1
|
Peruzzi E, Posocco B, Gerratana L, Nuti M, Orleni M, Gagno S, De Mattia E, Puglisi F, Cecchin E, Toffoli G, Roncato R. Exploring pharmacokinetic variability of palbociclib in HR+/HER2- metastatic breast cancer: a focus on age, renal function, and drug-gene interactions. Front Pharmacol 2024; 15:1420174. [PMID: 39309010 PMCID: PMC11412846 DOI: 10.3389/fphar.2024.1420174] [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: 04/19/2024] [Accepted: 07/15/2024] [Indexed: 09/25/2024] Open
Abstract
Palbociclib, an oral inhibitor of cyclin-dependent kinase 4 and 6, is approved for the treatment of metastatic breast cancer. This study investigated the influence of diverse clinical and biological factors-age, renal function, genetic variations, and concomitant medications (pharmacokinetic covariates)-on palbociclib pharmacokinetics. Employing a validated LC-MS/MS method, we analyzed the minimum plasma concentrations (Ctrough) of palbociclib in 68 women and determined the percentage deviations from the median Ctrough for each dosage group. Variations in a panel of absorption, distribution, metabolism, and excretion (ADME) genes were assessed using end-point allele-specific fluorescence detection and pyrosequencing. Two distinct patient cohorts were defined based on median values of age, creatinine, and eGFR, which exhibited statistically significant differences in percentage deviations (p = 0.0095, p = 0.0288, and p = 0.0005, respectively). Homozygous carriers of the PPARA variants displayed larger positive percentage deviations than the other group (p = 0.0292). Similarly, patients concurrently taking CYP3A and P-glycoprotein inhibitors alongside anticancer therapy exhibited significant variations (p = 0.0285 and p = 0.0334, respectively). Furthermore, exploring the drug-drug-gene interactions between inhibitors of CYP3A and P-glycoprotein with their respective genetic variants revealed two patient groups with statistically different percentage deviations (p = 0.0075, p = 0.0012, and p = 0.0191, respectively). These results could help address cases where pharmacokinetic covariates or subclinical conditions impair palbociclib adherence or response, aiming to offer tailored dosing strategies or monitoring for individual patients.
Collapse
Affiliation(s)
- Elena Peruzzi
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Bianca Posocco
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Lorenzo Gerratana
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
- Department of Medicine (DMED), University of Udine, Udine, Italy
| | - Margherita Nuti
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Marco Orleni
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
- Doctoral School in Pharmacological Sciences, University of Padua, Padova, Italy
| | - Sara Gagno
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Elena De Mattia
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Fabio Puglisi
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
- Department of Medicine (DMED), University of Udine, Udine, Italy
| | - Erika Cecchin
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Rossana Roncato
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
- Department of Medicine (DMED), University of Udine, Udine, Italy
| |
Collapse
|
2
|
Ferrer F, Tetu P, Dousset L, Lebbe C, Ciccolini J, Combarel D, Meyer N, Paci A, Bouchet S. Tyrosine kinase inhibitors in cancers: Treatment optimization - Part II. Crit Rev Oncol Hematol 2024; 200:104385. [PMID: 38810843 DOI: 10.1016/j.critrevonc.2024.104385] [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: 01/24/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024] Open
Abstract
Real-life populations are more heterogeneous than those included in prospective clinical studies. In cancer patients, comorbidities and co-medications favor the appearance of severe adverse effects which can significantly impact quality of life and treatment effectiveness. Most of tyrosine kinase inhibitors (TKI) have been developed with flat oral dosing exposing patients to the risk of poor adherence due to side effects. Additionally, genetic or physiological factors, differences in diet, and drug-drug interactions can lead to inter-individual variability affecting treatment outcomes and increasing the risk of adverse events. Knowledge of the different factors of variability allows individualized patient management. This review examines the effects of adherence, food intake, and pharmaceutical form on the pharmacokinetics of oral TKI, as well as evaluating pharmacokinetics considerations improving TKI management. Concentration-effectiveness and concentration-toxicity data are presented for the selected TKI, and a simple therapeutic drug monitoring schema is outlined to help individualize dosing of oral TKI.
Collapse
Affiliation(s)
- Florent Ferrer
- Department of Pharmacology, Clermont-Ferrand University Hospital, Clermont-Ferrand, France; SMARTc Unit, CRCM Inserm U1068, Aix Marseille Univ and APHM, Marseille, France; Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, Villejuif 94805, France
| | - Pauline Tetu
- Department of Dermatology, APHP Dermatology, Paris 7 Diderot University, INSERM U976, Hôpital Saint-Louis, Paris, France; Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, Villejuif 94805, France
| | - Léa Dousset
- Dermatology Department, Bordeaux University Hospital, Bordeaux, France; Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, Villejuif 94805, France
| | - Céleste Lebbe
- Department of Dermatology, APHP Dermatology, Paris 7 Diderot University, INSERM U976, Hôpital Saint-Louis, Paris, France; Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, Villejuif 94805, France
| | - Joseph Ciccolini
- SMARTc Unit, CRCM Inserm U1068, Aix Marseille Univ and APHM, Marseille, France; Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, Villejuif 94805, France
| | - David Combarel
- Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, Villejuif 94805, France; Service de Pharmacocinétique, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, Châtenay-Malabry 92 296, France
| | - Nicolas Meyer
- Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, Villejuif 94805, France; Université Paul Sabatier-Toulouse III, Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche, Toulouse 1037-CRCT, France
| | - Angelo Paci
- Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, Villejuif 94805, France; Service de Pharmacocinétique, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, Châtenay-Malabry 92 296, France
| | - Stéphane Bouchet
- Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, Villejuif 94805, France; Département de Pharmacologie, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France.
| |
Collapse
|
3
|
Zhao J, Yan D, Li Y, Xu X, Li F, Zhang S, Jin J, Qiu F. Simultaneous determination of 11 oral targeted antineoplastic drugs and 2 active metabolites by LC-MS/MS in human plasma and its application to therapeutic drug monitoring in cancer patients. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1237:124100. [PMID: 38547701 DOI: 10.1016/j.jchromb.2024.124100] [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: 01/23/2024] [Revised: 03/11/2024] [Accepted: 03/17/2024] [Indexed: 04/13/2024]
Abstract
Interindividual exposure differences have been identified in oral targeted antineoplastic drugs (OADs) owing to the pharmacogenetic background of the patients and their susceptibility to multiple factors, resulting in insufficient efficacy or adverse effects. Therapeutic drug monitoring (TDM) can prevent sub-optimal concentrations of OADs and improve their clinical treatment. This study aimed to develop and validate an LC-MS/MS method for the simultaneous quantification of 11 OADs (gefitinib, imatinib, lenvatinib, regorafenib, everolimus, osimertinib, sunitinib, tamoxifen, lapatinib, fruquintinib and sorafenib) and 2 active metabolites (N-desethyl sunitinib and Z-endoxifen) in human plasma. Protein precipitation was used to extract OADs from the plasma samples. Chromatographic separation was performed using an Eclipse XDB-C18 (4.6 × 150 mm, 5 μm) column with a gradient elution of the mobile phase composed of 2 mM ammonium acetate with 0.1 % formic acid in water (solvent A) and methanol (solvent B) at a flow rate of 0.8 mL/min. Mass analysis was performed using positive ion mode electrospray ionization in multiple-reaction monitoring mode. The developed method was validated following FDA bioanalytical guidelines. The calibration curves were linear over the range of 2-400 ng/mL for gefitinib, imatinib, lenvatinib, regorafenib, and everolimus; 1-200 ng/mL for osimertinib, sunitinib, N-desethyl sunitinib, tamoxifen, and Z-endoxifen; and 5-1000 ng/mL for lapatinib, fruquintinib, and sorafenib, with all coefficients of correlation above 0.99. The intra- and inter-day imprecision was below 12.81 %. This method was successfully applied to the routine TDM of gefitinib, lenvatinib, regorafenib, osimertinib, fruquintinib, and sorafenib to optimize the dosage regimens.
