrology & Hypertension, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Tejinder Kaur
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Debasis Nayak
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Zhiliang Xie
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Brenna Weadick
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Ruchika Raj
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Kasey Hill
- Pharmacoanalytic Shared Resource (PhASR), The Ohio State University, Columbus, OH, 43205, USA
| | - Nicole Abbott
- Pharmacoanalytic Shared Resource (PhASR), The Ohio State University, Columbus, OH, 43205, USA
| | - Arnav Joshi
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Nadeen Anabtawi
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Claire Bryant
- Center for Clinical & Translational Research, Nationwide Children's Hospital, Columbus, OH, 43210, USA
| | - Arpad Somogyi
- Campus Chemical Instrument Center Mass Spectrometry and Proteomics Facility, The Ohio State University, Columbus, OH, 43210, USA
| | - Zobeida Cruz-Monserrate
- Division of Gastroenterology, Hepatology, and Nutrition, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Foued Amari
- Genetically Engineered Mouse Modeling Core, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Vincenzo Coppola
- Genetically Engineered Mouse Modeling Core, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Alex Sparreboom
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Sharyn D Baker
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Jashvant D Unadkat
- Department of Pharmaceutics, College of Pharmacy, University of Washington, Seattle, WA, 98195, USA
- Translational Therapeutics, Ohio State University Comprehensive Cancer Center, Ohio State University, Columbus, OH, 43210, USA
| | - Mitch A Phelps
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
- Pharmacoanalytic Shared Resource (PhASR), The Ohio State University, Columbus, OH, 43205, USA
| | - Rajgopal Govindarajan
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA.
- Translational Therapeutics, Ohio State University Comprehensive Cancer Center, Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
2
|
van der Heijden L, van Nuland M, Beijnen J, Huitema A, Dorlo T. A naïve pooled data approach for extrapolation of Phase 0 microdose trials to therapeutic dosing regimens. Clin Transl Sci 2022; 16:258-268. [PMID: 36419385 PMCID: PMC9926085 DOI: 10.1111/cts.13446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/22/2022] [Accepted: 10/07/2022] [Indexed: 11/25/2022] Open
Abstract
Microdosing is a strategy to obtain knowledge of human pharmacokinetics prior to Phase I clinical trials. The most frequently used method to extrapolate microdose (≤100 μg) pharmacokinetics to therapeutic doses is based on linear extrapolation from a noncompartmental analysis (NCA) with a two-fold acceptance criterion between pharmacokinetic metrics of the extrapolated microdose and the therapeutic dose. The major disadvantage of NCA is the assumption of linear extrapolation of NCA metrics. In this study, we used a naïve pooled data (NPD) modeling approach to extrapolate microdose pharmacokinetics to therapeutic pharmacokinetics. Gemcitabine and anastrozole were used as examples of intravenous and oral drugs, respectively. Data from microdose studies were used to build a parent-metabolite model for gemcitabine and its metabolite 2',2'-difluorodeoxyuridine (dFdU) and a model for anastrozole. The pharmacokinetic microdose models were extrapolated to therapeutic doses. Extrapolation of the microdose showed differences in pharmacokinetic shape for gemcitabine and dFdU between the simulated and observed therapeutic concentrations, whereas the observed therapeutic concentrations for anastrozole were captured by the extrapolation. This study demonstrated the possible use and feasibility of an NPD modeling approach for the evaluation and application of microdose studies in early drug development. Last, physiologically-based pharmacokinetic modeling might be an alternative for microdose extrapolation of drugs with complex pharmacokinetics such as gemcitabine.
Collapse
Affiliation(s)
- Lisa van der Heijden
- Department of Pharmacy & PharmacologyAntoni van Leeuwenhoek/The Netherlands Cancer InstituteAmsterdamThe Netherlands,Division of PharmacologyAntoni van Leeuwenhoek/The Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Merel van Nuland
- Department of Pharmacy & PharmacologyAntoni van Leeuwenhoek/The Netherlands Cancer InstituteAmsterdamThe Netherlands,Division of PharmacologyAntoni van Leeuwenhoek/The Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Jos Beijnen
- Department of Pharmacy & PharmacologyAntoni van Leeuwenhoek/The Netherlands Cancer InstituteAmsterdamThe Netherlands,Division of PharmacologyAntoni van Leeuwenhoek/The Netherlands Cancer InstituteAmsterdamThe Netherlands,Division of Pharmaco‐epidemiology and Clinical Pharmacology, Faculty of Science, Department of Pharmaceutical SciencesUtrecht UniversityUtrechtThe Netherlands
| | - Alwin Huitema
- Department of Pharmacy & PharmacologyAntoni van Leeuwenhoek/The Netherlands Cancer InstituteAmsterdamThe Netherlands,Division of PharmacologyAntoni van Leeuwenhoek/The Netherlands Cancer InstituteAmsterdamThe Netherlands,Department of Clinical PharmacyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands,Department of PharmacologyPrincess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | - Thomas Dorlo
- Department of Pharmacy & PharmacologyAntoni van Leeuwenhoek/The Netherlands Cancer InstituteAmsterdamThe Netherlands,Division of PharmacologyAntoni van Leeuwenhoek/The Netherlands Cancer InstituteAmsterdamThe Netherlands
| |
Collapse
|
3
|
Is age just a number? A population pharmacokinetic study of gemcitabine. Cancer Chemother Pharmacol 2022; 89:697-705. [PMID: 35426526 DOI: 10.1007/s00280-022-04431-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 04/01/2022] [Indexed: 11/02/2022]
Abstract
PURPOSE Pharmacokinetic exposure to gemcitabine and its metabolite, 2',2'-difluorodeoxyuridine (dFdU), might be altered in elderly compared to their younger counterparts. It is unknown if age-based dose adjustments are necessary to reduce the development of treatment-induced adverse events. The aim of this study was to assess the impact of age on the pharmacokinetics of gemcitabine and dFdU. METHODS Pharmacokinetic sampling following a flexible limited sampling strategy was performed in patients ≥ 70 years after gemcitabine infusion. The data were supplemented with pharmacokinetic data in patients included in four previously conducted clinical trials. Nonlinear mixed effects modelling was performed on the pooled dataset to assess the impact of age on the pharmacokinetics of gemcitabine and dFdU. RESULTS In total, pharmacokinetic data were available of 197 patients, of whom 83 patients were aged ≥ 70 years (42%). A two-compartment model for both gemcitabine and dFdU with linear clearances from the central compartments described the data best. Age, tested as continuous and categorical (< 70 years versus ≥ 70 years) covariate, did not statistically affect the pharmacokinetics of gemcitabine and dFdU. CONCLUSION Age was not of influence on the pharmacokinetics of gemcitabine or its metabolite, dFdU. Age-related dose adjustments for gemcitabine based on pharmacokinetic considerations are not recommended. TRIAL REGISTRATION NUMBER NL39647.048.12, registered on May 3rd 2012.
