1
|
Overbeek JK, Guchelaar NA, Mohmaed Ali MI, Sark M, Hovenier C, Kievit W, Ligtenberg MJ, Ottevanger PB, Bloemendal HJ, Koolen SL, Mathijssen RH, Boere IA, Huitema AD, Sonke GS, Opdam FL, Heine RT, van Erp NP. Pharmacokinetic boosting of olaparib: Study protocol of a multicentre, open-label, randomised, non-inferiority trial (PROACTIVE-B). Contemp Clin Trials Commun 2025; 45:101477. [PMID: 40248173 PMCID: PMC12005849 DOI: 10.1016/j.conctc.2025.101477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/09/2025] [Accepted: 03/28/2025] [Indexed: 04/19/2025] Open
Abstract
Background Pharmacokinetic (PK) boosting is the intentional use of a drug-drug interaction to enhance systemic drug exposure. PK boosting of the anticancer drug olaparib, a CYP3A-substrate, has the potential to reduce PK variability, side effects and financial burden associated with this drug. After establishing adequate pharmacokinetic exposure with boosting in the PROACTIVE-A study, the PROACTIVE-B study is designed to evaluate non-inferiority for both efficacy and toxicity of the boosted therapy compared to the standard monotherapy of olaparib. Methods The PROACTIVE-B study is a nationwide, multicentre, prospective, randomized, non-inferiority trial. A total of 142 patients (128 patients with BRCA+, high-grade, FIGO III/IV ovarian cancer who receive olaparib as maintenance therapy; 14 patients with other approved indications for olaparib) who start olaparib treatment in line with the drug label will be randomized between the standard monotherapy of olaparib 300 mg twice daily (BID) and the boosted therapy of olaparib 100 mg BID with cobicistat 150 mg BID. The co-primary objectives are tolerability (dose reductions due to toxicity), and efficacy (progression-free survival at 12 months) in the ovarian cancer population. Secondary objectives include health status (EQ-5D-5L), patient satisfaction (Cancer Therapy Satisfaction Questionnaire (CTSQ)), and cost effectiveness using the institute for Medical Technology Assessment (iMTA) Productivity Cost Questionnaire (iPCQ) and iMTA Medical Consumption Questionnaire (iMCQ). Discussion PK boosting of olaparib is a potentially valuable strategy to reduce the olaparib dose and the variability in olaparib exposure with fewer side effects. Moreover, the lower costs related to the boosted therapy contribute to a durable and accessible anticancer treatment for all patients. Trial registration The PROACTIVE study has been published at ClinicalTrials.gov under NCT05078671 on October 14, 2021 and at EudraCT under 2021-004032-28 on August 24, 2021.
Collapse
Affiliation(s)
- Joanneke K. Overbeek
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Niels A.D. Guchelaar
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ma Ida Mohmaed Ali
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Muriëlle Sark
- Patient Advisory Group, Breast Cancer Research Group and Dutch Breast Cancer Association, Amsterdam, the Netherlands
| | | | - Wietske Kievit
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marjolijn J.L. Ligtenberg
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Haiko J. Bloemendal
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Stijn L.W. Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ron H.J. Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ingrid A. Boere
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Alwin D.R. Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Gabe S. Sonke
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Frans L. Opdam
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Rob ter Heine
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nielka P. van Erp
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
2
|
Overbeek JK, van Erp NP, Burger DM, den Broeder AA, Koolen SLW, Huitema ADR, Ter Heine R. Population Pharmacokinetics of Cobicistat and its Effect on the Pharmacokinetics of the Anticancer Drug Olaparib. Clin Pharmacokinet 2025; 64:425-435. [PMID: 39909979 PMCID: PMC11954716 DOI: 10.1007/s40262-025-01480-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2025] [Indexed: 02/07/2025]
Abstract
OBJECTIVES Pharmacokinetic (PK) boosting is the intentional use of strong inhibitors of metabolic enzymes or transporters to boost the systemic exposure of a therapeutic drug. PK boosting is expanding to therapeutic areas outside human immunodeficiency virus (HIV) therapy. Data on the PK of the booster cobicistat and its effect on CYP3A-substrates outside of HIV therapy are lacking. This study aimed to describe the PK of once- and twice-daily cobicistat regimens in healthy volunteers and patients with rheumatoid arthritis, cancer, or HIV infection and to investigate the interplay between cobicistat and the anticancer drug olaparib. METHODS Cobicistat levels from 683 samples from 66 subjects in four clinical trials were included in the analysis. For olaparib, 261 samples from 12 subjects from one trial were included. Population PK analysis was performed by nonlinear mixed-effects modelling. RESULTS Both cobicistat and olaparib PK were adequately described by a well-stirred liver model with one central compartment and Erlang type absorption. Cobicistat PK was similar across patient populations and dosing regimens. Cobicistat increased olaparib prehepatic bioavailability 1.65-fold (RSE 6%) and decreased intrinsic clearance 0.34-fold (RSE 6.5%). A correlation between olaparib PK and cobicistat exposure could not be identified. The interindividual variability in olaparib clearance was lower with cobicistat than without cobicistat. CONCLUSIONS The developed pharmacokinetic models adequately described cobicistat and olaparib plasma concentrations. PK boosting with cobicistat at 150 mg twice daily led to an increase in olaparib bioavailability and decrease in clearance. This effect was not correlated with cobicistat exposure, which may reflect saturation of the boosting effect of cobicistat at this dose. TRIAL REGISTRATION NUMBERS (DATE OF REGISTRATION) NCT02565888 (30-09-2015), NCT00825929 (19-01-2009), Netherlands Trial Register NL7766 (18-12-2018), NCT05078671 (22-09-2021).
Collapse
Affiliation(s)
- Joanneke K Overbeek
- Department of Pharmacy, Radboud University Medical Center, PO Box 9101, 6500HB, Nijmegen, The Netherlands.
| | - Nielka P van Erp
- Department of Pharmacy, Radboud University Medical Center, PO Box 9101, 6500HB, Nijmegen, The Netherlands
| | - David M Burger
- Department of Pharmacy, Radboud University Medical Center, PO Box 9101, 6500HB, Nijmegen, The Netherlands
| | | | - Stijn L W Koolen
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Rob Ter Heine
- Department of Pharmacy, Radboud University Medical Center, PO Box 9101, 6500HB, Nijmegen, The Netherlands
| |
Collapse
|
3
|
Knight W, Margaryan T, Sanai N, Tovmasyan A. A validated LC-MS/MS method for determination of neuro-pharmacokinetic behavior of niraparib in brain tumor patients. J Pharm Biomed Anal 2024; 245:116150. [PMID: 38657366 DOI: 10.1016/j.jpba.2024.116150] [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: 02/12/2024] [Revised: 04/12/2024] [Accepted: 04/14/2024] [Indexed: 04/26/2024]
Abstract
Niraparib is a potent and orally bioavailable inhibitor of poly (ADP-ribose) polymerase (PARP) with high specificity for isoforms 1 and 2. It has been approved by the U.S. Food and Drug Administration for ovarian cancer maintenance therapy and is currently under development for various cancers, including glioblastoma. To assess central nervous system (CNS) penetration of niraparib in glioblastoma patients, a novel bioanalytical method was developed to measure total and unbound niraparib levels in human brain tumor tissue and cerebrospinal fluid (CSF). The method was validated using plasma as a surrogate matrix over the concentration range of 1-10,000 nM on an LC-MS/MS system. The MS/MS detection was conducted in positive electrospray ionization mode, while chromatography was performed using a Kinetex™ PS C18 column with a total 3.5-minute gradient elution run time. The maximum coefficient of variation for both intra- and inter-day precision was 10.6%, with accuracy ranging from 92.8% - 118.5% across all matrices. Niraparib was stable in human brain homogenate for at least 6 hours at room temperature (RT) and 32 days at -20°C, as well as in stock and working solutions for at least 21 hours (RT) and 278 days (4°C). Equilibrium dialysis experiments revealed the fractions unbound of 0.05 and 0.16 for niraparib in human brain and plasma, respectively. The validated method is currently employed to assess niraparib levels in human glioblastoma tissue, CSF, and plasma in an ongoing trial on newly diagnosed glioblastoma and recurrent IDH1/2(+) ATRX mutant glioma patients (NCT05076513). Initial results of calculated total (Kp) and unbound (Kp,uu) tumor-to-plasma partition coefficients indicate significant brain penetration ability of niraparib in glioblastoma patients.
Collapse
Affiliation(s)
- William Knight
- Ivy Brain Tumor Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Tigran Margaryan
- Ivy Brain Tumor Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Nader Sanai
- Ivy Brain Tumor Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Artak Tovmasyan
- Ivy Brain Tumor Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA.