Collapse
Affiliation(s)
- Jing Zhao
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China
| | - Dongming Yan
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China
| | - Yue Li
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China
| | - Xiaoqing Xu
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China
| | - Fengling Li
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China
| | - Shuang Zhang
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China
| | - Jingyi Jin
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China.
| | - Furong Qiu
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China.
| |
Collapse
|
4
|
Philippe M, Guitton J, Goutelle S, Thoma Y, Favier B, Chtiba N, Michallet M, Belhabri A. Pharmacokinetic Consideration of Venetoclax in Acute Myeloid Leukemia Patients: A Potential Candidate for TDM? A Short Communication. Ther Drug Monit 2024; 46:127-131. [PMID: 37941111 DOI: 10.1097/ftd.0000000000001151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/05/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Venetoclax (VNX)-based regimens have demonstrated significantly favorable outcomes in patients with acute myeloid leukemia (AML) and are now becoming the standard treatment. Tyrosine kinase inhibitors are administered at a fixed dose, irrespective of body surface area or weight. For such orally targeted therapies, real-world data have highlighted a larger pharmacokinetic (PK) interindividual variability (IIV) than expected. Even if VNX PKs have been well characterized and described in the literature, only 1 clinical trial-based PK study has been conducted in patients with AML. This study aimed to evaluate the PK of VNX in AML patients. MATERIAL AND METHODS We retrospectively analyzed all patients treated with a combination of VNX-azacitidine between January and July 2022 at our center, using at least 1 available VNX blood sample. Based on a previously published population PK model, individual PK parameters were estimated to evaluate the exposure and IIV. RESULTS and Discussion. Twenty patients received VNX in combination with azacitidine, according to the PK data. A total of 93 plasma concentrations were collected. The dose of VNX was 400 mg, except in 7 patients who received concomitant posaconazole (VNX 70 mg). The patients' weight ranged from 49 kg to 108 kg (mean = 78 kg). Mean individual clearance was 13.5 ± 9.4 L/h with mean individual daily area under the concentration-time curves of 35.8 mg.h/L with significant IIV (coefficient of variation = 41.1%). Ten patients were still alive (8 in complete response), but all experienced at least 1 hematological toxicity of grade ≥ 3. CONCLUSIONS Based on the observed large PK variability in the data from our real-world AML patients, the risk of drug interactions and the recommended fixed-dosage regimen of VNX therapeutic drug monitoring may be useful.
Collapse
Affiliation(s)
| | - Jérôme Guitton
- Biochemistry and Pharmacology-Toxicology Laboratory, Lyon Sud Hospital, Pierre Benite, France
- ISPB, Faculté de Pharmacie de Lyon, Université Lyon 1, Lyon, France
| | - Sylvain Goutelle
- ISPB, Faculté de Pharmacie de Lyon, Université Lyon 1, Lyon, France
- Hospices Civils de Lyon, Groupement Hospitalier Nord, Service de Pharmacie, Lyon, France
- UMR CNRS 5558, Laboratoire de Biométrie et Biologie Evolutive, Université Lyon 1, Villeurbanne, France
| | - Yann Thoma
- School of Engineering and Management Vaud (HEIG-VD), HES-SO University of Applied Sciences and Arts Western Switzerland, Yverdon-les-Bains, Switzerland
| | | | - Nour Chtiba
- Faculty of Pharmacy of Monastir, University of Monastir, Monastir, Tunisia; and
| | | | | |
Collapse
|
5
|
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
|
6
|
Westerdijk K, Steeghs N, Tacke CSJ, van der Graaf WTA, van Erp NP, van Oortmerssen G, Hermens RPMG, Desar IME. Therapeutic drug monitoring to personalize dosing of imatinib, sunitinib, and pazopanib: A mixed methods study on barriers and facilitators. Cancer Med 2023; 12:21041-21056. [PMID: 37902257 PMCID: PMC10709747 DOI: 10.1002/cam4.6663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/20/2023] [Accepted: 10/03/2023] [Indexed: 10/31/2023] Open
Abstract
BACKGROUND Personalized dosing based on measurement of individual drug levels and adjusting the dose accordingly can improve efficacy and decrease unnecessary toxicity of oncological treatment. For imatinib, sunitinib, and pazopanib, this therapeutic drug monitoring (TDM)-guided dosing is, however, not routinely used, despite accumulating evidence favoring individualized dosing. Therefore, we aimed to identify and quantify (potential) barriers and facilitators in TDM-guided dosing for imatinib, sunitinib, and pazopanib. METHODS We performed a mixed methods study among all stakeholders involved: patients, healthcare professionals (HCPs), pharmaceutical companies, and health insurance companies. During the first qualitative part of this study, we performed semi-structured individual interviews and one focus group interview to identify all (potential) barriers and facilitators, and during the second quantitative part of this study, we used a web-based survey to quantify these findings. The interviews addressed the six domains of the implementation of change model of Grol and Wensing: (1) the innovation itself; (2) the HCP; (3) the patient; (4) social context; (5) organizational context; and (6) finances, law, and governance. RESULTS In the qualitative study, we interviewed 20 patients, 18 HCPs and 10 representatives of pharmaceutical and health insurance companies and identified 72 barriers and 90 facilitators. In the quantitative study, the survey was responded by 66 HCPs and 58 patients. Important barriers were on the domain of the HCP, such as a lack of experience with TDM (36.4%), on the domain of the patient, such as lack of awareness of TDM (39.7%), and the processing time for measurement and interpretation of the TDM result (40.9%) (organizational domain). Important facilitators were education of HCPs (95.5%), education of patients (87.9%) and facilitating an overview of when and where TDM measurements are being performed (86.4%). CONCLUSION We identified and quantified important barriers and facilitators for the implementation of TDM-guided dosing for imatinib, sunitinib, and pazopanib. Based on our results, the implementation strategy should mainly focus on educating both HCPs and patients and on the organizational aspect of TDM.
Collapse
Affiliation(s)
- Kim Westerdijk
- Department of Medical OncologyResearch Institute for Medical Innovation, Radboud University Medical CenterNijmegenThe Netherlands
| | - Neeltje Steeghs
- Department of Medical OncologyNetherlands Cancer Institute, Antoni van LeeuwenhoekAmsterdamThe Netherlands
| | - Casper S. J. Tacke
- Department of Medical OncologyResearch Institute for Medical Innovation, Radboud University Medical CenterNijmegenThe Netherlands
| | - Winette T. A. van der Graaf
- Department of Medical OncologyNetherlands Cancer Institute, Antoni van LeeuwenhoekAmsterdamThe Netherlands
- Department of Medical OncologyErasmus MC Cancer Institute, Erasmus Medical Center RotterdamRotterdamThe Netherlands
| | - Nielka P. van Erp
- Department of PharmacyResearch Institute for Medical Innovation, Radboud University Medical CenterNijmegenThe Netherlands
| | | | | | - Ingrid M. E. Desar
- Department of Medical OncologyResearch Institute for Medical Innovation, Radboud University Medical CenterNijmegenThe Netherlands
| | | |
Collapse
|
7
|
van Eerden RAG, IJzerman NS, van Meekeren M, Oomen-de Hoop E, Guchelaar NAD, Visser AMW, Matic M, van Schaik RHN, de Bruijn P, Moes DJAR, Jobse PA, Gelderblom H, Huitema ADR, Steeghs N, Mathijssen RHJ, Koolen SLW. CYP3A4*22 Genotype-Guided Dosing of Kinase Inhibitors in Cancer Patients. Clin Pharmacokinet 2023; 62:1129-1139. [PMID: 37310647 PMCID: PMC10386914 DOI: 10.1007/s40262-023-01260-4] [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: 05/03/2023] [Indexed: 06/14/2023]
Abstract
INTRODUCTION A genetic variant explaining a part of the exposure of many kinase inhibitors (KIs) is the single nucleotide polymorphism (SNP) CYP3A4*22, resulting in less CYP3A4 enzyme activity. The primary aim of this study was to investigate if the systemic exposure is non-inferior after a dose reduction of KIs metabolized by CYP3A4 in CYP3A4*22 carriers compared to patients without this SNP (i.e., wildtype patients) receiving the standard dose. METHODS In this multicenter, prospective, non-inferiority study, patients were screened for the presence of CYP3A4*22. Patients with the CYP3A4*22 SNP received a 20-33% dose reduction. At steady state, a pharmacokinetic (PK) analysis was performed and compared to the PK results from wildtype patients treated with the registered dose using a two-stage individual patient data meta-analysis approach. RESULTS In total, 207 patients were included in the final analysis. The CYP3A4*22 SNP was found in 16% of the patients in the final analysis (n = 34). Most of the included patients received imatinib (37%) or pazopanib (22%) treatment. The overall geometric mean ratio (GMR) comparing the exposure of the CYP3A4*22 carriers to the exposure of the wildtype CYP3A4 patients was 0.89 (90% confidence interval: 0.77-1.03). CONCLUSION Non-inferiority could not be proven for dose reduction of KIs metabolized by CYP3A4 in CYP3A4*22 carriers compared to the registered dose in wildtype patients. Therefore, an up-front dose reduction based upon the CYP3A4*22 SNP for all KIs does not seem an eligible new way of personalized therapy. TRIAL REGISTRATION International Clinical Trials Registry Platform Search Portal; number NL7514; registered 11/02/2019.