Collapse
|
4
|
Karuppaiah A, Babu D, Selvaraj D, Natrajan T, Rajan R, Gautam M, Ranganathan H, Siram K, Nesamony J, Sankar V. Building and behavior of a pH-stimuli responsive chitosan nanoparticles loaded with folic acid conjugated gemcitabine silver colloids in MDA-MB-453 metastatic breast cancer cell line and pharmacokinetics in rats. Eur J Pharm Sci 2021; 165:105938. [PMID: 34256103 DOI: 10.1016/j.ejps.2021.105938] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 10/20/2022]
Abstract
The pH-stimuli release behavior of nanoformulations may enhance the success rate of chemotherapeutic drugs in cancers by site-specific delivery of drugs to cancer tissues. The aim of the present study was to prepare chitosan (CS) nanoparticles (NPs) with previously synthesized folic acid (FA) capped silver nanoparticles (AgNPs) loaded with the anti-cancer drug gemcitabine (GEM) (FA-GEM-AgNPs). The CS-FA-GEM-AgNPs (CS-NPs) were characterized with dynamic light scattering (DLS), transmission electron microscopy (TEM), energy dispersive x-ray analysis (EDAX), selected area electron diffraction (SAED), and differential scanning calorimetric (DSC) analyses. The in-vitro drug release of GEM was evaluated in media of different pH. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was carried out to determine the cytotoxic effects of the prepared nanoformulations in media with various pH. The time- and pH-dependent apoptotic cell death induced by CS-NPs with MDA-MB-453 human breast cancer cell line was observed using acridine orange (AO)/ethidium bromide (EtBr) staining. The pharmacokinetic parameters were studied with high-performance liquid chromatography (HPLC) and atomic absorption spectroscopy (AAS). Two batches of CS-NPs formulations were prepared, one with AgNPs of particle size 143 nm and the other with 244 nm. The particle size for CS-NPs-I (FA-GEM-AgNPs-143 nm) and CS-NPs-II (FA-GEM-AgNPs-244 nm) was found to be 425 and 545 nm, respectively. The zeta potential was found to be 36.1 and 37.5 mV for CS-NPs-I and CS-NPs-II, respectively. CS-NPs-I and CS-NPs-II showed a polydispersity index (PDI) of 0.240 and 0.261, respectively. A TEM study confirmed the spherical nature of the NPs. The nanoformulations exerted pH-dependant effect against MDA-MB-453 cells with relatively higher cytotoxicity at the lower pH than at higher pH levels. The pharmacokinetic profile and tissue distribution of CS-NPs in rats exerted drug release in a pH-dependent manner with enhanced excretion of Ag+. An optimized nanoformulation for pH-stimuli responsive release of GEM was successfully developed for future therapeutic exploration.
Collapse
Affiliation(s)
- Arjunan Karuppaiah
- Department of Pharmaceutics, PSG College of Pharmacy, Coimbatore, Tamil Nadu, India
| | - Dinesh Babu
- Faculty of Pharmacy and Pharmaceutical Sciences, Katz Group Centre for Pharmacy and Health Research, University of Alberta, Edmonton, Alberta, Canada
| | - Divakar Selvaraj
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India; Department of Pharmacology, PSG College of Pharmacy, Coimbatore, Tamil Nadu, India
| | - Tamilselvan Natrajan
- Department of Pharmaceutics, KMCH College of Pharmacy, Coimbatore, Tamil Nadu, India
| | - Ravikumar Rajan
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, Tamil Nadu, India; Faculty of Medical Sciences, Arunachal University Studies, Namasi, Arunachal Pradesh, India
| | - Mrinmoy Gautam
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, Tamil Nadu, India
| | - Hariprasad Ranganathan
- Department of Pharmaceutical Analysis, PSG College of Pharmacy, Coimbatore, Tamil Nadu, India
| | - Karthik Siram
- Department of Pharmaceutics, PSG College of Pharmacy, Coimbatore, Tamil Nadu, India; Department of Biomedical and Pharmaceutical Sciences, Center for Translational Medicine, Skaggs School of Pharmacy, University of Montana, United States
| | - Jerry Nesamony
- Department of Pharmaceutics, Frederic and Mary Wolfe Center 114J, University of Toledo, United States
| | - Veintramuthu Sankar
- Department of Pharmaceutics, PSG College of Pharmacy, Coimbatore, Tamil Nadu, India.
| |
Collapse
|
5
|
LC-MS/MS method for quantitation of gemcitabine and its metabolite 2',2'-difluoro-2'-deoxyuridine in mouse plasma and brain tissue: Application to a preclinical pharmacokinetic study. J Pharm Biomed Anal 2021; 198:114025. [PMID: 33744463 DOI: 10.1016/j.jpba.2021.114025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 11/20/2022]
Abstract
A simple, sensitive, and relatively fast assay was developed and validated for the quantitation of gemcitabine (dFdC) and its major metabolite 2',2'-difluoro-2'-deoxyuridine (dFdU) in mouse plasma and brain tissue. The assay used a small sample (25 μL plasma and 5 mg brain) for extraction by protein precipitation. After dilution of the supernatant extract, 1 μL was injected into HPLC system for reverse phase chromatographic separation with a total run time of 8 min. Chromatographic resolution of dFdC and dFdU was achieved on a Gemini C18 column (50 × 4.6 mm, 3 μm) utilizing gradient elution. Multiple reaction monitoring (MRM) with positive/negative ion switching was performed for detection of dFdC and its internal standard (dFdC-IS) in positive ion mode and dFdU and its IS (dFdU-IS) in negative ion mode. Two calibration curves ranging from 5-2000 ng/mL and 250-50,000 ng/mL were generated for dFdC and dFdU in mouse plasma, respectively. For measurement of dFdC and dFdU in mouse brain tissue, another two curves were used ranging from 0.02 to 40 ng/mg and 1-40 ng/mg, respectively. This assay demonstrated excellent precision and accuracy within day and between days for simultaneous measurement of dFdC and dFdU at all the concentration levels in both matrices. The other parameters such as selectivity, sensitivity, matrix effects, recovery, and storage stability were also assessed for both analytes in each matrix. Compared to the previously reported methods, the sample extraction in the current assay was simplified significantly, and the analysis time was greatly shortened. We successfully applied the validated method to the analysis of dFdC and dFdU in mouse plasma, brain, and brain tumor tissue in a preclinical pharmacokinetic study.
Collapse
|
6
|
Van Nuland M, Rosing H, Thijssen B, Burgers JA, Huitema ADR, Marchetti S, Schellens JHM, Beijnen JH. Pilot Study to Predict Pharmacokinetics of a Therapeutic Gemcitabine Dose From a Microdose. Clin Pharmacol Drug Dev 2020; 9:929-937. [PMID: 31970932 DOI: 10.1002/cpdd.774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022]
Abstract
Microdose studies are exploratory trials to determine early drug pharmacokinetics in humans. In this trial we examined whether the pharmacokinetics of gemcitabine at a therapeutic dose could be predicted from the pharmacokinetics of a microdose. In this prospective, open-label microdosing study, a gemcitabine microdose (100 µg) was given intravenously to participants on day 1, followed by a therapeutic dose (1250 mg/m2 ) on day 2. Gemcitabine and its metabolite 2',2'-difluorodeoxyuracil (dFdU) were quantified in plasma and intracellularly by using liquid chromatography-mass spectrometry). Noncompartmental pharmacokinetic analysis was performed. Ten patients participated in this study. The mean area under the plasma concentration-time curve (AUC0-8 ) of gemcitabine after microdosing was 0.00074 h·mg/L and after therapeutic dosing was 16 h·mg/L. The mean AUC0-8 of dFdU following the microdose and therapeutic dose were 0.022 h·mg/L and 169 h·mg/L, respectively. Exposure to gemcitabine after the therapeutic dose was within 2-fold of the exposure following a microdose, when linearly extrapolated to 1250 mg/m2 . However, the shape of the concentration-time curve was different, as reflected by poor scalability in volume of distribution (939 L versus 222 L). Furthermore, intracellularly phosphorylated gemcitabine and phosphorylated dFdU levels could not be predicted from the microdose. The AUC0-8 of gemcitabine at therapeutic dose was accurately predicted by the pharmacokinetics of a microdose, when linearly extrapolated to 1250 mg/m2 . Volume of distribution, elimination rate constant, and intracellular pharmacokinetics of the therapeutic dose could not be predicted from the microdose, which demonstrates limitations of the microdose approach in this case.