| |
Collapse
|
4
|
Tan Z, Völler S, Yin A, Rieborn A, Gelderblom AJ, van der Hulle T, Knibbe CAJ, Moes DJAR. Population Pharmacokinetics of Cabozantinib in Metastatic Renal Cell Carcinoma Patients: Towards Drug Expenses Saving Regimens. Clin Pharmacokinet 2024; 63:857-869. [PMID: 38874883 PMCID: PMC11222182 DOI: 10.1007/s40262-024-01379-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: 04/18/2024] [Indexed: 06/15/2024]
Abstract
INTRODUCTION Cabozantinib is one of the preferred treatment options in the latest metastatic renal cell carcinoma (mRCC) guidelines. Cabozantinib is also associated with high drug expenses irrespective of the used dose, because a flat-prizing model has been implemented. In addition, concomitant intake with a high-fat meal increases its bioavailability on average by 57%. Combined with the long terminal half-life of cabozantinib (99 h), this creates possibilities to extend the dosing interval to reduce drug expenses whilst maintaining equivalent exposure. OBJECTIVES The primary objective was to evaluate the population pharmacokinetic (POPPK) model of cabozantinib developed for its registration using real-world patients' therapeutic drug monitoring (TDM) data. The secondary objective was to design, simulate, and evaluate alternative dose regimens with the aim to reduce drug expenses whilst maintaining comparable exposure. METHODS Retrospective TDM data from mRCC patients treated with cabozantinib were obtained. The data were evaluated using the published Food and Drug Administration (FDA) cabozantinib POPPK model, a two-compartment disposition model with a dual (fast and slow) lagged first-order absorption process derived from FDA registration documents, as a basis. Subsequently, simulations of alternative drug expenses saving regimens were evaluated. RESULTS Twenty-seven mRCC patients with 75 pharmacokinetic observations were included. Patients were treated for a median of 75 days with a median dose of 40 mg. Model evaluation results showed that the cabozantinib TDM concentrations were adequately predicted by the published FDA cabozantinib POPPK model, except for a slightly higher clearance (CL) of 3.11 L/h compared to the reported value (2.23 L/h). The simulation study indicated that an alternative dose regimen that consists of taking 60 mg of cabozantinib for 2 days and then skipping 1 day results in comparable average exposure when compared with a 40 mg daily dose, both without food interaction, while saving 33.3% of the total drug expenses per month. The food effect of a high-fat meal was also taken into account when simulating other alternative dose regimens; 40 mg every 72 h combined with a high-fat meal resulted in comparable exposure when compared with a 20 mg daily dose fasted, while saving 66.7% in drug expenses. CONCLUSIONS In this study, the optimized cabozantinib POPPK model resulted in adequate prediction of real-world cabozantinib pharmacokinetic data. Alternative dosing regimens with and without using known food interactions were proposed that resulted in potential strategies to significantly reduce cabozantinib drug expenses.
Collapse
Affiliation(s)
- Zhiyuan Tan
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Swantje Völler
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Anyue Yin
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Amy Rieborn
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - A J Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom van der Hulle
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Catherijne A J Knibbe
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Department of Clinical Pharmacy, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Dirk Jan A R Moes
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
5
|
Cetinkaya A, Kaya SI, Budak F, Ozkan SA. Current Analytical Methods for the Sensitive Assay of New-Generation Ovarian Cancer Drugs in Pharmaceutical and Biological Samples. Crit Rev Anal Chem 2024:1-17. [PMID: 38630637 DOI: 10.1080/10408347.2024.2339962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Ovarian cancer, which affects the female reproductive organs, is one of the most common types of cancer. Since this type of cancer has a high mortality rate from gynaecological cancers, the scientific community shows great interest in studies on its treatment. Chemotherapy, radiotherapy, and surgical treatment methods are used in its treatment. In the absence of targeted treatments in these treatment methods, side effects occur in patients, and patients show resistance to the drug. In addition, the underlying causes of ovarian cancer are still not fully known. The scientific world thinks that genetic factors, environmental conditions, and consumed foods may cause this cancer. The most important factor in the treatment of ovarian cancer is early diagnosis. Therefore, the drugs used in the treatment of ovarian cancer are platinum-based anticancer drugs. In addition to these drugs, the most preferred treatment method recently is targeted treatment approaches using poly(adenosine diphosphate ribose) polymerase (PARP) inhibitors. In this review, studies on the sensitive analysis of the treatment methods of these new-generation drugs used in the treatment of ovarian cancer have been comprehensively examined. In addition, the basic features, structural aspects, and biological data of analytical methods used in treatments with new-generation drugs are explained. Analytical studies carried out in the literature in recent years aim to show future developments in how these new-generation drugs are used today and to guide future studies by comprehensively examining and explaining the structure-activity relationship, mechanism of action, toxicity, and pharmacokinetic studies. Finally, in this study, the methods used in the analysis of drugs used in the treatment of ovarian cancer and the studies conducted between 2015 and 2023 were discussed in detail.
Collapse
Affiliation(s)
- Ahmet Cetinkaya
- Faculty of Pharmacy, Department of Analytical Chemistry, Ankara University, Ankara, Turkey
| | - S Irem Kaya
- Gulhane Faculty of Pharmacy, Department of Analytical Chemistry, University of Health Sciences, Ankara, Turkey
| | - Fatma Budak
- Faculty of Pharmacy, Department of Analytical Chemistry, Ankara University, Ankara, Turkey
- Graduate School of Health Sciences, Ankara University, Ankara, Turkey
| | - Sibel A Ozkan
- Faculty of Pharmacy, Department of Analytical Chemistry, Ankara University, Ankara, Turkey
| |
Collapse
|
6
|
Yasu T, Nishijima R, Ikuta R, Shirota M, Iwase H. Development of a simple high-performance liquid chromatography-ultraviolet detection method for olaparib in patients with ovarian cancer. Drug Discov Ther 2024; 17:428-433. [PMID: 38044120 DOI: 10.5582/ddt.2023.01074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Olaparib is a small-molecule inhibitor of poly(ADP)-ribose polymerase (PARP) used as maintenance therapy for recurrent ovarian cancer and newly diagnosed advanced ovarian cancer after initial chemotherapy. An exposure-toxicity correlation has been reported between the probability of anemia, a common adverse event associated with olaparib, and the steady-state minimum plasma concentration (Cmin) as well as the predicted maximum plasma concentration (Cmax). On the other hand, olaparib exhibits high interpatient variability with regard to the area under the concentration-time curve, Cmax, and Cmin. Therefore, we developed a simple and sensitive assay based on high-performance liquid chromatography with ultraviolet light (HPLC-UV) for the therapeutic drug monitoring of olaparib. The analysis was performed on an octadecylsilyl column with a mobile phase consisting of 0.5% KH2PO4 (pH 4.5) and acetonitrile (71:29, v/v), at a flow rate of 0.8 mL/min. Olaparib and an internal standard (imatinib) were well separated from the co-extracted material, with retention times of 13.6 and 11.5 min, respectively. The calibration curve for olaparib showed linearity over the concentration range of 0.10-10.0 μg/mL (r2 = 0.9998). The intra- and inter- day validation coefficients ranged from 1.79 to 4.13% and 1.37 to 3.55%, respectively. Measurement accuracy ranged from - 6.07 to 3.26%, with a recovery rate of more than 91.06%. The developed method was then applied to evaluate the plasma olaparib concentrations in patients with ovarian cancer. Our findings demonstrate that HPLC-UV is an economical, simple, and sensitive method for clinical application and holds promise for the effective drug monitoring of olaparib during ovarian cancer treatment.
Collapse
Affiliation(s)
- Takeo Yasu
- Department of Medicinal Therapy Research, Pharmaceutical Education and Research Center, Meiji Pharmaceutical University, Tokyo, Japan
- Department of Pharmacy, Tokyo Metropolitan Bokutoh Hospital, Tokyo, Japan
- Bokutoh Hospital-Meiji Pharmaceutical University Joint Research Center, Tokyo, Japan
| | - Ryosuke Nishijima
- Department of Medicinal Therapy Research, Pharmaceutical Education and Research Center, Meiji Pharmaceutical University, Tokyo, Japan
| | - Risa Ikuta
- Department of Clinical Laboratory, Tokyo Metropolitan Bokutoh Hospital, Tokyo, Japan
- Bokutoh Hospital-Meiji Pharmaceutical University Joint Research Center, Tokyo, Japan
| | - Mikio Shirota
- Department of Pharmacy, Tokyo Metropolitan Bokutoh Hospital, Tokyo, Japan
- Bokutoh Hospital-Meiji Pharmaceutical University Joint Research Center, Tokyo, Japan
| | - Haruko Iwase
- Department of Obstetrics and Gynecology, Tokyo Metropolitan Bokutoh Hospital, Tokyo, Japan
- Bokutoh Hospital-Meiji Pharmaceutical University Joint Research Center, Tokyo, Japan
| |
Collapse
|
7
|
Giraud EL, Te Brake LMH, van den Hombergh ECA, Desar IME, Kweekel DM, van Erp NP. Results of the first international quality control programme for oral targeted oncolytics. Br J Clin Pharmacol 2024; 90:336-343. [PMID: 37776845 DOI: 10.1111/bcp.15918] [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: 06/22/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/02/2023] Open
Abstract
AIMS With the rising number of oral targeted oncolytics and growing awareness of the benefits of therapeutic drug monitoring (TDM) within the field of oncology, it is expected that the requests for quantifying concentrations of these drugs will increase. It is important to (cross-)validate available assays and ensure its quality, as results may lead to altered dosing recommendations. Therefore, we aimed to evaluate the performance of laboratories measuring concentrations of targeted oral oncolytics in a one-time international quality control (QC) programme. METHODS Participating laboratories received a set of plasma samples containing low, medium and high concentrations of imatinib, sunitinib, desethylsunitinib, pazopanib, cabozantinib, olaparib, enzalutamide, desmethylenzalutamide and abiraterone, with the request to report their results back within five weeks after shipment. Accuracy was defined acceptable if measurements where within 85%-115% from the weighed-in reference concentrations. Besides descriptive statistics, an exploratory ANOVA was performed. RESULTS Seventeen laboratories from six countries reported 243 results. Overall, 80.7% of all measurements were within the predefined range of acceptable accuracy. Laboratories performed best in quantifying imatinib and poorest in quantifying desethylsunitinib (median absolute inaccuracy respectively 4.0% (interquartile range (IQR) 1.8%-6.5%) and 15.5% (IQR 8.8%-34.9%)). The poorest performance of desethylsunitinib might be caused by using the stable-isotope-labelled sunitinib instead of desethylsunitinib as an internal standard, or due to the light-induced cis(Z)/trans(E) isomerization of (desethyl)sunitinib. Overall, drug substance and performing laboratory seemed to influence the absolute inaccuracy (F = 16.4; p < 0.001 and F = 35.5; p < 0.001, respectively). CONCLUSION Considering this is the first evaluation of an international QC programme for oral targeted oncolytics, an impressive high percentage of measurements were within the predefined range of accuracy. Cross-validation of assays that are used for dose optimization of oncolytics will secure the performance and will protect patients from incorrect advices.