Collapse
Affiliation(s)
- Ruben A G van Eerden
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, PO box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Nikki S IJzerman
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, PO box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Milan van Meekeren
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Esther Oomen-de Hoop
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, PO box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Niels A D Guchelaar
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, PO box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Andrea M W Visser
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, PO box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Maja Matic
- Department of Clinical Chemistry, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ron H N van Schaik
- Department of Clinical Chemistry, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Peter de Bruijn
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, PO box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Dirk-Jan A R Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Pieter A Jobse
- Department of Internal Medicine, ADRZ, Goes, The Netherlands
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy and 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
| | - Neeltje Steeghs
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, PO box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Stijn L W Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, PO box 2040, 3000 CA, Rotterdam, The Netherlands.
- Department of Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
8
|
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
|
9
|
Sarah A, Dondi E, De Francia S. Tyrosine kinase inhibitors: the role of pharmacokinetics and pharmacogenetics. Expert Opin Drug Metab Toxicol 2023; 19:733-739. [PMID: 37916644 DOI: 10.1080/17425255.2023.2277758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/27/2023] [Indexed: 11/03/2023]
Affiliation(s)
- Allegra Sarah
- Department of Clinical and Biological Sciences, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, TO, Italy
| | - Emma Dondi
- Department of Clinical and Biological Sciences, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, TO, Italy
| | - Silvia De Francia
- Department of Clinical and Biological Sciences, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, TO, Italy
| |
Collapse
|
10
|
van Leuven J, Evans S, Kichenadasse G, Steeghs N, Bonevski B, Mikus G, van Dyk M. Framework for Implementing Individualised Dosing of Anti-Cancer Drugs in Routine Care: Overcoming the Logistical Challenges. Cancers (Basel) 2023; 15:3293. [PMID: 37444404 DOI: 10.3390/cancers15133293] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/26/2022] [Accepted: 12/26/2022] [Indexed: 07/15/2023] Open
Abstract
Precision medicine in oncology involves identifying the 'right drug', at the 'right dose', for the right person. Currently, many orally administered anti-cancer drugs, particularly kinase inhibitors (KIs), are prescribed at a standard fixed dose. Identifying the right dose remains one of the biggest challenges to optimal patient care. Recently the Precision Dosing Group established the Accurate Dosing of Anti-cancer Patient-centred Therapies (ADAPT) Program to address individualised dosing; thus, use existing anti-cancer drugs more safely and efficiently. In this paper, we outline our framework, based on the Medical Research Council (MRC) framework, with a simple 6-step process and strategies which have led to the successful implementation of the ADAPT program in South Australia. Implementation strategies in our 6-step process involve: (1) Evaluate the evidence and identify the cancer drugs: Literature review, shadowing other experts, establishing academic partnerships, adaptability/flexibility; (2) Establishment of analytical equipment for drug assays for clinical purposes: assessment for readiness, accreditation, feasibility, obtaining formal commitments, quality assurance to all stakeholders; (3) Clinical preparation and education: educational material, conducted educational meetings, involve opinion leaders, use of mass media, promote network weaving, conduct ongoing training; (4) Blood collection, sample preparation and analyses: goods received procedures, critical control points (transport time); (5) Interpret and release results with recommendations: facilitate the relay of clinical data to providers; (6) Clinical application: providing ongoing consultation, identify early adopters, identify, and prepare champions. These strategies were selected from the 73 implementation strategies outlined in the Expert Recommendations for Implementing Change (ERIC) study. The ADAPT program currently provides routine plasma concentrations for patients on several orally administered drugs in South Australia and is currently in its evaluation phase soon to be published. Our newly established framework could provide great potential and opportunities to advance individualised dosing of oral anti-cancer drugs in routine clinical care.
Collapse
Affiliation(s)
- Jason van Leuven
- College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
- Medical Oncology, Flinders Medical Centre, Adelaide 5042, Australia
| | - Simon Evans
- Implementation Science Unit, Department for Health and Wellbeing, Adelaide 5042, Australia
| | - Ganessan Kichenadasse
- College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
- Medical Oncology, Flinders Medical Centre, Adelaide 5042, Australia
| | - Neeltje Steeghs
- Antoni van Leeuwenhoek Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
- Department of Medical Oncology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Billie Bonevski
- College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
| | - Gerd Mikus
- College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Madelé van Dyk
- College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
- Medical Oncology, Flinders Medical Centre, Adelaide 5042, Australia
| |
Collapse
|
11
|
Therapeutic Drug Monitoring in Oncohematological Patients: A Fast and Accurate HPLC-UV Method for the Quantification of Nilotinib in Human Plasma and Its Clinical Application. Biomedicines 2023; 11:biomedicines11030947. [PMID: 36979926 PMCID: PMC10046071 DOI: 10.3390/biomedicines11030947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/11/2023] [Accepted: 03/14/2023] [Indexed: 03/22/2023] Open
Abstract
Nilotinib, a second-generation tyrosine kinase inhibitor, has demonstrated clinical activity in chronic myeloid leukemia. As an exposure–response relationship has been observed for nilotinib, its therapeutic drug monitoring could be a valuable tool in clinical practice. Therefore, the aim of this study was to develop and validate a selective and precise high performance liquid chromatography–ultraviolet method for the measurement of nilotinib in plasma from patients with cancer. After protein precipitation extraction with acetonitrile, nilotinib and rilpivirine were separated using isocratic elution on a Tracer Excel 120 ODS C18 column using a mobile phase consisting of a mixture of potassium dihydrogen phosphate-buffered solution (pH 5.5; 0.037 M)–methanol–acetonitrile (45:45:10, v/v/v), pumped at a flow rate of 1.7 mL·min−1. A wavelength of 254 nm was selected for the quantification of the analyte and the internal standard (IS). The technique was validated following the guidelines for the validation of analytical methods of regulatory agencies (Food and Drug Administration (FDA) and the European Medicines Agency (EMA)). Linearity was established in a concentration range between 125 and 7000 ng/mL. The detection limit was 90 ng/mL, and the lower limit of quantification was 125 ng/mL. For all concentrations in the calibration curve, the intraday and interday coefficients of variation were less than 4.1%. Median recovery of nilotinib from plasma was ≥65.1% (±21.4%). The method described is sensitive, selective, reproducible, and rapid, and can be used for the accurate determination of nilotinib in human plasma for pharmacokinetics studies and for therapeutic drug monitoring (TDM) of nilotinib in routine clinical practice.
Collapse
|
12
|
Dalle Fratte C, Gagno S, Roncato R, Polesel J, Zanchetta M, Buzzo M, Posocco B, De Mattia E, Borsatti R, Puglisi F, Foltran L, Guardascione M, Buonadonna A, Cecchin E, Toffoli G. CYP2D6 and CYP2C8 pharmacogenetics and pharmacological interactions to predict imatinib plasmatic exposure in GIST patients. Br J Clin Pharmacol 2023; 89:1089-1098. [PMID: 36178950 DOI: 10.1111/bcp.15551] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/14/2022] [Accepted: 09/22/2022] [Indexed: 11/30/2022] Open
Abstract
AIMS Patients on treatment with oral fixed dose imatinib are frequently under- or overexposed to the drug. We investigated the association between the gene activity score (GAS) of imatinib-metabolizing cytochromes (CYP3A4, CYP3A5, CYP2D6, CYP2C9, CYP2C19, CYP2C8) and imatinib and nor-imatinib exposure. We also investigated the impact of concurrent drug-drug-interactions (DDIs) on the association between GAS and imatinib exposure. METHODS Serial plasma samples were collected from 33 GIST patients treated with imatinib 400 mg daily within a prospective clinical trial. Imatinib and nor-imatinib Ctrough were quantified by liquid chromatography with tandem mass spectrometry (LC-MS/MS). Genetic polymorphisms with a functional impact on imatinib-metabolizing cytochromes were identified and a GAS was calculated for each gene. A DDI-adjusted GAS was also generated. RESULTS Imatinib and nor-imatinib Ctrough were measured in 161 plasma samples. CYP2D6 GAS and metabolizer status based on genotype were associated with imatinib and (imatinib + nor-imatinib) Ctrough . CYP2D6 poor and intermediate metabolizers were predicted to have a lower nor-imatinib/imatinib metabolic ratio than normal metabolizers (0.197 and 0.193 vs. 0.247, P = .0205), whereas CYP2C8*3 carriers had a higher ratio than CYP2C8*1/*1 patients (0.263 vs. 0.201, P = .0220). CYP2C9 metabolizer status was inversely related to the metabolic ratio with an effect probably driven by the linkage disequilibrium between CYP2C9*2 and CYP2C8*3. The CYP2D6 DDI-adjusted GAS was still predictive of imatinib exposure. CONCLUSIONS These findings highlight that CYP2D6 plays a major role in imatinib pharmacokinetics, but other players (i.e., CYP2C8) may influence imatinib exposure. These findings could drive the selection of patients more susceptible to imatinib under- or overexposure who could be candidates for personalized treatment and intensified monitoring strategies.