Collapse
Affiliation(s)
- M Van Nuland
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - H Rosing
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - B Thijssen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - J A Burgers
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - A D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.,Division of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.,Department of Clinical Pharmacy University Medical Center Utrecht, Utrecht University, the Netherlands
| | - S Marchetti
- Division of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - J H M Schellens
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - J H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.,Division of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.,Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
7
|
A Phase Ib Study of the FGFR/VEGFR Inhibitor Dovitinib With Gemcitabine and Capecitabine in Advanced Solid Tumor and Pancreatic Cancer Patients. Am J Clin Oncol 2019; 42:184-189. [PMID: 30418178 DOI: 10.1097/coc.0000000000000492] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVES Preclinical studies demonstrated antitumor activity of dovitinib in pancreatic cancer models. This phase Ib study aimed to determine the maximum tolerated dose (MTD) of dovitinib in combination with gemcitabine and capecitabine and to characterize the safety and pharmacokinetic profile in patients with advanced pancreatic and biliary tract cancers and solid malignancies. MATERIALS AND METHODS Patients received gemcitabine 1000 mg/m² intravenously on days 1 and 8, capecitabine 1300 mg/m² oral daily from day 1 to 14, and dovitinib oral daily 5 days on and 2 days off, every 21-day cycle. The standard 3+3 dose escalation design was utilized and the study expanded to treat an additional 20 advanced pancreatic and biliary tract cancers patients at MTD. RESULTS A total of 29 patients were enrolled. One patient experienced dose-limiting grade 3 colitis. Two patients developed clinically significant neuropathy after the first cycle requiring dose reduction. The MTD was not reached and dovitinib 300 mg was declared the recommended dose for expansion. The most frequent grade 2 or worse adverse events were fatigue (45%), neutropenia (41%), thrombocytopenia (34%), anemia (24%), nausea (24%), and palmer-plantar erythrodysaesthesia syndrome (21%). Partial responses were observed in 5 patients. Pharmacokinetic studies showed no drug-drug interaction between dovitinib, capecitabine and gemcitabine. Fibroblast growth factor 23 plasma level increased in 4 of 5 patients during the first cycle of treatment. CONCLUSIONS Dovitinib 300 mg daily is the recommended dose when combined with gemcitabine and capecitabine, achieving clinically relevant plasma concentrations. The study combination demonstrated encouraging efficacy signals in advanced pancreatic cancer.
Collapse
|
8
|
Derissen EJB, Huitema ADR, Rosing H, Schellens JHM, Beijnen JH. Intracellular pharmacokinetics of gemcitabine, its deaminated metabolite 2',2'-difluorodeoxyuridine and their nucleotides. Br J Clin Pharmacol 2018; 84:1279-1289. [PMID: 29451684 DOI: 10.1111/bcp.13557] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 01/01/2018] [Accepted: 01/28/2018] [Indexed: 12/17/2022] Open
Abstract
AIMS Gemcitabine (2',2'-difluoro-2'-deoxycytidine; dFdC) is a prodrug that has to be phosphorylated within the tumour cell to become active. Intracellularly formed gemcitabine diphosphate (dFdCDP) and triphosphate (dFdCTP) are considered responsible for the antineoplastic effects of gemcitabine. However, a major part of gemcitabine is converted into 2',2'-difluoro-2'-deoxyuridine (dFdU) by deamination. In the cell, dFdU can also be phosphorylated to its monophosphate (dFdUMP), diphosphate (dFdUDP) and triphosphate (dFdUTP). In vitro data suggest that these dFdU nucleotides might also contribute to the antitumour effects, although little is known about their intracellular pharmacokinetics (PK). Therefore, the objective of the present study was to gain insight into the intracellular PK of all dFdC and dFdU nucleotides formed during gemcitabine treatment. METHODS Peripheral blood mononuclear cell (PBMC) samples were collected from 38 patients receiving gemcitabine, at multiple time points after infusion. Gemcitabine, dFdU and their nucleotides were quantified in PBMCs. In addition, gemcitabine and dFdU plasma concentrations were monitored. The individual PK parameters in plasma and in PBMCs were determined. RESULTS Both in plasma and in PBMCs, dFdU was present in higher concentrations than gemcitabine [mean intracellular area under the concentration-time curve from time zero to 24 h (AUC0-24 h ) 1650 vs. 95 μM*h]. However, the dFdUMP, dFdUDP and dFdUTP concentrations in PBMCs were much lower than the dFdCDP and dFdCTP concentrations. The mean AUC0-24 h for dFdUTP was 312 μM*h vs. 2640 μM*h for dFdCTP. CONCLUSIONS The study provides the first complete picture of all nucleotides that are formed intracellularly during gemcitabine treatment. Low intracellular dFdU nucleotide concentrations were found, which calls into question the relevance of these nucleotides for the cytotoxic effects of gemcitabine.
Collapse
Affiliation(s)
- Ellen J B Derissen
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek Hospital - The Netherlands Cancer Institute and MC Slotervaart, Louwesweg, 6, 1066, EC, Amsterdam, The Netherlands.,Department of Clinical Pharmacology and Pharmacy, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek Hospital - The Netherlands Cancer Institute and MC Slotervaart, Louwesweg, 6, 1066, EC, Amsterdam, The Netherlands.,Department of Clinical Pharmacy, University Medical Center Utrecht, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands
| | - Hilde Rosing
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek Hospital - The Netherlands Cancer Institute and MC Slotervaart, Louwesweg, 6, 1066, EC, Amsterdam, The Netherlands
| | - Jan H M Schellens
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands.,Science Faculty, Utrecht Institute for Pharmaceutical Sciences (UIPS), Division of Pharmaco-epidemiology & Clinical Pharmacology, Utrecht University, P.O. Box 80082, 3508, TB, Utrecht, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek Hospital - The Netherlands Cancer Institute and MC Slotervaart, Louwesweg, 6, 1066, EC, Amsterdam, The Netherlands.,Science Faculty, Utrecht Institute for Pharmaceutical Sciences (UIPS), Division of Pharmaco-epidemiology & Clinical Pharmacology, Utrecht University, P.O. Box 80082, 3508, TB, Utrecht, The Netherlands
| |
Collapse
|
9
|
van Nuland M, Hillebrand MJX, Rosing H, Burgers JA, Schellens JHM, Beijnen JH. Ultra-sensitive LC-MS/MS method for the quantification of gemcitabine and its metabolite 2',2'-difluorodeoxyuridine in human plasma for a microdose clinical trial. J Pharm Biomed Anal 2017; 151:25-31. [PMID: 29294409 DOI: 10.1016/j.jpba.2017.12.048] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/15/2017] [Accepted: 12/22/2017] [Indexed: 10/18/2022]
Abstract
In microdose clinical trials a maximum of 100 μg of drug substance is administered to participants, in order to determine the pharmacokinetic properties of the agents. Measuring low plasma concentrations after administration of a microdose is challenging and requires the use of ulta-sensitive equipment. Novel liquid chromatography-mass spectrometry (LC-MS/MS) platforms can be used for quantification of low drug plasma levels. Here we describe the development and validation of an LC-MS/MS method for quantification of gemcitabine and its metabolite 2',2'-difluorodeoxyuridine (dFdU) in the low picogram per milliliter range to support a microdose trial. The validated assay ranges from 2.5-500 pg/mL for gemcitabine and 250-50,000 pg/mL for dFdU were linear, with a correlation coefficient (r2) of 0.996 or better. Sample preparation with solid phase extraction provided a good and reproducible recovery. All results were within the acceptance criteria of the latest US FDA guidance and EMA guidelines. In addition, the method was successfully applied to measure plasma concentrations of gemcitabine in a patient after administration of a microdose of gemcitabine.