Collapse
Affiliation(s)
- Eline L Giraud
- Department of Pharmacy, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Lindsey M H Te Brake
- Department of Pharmacy, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Erik C A van den Hombergh
- Department of Pharmacy, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Ingrid M E Desar
- Department of Medical Oncology, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Dina M Kweekel
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, The Netherlands
- Drug Analysis and Toxicology division (KKGT) of the Dutch Foundation for Quality Assessment in Medical Laboratories (SKML), Utrecht, The Netherlands
| | - Nielka P van Erp
- Department of Pharmacy, Research Institute for Medical Innovation, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
8
|
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
|
9
|
Abdelgalil AA, Alkahtani HM. Regorafenib: A comprehensive drug profile. PROFILES OF DRUG SUBSTANCES, EXCIPIENTS, AND RELATED METHODOLOGY 2023; 49:41-79. [PMID: 38423709 DOI: 10.1016/bs.podrm.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Regorafenib is a small molecule tyrosine kinase inhibitor administered orally drug, act by inhibiting the activity of the VEGF receptors. It is used for the treatment of patients with metastatic colorectal cancer (CRC), advanced gastrointestinal stromal tumors, and hepatocellular carcinoma. This comprehensive profile on regorafenib includes an original data as well as data collected from the literature on Profiles of Methods of Drug Synthesis, different Physical Drug Profiles, Drug Analytical methods and Pharmacological profile (ADME). This chapter is divided into five main sections: General Description of the drug, Physical Characteristics, Methods of Preparation, Methods of Analysis, Pharmacology and List of References. These main sections are further divided to many sub-titles to cover most aspect of the drug in the light of the available literature. Among these sub-titles are the formulae, Elemental Analysis, physical characteristics which include constant of ionization, solubility, X-ray powder diffraction pattern, TGA, thermal conduct and spectroscopic and stability. Additionally, analytical techniques including Electrochemical, Spectrophotometric and chromatographic methods, ADME profiles and pharmacological effects were also discussed. Furthermore, methods and schemes are outlined for the preparation of the drug substance.
Collapse
Affiliation(s)
- Ahmed A Abdelgalil
- Central Laboratory, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia.
| | - Hamad M Alkahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
10
|
Overbeek JK, Guchelaar NAD, Mohmaed Ali MI, Ottevanger PB, Bloemendal HJ, Koolen SLW, Mathijssen RHJ, Boere IA, Hamberg P, Huitema ADR, Sonke GS, Opdam FL, Ter Heine R, van Erp NP. Pharmacokinetic boosting of olaparib: A randomised, cross-over study (PROACTIVE-study). Eur J Cancer 2023; 194:113346. [PMID: 37806255 DOI: 10.1016/j.ejca.2023.113346] [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: 08/03/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Pharmacokinetic (PK) boosting is the intentional use of a drug-drug interaction to enhance systemic drug exposure. PK boosting of olaparib, a CYP3A-substrate, has the potential to reduce PK variability and financial burden. The aim of this study was to investigate equivalence of a boosted, reduced dose of olaparib compared to the non-boosted standard dose. METHODS This cross-over, multicentre trial compared olaparib 300 mg twice daily (BID) with olaparib 100 mg BID boosted with the strong CYP3A-inhibitor cobicistat 150 mg BID. Patients were randomised to the standard therapy followed by the boosted therapy, or vice versa. After seven days of each therapy, dense PK sampling was performed for noncompartmental PK analysis. Equivalence was defined as a 90% Confidence Interval (CI) of the geometric mean ratio (GMR) of the boosted versus standard therapy area under the plasma concentration-time curve (AUC0-12 h) within no-effect boundaries. These boundaries were set at 0.57-1.25, based on previous pharmacokinetic studies with olaparib capsules and tablets. RESULTS Of 15 included patients, 12 were eligible for PK analysis. The GMR of the AUC0-12 h was 1.45 (90% CI 1.27-1.65). No grade ≥3 adverse events were reported during the study. CONCLUSIONS Boosting a 100 mg BID olaparib dose with cobicistat increases olaparib exposure 1.45-fold, compared to the standard dose of 300 mg BID. Equivalence of the boosted olaparib was thus not established. Boosting remains a promising strategy to reduce the olaparib dose as cobicistat increases olaparib exposure Adequate tolerability of the boosted therapy with higher exposure should be established.
Collapse
Affiliation(s)
- Joanneke K Overbeek
- Department of Pharmacy, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Gelderland, the Netherlands.
| | - Niels A D Guchelaar
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, South Holland, the Netherlands
| | - Ma Ida Mohmaed Ali
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, North Holland, the Netherlands
| | - Petronella B Ottevanger
- Department of Medical Oncology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Gelderland, the Netherlands
| | - Haiko J Bloemendal
- Department of Medical Oncology, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Gelderland, the Netherlands
| | - Stijn L W Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, South Holland, the Netherlands; Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, South Holland, the Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, South Holland, the Netherlands
| | - Ingrid A Boere
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, South Holland, the Netherlands
| | - Paul Hamberg
- Department of Internal Medicine, Franciscus Gasthuis and Vlietland, Rotterdam, South Holland, the Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, North Holland, the Netherlands; Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, Utrecht, the Netherlands; Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, Utrecht, the Netherlands
| | - Gabe S Sonke
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, North Holland, the Netherlands
| | - Frans L Opdam
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, North Holland, the Netherlands
| | - Rob Ter Heine
- Department of Pharmacy, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Gelderland, the Netherlands
| | - Nielka P van Erp
- Department of Pharmacy, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, Gelderland, the Netherlands
| |
Collapse
|
11
|
Myszkiewicz MF, Puzanov I, Goey AKL. Development and validation of an LC-MS/MS method to measure the BRAF inhibitors dabrafenib and encorafenib quantitatively and four major metabolites semi-quantitatively in human plasma. J Pharm Biomed Anal 2023; 234:115594. [PMID: 37478552 PMCID: PMC10528671 DOI: 10.1016/j.jpba.2023.115594] [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: 04/26/2023] [Revised: 06/28/2023] [Accepted: 07/16/2023] [Indexed: 07/23/2023]
Abstract
This article describes the development and validation of a liquid-chromatography coupled with tandem mass spectrometry (LC-MS/MS) assay for the simultaneous quantitation of the BRAF inhibitors dabrafenib and encorafenib, and semi-quantitation of their major metabolites (i.e., carboxy-dabrafenib, desmethyl-dabrafenib, hydroxy-dabrafenib, M42.5A) in human plasma. Analytes were extracted from human plasma by protein precipitation, followed by reversed phase high-performance liquid chromatography. Analyte detection was performed using tandem mass spectrometry with heated electrospray ionization operating in positive ion mode. The assay was validated in accordance with the current U.S. Food and Drug Administration Guidance on Bioanalytical Method Validation. Results showed that measurements were both accurate (94.6-112.0 %) and precise (within-run: 1.9-3.4 %; between-run: 1.7-12.0 %) spanning a concentration range of 5 to 2000 ng/mL for dabrafenib and 10 to 4000 ng/mL for encorafenib. Recoveries for these analytes were consistent with mean values ranging from 85.6 % to 90.9 %. The mean internal standard-normalized matrix factors for each drug ranged between 0.87 and 0.98 and were found to be precise (% RSD <6.4 %). Dabrafenib and encorafenib were stable in the final extract and in human plasma held under various storage conditions. The metabolites also passed the validation criteria for precision and selectivity. Finally, the clinical applicability of the assay was confirmed by (semi-)quantitation of all six analytes in plasma samples from cancer patients receiving standard-of-care treatment with dabrafenib and encorafenib. Reproducibility of the measured analyte concentrations in study samples was confirmed successfully by incurred sample reanalysis. In conclusion, this sensitive LC-MS/MS assay has been validated successfully and is suitable for therapeutic drug monitoring of dabrafenib and encorafenib and clinical pharmacokinetic studies with these BRAF inhibitors.
Collapse
Affiliation(s)
- Melody F Myszkiewicz
- Bioanalytics, Metabolomics, and Pharmacokinetics Shared Resource, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Andrew K L Goey
- Bioanalytics, Metabolomics, and Pharmacokinetics Shared Resource, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA; Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| |
Collapse
|
12
|
Al Shirity ZN, Westra N, Hateren KV, Munnink THO, Kosterink JGW, Mian P, Hooge MNLD, Touw DJ, Gareb B. Validation of an LC-MS/MS assay for rapid and simultaneous quantification of 21 kinase inhibitors in human plasma and serum for therapeutic drug monitoring. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1229:123872. [PMID: 37716342 DOI: 10.1016/j.jchromb.2023.123872] [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: 06/19/2023] [Revised: 08/30/2023] [Accepted: 09/02/2023] [Indexed: 09/18/2023]
Abstract
Kinase inhibitors have revolutionized cancer treatment in the past 25 years and currently form the cornerstone of many treatments. Due to the increasing evidence for therapeutic drug monitoring (TDM) of kinase inhibitors, the need is growing for new assays to rapidly evaluate kinase inhibitor plasma concentrations. In this study, we developed an LC-MS/MS assay for the rapid and simultaneous quantification of 21 kinase inhibitors. First, a literature search was conducted to ensure that the linear ranges of the analytes were in line with the reported therapeutic windows and/or TDM reference values. Subsequently, the assay was validated according to FDA and EMA guidelines for linearity, selectivity, carry-over, accuracy, precision, dilution integrity, matrix effect, recovery, and stability. The assay was fast, with a short run-time of 2 min per sample. Sample pre-treatment consisted of protein precipitation with methanol enriched with stable isotope-labeled internal standards (SIL-IS), and the mixture was vortexed and centrifuged before sample injection. Separation was achieved using a C18 column (3 μm,50 × 2.1 mm) with a gradient of two mobile phases (ammonium formate buffer pH 3.5 and acetonitrile). Analyte detection was conducted in positive ionization mode using selected reaction monitoring. The assay was accurate and precise in plasma as well as in serum. Extraction recovery ranged between 95.0% and 106.0%, and the matrix effect was 95.7%-105.2%. The stability of the analytes varied at room temperature and in refrigerated conditions. However, all drugs were found to be stable for 7 days in the autosampler. The clinical applicability of the analytical method (486 analyzed samples between 1 July 2022-1 July 2023) as well as external quality control testing results were evaluated. Taken together, the results demonstrate that the analytical method was validated and applicable for routine analyses in clinical practice.