Collapse
Affiliation(s)
- Chiara Dalle Fratte
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Sara Gagno
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Rossana Roncato
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Jerry Polesel
- Unit of Cancer Epidemiology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Martina Zanchetta
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Mauro Buzzo
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Bianca Posocco
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Elena De Mattia
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Rachele Borsatti
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Fabio Puglisi
- Department of Medical Oncology, Unit of Medical Oncology and Cancer Prevention, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy.,Department of Medicine, University of Udine, Udine, Italy
| | - Luisa Foltran
- Department of Medical Oncology, Unit of Medical Oncology and Cancer Prevention, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Michela Guardascione
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy.,Department of Medical Oncology, Unit of Medical Oncology and Cancer Prevention, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Angela Buonadonna
- Department of Medical Oncology, Unit of Medical Oncology and Cancer Prevention, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Erika Cecchin
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| |
Collapse
|
13
|
Roušarová J, Šíma M, Slanař O. Therapeutic Drug Monitoring of Protein Kinase Inhibitors in Breast Cancer Patients. Prague Med Rep 2021; 122:243-256. [PMID: 34924102 DOI: 10.14712/23362936.2021.22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Protein kinase inhibitors (PKIs) represent up-to-date therapeutic approach in breast cancer treatment. Although cancer is a rapidly progressive disease, many substances, including PKIs, are usually used at fixed doses without regard to each patient's individuality. Therapeutic drug monitoring (TDM) is a tool that allows individualization of therapy based on drug plasma levels. For TDM conduct, exposure-response relationships of drug substances are required. The pharmacokinetic data and exposure-response evidence supporting the use of TDM for 6 PKIs used in breast cancer treatment, one of the most common female tumour diseases, are discussed in this review.
Collapse
Affiliation(s)
- Jaroslava Roušarová
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.
| | - Martin Šíma
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Ondřej Slanař
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| |
Collapse
|
14
|
Developing a Nationwide Infrastructure for Therapeutic Drug Monitoring of Targeted Oral Anticancer Drugs: The ON-TARGET Study Protocol. Cancers (Basel) 2021; 13:cancers13246281. [PMID: 34944899 PMCID: PMC8699239 DOI: 10.3390/cancers13246281] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 12/28/2022] Open
Abstract
Simple Summary Relationships between drug concentrations in blood and efficacy and/or toxicity have been reported for up to 80% of oral anticancer drugs (OADs). Most OADs exhibit highly variable drug concentrations at the approved dose. This may result in a significant proportion of patients with suboptimal drug concentrations. Therapeutic Drug Monitoring (TDM), which is dose optimization based on measured drug concentrations, can be used to personalize drug dosing with the overall goal to improve the benefit-risk ratio of anticancer drug treatment. The ON-TARGET study aims to investigate the feasibility of TDM in patients receiving either axitinib or cabozantinib for the treatment of renal-cell carcinoma with the main objective to improve severe tyrosine kinase inhibitor associated toxicity. Additionally, the feasibility of volumetric absorptive microsampling (VAMS), a novel minimally invasive and easy to handle blood sampling technique, for TDM sample collection is investigated. Abstract Exposure-efficacy and/or exposure-toxicity relationships have been identified for up to 80% of oral anticancer drugs (OADs). Usually, OADs are administered at fixed doses despite their high interindividual pharmacokinetic variability resulting in large differences in drug exposure. Consequently, a substantial proportion of patients receive a suboptimal dose. Therapeutic Drug Monitoring (TDM), i.e., dosing based on measured drug concentrations, may be used to improve treatment outcomes. The prospective, multicenter, non-interventional ON-TARGET study (DRKS00025325) aims to investigate the potential of routine TDM to reduce adverse drug reactions in renal cell carcinoma patients receiving axitinib or cabozantinib. Furthermore, the feasibility of using volumetric absorptive microsampling (VAMS), a minimally invasive and easy to handle blood sampling technique, for sample collection is examined. During routine visits, blood samples are collected and sent to bioanalytical laboratories. Venous and VAMS blood samples are collected in the first study phase to facilitate home-based capillary blood sampling in the second study phase. Within one week, the drug plasma concentrations are measured, interpreted, and reported back to the physician. Patients report their drug intake and toxicity using PRO-CTCAE-based questionnaires in dedicated diaries. Ultimately, the ON-TARGET study aims to develop a nationwide infrastructure for TDM for oral anticancer drugs.
Collapse
|
15
|
Fahmy A, Hopkins AM, Sorich MJ, Rowland A. Evaluating the utility of therapeutic drug monitoring in the clinical use of small molecule kinase inhibitors: a review of the literature. Expert Opin Drug Metab Toxicol 2021; 17:803-821. [PMID: 34278936 DOI: 10.1080/17425255.2021.1943357] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction: Orally administered small molecule kinase inhibitors (KI) are a key class of targeted anti-cancer medicines that have contributed substantially to improved survival outcomes in patients with advanced disease. Since the introduction of KIs in 2001, there has been a building body of evidence that the benefit derived from these drugs may be further enhanced by individualizing dosing on the basis of concentration.Areas covered: This review considers the rationale for individualized KI dosing and the requirements for robust therapeutic drug monitoring (TDM). Current evidence supporting TDM-guided KI dosing is presented and critically evaluated, and finally potential approaches to address translational challenges for TDM-guided KI dosing and alternate approaches to support individualization of KI dosing are discussed.Expert opinion: Intuitively, the individualization of KI dosing through an approach such as TDM-guided dosing has great potential to enhance the effectiveness and tolerability of these drugs. However, based on current literature evidence it is unrealistic to propose that TDM-guided KI dosing should be routinely implemented into clinical practice.
Collapse
Affiliation(s)
- Alia Fahmy
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Ashley M Hopkins
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Michael J Sorich
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Andrew Rowland
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
16
|
Panetta JC, Campagne O, Gartrell J, Furman W, Stewart CF. Pharmacokinetically guided dosing of oral sorafenib in pediatric hepatocellular carcinoma: A simulation study. Clin Transl Sci 2021; 14:2152-2160. [PMID: 34060723 PMCID: PMC8604221 DOI: 10.1111/cts.13069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/05/2021] [Accepted: 04/04/2021] [Indexed: 02/01/2023] Open
Abstract
Sorafenib improves outcomes in adult hepatocellular carcinoma; however, hand foot skin reaction (HFSR) is a dose limiting toxicity of sorafenib that limits its use. HFSR has been associated with sorafenib systemic exposure. The objective of this study was to use modeling and simulation to determine whether using pharmacokinetically guided dosing to achieve a predefined sorafenib target range could reduce the rate of HFSR. Sorafenib steady‐state exposures (area under the concentration curve from 0 to 12‐h [AUC0–>12 h]) were simulated using published sorafenib pharmacokinetics at either a fixed dosage (90 mg/m2/dose) or a pharmacokinetically guided dose targeting an AUC0–>12 h between 20 and 55 h µg/ml. Dosages were either rounded to the nearest quarter of a tablet (50 mg) or capsule (10 mg). A Cox proportional hazard model from a previously published study was used to quantify HFSR toxicity. Simulations showed that in‐target studies increased from 50% using fixed doses with tablets to 74% using pharmacokinetically guided dosing with capsules. The power to observe at least 4 of 6 patients in the target range increased from 33% using fixed dosing with tablets to 80% using pharmacokinetically guided with capsules. The expected HFSR toxicity rate decreased from 22% using fixed doses with tablets to 16% using pharmacokinetically guided dosing with capsules. The power to observe less than 6 of 24 studies with HFSR toxicity increased from 51% using fixed dosing with tablets to 88% using pharmacokinetically guided with capsules. Our simulations provide the rationale to use pharmacokinetically guided sorafenib dosing to maintain effective exposures that potentially improve tolerability in pediatric clinical trials.