Collapse
Affiliation(s)
- M van Nuland
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek/The Netherlands Cancer Institute and MC Slotervaart, Amsterdam, The Netherlands; Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| | - M J X Hillebrand
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek/The Netherlands Cancer Institute and MC Slotervaart, Amsterdam, The Netherlands
| | - H Rosing
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek/The Netherlands Cancer Institute and MC Slotervaart, Amsterdam, The Netherlands
| | - J A Burgers
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J H M Schellens
- Division of Clinical Pharmacology, Department of Medical Oncology, Antoni van Leeuwenhoek/The Netherlands Cancer Institute, Amsterdam, The Netherlands; Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands; Division of Pharmacology, Antoni van Leeuwenhoek/The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J H Beijnen
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek/The Netherlands Cancer Institute and MC Slotervaart, Amsterdam, The Netherlands; Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands; Division of Pharmacology, Antoni van Leeuwenhoek/The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
10
|
Epstein–Barr virus-targeted therapy in nasopharyngeal carcinoma. J Cancer Res Clin Oncol 2015; 141:1845-57. [DOI: 10.1007/s00432-015-1969-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/30/2015] [Indexed: 01/28/2023]
|
11
|
Keizer RJ, Jansen RS, Rosing H, Thijssen B, Beijnen JH, Schellens JHM, Huitema ADR. Incorporation of concentration data below the limit of quantification in population pharmacokinetic analyses. Pharmacol Res Perspect 2015; 3:e00131. [PMID: 26038706 PMCID: PMC4448983 DOI: 10.1002/prp2.131] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 02/09/2015] [Accepted: 02/11/2015] [Indexed: 11/30/2022] Open
Abstract
Handling of data below the lower limit of quantification (LLOQ), below the limit of quantification (BLOQ) in population pharmacokinetic (PopPK) analyses is important for reducing bias and imprecision in parameter estimation. We aimed to evaluate whether using the concentration data below the LLOQ has superior performance over several established methods. The performance of this approach (“All data”) was evaluated and compared to other methods: “Discard,” “LLOQ/2,” and “LIKE” (likelihood-based). An analytical and residual error model was constructed on the basis of in-house analytical method validations and analyses from literature, with additional included variability to account for model misspecification. Simulation analyses were performed for various levels of BLOQ, several structural PopPK models, and additional influences. Performance was evaluated by relative root mean squared error (RMSE), and run success for the various BLOQ approaches. Performance was also evaluated for a real PopPK data set. For all PopPK models and levels of censoring, RMSE values were lowest using “All data.” Performance of the “LIKE” method was better than the “LLOQ/2” or “Discard” method. Differences between all methods were small at the lowest level of BLOQ censoring. “LIKE” method resulted in low successful minimization (<50%) and covariance step success (<30%), although estimates were obtained in most runs (∼90%). For the real PK data set (7.4% BLOQ), similar parameter estimates were obtained using all methods. Incorporation of BLOQ concentrations showed superior performance in terms of bias and precision over established BLOQ methods, and shown to be feasible in a real PopPK analysis.
Collapse
Affiliation(s)
- Ron J Keizer
- Department of Bioengineering & Therapeutic Sciences, University of California San Francisco Box 2911, San Francisco, California, 94143 ; Department of Pharmacy & Pharmacology, Slotervaart Hospital/The Netherlands Cancer Institute Louwesweg 6, 1066 EC, Amsterdam, The Netherlands
| | - Robert S Jansen
- Department of Pharmacy & Pharmacology, Slotervaart Hospital/The Netherlands Cancer Institute Louwesweg 6, 1066 EC, Amsterdam, The Netherlands
| | - Hilde Rosing
- Department of Pharmacy & Pharmacology, Slotervaart Hospital/The Netherlands Cancer Institute Louwesweg 6, 1066 EC, Amsterdam, The Netherlands
| | - Bas Thijssen
- Department of Pharmacy & Pharmacology, Slotervaart Hospital/The Netherlands Cancer Institute Louwesweg 6, 1066 EC, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, Slotervaart Hospital/The Netherlands Cancer Institute Louwesweg 6, 1066 EC, Amsterdam, The Netherlands ; Division of Drug Toxicology, Section of Biomedical Analysis, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University Utrecht, The Netherlands
| | - Jan H M Schellens
- Division of Drug Toxicology, Section of Biomedical Analysis, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University Utrecht, The Netherlands ; Division of Clinical Pharmacology, Department of Medical Oncology, Antoni van Leeuwenhoek, Hospital/the Netherlands Cancer Institute Amsterdam, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, Slotervaart Hospital/The Netherlands Cancer Institute Louwesweg 6, 1066 EC, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Stability-indicating HPLC determination of gemcitabine in pharmaceutical formulations. Int J Anal Chem 2015; 2015:862592. [PMID: 25838825 PMCID: PMC4370102 DOI: 10.1155/2015/862592] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 01/23/2015] [Accepted: 01/26/2015] [Indexed: 11/17/2022] Open
Abstract
A simple, sensitive, inexpensive, and rapid stability indicating high performance liquid chromatographic method has been developed for determination of gemcitabine in injectable dosage forms using theophylline as internal standard. Chromatographic separation was achieved on a Phenomenex Luna C-18 column (250 mm × 4.6 mm; 5μ) with a mobile phase consisting of 90% water and 10% acetonitrile (pH 7.00 ± 0.05). The signals of gemcitabine and theophylline were recorded at 275 nm. Calibration curves were linear in the concentration range of 0.5–50 μg/mL. The correlation coefficient was 0.999 or higher. The limit of detection and limit of quantitation were 0.1498 and 0.4541 μg/mL, respectively. The inter- and intraday precision were less than 2%. Accuracy of the method ranged from 100.2% to 100.4%. Stability studies indicate that the drug was stable to sunlight and UV light. The drug gives 6 different hydrolytic products under alkaline stress and 3 in acidic condition. Aqueous and oxidative stress conditions also degrade the drug. Degradation was higher in the alkaline condition compared to other stress conditions. The robustness of the methods was evaluated using design of experiments. Validation reveals that the proposed method is specific, accurate, precise, reliable, robust, reproducible, and suitable for the quantitative analysis.