Collapse
Affiliation(s)
- Zaid N Al Shirity
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands.
| | - Niels Westra
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Kai van Hateren
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Thijs H Oude Munnink
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Jos G W Kosterink
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands; Department of PharmacoTherapy, -Epidemiology and -Economics, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands
| | - Paola Mian
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Marjolijn N Lub-de Hooge
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands; Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Daan J Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands; Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands
| | - Bahez Gareb
- Department of Clinical Pharmacy and Pharmacology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
13
|
Chen F, Chen W, Wang Z, Peng Y, Wang B, Pan B, Guo W. Development and clinical application of a liquid chromatography-tandem mass spectrometry-based assay to quantify eight tyrosine kinase inhibitors in human plasma. J Mass Spectrom Adv Clin Lab 2023; 29:2-8. [PMID: 37234251 PMCID: PMC10205537 DOI: 10.1016/j.jmsacl.2023.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 04/13/2023] [Accepted: 05/01/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction Tyrosine kinase inhibitors (TKIs) are widely used in tumor treatment. The detection of these medicines by liquid chromatography-tandem mass spectrometry (LC-MS/MS) can avoid the interference of structurally similar compounds. Objectives This study aimed to develop and validate a new LC-MS/MS assay for the quantification of eight tyrosine kinase inhibitors in human plasma and to preliminarily evaluate the clinical utility of the therapeutic drug monitoring method. Methods Plasma samples were prepared by simple protein precipitation and separated using an ultra-high-performance reversed phase column. Detection was achieved using a triple quadrupole mass spectrometer in the positive ionization mode. The assay was validated against standard guidelines. We reviewed and analyzed the results of 268 plasma samples obtained from patients administered imatinib and other TKIs collected from January 2020 to November 2021 at Zhongshan Hospital. The analytes were separated and quantified within 3.5 min. Results The newly developed method demonstrated linearity for the detected drug concentration in the range of 20 to 2000 ng/ml for gefitinib (r2 = 0.991) and crizotinib (r2 = 0.992), 50 to 5000 ng/ml for nilotinib (r2 = 0.991) and imatinib (r2 = 0.995), 1500-150,000 ng/ml for vemurafenib (r2 = 0.998), 1000-100,000 ng/ml for pazopanib (r2 = 0.993), 0.5-100 ng/ml for axitinib (r2 = 0.992) and 5-500 ng/ml for sunitinib (r2 = 0.991) and N-desethyl sunitinib (r2 = 0.998). The lower limit of quantification (LLOQ) was 20 ng/ml for gefitinib and crizotinib, 50 ng/ml for nilotinib and imatinib, 1500 ng/ml for vemurafenib, 1000 ng/ml for pazopanib, 0.5, and 5 ng/ml for sunitinib and N-desethyl sunitinib, respectively. Specificity, precision, accuracy, and stability were tested, and met the requirements of the guidelines. At the same dose, there was no significant difference in plasma drug concentration between the original imatinib medicine and the generic medicine after patent expiration. Conclusion We developed a sensitive and reliable method for the quantification of eight TKIs.
Collapse
|
14
|
Canil G, Orleni M, Posocco B, Gagno S, Bignucolo A, Montico M, Roncato R, Corsetti S, Bartoletti M, Toffoli G. LC-MS/MS Method for the Quantification of PARP Inhibitors Olaparib, Rucaparib and Niraparib in Human Plasma and Dried Blood Spot: Development, Validation and Clinical Validation for Therapeutic Drug Monitoring. Pharmaceutics 2023; 15:pharmaceutics15051524. [PMID: 37242766 DOI: 10.3390/pharmaceutics15051524] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Poly (ADP-ribose) polymerase inhibitors (PARPis) are becoming increasingly meaningful in oncology, and their therapeutic drug monitoring (TDM) might be beneficial for patients. Several bioanalytical methods have been reported for PARPis quantification in human plasma, but advantages might be obtained using dried blood spot (DBS) as a sampling technique. Our aim was to develop and validate a liquid chromatography-tandem mass spectrometric (LC-MS/MS) method for olaparib, rucaparib, and niraparib quantification in both human plasma and DBS matrices. Additionally, we aimed to assess the correlation between the drug concentrations measured in these two matrices. DBS from patients was obtained using Hemaxis DB10 for volumetric sampling. Analytes were separated on a Cortecs-T3 column and detected with electrospray ionization (ESI)-MS in positive ionization mode. Validation was performed according to the latest regulatory guidelines, in the range (ng/mL) 140-7000 for olaparib, 100-5000 for rucaparib, and 60-3000 for niraparib, within the hematocrit (Hct) range 29-45%. The Passing-Bablok and Bland-Altman statistical analyses revealed a strong correlation between plasma and DBS for olaparib and niraparib. However, due to the limited amount of data, it was challenging to establish a robust regression analysis for rucaparib. To ensure a more reliable assessment, additional samples are required. The DBS-to-plasma ratio was used as a conversion factor (CF) without considering any patient-related hematological parameters. These results provide a solid basis for the feasibility of PARPis TDM using both plasma and DBS matrices.
Collapse
Affiliation(s)
- Giovanni Canil
- Experimental and Clinical Pharmacology Unit, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
| | - Marco Orleni
- Experimental and Clinical Pharmacology Unit, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
- Doctoral School in Pharmacological Sciences, University of Padua, 35131 Padova, Italy
| | - Bianca Posocco
- Experimental and Clinical Pharmacology Unit, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
| | - Sara Gagno
- Experimental and Clinical Pharmacology Unit, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
| | - Alessia Bignucolo
- Experimental and Clinical Pharmacology Unit, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
| | - Marcella Montico
- Clinical Trial Office, CRO Aviano, National Cancer Institute, IRCSS, 33081 Aviano, Italy
| | - Rossana Roncato
- Experimental and Clinical Pharmacology Unit, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
| | - Serena Corsetti
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
| | - Michele Bartoletti
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
| |
Collapse
|
15
|
Nanomaterial-based electrochemical sensing platform for the determination of Olaparib. Electrochim Acta 2023. [DOI: 10.1016/j.electacta.2023.142198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
|
16
|
Bioanalytical Methods for Poly(ADP-Ribose) Polymerase Inhibitor Quantification: A Review for Therapeutic Drug Monitoring. Ther Drug Monit 2023; 45:306-317. [PMID: 36728223 PMCID: PMC10168115 DOI: 10.1097/ftd.0000000000001081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/29/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND Therapeutic drug monitoring (TDM) of poly(ADP-ribose) polymerase inhibitors (PARPis) is an exploratory practice aimed at improving the quality of treatment through personalized therapy. Currently, there are 4 European Medicines Agency-approved and US Food and Drug Administration-approved PARPis available clinically whose quantification requires validated analytical methods: olaparib, niraparib, rucaparib, and talazoparib. The purpose of this literature review was to highlight the pharmacological features of PARPis that could support their TDM practice and provide a detailed discussion of the available liquid chromatography coupled with tandem mass spectrometry methods for their quantification. METHODS Using several Medical Subject Heading terms, the literature was searched using several research engines, including SciFinder, Web of Science, Google Scholar, and PubMed, to find articles published before August 2022. RESULTS Exposure-efficacy and exposure-safety profiles, drug-drug interactions, and hepatic/renal impairment of PARPis provide the potential rationale to monitor their concentrations through TDM. Several bioanalytical methods for their quantification have been reported and compared, and a great deal of heterogeneity has been found among methods, regarding both their analytical and regulatory aspects. CONCLUSIONS In addition to reducing toxicity and increasing the efficacy of PARPis therapy, TDM could be beneficial to thoroughly investigate the exposure-response relationships of PARPis and to establish pharmacokinetic thresholds for clinical decisions. Based on the comparison of published bioanalytical methods, their transferability and validation both play a key role in method selection. For future use in clinical TDM, we anticipate that bioanalytical methods should address every analytical need more thoroughly and should be validated with standardized guidelines.
Collapse
|
17
|
Nieto López de la Nieta L, Guzmán Bernardo FJ, Castañeda Peñalvo G, Rodríguez Flores J. Solid phase extraction prior to non-aqueous capillary electrophoresis with ultraviolet detection as a valuable strategy for therapeutic drug monitoring of cabozantinib. Microchem J 2022. [DOI: 10.1016/j.microc.2022.107830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
18
|
Jiang W, Zhao T, Zhen X, Jin C, Li H, Ha J. Rapid Determination of 9 Tyrosine Kinase Inhibitors for the Treatment of Hepatocellular Carcinoma in Human Plasma by QuEChERS-UPLC-MS/MS. Front Pharmacol 2022; 13:920436. [PMID: 35800447 PMCID: PMC9253689 DOI: 10.3389/fphar.2022.920436] [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: 04/14/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
A reliable and rapid method employing QuEChERS (Quick, Easy, Cheap, Effective, Rugged, and Safe) pretreatment coupled with ultra-performance liquid chromatography–tandem mass spectrometry (UPLC–MS/MS) was successfully developed and validated for the analysis of nine tyrosine kinase inhibitors (TKIs) in human plasma. Biological samples were extracted with acetonitrile and salted out with 350 mg of anhydrous magnesium sulfate (MgSO4), followed by purification with 40 mg of ethyl enediamine-N-propylsilane (PSA) adsorbents. All analytes and internal standards (IS) were separated on the Hypersil GOLD VANQUISH C18 (2.1 mm × 100 mm, 1.9 μM) column using the mobile phases composed of acetonitrile (phase A) and 0.1% formic acid in water (phase B) for 8.0 min. Detection was performed by selection reaction monitoring (SRM) in the positive ion electrospray mode. Lenvatinib, sorafenib, cabozantinib, apatinib, gefitinib, regorafenib, and anlotinib rendered good linearity over the range of 0.1–10 ng/ml, and 1–100 ng/ml for tivantinib and galunisertib. All linear correlation coefficients for all standard curves were ≥ 0.9966. The limits of detection (LOD) and the limits of quantitation (LOQ) ranged from 0.003 to 0.11 ng/ml and 0.01–0.37 ng/ml, respectively. The method was deemed satisfactory with an accuracy of -7.34–6.64%, selectivity, matrix effect (ME) of 90.48–107.77%, recovery, and stability. The proposed method is simple, efficient, reliable, and applicable for the detection of TKIs in human plasma samples as well as for providing a reference for the clinical adjustment of drug administration regimen by monitoring the drug concentrations in the plasma of patients.