Collapse
Affiliation(s)
- John C Panetta
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Olivia Campagne
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jessica Gartrell
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Wayne Furman
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Clinton F Stewart
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
17
|
Therapeutic Drug Monitoring of Targeted Anticancer Protein Kinase Inhibitors in Routine Clinical Use: A Critical Review. Ther Drug Monit 2021; 42:33-44. [PMID: 31479043 DOI: 10.1097/ftd.0000000000000699] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Therapeutic response to oral targeted anticancer protein kinase inhibitors (PKIs) varies widely between patients, with insufficient efficacy of some of them and unacceptable adverse reactions of others. There are several possible causes for this heterogeneity, such as pharmacokinetic (PK) variability affecting blood concentrations, fluctuating medication adherence, and constitutional or acquired drug resistance of cancer cells. The appropriate management of oncology patients with PKI treatments thus requires concerted efforts to optimize the utilization of these drug agents, which have probably not yet revealed their full potential. METHODS An extensive literature review was performed on MEDLINE on the PK, pharmacodynamics, and therapeutic drug monitoring (TDM) of PKIs (up to April 2019). RESULTS This review provides the criteria for determining PKIs suitable candidates for TDM (eg, availability of analytical methods, observational PK studies, PK-pharmacodynamics relationship analysis, and randomized controlled studies). It reviews the major characteristics and limitations of PKIs, the expected benefits of TDM for cancer patients receiving them, and the prerequisites for the appropriate utilization of TDM. Finally, it discusses various important practical aspects and pitfalls of TDM for supporting better implementation in the field of cancer treatment. CONCLUSIONS Adaptation of PKIs dosage regimens at the individual patient level, through a rational TDM approach, could prevent oncology patients from being exposed to ineffective or unnecessarily toxic drug concentrations in the era of personalized medicine.
Collapse
|
18
|
Demlová R, Turjap M, Peš O, Kostolanská K, Juřica J. Therapeutic Drug Monitoring of Sunitinib in Gastrointestinal Stromal Tumors and Metastatic Renal Cell Carcinoma in Adults-A Review. Ther Drug Monit 2021; 42:20-32. [PMID: 31259881 DOI: 10.1097/ftd.0000000000000663] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Sunitinib is an inhibitor of multiple receptor tyrosine kinases and is a standard-of-care treatment for advanced and metastatic renal cell carcinoma and a second-line treatment in locally advanced inoperable and metastatic gastrointestinal stromal tumors. A fixed dose of the drug, however, does not produce a uniform therapeutic outcome in all patients, and many face adverse effects and/or toxicity. One of the possible causes of the interindividual variability in the efficacy and toxicity response is the highly variable systemic exposure to sunitinib and its active metabolite. This review aims to summarize all available clinical evidence of the treatment of adult patients using sunitinib in approved indications, addressing the necessity to introduce proper and robust therapeutic drug monitoring (TDM) of sunitinib and its major metabolite, N-desethylsunitinib. METHODS The authors performed a systematic search of the available scientific literature using the PubMed online database. The search terms were "sunitinib" AND "therapeutic drug monitoring" OR "TDM" OR "plasma levels" OR "concentration" OR "exposure." The search yielded 520 journal articles. In total, 447 publications were excluded because they lacked sufficient relevance to the reviewed topic. The remaining 73 articles were, together with currently valid guidelines, thoroughly reviewed. RESULTS There is sufficient evidence confirming the concentration-efficacy and concentration-toxicity relationship in the indications of gastrointestinal stromal tumors and metastatic renal clear-cell carcinoma. For optimal therapeutic response, total (sunitinib + N-desethylsunitinib) trough levels of 50-100 ng/mL serve as a reasonable target therapeutic range. To avoid toxicity, the total trough levels should not exceed 100 ng/mL. CONCLUSIONS According to the current evidence presented in this review, a TDM-guided dose modification of sunitinib in selected groups of patients could provide a better treatment outcome while simultaneously preventing sunitinib toxicity.
Collapse
Affiliation(s)
- Regina Demlová
- Department of Pharmacology, Medical Faculty, Masaryk University, Brno
| | - Miroslav Turjap
- Department of Clinical Pharmacy, University Hospital Ostrava, Ostrava
| | - Ondřej Peš
- Department of Biochemistry, Medical Faculty, Masaryk University
| | | | - Jan Juřica
- Department of Pharmacology, Medical Faculty, Masaryk University, Masaryk Memorial Cancer Institute; and.,Department of Human Pharmacology and Toxicology, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| |
Collapse
|
19
|
Mueller-Schoell A, Groenland SL, Scherf-Clavel O, van Dyk M, Huisinga W, Michelet R, Jaehde U, Steeghs N, Huitema ADR, Kloft C. Therapeutic drug monitoring of oral targeted antineoplastic drugs. Eur J Clin Pharmacol 2021; 77:441-464. [PMID: 33165648 PMCID: PMC7935845 DOI: 10.1007/s00228-020-03014-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE This review provides an overview of the current challenges in oral targeted antineoplastic drug (OAD) dosing and outlines the unexploited value of therapeutic drug monitoring (TDM). Factors influencing the pharmacokinetic exposure in OAD therapy are depicted together with an overview of different TDM approaches. Finally, current evidence for TDM for all approved OADs is reviewed. METHODS A comprehensive literature search (covering literature published until April 2020), including primary and secondary scientific literature on pharmacokinetics and dose individualisation strategies for OADs, together with US FDA Clinical Pharmacology and Biopharmaceutics Reviews and the Committee for Medicinal Products for Human Use European Public Assessment Reports was conducted. RESULTS OADs are highly potent drugs, which have substantially changed treatment options for cancer patients. Nevertheless, high pharmacokinetic variability and low treatment adherence are risk factors for treatment failure. TDM is a powerful tool to individualise drug dosing, ensure drug concentrations within the therapeutic window and increase treatment success rates. After reviewing the literature for 71 approved OADs, we show that exposure-response and/or exposure-toxicity relationships have been established for the majority. Moreover, TDM has been proven to be feasible for individualised dosing of abiraterone, everolimus, imatinib, pazopanib, sunitinib and tamoxifen in prospective studies. There is a lack of experience in how to best implement TDM as part of clinical routine in OAD cancer therapy. CONCLUSION Sub-therapeutic concentrations and severe adverse events are current challenges in OAD treatment, which can both be addressed by the application of TDM-guided dosing, ensuring concentrations within the therapeutic window.
Collapse
Affiliation(s)
- Anna Mueller-Schoell
- Dept. of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Berlin, Germany
- Graduate Research Training Program, PharMetrX, Berlin/Potsdam, Germany
| | - Stefanie L Groenland
- Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Oliver Scherf-Clavel
- Institute of Pharmacy and Food Chemistry, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Madelé van Dyk
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Wilhelm Huisinga
- Institute of Mathematics, University of Potsdam, Potsdam, Germany
| | - Robin Michelet
- Dept. of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Berlin, Germany
| | - Ulrich Jaehde
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Bonn, Bonn, Germany
| | - Neeltje Steeghs
- Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
- Department of Clinical Pharmacy, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | - Charlotte Kloft
- Dept. of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Berlin, Germany.
| |
Collapse
|
20
|
Hohenegger M. Pharmacokinetic considerations in geriatric cancer patients. MEMO - MAGAZINE OF EUROPEAN MEDICAL ONCOLOGY 2021; 14:11-14. [DOI: 10.1007/s12254-020-00668-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/11/2020] [Indexed: 08/30/2023]
Abstract
SummaryPharmacological anticancer therapy in elderly people has to account for pharmacokinetic aspects in view of age-related changes in organ function and disease-related alterations. Age-related changes in organ function might still be physiological and have to be discriminated from concomitant diseases and their pharmacotherapy. Although efficacy is retained with pharmacological anticancer therapies in elderly patients, plasma drug concentrations and the incidence of adverse reactions often increase. Thus, altered organ function in elderly will be reviewed with respect to clinically relevant outcomes. Furthermore, possible consequences of therapeutic drug monitoring will be discussed focusing on novel targeted therapies with small molecules. Examples of therapeutic drug monitoring during targeted therapies may represent an easy tool to overcome the individual pharmacokinetic situation of elderly cancer patients and may contribute to enhanced safety, when implemented in clinical routine.