Collapse
|
13
|
Leijen S, Burgers SA, Baas P, Pluim D, Tibben M, van Werkhoven E, Alessio E, Sava G, Beijnen JH, Schellens JHM. Phase I/II study with ruthenium compound NAMI-A and gemcitabine in patients with non-small cell lung cancer after first line therapy. Invest New Drugs 2014; 33:201-14. [PMID: 25344453 DOI: 10.1007/s10637-014-0179-1] [Citation(s) in RCA: 305] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/15/2014] [Indexed: 11/30/2022]
Abstract
BACKGROUND This phase I/II study determined the maximal tolerable dose, dose limiting toxicities, antitumor activity, the pharmacokinetics and pharmacodynamics of ruthenium compound NAMI-A in combination with gemcitabine in Non-Small Cell Lung Cancer patients after first line treatment. METHODS Initial dose escalation of NAMI-A was performed in a 28 day cycle: NAMI-A as a 3 h infusion through a port-a-cath at a starting dose of 300 mg/m(2) at day 1, 8 and 15, in combination with gemcitabine 1,000 mg/m(2) at days 2, 9 and 16. Subsequently, dose escalation of NAMI-A in a 21 day schedule was explored. At the maximal tolerable dose level of this schedule an expansion group was enrolled of which 15 patients were evaluable for response. RESULTS Due to frequent neutropenic dose interruptions in the third week, the 28 day schedule was amended into a 21 day schedule. The maximal tolerable dose was 300 and 450 mg/m(2) of NAMI-A (21 day schedule). Main adverse events consisted of neutropenia, anemia, elevated liver enzymes, transient creatinine elevation, nausea, vomiting, constipation, diarrhea, fatigue, and renal toxicity. CONCLUSION NAMI-A administered in combination with gemcitabine is only moderately tolerated and less active in NSCLC patients after first line treatment than gemcitabine alone.
Collapse
Affiliation(s)
- Suzanne Leijen
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Veeraraghavan S, Thappali S, Viswanadha S, Nalla S, Chennupati S, Golla M, Vakkalanka S, Rangasamy M. Simultaneous quantification of idelalisib, fludarabine and lenalidomide in rat plasma by using high-performance liquid chromatography coupled with heated electrospray ionization tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2014; 949-950:63-9. [DOI: 10.1016/j.jchromb.2014.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 11/29/2013] [Accepted: 01/01/2014] [Indexed: 10/25/2022]
|
15
|
Joerger M, Huitema ADR, Koeberle D, Rosing H, Beijnen JH, Hitz F, Cerny T, Schellens JHM, Gillessen S. Safety and pharmacology of gemcitabine and capecitabine in patients with advanced pancreatico-biliary cancer and hepatic dysfunction. Cancer Chemother Pharmacol 2013; 73:113-24. [PMID: 24166106 DOI: 10.1007/s00280-013-2327-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 10/15/2013] [Indexed: 10/26/2022]
Abstract
PURPOSE We assessed the impact of hepatic dysfunction on the safety and pharmacology of gemcitabine/capecitabine in patients with advanced pancreatico-biliary cancer. METHODS We included 12 patients receiving 3 weekly gemcitabine 1,000 mg/m(2) day 1, 8 and oral capecitabine 650 mg/m(2) b.i.d. over 2 weeks until disease progression or intolerable toxicity. Patients were included into one normal hepatic function cohort [total bilirubin (TB) ≤15 μmol/L] and 3 cohorts with increasing TB (16-39, 40-80, >80 μmol/L). Three patients with a creatinine clearance <60 ml/min were also included. Patients were sampled for gemcitabine, difluoro-deoxy uridine, intracellular gemcitabine triphosphates, capecitabine, 5'-deoxy-5-fluorocytidine, 5'-deoxy-5-fluorouridine and 5-fluorouracil up to 4 h after initiation of chemotherapy on day 1, and up to 90 min on day 8. All compounds were analyzed using validated liquid chromatography-tandem mass spectrometry. Nonlinear mixed-effect modeling was used for population analysis. RESULTS Hepatic dysfunction was caused by intrahepatic cholestasis in 4 out of 8 patients (50 %) and extrahepatic cholestasis in another 4 patients (50 %). Dose-limiting toxicity was increasing hyperbilirubinemia and severe neutropenia in 2 patients each. Hepatic dysfunction was not associated with dose-limiting toxicity or severe hematological or non-hematological toxicity. However, hepatic dysfunction was associated with low clearance of both gemcitabine (p = 10(-3)) and capecitabine (p = 10(-5)), and low intracellular gemcitabine triphosphate concentrations (p = 10(-3)). CONCLUSIONS Gemcitabine/capecitabine can be given at the standard dose in patients with severe hyperbilirubinemia, though the present data suggest that gemcitabine's activity may be limited due to poor intracellular activation. In patients with severe hyperbilirubinemia, initial monotherapy with capecitabine should be considered, followed by the addition of gemcitabine with improving hyperbilirubinemia.
Collapse
Affiliation(s)
- M Joerger
- Department of Medical Oncology and Hematology, Cantonal Hospital, Rorschacherstrasse 95, 9007, St. Gallen, Switzerland,
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Wickremsinhe ER, Lee LB, Schmalz CA, Torchia J, Ruterbories KJ. High sensitive assay employing column switching chromatography to enable simultaneous quantification of an amide prodrug of gemcitabine (LY2334737), gemcitabine, and its metabolite dFdU in human plasma by LC-MS/MS. J Chromatogr B Analyt Technol Biomed Life Sci 2013; 932:117-22. [PMID: 23831704 DOI: 10.1016/j.jchromb.2013.06.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 05/30/2013] [Accepted: 06/07/2013] [Indexed: 10/26/2022]
Abstract
In this study we report a high sensitive method for the simultaneous analysis of LY2334737 (2'-deoxy-2',2'-difluoro-N-(1-oxo-2-propylpentyl)-cytidine), an amide prodrug of gemcitabine (2', 2'-difluoro-deoxycytidine), along with its active drug gemcitabine and its major metabolite dFdU (2',2'-difluoro-deoxyuridine) by LC-MS/MS. Quantification of all three analytes within a single analysis was challenging because the physio-chemical properties of LY2334737 were significantly different from gemcitabine and dFdU and was accomplished by incorporating column-switching. The assay was fully validated to quantify LY2334737 from 0.1 to 100ng/mL, gemcitabine from 0.25 to 100ng/mL and dFdU from 1 to 1000ng/mL in order to cover the diverse concentration ranges expected in clinical samples. A 25-fold dilution was also validated to accommodate any samples outside this range. Overall, the assay had good accuracy (ranging from -7.0 to 1.2% relative error) and precision (ranging from 2.1 to 8.4% relative standard deviation). Extraction efficiency was greater than 80% for all three analytes and there were no matrix effects. Plasma samples were stable for 24h at room temperature, 660 days in frozen storage, and at least 4 freeze-thaw cycles, at both -20 and -70°C. Data from clinical trials showed that plasma concentrations for LY2334737, gemcitabine, and dFdU were successfully quantified from a single LC-MS/MS analysis and that the assay ranges selected for the three analytes were appropriate and minimized the need for reanalysis.