Collapse
Affiliation(s)
- Wen Jiang
- College of Chemistry and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Tingting Zhao
- College of Pharmacy, Hebei Medical University, Shijiazhuang, China
| | - Xiaolan Zhen
- Hebei Institute of Drug and Medical Device Inspection, Shijiazhuang, China
| | - Chengcheng Jin
- College of Chemistry and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Hui Li
- Hebei Institute of Drug and Medical Device Inspection, Shijiazhuang, China
- *Correspondence: Hui Li, ; Jing Ha,
| | - Jing Ha
- College of Chemistry and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- *Correspondence: Hui Li, ; Jing Ha,
| |
Collapse
|
19
|
Comparison of a newly developed high performance liquid chromatography method with diode array detection to a liquid chromatography tandem mass spectrometry method for the quantification of cabozantinib, dabrafenib, nilotinib and osimertinib in human serum - Application to therapeutic drug monitoring. Clin Biochem 2022; 105-106:35-43. [PMID: 35483452 DOI: 10.1016/j.clinbiochem.2022.04.011] [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] [Revised: 04/13/2022] [Accepted: 04/23/2022] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Liquid chromatography tandem mass spectrometry (LC-MS/MS) is a highly selective and sensitive method for the quantification of kinase inhibitors, yet not widely available in clinical routine for therapeutic drug monitoring (TDM). To provide a more accessible alternative, a high-performance liquid chromatography method with ultraviolet/diode array detection (HPLC-UV/DAD) to quantify cabozantinib, dabrafenib, nilotinib and osimertinib, was developed and validated. Results were compared to LC-MS/MS. METHOD After liquid-liquid-extraction and reconstitution of the residue in 20 mM potassium dihydrogen phosphate (KH2PO4) (pH4.6), acetonitrile and methanol (50:25:25,v/v/v), chromatographic separation was achieved in 20.0 min using a Luna® C18(2)-HST column (100 × 2 mm, 2.5 μm), protected by a C18 guard column (4 × 2 mm) (column temperature: 30 °C, autosampler: 10 °C). Mobile phase A and B consisted of 20 mM KH2PO4 (pH4.9) and acetonitrile (9:1,v/v) and acetonitrile:20 mM KH2PO4 (pH4.9) (7:3,v/v), respectively. Gradient elution was performed at 200 µL/min. Analytes were quantified at 250, 280 and 330 nm, using sorafenib as internal standard. RESULTS Calibration curves were linear (35-2,000 ng/mL). Method validation assays met requirements by U.S. Food and Drug Administration and European Medicines Agency. Compared to the more sensitive and specific LC-MS/MS, HPLC-UV/DAD showed a good correlation and a strong positive association (Kendall's tau 0.811¬-0.963, p < 0.05). Bland-Altman-plots revealed 100% (cabozantinib), 98.6% (dabrafenib), 98.6% (nilotinib) and 96.2% (osimertinib) of relative differences inside the limits of agreement. Regulatory agency criteria for sample reanalysis and cross validation were met (±20%-criterion:100% (cabozantinib), 94.3% (dabrafenib), 92% (nilotinib) and 84.6% (osimertinib). CONCLUSION The developed HPLC-UV/DAD method is "fit-for-TDM" in clinical routine and serves as a genuine alternative to LC-MS/MS.
Collapse
|
20
|
Jones R, Holleran J, Parise RA, Rudek MA, Chan J, Wen Y, Gobburu J, Lewis LD, Beumer JH. Quantitation of Cabozantinib in Human Plasma by LC-MS/MS. J Chromatogr Sci 2022; 60:274-279. [PMID: 34240176 PMCID: PMC8946687 DOI: 10.1093/chromsci/bmab090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Indexed: 12/14/2022]
Abstract
To support a phase III randomized trial of the multi-targeted tyrosine kinase inhibitor cabozantinib in neuroendocrine tumors, we developed a high-performance liquid chromatography mass spectrometry method to quantitate cabozantinib in 50 μL of human plasma. After acetonitrile protein precipitation, chromatographic separation was achieved with a Phenomenex synergy polar reverse phase (4 μm, 2 × 50 mm) column and a gradient of 0.1% formic acid in acetonitrile and 0.1% formic acid in water over a 5-min run time. Detection was performed on a Quattromicro quadrupole mass spectrometer with electrospray, positive-mode ionization. The assay was linear over the concentration range 50-5000 ng/mL and proved to be accurate (103.4-105.4%) and precise (<5.0%CV). Hemolysis (10% RBC) and use of heparin as anticoagulant did not impact quantitation. Recovery from plasma varied between 103.0-107.7% and matrix effect was -47.5 to -41.3%. Plasma freeze-thaw stability (97.7-104.9%), stability for 3 months at -80°C (103.4-111.4%), and stability for 4 h at room temperature (100.1-104.9%) were all acceptable. Incurred sample reanalysis of (N = 64) passed: 100% samples within 20% difference, -0.7% median difference and 1.1% median absolute difference. External validation showed a bias of less than 1.1%. This assay will help further define the clinical pharmacokinetics of cabozantinib.
Collapse
Affiliation(s)
- Reyna Jones
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, 5115 Centre Ave, Pittsburgh, PA 15232 , USA
| | - Julianne Holleran
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, 5115 Centre Ave, Pittsburgh, PA 15232 , USA
| | - Robert A Parise
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, 5115 Centre Ave, Pittsburgh, PA 15232 , USA
| | - Michelle A Rudek
- Department of Oncology and Medicine, Johns Hopkins University School of Medicine and Johns Hopkins Sidney Kimmel Cancer Center, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Jennifer Chan
- Dana Farber/Partners CancerCare, Dana-Farber Cancer Institute 450 Brookline Ave. Boston, MA 02215-5450, USA
| | - Yujia Wen
- Alliance for Clinical Trials in Oncology, 125 S. Wacker Drive, Suite 1600, Chicago, IL 60606 , USA
| | - Joga Gobburu
- Center for Translational Medicine, University of Maryland, 20 North Pine Street Baltimore, Maryland 21201 , USA
| | - Lionel D Lewis
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, The Geisel School of Medicine at Dartmouth, 1 Medical Center Dr, Lebanon, NH 03766, USA
| | - Jan H Beumer
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, 5115 Centre Ave, Pittsburgh, PA 15232 , USA
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, 5115 Centre Ave, Pittsburgh, PA 15232, USA
- Department of Pharmaceutical Sciences, University of Pittsburgh, School of Pharmacy, 3501 Terrace St Pittsburgh, PA 15261, USA
| |
Collapse
|
21
|
Krens SD, van Erp NP, Groenland SL, Moes DJAR, Mulder SF, Desar IME, van der Hulle T, Steeghs N, van Herpen CML. Exposure-response analyses of cabozantinib in patients with metastatic renal cell cancer. BMC Cancer 2022; 22:228. [PMID: 35236333 PMCID: PMC8892746 DOI: 10.1186/s12885-022-09338-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/17/2022] [Indexed: 11/10/2022] Open
Abstract
Aim In the registration trial, cabozantinib exposure ≥ 750 ng/mL correlated to improved tumor size reduction, response rate and progression free survival (PFS) in patients with metastatic renal cell cancer (mRCC). Because patients in routine care often differ from patients in clinical trials, we explored the cabozantinib exposure–response relationship in patients with mRCC treated in routine care. Methods Cabozantinib trough concentrations (Cmin) were collected and average exposure was calculated per individual. Exposure–response analyses were performed using the earlier identified target of Cmin > 750 ng/mL and median Cmin. In addition, the effect of dose reductions on response was explored. PFS was used as measure of response. Results In total, 59 patients were included:10% were classified as favourable, 61% as intermediate and 29% as poor IMDC risk group, respectively. Median number of prior treatment lines was 2 (0–5). Starting dose was 60 mg in 46%, 40 mg in 42% and 20 mg in 12% of patients. Dose reductions were needed in 58% of patients. Median Cmin was 572 ng/mL (IQR: 496–701). Only 17% of patients had an average Cmin ≥ 750 ng/mL. Median PFS was 52 weeks (95% CI: 40–64). No improved PFS was observed for patients with Cmin ≥ 750 ng/mL or ≥ 572 ng/ml. A longer PFS was observed for patients with a dose reduction vs. those without (65 vs. 31 weeks, p = .001). After incorporating known covariates (IMDC risk group and prior treatment lines (< 2 vs. ≥ 2)) in the multivariable analysis, the need for dose reduction remained significantly associated with improved PFS (HR 0.32, 95% CI:0.14–0.70, p = .004). Conclusion In these explorative analyses, no clear relationship between increased cabozantinib exposure and improved PFS was observed. Average cabozantinib exposure was below the previously proposed target in 83% of patients. Future studies should focus on validating the cabozantinib exposure required for long term efficacy. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09338-1.