Collapse
|
21
|
Aghai F, Zimmermann S, Kurlbaum M, Jung P, Pelzer T, Klinker H, Isberner N, Scherf-Clavel O. Development and validation of a sensitive liquid chromatography tandem mass spectrometry assay for the simultaneous determination of ten kinase inhibitors in human serum and plasma. Anal Bioanal Chem 2020; 413:599-612. [PMID: 33155133 PMCID: PMC7644392 DOI: 10.1007/s00216-020-03031-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/02/2020] [Accepted: 10/27/2020] [Indexed: 11/29/2022]
Abstract
A liquid chromatography tandem mass spectrometry method for the analysis of ten kinase inhibitors (afatinib, axitinib, bosutinib, cabozantinib, dabrafenib, lenvatinib, nilotinib, osimertinib, ruxolitinib, and trametinib) in human serum and plasma for the application in daily clinical routine has been developed and validated according to the US Food and Drug Administration and European Medicines Agency validation guidelines for bioanalytical methods. After protein precipitation of plasma samples with acetonitrile, chromatographic separation was performed at ambient temperature using a Waters XBridge® Phenyl 3.5 μm (2.1 × 50 mm) column. The mobile phases consisted of water-methanol (9:1, v/v) with 10 mM ammonium bicarbonate as phase A and methanol-water (9:1, v/v) with 10 mM ammonium bicarbonate as phase B. Gradient elution was applied at a flow rate of 400 μL/min. Analytes were detected and quantified using multiple reaction monitoring in electrospray ionization positive mode. Stable isotopically labeled compounds of each kinase inhibitor were used as internal standards. The acquisition time was 7.0 min per run. All analytes and internal standards eluted within 3.0 min. The calibration curves were linear over the range of 2–500 ng/mL for afatinib, axitinib, bosutinib, lenvatinib, ruxolitinib, and trametinib, and 6–1500 ng/mL for cabozantinib, dabrafenib, nilotinib, and osimertinib (coefficients of correlation ≥ 0.99). Validation assays for accuracy and precision, matrix effect, recovery, carryover, and stability were appropriate according to regulatory agencies. The rapid and sensitive assay ensures high throughput and was successfully applied to monitor concentrations of kinase inhibitors in patients. Graphical abstract ![]()
Collapse
Affiliation(s)
- Fatemeh Aghai
- Department of Internal Medicine II, University of Würzburg Medical Center, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Sebastian Zimmermann
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Max Kurlbaum
- Department of Internal Medicine I, University of Würzburg Medical Center, Oberdürrbacher Str. 6, 97080, Würzburg, Germany.,Core Unit Clinical Mass Spectrometry, University of Würzburg Medical Center, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Pius Jung
- Department of Internal Medicine I, Division of Pneumonology, University of Würzburg Medical Center, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Theo Pelzer
- Department of Internal Medicine I, Division of Pneumonology, University of Würzburg Medical Center, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Hartwig Klinker
- Department of Internal Medicine II, University of Würzburg Medical Center, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Nora Isberner
- Department of Internal Medicine II, University of Würzburg Medical Center, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Oliver Scherf-Clavel
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074, Würzburg, Germany.
| |
Collapse
|
22
|
Labeur TA, Hofsink Q, Takkenberg RB, van Delden OM, Mathôt RAA, Schinner R, Malfertheiner P, Amthauer H, Schütte K, Basu B, Kuhl C, Mayerle J, Ricke J, Klümpen HJ. The value of sorafenib trough levels in patients with advanced hepatocellular carcinoma - a substudy of the SORAMIC trial. Acta Oncol 2020; 59:1028-1035. [PMID: 32366155 DOI: 10.1080/0284186x.2020.1759826] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Background: Sorafenib for advanced hepatocellular carcinoma (HCC) is dose adjusted by toxicity. Preliminary studies have suggested an association between plasma concentrations of sorafenib and its main metabolite (M2) and clinical outcomes. This study aimed to validate these findings and establish target values for sorafenib trough concentrations.Methods: Patients with advanced HCC were prospectively recruited within a multicenter phase II study (SORAMIC). Patients with blood samples available at trough level were included for this pharmacokinetic (PK) substudy. Trough plasma concentrations of sorafenib and its main metabolite (M2) were associated with sorafenib-related toxicity and overall survival (OS).Results: Seventy-four patients were included with a median OS of 19.7 months (95% CI 16.1-23.3). Patients received sorafenib for a median of 51 weeks (IQR 27-62) and blood samples were drawn after a median of 25 weeks (IQR 10-42). Patients had a median trough concentration of 3217 ng/ml (IQR 2166-4526) and 360 ng/ml (IQR 190-593) with coefficients of variation of 65% and 146% for sorafenib and M2, respectively. Patients who experienced severe sorafenib-related toxicity received a lower average daily dose (551 vs 730 mg/day, p = .003), but showed no significant differences in sorafenib (3298 vs 2915 ng/ml, p = .442) or M2 trough levels (428 vs 283 ng/ml, p = .159). Trough levels of sorafenib or M2 showed no significant association with OS.Conclusions: In patients with advanced HCC treated with sorafenib, the administered dose, trough levels of sorafenib or M2, and clinical outcomes were poorly correlated. Toxicity-adjusted dosing remains the standard for sorafenib treatment.
Collapse
Affiliation(s)
- Tim A. Labeur
- Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Quincy Hofsink
- Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - R. Bart Takkenberg
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Otto M. van Delden
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Ron A. A. Mathôt
- Hospital Pharmacy, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Regina Schinner
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | | | - Holger Amthauer
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Kerstin Schütte
- Department of Internal Medicine and Gastroenterology, Niels-Stensen-Kliniken, Marienhospital Osnabrück, Osnabrück, Germany
| | - Bristi Basu
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Christiane Kuhl
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Julia Mayerle
- Department of Medicine II, University Hospital, LMU Munich, Munich, Germany
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Heinz-Josef Klümpen
- Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Castellano D, Pablo Maroto J, Benzaghou F, Taguieva N, Nguyen L, Clary DO, Jonasch E. Exposure-response modeling of cabozantinib in patients with renal cell carcinoma: Implications for patient care. Cancer Treat Rev 2020; 89:102062. [DOI: 10.1016/j.ctrv.2020.102062] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 01/17/2023]
|
24
|
Ferrer F, Fanciullino R, Milano G, Ciccolini J. Towards Rational Cancer Therapeutics: Optimizing Dosing, Delivery, Scheduling, and Combinations. Clin Pharmacol Ther 2020; 108:458-470. [PMID: 32557660 DOI: 10.1002/cpt.1954] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/30/2020] [Indexed: 12/16/2022]
Abstract
The current trend to personalize anticancer therapies mostly relies on selecting the best drug or combination of drugs to achieve optimal efficacy in patients. In addition to the comprehensive genetic and molecular knowledge of each tumor before choosing the drugs to be given, there is probably much room left for improvement by further personalizing the very modes by which the drugs are given, once they have been carefully selected. In particular, shifting from standard dosing to tailored dosing should help in maintaining drug exposure levels in the right therapeutic window, thus ensuring that the efficacy/toxicity balance is optimal. This paper covers the current knowledge regarding pharmacokinetic/pharmacodynamic relationships of anticancer agents, from decades-old cytotoxics to the latest immune checkpoint inhibitors, the most frequent sources for long-neglected interpatient variability impacting on drug exposure levels, and what could be done to achieve real personalized medicine in oncology such as implementing therapeutic drug monitoring with adaptive dosing strategies or using model-driven modalities for personalized dosing and scheduling.