Collapse
|
17
|
Leijen S, Veltkamp SA, Huitema ADR, van Werkhoven E, Beijnen JH, Schellens JHM. Phase I dose-escalation study and population pharmacokinetic analysis of fixed dose rate gemcitabine plus carboplatin as second-line therapy in patients with ovarian cancer. Gynecol Oncol 2013; 130:511-7. [PMID: 23665458 DOI: 10.1016/j.ygyno.2013.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Revised: 04/30/2013] [Accepted: 05/02/2013] [Indexed: 01/15/2023]
Abstract
OBJECTIVE This phase I study of fixed dose rate (FDR) gemcitabine and carboplatin assessed the maximum tolerated dose (MTD), dose-limiting toxicities (DLTs), safety, pharmacokinetic (PK)/pharmacodynamic (PD) profile and preliminary anti-tumor activity in patients with recurrent ovarian cancer (OC). METHODS Patients with recurrent OC after first line treatment were treated with carboplatin and FDR gemcitabine (infusion speed 10mg/m(2)/min) on days 1, 8 and 15, every 28 days. Pharmacokinetics included measurement of platinum concentrations in plasma ultrafiltrate (pUF) and plasma concentrations of gemcitabine (dFdC) and metabolite dFdU. Intracellular levels of dFdC triphosphate (dFdC-TP), the most active metabolite of gemcitabine, were determined in peripheral blood mononuclear cells (PBMCs). Population pharmacokinetic modeling and simulation were performed to further investigate the optimal schedule. RESULTS Twenty three patients were enrolled. Initial dose escalation was performed using FDR gemcitabine 300 mg/m(2) (administered at infusion speed of 10 mg/m(2)/min) combined with carboplatin AUC 2.5 and 3. Excessive bone marrow toxicity led to a modified dose escalation schedule: carboplatin AUC 2 and dose escalation of FDR gemcitabine (300 mg/m(2), 450 mg/m(2), 600 mg/m(2) and 800 mg/m(2)). DLT criteria as defined per protocol prior to the study were not met with carboplatin AUC 2 in combination with FDR gemcitabine 300-800 mg/m(2) because of myelosuppressive dose-holds (especially thrombocytopenia and neutropenia). CONCLUSIONS FDR gemcitabine in combination with carboplatin administered in this 28 days schedule resulted in increased grade 3/4 toxicity compared to conventional 30-minute infused gemcitabine. A two weekly schedule (chemotherapy on days 1 and 8) would be more appropriate.
Collapse
Affiliation(s)
- Suzanne Leijen
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
18
|
Liquid chromatography coupled to tandem mass spectrometry and high resolution mass spectrometry as analytical tools to characterize multi-class cytostatic compounds. J Chromatogr A 2013; 1276:78-94. [DOI: 10.1016/j.chroma.2012.12.031] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 12/14/2012] [Accepted: 12/17/2012] [Indexed: 11/19/2022]
|
19
|
Sun Y, Tang D, Chen H, Zhang F, Fan B, Zhang B, Fang S, Lu Q, Wei Y, Yin J, Yin X. Determination of gemcitabine and its metabolite in extracellular fluid of rat brain tumor by ultra performance liquid chromatography-tandem mass spectrometry using microdialysis sampling after intralesional chemotherapy. J Chromatogr B Analyt Technol Biomed Life Sci 2013; 919-920:10-9. [PMID: 23396113 DOI: 10.1016/j.jchromb.2012.12.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 11/26/2012] [Accepted: 12/09/2012] [Indexed: 10/27/2022]
Abstract
The cytotoxic agent Gemcitabine (2',2'-difluoro-2'-deoxycytidine) has been proved to be effective in the treatment of malignant gliomas. A rapid, sensitive and specific ultra performance liquid chromatography with tandem mass spectrometry (UPLC-MS/MS) assay using microdialysis sampling was developed and validated to quantify gemcitabine and its major metabolite 2',2'-difluoro-2'-deoxyuridine (dFdU) in Sprague-Dawley rat bearing 9L glioma. Microdialysis probes were surgically implanted into the area of rat brain tumor in the striatal hemisphere, and artificial cerebrospinal fluid was used as a perfusion medium. The samples were analyzed directly by UPLC-MS/MS after the addition of 5-bromouracil as an internal standard (IS). Separation was achieved on Agilent SB-C(18) (50 mm × 2.1mm I.D., 1.8 μm) column at 40 °C using an isocratic elution method with acetonitrile and 0.1% formic acid (4:96, v/v) at a flow rate of 0.2 mL/min. Detection was performed using electrospray ionization in positive ion selected reaction monitoring mode by monitoring the following ion transitions m/z 264.0→112.0 (gemcitabine), m/z 265.1→113.0 (dFdU) and m/z 190.9→173.8 (IS). The calibration curves of gemcitabine and dFdU were linear in the concentration range of 0.66-677.08 ng/mL and 0.31-312.00 ng/mL, respectively. The lower limit of quantification of gemcitabine and dFdU were 0.66 ng/mL and 0.31 ng/mL, respectively. The lower limit of detection of gemcitabine and dFdU were calculated to be 0.2 ng/mL and 0.1 ng/mL, respectively. All the validation data, such as intra- and inter-day precision, accuracy, selectivity and stability, were within the required limits. The validated method was simple, precise and accurate, which was successfully employed to determinate the concentrations of gemcitabine and dFdU in the extracellular fluid of rat brain tumor.
Collapse
Affiliation(s)
- Ying Sun
- Key Laboratory of New Drug and Clinical Application, Xuzhou Medical College, No. 209 Tongshan Road, Xuzhou 221004, Jiangsu, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Zhou J, Gao S, Zhang F, Jiang B, Zhan Q, Cai F, Li J, Chen W. Liquid chromatography–tandem mass spectrometry method for simultaneous determination of seven commonly used anticancer drugs in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2012; 906:1-8. [DOI: 10.1016/j.jchromb.2012.07.033] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 07/21/2012] [Accepted: 07/24/2012] [Indexed: 12/17/2022]
|
21
|
Guo F, Mao X, Wang J, Luo F, Wang Z. Gemcitabine adsorbed onto carbon particles increases drug concentrations at the injection site and in the regional lymph nodes in an animal experiment and a clinical study. J Int Med Res 2012; 39:2217-27. [PMID: 22289537 DOI: 10.1177/147323001103900618] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
This study investigated whether gemcitabine, adsorbed onto activated carbon particles (GEM-AC), increased the concentration of gemcitabine at the injection site and in the regional lymph nodes in an experimental animal model and a clinical study. The adsorption isotherm for GEM-AC was defined, and the concentration and distribution of gemcitabine in rats (n = 50) and in patients with pancreatic cancer (n = 8) was investigated. Drug concentrations in plasma, tumour samples, lymph nodes and at the injection site were measured after GEM-AC or gemcitabine solution (GEM-Sol) were subcutaneously injected into the left hind foot pad in rats, or into pancreatic tumours in patients. These experiments showed that GEM-AC was selectively delivered to the regional lymph nodes and the injection site, from which it slowly released greater amounts of gemcitabine to maintain the free concentration of gemcitabine at a relatively high level for a long period of time. The administration of GEM-AC might enhance the anticancer effects of gemcitabine.