Collapse
Affiliation(s)
- Stefanie D Krens
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands.
| | - Nielka P van Erp
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Stefanie L Groenland
- Division of Medical Oncology, Department of Clinical Pharmacology, The Netherlands Cancer Institute-Antoni Van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Dirk Jan A R Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Sasja F Mulder
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Ingrid M E Desar
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Tom van der Hulle
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Neeltje Steeghs
- Division of Medical Oncology, Department of Clinical Pharmacology, The Netherlands Cancer Institute-Antoni Van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Carla M L van Herpen
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| |
Collapse
|
22
|
An Easily Expandable Multi-Drug LC-MS Assay for the Simultaneous Quantification of 57 Oral Antitumor Drugs in Human Plasma. Cancers (Basel) 2021; 13:cancers13246329. [PMID: 34944950 PMCID: PMC8699473 DOI: 10.3390/cancers13246329] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/06/2021] [Accepted: 12/13/2021] [Indexed: 11/25/2022] Open
Abstract
Simple Summary Oral antitumor therapy has significantly improved clinical outcomes in multiple tumor entities. However, following a standard dosing regime, strong interindividual variability in patients’ plasma concentrations can be observed for many oral antitumor drugs. This results in risks of reduced therapeutic effect and increased side effects. Monitoring these variable plasma concentrations is an important tool in evaluating multiple factors influencing drug exposure and, if necessary, adjusting therapeutic doses. Here, we developed a method for the simultaneous measurement of 57 oral antitumor drug plasma concentrations. Detection and quantification were achieved using liquid chromatography coupled to an Orbitrap mass spectrometer, which can be easily expanded to newly approved oral antitumor drugs in the future. Applicability of the method was proven by measuring 71 plasma samples from 39 patients undergoing oral antitumor therapy. In summary, the developed method provides an important tool for exposure measurements of oral antitumor drugs. Abstract Oral anticancer drugs have led to significant improvements in the treatment of multiple tumor entities. However, in patients undergoing oral antitumor therapy, plasma concentrations are highly variable, resulting in risks of reduced therapeutic effects or an increase in side effects. One important tool to reduce this variability is therapeutic drug monitoring. In this work we describe a method to simultaneously quantify the plasma concentrations of 57 oral antitumor agents. Quantification of these drugs was achieved using liquid chromatography coupled to an Orbitrap mass spectrometer. The method was fully validated according to the FDA guidelines and constitutes a simple and robust way for exposure monitoring of a wide variety of oral anticancer drugs. Applicability to clinical routine was demonstrated by the analysis of 71 plasma samples taken from 39 patients. In summary, this new multi-drug method allows simultaneous quantification of 57 oral antitumor drugs, which can be applied to exposure monitoring in clinical studies, taking into account the broad variety of oral antitumor drugs prescribed in clinical routine.
Collapse
|
23
|
Grover P, Bhardwaj M, Mehta L. LC-MS/MS studies for identification and characterization of new forced degradation products of dabrafenib and establishment of their degradation pathway. J Pharm Biomed Anal 2021; 206:114351. [PMID: 34509659 DOI: 10.1016/j.jpba.2021.114351] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/08/2021] [Accepted: 08/27/2021] [Indexed: 01/01/2023]
Abstract
Dabrafenib (Tafinlar) is used for the treatment of patients with BRAF V600 mutation positive unresectable or metastatic melanoma. Forced degradation study of the drug product and drug substance is very much important in drug development and drug discovery to establish the intrinsic stability and understand its behaviors towards different stress conditions. In the current study, compressive stress testing of dabrafenib has been performed as per the recommendation of ICH guidelines to identify and characterize all major degradation products of dabrafenib (DPD) formed. Drug substances were exposed to different stressed conditions as per ICH recommendations. The present study observed that the dabrafenib drug substance is very much sensitive when exposed to oxidative degradation conditions at 80 °C temperature conditions and also sensitive to photolytic degradation conditions. Dabrafenib is stable when treated in acidic, alkaline, neutral and thermal degradation environments as there is no degradation observed in signification percentage under these stressed conditions. The best separation of eight degradation products and dabrafenib drug substance was obtained in Waters BEH (Ethylene Bridge Hybrid) C-18 column (1.7 µm, 100 mm × 2.1 mm) having mobile phase composed of Formic acid (0.1%) and methanol as Eluent A and Eluent B respectively using 225 nm wavelengths. The volume of injection (5 µL) and flow rate (0.3 mL/min) was set throughout the study. Dabrafenib is highly unstable to oxidative stressed conditions as five major degradation products (DPD-II, DPD-III, DPD-IV, DPD-V and DPD-VII) were obtained when exposed to hydrogen peroxide. When dabrafenib is treated under photolytic degradation conditions, three major DPs were formed (DPD-I, DPD-VI and DP-VIII). These DPs were further identified and characterized on sophisticated HRMS/MS/TOF technique for accurate mass measurement. Characterization of all the degradation products was carried out in the ESI positive mode of ionization. The establishment of the degradation pathway of drug substance and fragmentation pathway of DPs were explained in the present study which was never reported in any literature.
Collapse
Affiliation(s)
- Parul Grover
- KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad 201206, India.
| | - Monika Bhardwaj
- Natural Product Chemistry Division, Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Lovekesh Mehta
- Amity Institute of Pharmacy, Amity University, Noida 201301, India
| |
Collapse
|
24
|
Zimmermann S, Kurlbaum M, Mayer S, Fassnacht M, Kroiss M, Scherf-Clavel O. Simulation-Based Interpretation of Therapeutically Monitored Cabozantinib Plasma Concentration in Advanced Adrenocortical Carcinoma with Hemodialysis. Ther Drug Monit 2021; 43:706-711. [PMID: 34001696 DOI: 10.1097/ftd.0000000000000905] [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: 03/10/2021] [Accepted: 04/30/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Adrenocortical carcinoma is an orphan but aggressive malignancy with limited treatment options. Cabozantinib (CAB), a tyrosine kinase inhibitor, has emerged as a new potential treatment. However, no data are available on whether and how CAB can be administered to patients undergoing hemodialysis. METHODS An liquid chromatography with tandem mass spectrometry detection method was developed and validated according to the European Medicines Agency and United States Food and Drug Administration guidelines for bioanalytical method validation. The samples were prepared using protein precipitation and online solid-phase extraction. The method was applied to clinical samples of an adrenocortical carcinoma patient receiving CAB treatment (80 mg daily). During the 10 days of observation, the patient received periodic hemodialysis on 7 days. Pharmacokinetic (PK) simulations were performed using Bayesian forecasting according to an existing population PK model for CAB. RESULTS Based on the PK simulation, a mean plasma trough concentration of 1375 ng/mL [90% prediction interval (PI), 601-2602 ng/mL] in the steady state at a daily dose of 80 mg was expected for CAB. However, an individual simulation involving the measured plasma levels of the patient resulted in a mean trough concentration of 348 ng/mL (90% PI, 278-430 ng/mL). The model based on individual PK parameters estimated accessible plasma levels of 521, 625, and 834 ng/mL by dose adjustment to 100, 120, and 160 mg, respectively. CONCLUSIONS After establishing an liquid chromatography with tandem mass spectrometry detection method for therapeutic drug monitoring of CAB, our analyses involving a single patient undergoing hemodialysis indicated that higher than expected doses of CAB were required to achieve reasonable plasma concentrations. Our study demonstrates the usefulness of therapeutic drug monitoring for the evaluation of "new" drugs in patients with renal impairment.
Collapse
Affiliation(s)
- Sebastian Zimmermann
- Department of Clinical Pharmacy Institute for Pharmacy and Food Chemistry, University of Würzburg, Würzburg, Germany
| | - Max Kurlbaum
- Department of Internal Medicine I, Division of Endocrinology/Diabetology, University Hospital, University of Würzburg, Würzburg, Germany . Dr. Kroiss is now with the Department of Medicine IV, University Hospital Munich, Ludwig- Maximilians-Universität München, Munich, Germany
- Core Unit Clinical Mass Spectrometry, University Hospital, University of Würzburg, Würzburg, Germany
| | - Stefanie Mayer
- Department of Internal Medicine I, Division of Nephrology, University Hospital, University of Würzburg, Würzburg, Germany; and
| | - Martin Fassnacht
- Department of Internal Medicine I, Division of Endocrinology/Diabetology, University Hospital, University of Würzburg, Würzburg, Germany . Dr. Kroiss is now with the Department of Medicine IV, University Hospital Munich, Ludwig- Maximilians-Universität München, Munich, Germany
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| | - Matthias Kroiss
- Department of Internal Medicine I, Division of Endocrinology/Diabetology, University Hospital, University of Würzburg, Würzburg, Germany . Dr. Kroiss is now with the Department of Medicine IV, University Hospital Munich, Ludwig- Maximilians-Universität München, Munich, Germany
- Core Unit Clinical Mass Spectrometry, University Hospital, University of Würzburg, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| | - Oliver Scherf-Clavel
- Department of Clinical Pharmacy Institute for Pharmacy and Food Chemistry, University of Würzburg, Würzburg, Germany
| |
Collapse
|
25
|
Krens SD, van Boxtel W, Uijen MJM, Jansman FGA, Desar IME, Mulder SF, van Herpen CML, van Erp NP. Exposure-toxicity relationship of cabozantinib in patients with renal cell cancer and salivary gland cancer. Int J Cancer 2021; 150:308-316. [PMID: 34494665 PMCID: PMC9291492 DOI: 10.1002/ijc.33797] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/23/2021] [Accepted: 08/17/2021] [Indexed: 01/18/2023]
Abstract
Cabozantinib is registered in fixed 60 mg dose. However, 46% to 62% of patients in the registration studies needed a dose reduction due to toxicity. Improved clinical efficacy has been observed in renal cell carcinoma patients (RCC) with a cabozantinib exposure greater than 750 μg/L. In our study we explored the cabozantinib exposure in patients with different tumour types. We included RCC patients from routine care and salivary gland carcinoma (SGC) patients from a phase II study with ≥1 measured Cmin at steady‐state. The geometric mean (GM) Cmin at the starting dose, at 40 mg and at best tolerated dose (BTD) were compared between both tumour types. Forty‐seven patients were included. All SGC patients (n = 22) started with 60 mg, while 52% of RCC patients started with 40 mg. GM Cmin at the start dose was 1456 μg/L (95% CI: 1185‐1789) vs 682 μg/L (95% CI: 572‐812) (P < .001) for SGC and RCC patients, respectively. When dose‐normalised to 40 mg, SGC patients had a significantly higher cabozantinib exposure compared to RCC patients (Cmin 971 μg/L [95% CI: 790‐1193] vs 669 μg/L [95% CI: 568‐788]) (P = .005). Dose reductions due to toxicity were needed in 91% and 60% of SGC and RCC patients, respectively. Median BTD was between 20 to 30 mg for SGC and 40 mg for RCC patients. GM Cmin at BTD were comparable between the SGC and the RCC group, 694 μg/L (95% CI: 584‐824) vs 583 μg/L (95% CI: 496‐671) (P = .1). The observed cabozantinib exposure at BTD of approximately 600 μg/L is below the previously proposed target. Surprisingly, a comparable exposure at BTD was reached at different dosages of cabozantinib for SGC patients compared to RCC patients Further research is warranted to identify the optimal exposure and starting dose to balance efficacy and toxicity.