Collapse
Affiliation(s)
- Florent Ferrer
- SMARTc Unit, CRCM Inserm U1068, Aix Marseille Univ and APHM, Marseille, France
| | | | - Gérard Milano
- Onco-Pharmacology Unit, Centre Antoine Lacassagne, Nice, France
| | - Joseph Ciccolini
- SMARTc Unit, CRCM Inserm U1068, Aix Marseille Univ and APHM, Marseille, France
| |
Collapse
|
25
|
Hopkins AM, Menz BD, Wiese MD, Kichenadasse G, Gurney H, McKinnon RA, Rowland A, Sorich MJ. Nuances to precision dosing strategies of targeted cancer medicines. Pharmacol Res Perspect 2020; 8:e00625. [PMID: 32662214 PMCID: PMC7358594 DOI: 10.1002/prp2.625] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 01/05/2023] Open
Abstract
Selecting the dose of a targeted cancer medicine that is most appropriate for a specific individual is a rational approach to maximize therapeutic outcomes and minimize toxicity. There are many different options for optimizing the dose of targeted cancer medicines and the purpose of this review is to provide a comprehensive comparison of the main options explored in prospective studies. Precision initial dose selection of targeted cancer therapies has been minimally explored to date; however, concentration, toxicity, and therapeutic outcome markers are used to guide on-therapy dose adaption of targeted cancer therapies across several medicines and cancers. While a specific concentration, toxicity, or therapeutic outcome marker commonly dominates an investigated precision on-therapy dose adaption strategy, greater attention to simultaneously account for exposure, toxicity, therapeutic outcomes, disease status, time since treatment initiation and patient preferences are required for optimal patient outcomes. To enable successful implementation of precision dosing strategies for targeted cancer medicines into clinical practice, future prospective studies aiming to develop strategies should consider these elements in their design.
Collapse
Affiliation(s)
- Ashley M. Hopkins
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Bradley D. Menz
- Division of PharmacySouthern Adelaide Local Health Network, Flinders Medical CentreAdelaideSouth AustraliaAustralia
| | - Michael D. Wiese
- School of Pharmacy and Medical SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Ganessan Kichenadasse
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Howard Gurney
- Department of Medical OncologyWestmead HospitalSydneyNew South WalesAustralia
| | - Ross A. McKinnon
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Andrew Rowland
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Michael J. Sorich
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| |
Collapse
|
26
|
Ackland SP, Michael M, Souza P, Martin JH, Clarke SJ, Francis K, Karapetis CS, Gurney H. Science and art of
anticancer
drug dosing: nine steps to personalised therapy. Intern Med J 2020. [DOI: 10.1111/imj.14948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Stephen P. Ackland
- Faculty of Health and MedicineUniversity of Newcastle Newcastle New South Wales Australia
- Department of Medical OncologyCalvary Mater Newcastle Newcastle New South Wales Australia
- Hunter Medical Research Institute Newcastle New South Wales Australia
| | - Michael Michael
- Department of Medical OncologyPeter MacCallum Cancer Centre Melbourne Victoria Australia
| | - Paul Souza
- School of MedicineUniversity of Wollongong Wollongong New South Wales Australia
| | - Jennifer H. Martin
- Hunter Medical Research Institute Newcastle New South Wales Australia
- Centre for Human Drug Research, School of Medicine and Public Health, Faculty of Health and MedicineUniversity of Newcastle Newcastle New South Wales Australia
| | - Stephen J. Clarke
- Department of Medical OncologyRoyal North Shore Hospital Sydney New South Wales Australia
- Sydney Medical SchoolUniversity of Sydney Sydney New South Wales Australia
| | - Kay Francis
- Medical Oncology Group of Australia Sydney New South Wales Australia
| | - Christos S. Karapetis
- Medical Oncology Group of Australia Sydney New South Wales Australia
- Flinders Centre for Innovation in CancerFlinders University and Flinders Medical Centre Adelaide South Australia Australia
| | - Howard Gurney
- Crown Princess Mary Cancer CentreWestmead Hospital Sydney New South Wales Australia
- Faculty of Medicine and Health SciencesMacquarie University Sydney New South Wales Australia
| |
Collapse
|
27
|
Lyashchenko AK, Cremers S. On precision dosing of oral small molecule drugs in oncology. Br J Clin Pharmacol 2020; 87:263-270. [PMID: 32621551 DOI: 10.1111/bcp.14454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/06/2020] [Accepted: 06/17/2020] [Indexed: 12/17/2022] Open
Abstract
Personalization of oral small molecule anticancer drug doses based on individual patient blood drug levels, also known as therapeutic drug monitoring (TDM), has the potential to significantly improve the effectiveness of treatment by maximizing drug efficacy and minimize toxicity. However, this option has not yet been widely embraced by the oncology community. Some reasons for this include increased logistical complexity of dose individualization, the lack of clinical laboratories that measure small molecule drug concentrations in support of patient care, and the lack of reimbursement of costs. However, the main obstacle may be the lack of studies clearly demonstrating that monitoring of oral small molecule anticancer drug levels actually improves clinical outcomes. Without unequivocal evidence in support of TDM-guided dose individualization, especially demonstration of improved survival with TDM in randomized controlled trials, wide acceptance of this approach by oncologists and reimbursement by insurance companies is unlikely, and patients may continue to suffer as a result of receiving incorrect drug doses. This article reviews the current status of TDM of oral small molecule drugs in oncology and intends to provide strategic insights into the design of studies for evaluating the utility of TDM in this clinical context.
Collapse
Affiliation(s)
- Alex K Lyashchenko
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Serge Cremers
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
28
|
Kovacevic T, Kovacevic SV, Stanetic M, Kovacevic P, Miljkovic B. Impact of pharmacist's intervention on decreasing erlotinib interactions in the treatment of lung cancer patients in low resource settings. J Oncol Pharm Pract 2020; 27:350-358. [PMID: 32349642 DOI: 10.1177/1078155220921545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND This study aimed to demonstrate that having clinical pharmacist as a member of oncology team in low and middle income countries might lead to significant reduction in the number of erlotinib interactions in the treatment of non-small cell lung cancer patients. METHODS A group of 44 patients was labeled as intervention group and they were analyzed prospectively in the period from 1 January 2017 to 1 May 2018 during clinical pharmacist's participation in regular weekly multidisciplinary oncology team meetings. The control group consisted of 44 out of 110 patients treated with erlotinib before the involvement of a clinical pharmacist in oncology team, match paired with 44 patients in intervention group. RESULTS Clinically significant interactions were identified in two-thirds of studied patients (57 out of 88). Most drug interactions, 38%, potentially result in decrease of serum concentration of erlotinib. Clinical pharmacist provided therapy modification suggestions for 32 out of 44 (72.72%) patients in the intervention group, most of which were accepted by doctors. In the intervention group, there were significantly less clinically significant interactions compared to the control group (10 versus 24, p = 0.002). Progression-free survival was significantly longer in the pharmacist's intervention group (p = 0.001). CONCLUSIONS Clinical pharmacist's intervention led to significant decrease in erlotinib interactions which may result in treatment optimization of lung cancer patients.
Collapse
Affiliation(s)
- Tijana Kovacevic
- Pharmacy Department, University Clinical Centre Republic of Srpska, Banja Luka, Bosnia and Herzegovina.,Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Sandra Vezmar Kovacevic
- Department of Pharmacokinetics and Clinical Pharmacy, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Mirko Stanetic
- Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina.,Lung Disease Clinic, University Clinical Centre Republic of Srpska, Banja Luka, Bosnia and Herzegovina
| | - Pedja Kovacevic
- Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina.,Medical Intensive Care Unit, University Clinical Centre Republic of Srpska, Banja Luka, Bosnia and Herzegovina
| | - Branislava Miljkovic
- Department of Pharmacokinetics and Clinical Pharmacy, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
29
|
Corn PG, Zhang M, Nogueras-Gonzalez GM, Xiao L, Zurita AJ, Subudhi SK, Tu SM, Aparicio AM, Coarfa C, Rajapakshe K, Huang S, Navone NM, Lin SH, Wang G, Ramachandran S, Titus MA, Panaretakis T, Gallick GE, Efstathiou E, Troncoso P, Logothetis C. A Phase II Study of Cabozantinib and Androgen Ablation in Patients with Hormone-Naïve Metastatic Prostate Cancer. Clin Cancer Res 2020; 26:990-999. [PMID: 31941830 DOI: 10.1158/1078-0432.ccr-19-2389] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/23/2019] [Accepted: 11/08/2019] [Indexed: 02/03/2023]
Abstract
PURPOSE Cabozantinib, an oral inhibitor of c-MET/VEGFR2 signaling, improved progression-free survival (mPFS) but not overall survival (OS) in metastatic castrate-resistant prostate cancer. We evaluated cabozantinib plus androgen deprivation therapy (ADT) in hormone-naïve metastatic prostate cancer (HNMPCa). PATIENTS AND METHODS Patients received ADT plus cabozantinib starting at 60 mg daily. The primary endpoint was castrate-resistant PFS by radiographic criteria, clinical progression, or receipt of additional therapy. Secondary endpoints included OS, safety, radiographic responses, and biomarker modulation. RESULTS Sixty-two patients received treatment. With a median follow-up of 31.2 months, the mPFS was 16.1 months (95% CI, 14.6-22.7 months), and mOS was not reached. Reductions in PSA ≥ 90%, bone-specific alkaline phosphatase ≥ 50%, and urine N-telopeptides ≥ 50% occurred in 83%, 87%, and 86% of evaluable patients, respectively. Responses in bone scan and measurable disease were observed in 81% of and 90% of evaluable patients, respectively. Most common grade 3 adverse events were hypertension (19%), diarrhea (6%), and thromboembolic events (6%), and dose reductions occurred in 85% of patients. Analysis of baseline cytokine and angiogenic factors (CAFs) revealed that higher plasma concentrations of Lumican, CXCL5, CD25, and CD30 were associated with shorter PFS as was high tumor expression of pFGFR1. CONCLUSIONS Cabozantinib plus ADT has promising clinical activity in HNMPCa. CAF profiles and tissue markers suggest candidate prognostic and predictive markers of cabozantinib benefit and provide insights for rational therapy combinations.