Collapse
Affiliation(s)
- F Guo
- Department of General Surgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | | | |
Collapse
|
22
|
Saiki Y, Yoshino Y, Fujimura H, Manabe T, Kudo Y, Shimada M, Mano N, Nakano T, Lee Y, Shimizu S, Oba S, Fujiwara S, Shimizu H, Chen N, Nezhad ZK, Jin G, Fukushige S, Sunamura M, Ishida M, Motoi F, Egawa S, Unno M, Horii A. DCK is frequently inactivated in acquired gemcitabine-resistant human cancer cells. Biochem Biophys Res Commun 2012; 421:98-104. [PMID: 22490663 DOI: 10.1016/j.bbrc.2012.03.122] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2012] [Accepted: 03/27/2012] [Indexed: 12/11/2022]
Abstract
Although gemcitabine is the most effective chemotherapeutic agent against pancreatic cancer, a growing concern is that a substantial number of patients acquire gemcitabine chemoresistance. To elucidate the mechanisms of acquisition of gemcitabine resistance, we developed gemcitabine-resistant cell lines from six human cancer cell lines; three pancreatic, one gastric, one colon, and one bile duct cancer. We first analyzed gemcitabine uptake using three paired parental and gemcitabine resistant pancreatic cancer cell lines (PK-1 and RPK-1, PK-9 and RPK-9, PK-59 and RPK-59) and found that uptake of gemcitabine was rapid. However, no DNA damage was induced in resistant cells. We further examined the microarray-based expression profiles of the cells to identify genes associated with gemcitabine resistance and found a remarkable reduction in the expression of deoxycytidine kinase (DCK). DCK is a key enzyme that activates gemcitabine by phosphorylation. Genetic alterations and expression of DCK were studied in these paired parental and derived gemcitabine-resistant cell lines, and inactivating mutations were found only in gemcitabine-resistant cell lines. Furthermore, siRNA-mediated knockdown of DCK in the parental cell lines yielded gemcitabine resistance, and introduction of DCK into gemcitabine-resistant cell lines invariably restored gemcitabine sensitivities. Mutation analyses were expanded to three other different paired cell lines, DLD-1 and RDLD-1 (colon cancer cell line), MKN-28 and RMKN-28 (gastric cancer cell line), and TFK-1 and RTFK -1 (cholangiocarcinoma cell line). We found inactivating mutations in RDLD-1 and RTFK-1 and decreased expression of DCK in RMKN-28. These results indicate that the inactivation of DCK is one of the crucial mechanisms in acquisition of gemcitabine resistance.
Collapse
Affiliation(s)
- Yuriko Saiki
- Department of Molecular Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Bapiro TE, Richards FM, Goldgraben MA, Olive KP, Madhu B, Frese KK, Cook N, Jacobetz MA, Smith DM, Tuveson DA, Griffiths JR, Jodrell DI. A novel method for quantification of gemcitabine and its metabolites 2',2'-difluorodeoxyuridine and gemcitabine triphosphate in tumour tissue by LC-MS/MS: comparison with (19)F NMR spectroscopy. Cancer Chemother Pharmacol 2011; 68:1243-53. [PMID: 21431415 PMCID: PMC3215866 DOI: 10.1007/s00280-011-1613-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Accepted: 03/04/2011] [Indexed: 02/06/2023]
Abstract
PURPOSE To develop a sensitive analytical method to quantify gemcitabine (2',2'-difluorodeoxycytidine, dFdC) and its metabolites 2',2'-difluorodeoxyuridine (dFdU) and 2',2'-difluorodeoxycytidine-5'-triphosphate (dFdCTP) simultaneously from tumour tissue. METHODS Pancreatic ductal adenocarcinoma tumour tissue from genetically engineered mouse models of pancreatic cancer (KP ( FL/FL ) C and KP ( R172H/+) C) was collected after dosing the mice with gemcitabine. (19)F NMR spectroscopy and LC-MS/MS protocols were optimised to detect gemcitabine and its metabolites in homogenates of the tumour tissue. RESULTS A (19)F NMR protocol was developed, which was capable of distinguishing the three analytes in tumour homogenates. However, it required at least 100 mg of the tissue in question and a long acquisition time per sample, making it impractical for use in large PK/PD studies or clinical trials. The LC-MS/MS protocol was developed using porous graphitic carbon to separate the analytes, enabling simultaneous detection of all three analytes from as little as 10 mg of tissue, with a sensitivity for dFdCTP of 0.2 ng/mg tissue. Multiple pieces of tissue from single tumours were analysed, showing little intra-tumour variation in the concentrations of dFdC or dFdU (both intra- and extra-cellular). Intra-tumoural variation was observed in the concentration of dFdCTP, an intra-cellular metabolite, which may reflect regions of different cellularity within a tumour. CONCLUSION We have developed a sensitive LC-MS/MS method capable of quantifying gemcitabine, dFdU and dFdCTP in pancreatic tumour tissue. The requirement for only 10 mg of tissue enables this protocol to be used to analyse multiple areas from a single tumour and to spare tissue for additional pharmacodynamic assays.
Collapse
Affiliation(s)
- Tashinga E. Bapiro
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Box 278, Cambridge, CB2 0RE UK
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Frances M. Richards
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Box 278, Cambridge, CB2 0RE UK
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Mae A. Goldgraben
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Box 278, Cambridge, CB2 0RE UK
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Kenneth P. Olive
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Box 278, Cambridge, CB2 0RE UK
- Present Address: Herbert Irving Comprehensive Cancer Center and Departments of Medicine and Pathology, Columbia University, New York, NY 10032 USA
| | - Basetti Madhu
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Box 278, Cambridge, CB2 0RE UK
| | - Kristopher K. Frese
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Box 278, Cambridge, CB2 0RE UK
| | - Natalie Cook
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Box 278, Cambridge, CB2 0RE UK
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Michael A. Jacobetz
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Box 278, Cambridge, CB2 0RE UK
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Donna-Michelle Smith
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Box 278, Cambridge, CB2 0RE UK
| | - David A. Tuveson
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Box 278, Cambridge, CB2 0RE UK
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - John R. Griffiths
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Box 278, Cambridge, CB2 0RE UK
| | - Duncan I. Jodrell
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Box 278, Cambridge, CB2 0RE UK
- Department of Oncology, University of Cambridge, Cambridge, UK
| |
Collapse
|
24
|
Analysis of anticancer drugs: a review. Talanta 2011; 85:2265-89. [PMID: 21962644 DOI: 10.1016/j.talanta.2011.08.034] [Citation(s) in RCA: 343] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 08/15/2011] [Accepted: 08/16/2011] [Indexed: 01/05/2023]
Abstract
In the last decades, the number of patients receiving chemotherapy has considerably increased. Given the toxicity of cytotoxic agents to humans (not only for patients but also for healthcare professionals), the development of reliable analytical methods to analyse these compounds became necessary. From the discovery of new substances to patient administration, all pharmaceutical fields are concerned with the analysis of cytotoxic drugs. In this review, the use of methods to analyse cytotoxic agents in various matrices, such as pharmaceutical formulations and biological and environmental samples, is discussed. Thus, an overview of reported analytical methods for the determination of the most commonly used anticancer drugs is given.