What's new?
Cabozantinib, a potent tyrosine kinase inhibitor that targets multiple signaling pathways, is approved for use against advanced renal cell carcinoma (RCC). Variations in cabozantinib clearance, however, warrant further investigation. Here, the authors evaluated cabozantinib exposure in RCC patients and in patients with salivary gland cancer (SGC). SGC patients were found to have significantly higher cabozantinib exposure compared to RCC patients following a 40 mg dose. However, the best‐tolerated cabozantinib exposure was equivalent (~600 μg/L) for both tumor types and was substantially below the previously proposed target. The findings offer insight on exposure, dose, and the balance between efficacy and toxicity for cabozantinib.
Collapse
Affiliation(s)
- Stefanie D Krens
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wim van Boxtel
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maike J M Uijen
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frank G A Jansman
- Department of Clinical Pharmacy, Deventer Hospital, Deventer, The Netherlands.,Unit of Pharmacotherapy, Pharmacoepidemiology and Pharmacoeconomics, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Ingrid M E Desar
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sasja F Mulder
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carla M L van Herpen
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nielka P van Erp
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
26
|
Nair S, Davis A, Campagne O, Schuetz JD, Stewart CF. Development and validation of an LC-MS/MS method to quantify the bromodomain and extra-terminal (BET) inhibitor JQ1 in mouse plasma and brain microdialysate: Application to cerebral microdialysis study. J Pharm Biomed Anal 2021; 204:114274. [PMID: 34311284 DOI: 10.1016/j.jpba.2021.114274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 11/30/2022]
Abstract
JQ1, is a cell-permeable small-molecule inhibitor of bromodomain and extra-terminal protein (BET) function with reportedly good CNS penetration, however, unbound and pharmacologically active CNS JQ1 exposures have not been characterized. Additionally, no quantitative bioanalytical methods for JQ1 have been described in the literature to support the CNS penetration studies. In the present article, we discuss the development and validation of a sensitive and reliable liquid chromatography-tandem mass spectrometry (LC-MS/MS) quantitative methods to determine JQ1 in mouse plasma and brain microdialysate. JQ1 and the internal standard, dabrafenib (ISTD), were extracted from plasma and microdialysate samples using a simple solid phase extraction protocol performed on an Oasis HLB μElution plate. Chromatographic separation of JQ1 and ISTD was achieved on a reversed phase C12 analytical column with gradient elution profile of mobile phases (MP A: water containing 0.1 % formic acid and MP B: acetonitrile containing 0.1 % formic acid) at a flow rate of 0.6 mL/min. The mass spectrometric detection was performed in the positive MRM ion mode by monitoring the transitions 457.40 > 341.30 (JQ1) and 520.40 > 307.20 (ISTD). The calibration curves demonstrated good linearities over the concentration range of 5-1000 ng/mL for the mouse plasma method (r2 ≥ 0.99) and 0.5-500 ng/mL for the microdialysate method (r2 ≥ 0.99). The experimental limit of quantification obtained was 5 and 0.5 ng/mL for the mouse plasma and microdialysate method, respectively, with the coefficient of variation less than 10 % for the analyte peak area. All the other validation parameters, including intra-and inter-day accuracy and precision, matrix effect, selectivity, carryover effect, and stability, were within the USFDA bioanalytical guidelines acceptance limits. The LC-MS/MS method was successfully applied to a mouse pharmacokinetic and cerebral microdialysis study to characterize the unbound JQ1 exposure in brain extracellular fluid and plasma.
Collapse
Affiliation(s)
- Sreenath Nair
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Abigail Davis
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Olivia Campagne
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - John D Schuetz
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Clinton F Stewart
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
27
|
Concurrent detection of cabozantinib as an anticancer agent and its major metabolites in human serum using fluorescence-coupled micellar liquid chromatography. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2021.103206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
28
|
Zhou L, Wang S, Chen M, Huang S, Zhang M, Bao W, Bao A, Zhang P, Guo H, Liu Z, Xie G, Gao J, Wu Z, Lou Y, Fan G. Simultaneous and rapid determination of 12 tyrosine kinase inhibitors by LC-MS/MS in human plasma: Application to therapeutic drug monitoring in patients with non-small cell lung cancer. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1175:122752. [PMID: 33991955 DOI: 10.1016/j.jchromb.2021.122752] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022]
Abstract
In recent years, more than 50 tyrosine kinase inhibitors (TKIs) was indicated against numerous cancers, especially outstanding advantages in the treatment of non-small cell lung cancer (NSCLC), and several studies have shown that therapeutic drug monitoring (TDM) of TKIs can improve treatment efficacy and safety. The present study aimed to develop and validate a LC-MS/MS method for the TDM of 12 TKIs (gefitinib, erlotinib, afatinib, dacomitinib, icotinib, osimertinib, crizotinib, ceritinib, alectinib, dabrafenib, trametinib, anlotinib) in patients with NSCLC. The analytes of interest and internal standard were extracted from human plasma. Salting-out assisted liquid-liquid extraction (SALLE) with 5 M ammonium acetate solution was optimized for method validation and compared to simple protein precipitation (PPT). Chromatographic separation was conducted on Waters X bridge C18 column (100 × 4.6 mm, 3.5 μm) using a gradient elution of acetonitrile/5mM ammonium acetate in pure water with 0.1% (v/v) formic acid at 40 °C within 6 min. The total flow was maintained at 1 mL/min, 30% of the post column flow was split into the mass spectrometer and the rest to waste via a 3-way tee. The mass analysis was performed by positive ion electrospray ionization (ESI) in multiple-reaction monitoring (MRM) mode. The assay was validated based on the guidelines on bioanalytical methods by FDA. This quantification method was proved to be satisfactory in selectivity, accuracy, precision, linearity (r2 > 0.995), recovery, matrix effect and stability and the accuracy was further assessed in plasma with a degree of hemolysis of 4%. The described method to simultaneously quantify the 12 selected anticancer drugs in human plasma was successfully validated and applied to routine TDM of gefitinib, erlotinib, icotinib, osimertinib, crizotinib and anlotinib in cancer patients. TKIs plasma monitoring helps to individualize dose adjustment and manage adverse effects in NSCLC patients.
Collapse
Affiliation(s)
- Lijuan Zhou
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Shuowen Wang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Ming Chen
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China
| | - Shiqi Huang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530001, PR China
| | - Min Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Wuping Bao
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Aihua Bao
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Pengyu Zhang
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Haiying Guo
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Zhenwei Liu
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Guogang Xie
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Jianwei Gao
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Zhenghua Wu
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China.
| | - Yuefen Lou
- Department of Pharmacy, Shanghai Fourth People's Hospital, Affiliated to Tongji University School of Medicine, Shanghai 200434, PR China.
| | - Guorong Fan
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, PR China.
| |
Collapse
|
29
|
Balakirouchenane D, Khoudour N, Guégan S, Kramkimel N, Franck N, Rodier T, Goldwasser F, Dupin N, Aractingi S, Vidal M, Blanchet B. Simultaneous quantification of dabrafenib, hydroxy-dabrafenib and trametinib in human plasma by liquid chromatography-tandem mass spectrometry. J Pharm Biomed Anal 2020; 193:113718. [PMID: 33166838 DOI: 10.1016/j.jpba.2020.113718] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/16/2020] [Accepted: 10/17/2020] [Indexed: 12/21/2022]
Abstract
A new liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for the simultaneous quantification of dabrafenib (DAB), its main metabolite hydroxy-dabrafenib (OHD) and trametinib (TRA) in human plasma has been developed and validated. After addition of internal standard (dabrafenib-d9), extraction was achieved after protein precipitation with acetonitrile containing 1 % (v/v) formic acid. Chromatographic separation was performed on an Accucore® C18 (2.1 × 50 mm; 2.6 μm) column using a gradient elution of water acidified with 0.1 % (v/v) formic acid (A) and acetonitrile containing 0.1 % (v/v) formic acid (B) at a flow rate of 500 μL/min. The calibration ranged from 10 to 2000 ng/mL for DAB and OHD and from 5 to 50 ng/mL for TRA. This method was validated with satisfactory results including good precision (intra- and inter-assay coefficient of variation from 2.0 %-14.9 %) and good accuracy (inter- and intra-day bias between -1.2 % and 10.9 %), as well as long term stability in unprocessed plasma at -20 °C. This newly proposed method is useful for clinical research purposes as well as therapeutic drug monitoring for patients with a Rapidly Accelerated Fibrosarcoma kinase B (BRAF)-mutated cancer.