Collapse
|
25
|
Joerger M, Burgers JA, Baas P, Doodeman VD, Smits PHM, Jansen RS, Vainchtein LD, Rosing H, Huitema ADR, Beijnen JH, Schellens JHM. Gene polymorphisms, pharmacokinetics, and hematological toxicity in advanced non-small-cell lung cancer patients receiving cisplatin/gemcitabine. Cancer Chemother Pharmacol 2011; 69:25-33. [PMID: 21590444 DOI: 10.1007/s00280-011-1670-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Accepted: 05/03/2011] [Indexed: 11/25/2022]
Abstract
BACKGROUND This study quantified the impact of drug pathway-associated genetic variants on the pharmacokinetics (PK) of gemcitabine and cisplatin in patients with advanced non-small-cell lung cancer (NSCLC). METHODS Thirty-seven patients with advanced NSCLC were sampled for plasma concentrations of gemcitabine, difluoro-deoxy uridine (dFdU), intracellular gemcitabine triphosphates (dFdCTP), and unbound platinum concentrations after gemcitabine 1,250 mg/m(2) i.v. followed by cisplatin 75 mg/m(2). We analyzed 13 germline single nucleotide polymorphisms and one deletion-glutathione S-transferase (GST) M1-within six drug pathway-associated genes (GSTM1, GSTP1, cytidine deaminase (CDA), solute carrier (SLC) 28A1, SLC28A2, and deoxycytidine kinase). PK models were fitted to the data using nonlinear mixed-effects modeling, and genetic data were tested on drug PK and hematological toxicity. RESULTS Patients carrying the nonsynonymous CDA SNP 79A >C (CDA*2) had a 21% lower gemcitabine clearance as compared to wild-type patients (outcomes and complications.0.0009), but the risk for chemotherapy-associated neutropenia (61% vs. 32%, P = 0.07) and severe neutropenia (17% vs. 5%, P = 0.26) was not significantly higher. Other gene polymorphisms were not associated with drug PK parameters or hematological toxicity. The known functional mutant variant CDA*3 was not found in any of the patients. CONCLUSIONS Although the mutant CDA*2 allele results in an increased exposure to gemcitabine in Caucasian patients, this study gives no definite conclusion on the clinical relevance of this finding. Further studies should look into the relationship between CDA genotypes, plasmatic CDA activity, and clinical outcome in patients receiving gemcitabine-based chemotherapy.
Collapse
Affiliation(s)
- M Joerger
- Department of Pharmacy and Pharmacology, Slotervaart Hospital/The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
O'Sullivan GP, Scully NM, Glennon JD. Polar-Embedded and Polar-Endcapped Stationary Phases for LC. ANAL LETT 2010. [DOI: 10.1080/00032711003653973] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
27
|
Jansen RS, Rosing H, Schellens JHM, Beijnen JH. Simultaneous quantification of 2',2'-difluorodeoxycytidine and 2',2'-difluorodeoxyuridine nucleosides and nucleotides in white blood cells using porous graphitic carbon chromatography coupled with tandem mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2009; 23:3040-3050. [PMID: 19705384 DOI: 10.1002/rcm.4212] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
A novel assay for the simultaneous quantification of the widely used anticancer agent 2',2'-difluorodeoxycytidine (gemcitabine; dFdC), its deaminated metabolite 2',2'-difluorodeoxyuridine (dFdU) and their mono-, di- and triphosphates (dFdCMP, dFdCDP, dFdCTP, dFdUMP, dFdUDP and dFdUTP) in peripheral blood mononuclear cells (PBMCs) is described. Separation of all eight compounds was achieved within 15 min using a porous graphitic carbon column (Hypercarb) with a gradient from 0 to 25 mM ammonium bicarbonate in acetonitrile/water (15:85, v/v). Calibration ranges in PBMC lysate from 4.29 to 429, 29.0 to 2900, 31.4 to 3140 and 36.9 to 3690 nM for dFdC, dFdCMP, dFdCDP and dFdCTP and from 42.1 to 4210, 25.4 to 2540, 43.2 to 4320 and 52.7 to 5270 nM for dFdU, dFdUMP, dFdUDP and dFdUTP, respectively, were validated. Accuracies were within 82.3-119% at the lower limit of quantification (LLOQ) and the precisions were less than 20.0%. At the other tested levels accuracies were within 91.4-114% and precisions less than 14.9%. Mixtures of (13)C,(15)N(2)-labeled dFdC and dFdU nucleotides were synthesized and used as internal standards. Whole blood samples showed extensive ongoing dFdC metabolism when stored at room temperature, but not on ice-water, which made the addition of enzyme inhibitors unnecessary. Stock solutions and samples were stable under all analytically relevant conditions. The method was successfully applied to clinical samples.
Collapse
Affiliation(s)
- Robert S Jansen
- Department of Pharmacy & Pharmacology, Slotervaart Hospital/The Netherlands Cancer Institute, Louwesweg 6, 1066 EC Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
28
|
Koolen SLW, Huitema ADR, Jansen RS, van Voorthuizen T, Beijnen JH, Smit WM, Schellens JHM. Pharmacokinetics of gemcitabine and metabolites in a patient with double-sided nephrectomy: a case report and review of the literature. Oncologist 2009; 14:944-8. [PMID: 19726456 DOI: 10.1634/theoncologist.2009-0111] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
CASE A patient with complete renal failure as a result of urothelial cell carcinoma-related nephrectomy of both kidneys received palliative chemotherapy with carboplatin and gemcitabine. TREATMENT The patient received gemcitabine at 1,000 mg/m(2) followed by carboplatin at 100 mg. Shortly after, he underwent hemodialysis. The pharmacokinetics of gemcitabine and metabolites in plasma and in peripheral blood mononuclear cells were monitored. RESULTS Double-sided nephrectomy and hemodialysis had no influence on gemcitabine pharmacokinetics; however, a high exposure was seen for the main metabolite, difluordeoxyuridine (dFdU) (area under the concentration-time curve, 0-51 hours, 844 microg/ml.hour). During hemodialysis, plasma concentrations of dFdU were reduced by 50%. High concentrations of intracellular phosphorylated metabolites (gemcitabine triphosphate and dFdU triphosphate) were observed: 228 pmol/10(6) cells and 47 pmol/10(6) cells, respectively. The patient tolerated the regimen poorly; adverse events included grade 4 thrombocytopenia. CONCLUSION Hemodialysis effectively reduced plasma concentrations of dFdU. Furthermore, high concentrations of intracellular phosphorylated metabolites may be related to double-sided nephrectomy, resulting in poor tolerability of gemcitabine.
Collapse
Affiliation(s)
- Stijn L W Koolen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute/Slotervaart Hospital, Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
29
|
Quantification of polar drugs in human plasma with liquid chromatography–tandem mass spectrometry. Bioanalysis 2009; 1:187-203. [DOI: 10.4155/bio.09.19] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Liquid chromatography–tandem mass spectrometry (LC–MS/MS) has played an important role in quantitative bioanalytical assays. This review summarizes the recent progress on quantification of polar drugs in plasma with LC–MS/MS. Various types of polar analytes were extracted using protein precipitation or solid-phase extraction and precolumn derivatization was utilized in some cases. The analytes were then separated using different types of chromatographic method, which included reversed-phase chromatography, aqueous normal-phase chromatography, hydrophilic interaction liquid chromatography and ion-pairing chromatography. Stationary phases of mixed mode and porous graphitic carbon materials are gaining acceptance in bioanalytical applications. These technologies can be valuable supplements in the quantification of polar drugs in human plasma with LC–MS/MS. Matrix effects have also been discussed in this review.
Collapse
|