Collapse
Affiliation(s)
- David Balakirouchenane
- Department of Pharmacokinetics and Pharmacochemistry, Cochin Hospital, AP-HP, CARPEM, 75014 Paris, France; UMR8038 CNRS, U1268 INSERM, Faculty of Pharmacy, University of Paris, PRES Sorbonne Paris Cité, CARPEM, 75006 Paris, France.
| | - Nihel Khoudour
- Department of Pharmacokinetics and Pharmacochemistry, Cochin Hospital, AP-HP, CARPEM, 75014 Paris, France
| | - Sarah Guégan
- Department of Dermatology, Cochin Hospital AP-HP, 75014 Paris, France; Cochin Institute, INSERM U1016, University of Paris, 75014 Paris, France
| | - Nora Kramkimel
- Department of Dermatology, Cochin Hospital AP-HP, 75014 Paris, France
| | - Nathalie Franck
- Department of Dermatology, Cochin Hospital AP-HP, 75014 Paris, France
| | - Thomas Rodier
- Department of Pharmacokinetics and Pharmacochemistry, Cochin Hospital, AP-HP, CARPEM, 75014 Paris, France; UMR8038 CNRS, U1268 INSERM, Faculty of Pharmacy, University of Paris, PRES Sorbonne Paris Cité, CARPEM, 75006 Paris, France
| | | | - Nicolas Dupin
- Department of Dermatology, Cochin Hospital AP-HP, 75014 Paris, France; Cochin Institute, INSERM U1016, University of Paris, 75014 Paris, France
| | - Selim Aractingi
- Department of Dermatology, Cochin Hospital AP-HP, 75014 Paris, France; Cochin Institute, INSERM U1016, University of Paris, 75014 Paris, France
| | - Michel Vidal
- Department of Pharmacokinetics and Pharmacochemistry, Cochin Hospital, AP-HP, CARPEM, 75014 Paris, France; UMR8038 CNRS, U1268 INSERM, Faculty of Pharmacy, University of Paris, PRES Sorbonne Paris Cité, CARPEM, 75006 Paris, France
| | - Benoit Blanchet
- Department of Pharmacokinetics and Pharmacochemistry, Cochin Hospital, AP-HP, CARPEM, 75014 Paris, France; UMR8038 CNRS, U1268 INSERM, Faculty of Pharmacy, University of Paris, PRES Sorbonne Paris Cité, CARPEM, 75006 Paris, France
| |
Collapse
|
30
|
Sulochana SP, Trivedi RK, Srinivas NR, Mullangi R. A concise review of bioanalytical methods of small molecule immuno-oncology drugs in cancer therapy. Biomed Chromatogr 2020; 35:e4996. [PMID: 33047346 DOI: 10.1002/bmc.4996] [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: 08/07/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 01/13/2023]
Abstract
Immuno-oncology (IO) is an emerging option to treat cancer malignancies. In the last two years, IO has accounted for more than 90% of the new active drugs in various therapeutic indications of oncology drug development. Bioanalytical methods used for the quantitation of various IO small molecule drugs have been summarized in this review. The most commonly used are HPLC and LC-MS/MS methods. Determination of IO drugs from biological matrices involves drug extraction from the biological matrix, which is mostly achieved by simple protein precipitation, liquid-liquid extraction and solid-phase extraction. Subsequently, quantitation is usually achieved by LC-MS/MS, but HPLC-UV has also been employed. The bioanalytical methods reported for each drug are briefly discussed and tabulated for easy access. Our review indicates that LC-MS/MS is a versatile and reliable tool for the sensitive, rapid and robust quantitation of IO drugs.
Collapse
Affiliation(s)
- Suresh P Sulochana
- Pharmacokinetics and Drug Metabolism Group, University of Mississippi, MS, USA
| | | | | | | |
Collapse
|
31
|
Yang H, Ren L, Wang Y, Bi X, Li X, Wen M, Zhang Q, Yang Y, Jia Y, Li Y, Zang A, Wei Y, Dai G. FBI-1 enhanced the resistance of triple-negative breast cancer cells to chemotherapeutic agents via the miR-30c/PXR axis. Cell Death Dis 2020; 11:851. [PMID: 33051436 PMCID: PMC7554048 DOI: 10.1038/s41419-020-03053-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022]
Abstract
The factor that binds to the inducer of short transcripts-1 (FBI-1) is a transcription suppressor and an important proto-oncogene that plays multiple roles in carcinogenesis and therapeutic resistance. In the present work, our results indicated that FBI-1 enhanced the resistance of triple-negative breast cancer (TNBC) cells to chemotherapeutic agents by repressing the expression of micoRNA-30c targeting the pregnane X receptor (PXR). The expression of FBI-1 was positively related to PXR and its downstream drug resistance-related genes in TNBC tissues. FBI-1 enhanced the expression of PXR and enhanced the activation of the PXR pathway. The miR-30c decreased the expression of PXR by targeting the 3'-UTR of PXR, and FBI-1 increased the expression of PXR by repressing miR-30c's expression. Through the miR-30c/PXR axis, FBI-1 accelerated the clearance or elimination of antitumor agents in TNBC cells (the TNBC cell lines or the patients derived cells [PDCs]) and induced the resistance of cells to antitumor agents. Therefore, the results indicated that the miR-30c/PXR axis participates in the FBI-1-mediated drug-resistance of TNBC cells.
Collapse
Affiliation(s)
- Hua Yang
- Department of the Medical Oncology, the PLA General Hospital, Beijing, 100853, China.,Department of the Medical Oncology/the Hebei Key Laboratory of the Cancer Radiotherapy and Chemotherapy, Baoding City, 071000, Hebei province, P.R. China
| | - Lili Ren
- Department of the Medical Oncology/the Hebei Key Laboratory of the Cancer Radiotherapy and Chemotherapy, Baoding City, 071000, Hebei province, P.R. China
| | - Yanan Wang
- Department of the Medical Oncology/the Hebei Key Laboratory of the Cancer Radiotherapy and Chemotherapy, Baoding City, 071000, Hebei province, P.R. China
| | - Xuebing Bi
- Department of the Medical Oncology/the Hebei Key Laboratory of the Cancer Radiotherapy and Chemotherapy, Baoding City, 071000, Hebei province, P.R. China
| | - Xiaoli Li
- Department of the Medical Oncology/the Hebei Key Laboratory of the Cancer Radiotherapy and Chemotherapy, Baoding City, 071000, Hebei province, P.R. China
| | - Ming Wen
- Department of the Gastrointestinal Surgery, the Affiliated Hospital of Hebei University, Baoding City, 071000, Hebei province, P.R. China
| | - Qian Zhang
- Department of the Medical Oncology/the Hebei Key Laboratory of the Cancer Radiotherapy and Chemotherapy, Baoding City, 071000, Hebei province, P.R. China
| | - Yang Yang
- Department of the Medical Oncology/the Hebei Key Laboratory of the Cancer Radiotherapy and Chemotherapy, Baoding City, 071000, Hebei province, P.R. China
| | - Youchao Jia
- Department of the Medical Oncology/the Hebei Key Laboratory of the Cancer Radiotherapy and Chemotherapy, Baoding City, 071000, Hebei province, P.R. China
| | - Yumiao Li
- Department of the Medical Oncology/the Hebei Key Laboratory of the Cancer Radiotherapy and Chemotherapy, Baoding City, 071000, Hebei province, P.R. China
| | - Aimin Zang
- Department of the Medical Oncology/the Hebei Key Laboratory of the Cancer Radiotherapy and Chemotherapy, Baoding City, 071000, Hebei province, P.R. China
| | - Yaning Wei
- Department of the Medical Oncology/the Hebei Key Laboratory of the Cancer Radiotherapy and Chemotherapy, Baoding City, 071000, Hebei province, P.R. China.
| | - Guanghai Dai
- Department of the Medical Oncology, the PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
32
|
Krens SD, van der Meulen E, Jansman FGA, Burger DM, van Erp NP. Quantification of cobimetinib, cabozantinib, dabrafenib, niraparib, olaparib, vemurafenib, regorafenib and its metabolite regorafenib M2 in human plasma by UPLC-MS/MS. Biomed Chromatogr 2020; 34:e4758. [PMID: 31758580 PMCID: PMC7065026 DOI: 10.1002/bmc.4758] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/13/2019] [Accepted: 11/19/2019] [Indexed: 12/30/2022]
Abstract
A sensitive and selective ultra‐high performance liquid chromatography–tandem mass spectrometry (UPLC–MS/MS) method for the simultaneous determination of seven oral oncolytics (two PARP inhibitors, i.e. olaparib and niraparib, and five tyrosine kinase inhibitors, i.e. cobimetinib, cabozantinib, dabrafenib, vemurafenib and regorafenib, plus its active metabolite regorafenib M2) in EDTA plasma was developed and validated. Stable isotope‐labelled internal standards were used for each analyte. A simple protein precipitation method was performed with acetonitrile. The LC–MS/MS system consisted of an Acquity H‐Class UPLC system, coupled to a Xevo TQ‐S micro tandem mass spectrometer. The compounds were separated on a Waters CORTECS UPLC C18 column (2.1 × 50 mm, 1.6 μm particle size) and eluted with a gradient elution system. The ions were detected in the multiple reaction monitoring mode. The method was validated for cobimetinib, cabozantinib, dabrafenib, niraparib, olaparib, vemurafenib, regorafenib and regorafenib M2 over the ranges 6–1000, 100–5000, 10–4000, 200–2000, 200–20,000, 5000–100,000, 500–10,000 and 500–10,000 μg/L, respectively. Within‐day accuracy values for all analytes ranged from 86.8 to 115.0% with a precision of <10.4%. Between‐day accuracy values ranged between 89.7 and 111.9% with a between‐day precision of <7.4%. The developed method was successfully used for guiding therapy with therapeutic drug monitoring in cancer patients and clinical research programs in our laboratory.
Collapse
Affiliation(s)
- Stefanie D Krens
- Department of Pharmacy, Radboud University Medical Center , Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Eric van der Meulen
- Department of Pharmacy, Radboud University Medical Center , Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Frank G A Jansman
- Department of Pharmacy, Deventer Hospital, Deventer, The Netherlands.,Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands
| | - David M Burger
- Department of Pharmacy, Radboud University Medical Center , Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Nielka P van Erp
- Department of Pharmacy, Radboud University Medical Center , Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| |
Collapse
|