Collapse
Affiliation(s)
- Paul G Corn
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Miao Zhang
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Lianchun Xiao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amado J Zurita
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sumit K Subudhi
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shi-Ming Tu
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ana M Aparicio
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Shixia Huang
- Department of Molecular and Cellular Oncology, Baylor College of Medicine, Houston, Texas
| | - Nora M Navone
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sue-Hwa Lin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Guocan Wang
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sumankalai Ramachandran
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark A Titus
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Theocharis Panaretakis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gary E Gallick
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eleni Efstathiou
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patricia Troncoso
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christopher Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
30
|
Effective quantification of 11 tyrosine kinase inhibitors and caffeine in human plasma by validated LC-MS/MS method with potent phospholipids clean-up procedure. Application to therapeutic drug monitoring. Talanta 2020; 208:120450. [DOI: 10.1016/j.talanta.2019.120450] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/30/2019] [Accepted: 10/04/2019] [Indexed: 12/11/2022]
|
31
|
Labeur TA, Achterbergh R, Takkenberg B, Van Delden O, Mathôt R, Klümpen H. Sorafenib for Patients with Hepatocellular Carcinoma and Child-Pugh B Liver Cirrhosis: Lessons Learned from a Terminated Study. Oncologist 2019; 25:e1274-e1279. [PMID: 31645371 PMCID: PMC7485346 DOI: 10.1634/theoncologist.2019-0718] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 09/15/2019] [Indexed: 12/22/2022] Open
Abstract
Lessons Learned Recruitment of patients with advanced hepatocellular carcinoma and Child‐Pugh B for sorafenib treatment and additional pharmacokinetic studies is challenging. Patients with Child‐Pugh B liver cirrhosis have high rates of cirrhosis‐related adverse events.
Background Few data are available on the pharmacokinetics (PK) of sorafenib in patients with advanced hepatocellular carcinoma (HCC) and Child‐Pugh B liver cirrhosis. This study aimed to explore the sorafenib PK and its relationship with efficacy and toxicity in these patients. Methods Patients with advanced HCC and Child‐Pugh B7‐8 liver function were prospectively recruited at a tertiary center. Adverse events (AEs), progression‐free survival (PFS), and overall survival (OS) were recorded. Patients received a starting dose of 200 b.i.d. with toxicity‐adjusted dose escalation to a target dose of 400 mg b.i.d. with PK sampling at fixed time points. Results Between May 2014 and March 2017, 12 patients were screened, of whom 7 progressed to a terminal stage during the screening (n = 6) or shortly after recruitment (n = 1). The five included patients had median PFS of 3.8 months (range, 1.7–10.8) and OS of 7.4 months (range, 1.7–25.8). Three patients had severe AEs and one patient had a partial response with an OS of 25.8 months. In 2017, the trial was aborted for lack of accrual. Conclusion Because of low accrual, no conclusion can be drawn on the sorafenib PK in patients with advanced HCC and Child‐Pugh B liver cirrhosis. The poor survival and frequent cirrhosis‐related AEs suggest limited benefit for most of these patients.
Collapse
Affiliation(s)
- Tim A. Labeur
- Department of Medical Oncology, Amsterdam University Medical Centers, University of AmsterdamMeibergdreef 9, AmsterdamThe Netherlands
- Cancer Center AmsterdamAmsterdamThe Netherlands
| | - Roos Achterbergh
- Hospital Pharmacy, Amsterdam University Medical Centers, University of AmsterdamMeibergdreef 9, AmsterdamThe Netherlands
| | - Bart Takkenberg
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, University of AmsterdamMeibergdreef 9, AmsterdamThe Netherlands
| | - Otto Van Delden
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, University of AmsterdamMeibergdreef 9, AmsterdamThe Netherlands
| | - Ron Mathôt
- Hospital Pharmacy, Amsterdam University Medical Centers, University of AmsterdamMeibergdreef 9, AmsterdamThe Netherlands
| | - Heinz‐Josef Klümpen
- Department of Medical Oncology, Amsterdam University Medical Centers, University of AmsterdamMeibergdreef 9, AmsterdamThe Netherlands
- Cancer Center AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
32
|
Ornstein MC, Pal SK, Wood LS, Tomer JM, Hobbs BP, Jia XS, Allman KD, Martin A, Olencki T, Davis NB, Gilligan TD, Mortazavi A, Rathmell WK, Garcia JA, Rini BI. Individualised axitinib regimen for patients with metastatic renal cell carcinoma after treatment with checkpoint inhibitors: a multicentre, single-arm, phase 2 study. Lancet Oncol 2019; 20:1386-1394. [PMID: 31427205 DOI: 10.1016/s1470-2045(19)30513-3] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/13/2019] [Accepted: 06/13/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Checkpoint inhibitor therapy is a standard of care for patients with metastatic renal cell carcinoma. Treatment options after checkpoint inhibitor therapy include vascular endothelial growth factor receptor (VEGF-R) tyrosine kinase inhibitors, although no prospective data regarding their use in this setting exist. Axitinib is a VEGF-R inhibitor with clinical data supporting increased activity with dose titration. We aimed to investigate the activity of dose titrated axitinib in patients with metastatic renal cell carcinoma who were previously treated with checkpoint inhibitor. METHODS We did a multicentre, phase 2 trial of axitinib given on an individualised dosing algorithm. Patients at least 18 years of age with histologically or cytologically confirmed locally recurrent or metastatic renal cell carcinoma with clear cell histology, a Karnofsky Performance Status of 70% or more, and measurable disease who received checkpoint inhibitor therapy as the most recent treatment were eligible. There was no limit on number of previous therapies received. Patients received oral axitinib at a starting dose of 5 mg twice daily with dose titration every 14 days in 1 mg increments (ie, 5 mg twice daily to 6 mg twice daily, up to 10 mg twice daily maximum dose) if there was no axitinib-related grade 2 or higher mucositis, diarrhoea, hand-foot syndrome, or fatigue. If one or more of these grade 2 adverse events occurred, axitinib was withheld for 3 days before the same dose was resumed. Dose reductions were made if recurrent grade 2 adverse events despite treatment breaks or grade 3-4 adverse events occurred. The primary outcome was progression-free survival. Analyses were done per protocol in all patients who received at least one dose of axitinib. Recruitment has been completed and the trial is ongoing. This trial is registered with ClincalTrials.gov, number NCT02579811. FINDINGS Between Jan 5, 2016 and Feb 21, 2018, 40 patients were enrolled and received at least one dose of study treatment. With a median follow-up of 8·7 months (IQR 3·7-14·2), the median progression-free survival was 8·8 months (95% CI 5·7-16·6). Fatigue (83%) and hypertension (75%) were the most common all-grade adverse events. The most common grade 3 adverse event was hypertension (24 patients [60%]). There was one (3%) grade 4 adverse event (elevated lipase) and no treatment-related deaths occurred. Serious adverse events that were likely related to therapy occurred in eight (20%) patients; the most common were dehydration (n=4) and diarrhoea (n=2). INTERPRETATION Individualised axitinib dosing in patients with metastatic renal cell inoma previously treated with checkpoint inhibitors did not meet the prespecified threshold for progression free survival, but these data show that this individualised titration scheme is feasible and has robust clinical activity. These prospective results warrant consideration of axitinib in this setting. FUNDING Pfizer.
Collapse
Affiliation(s)
- Moshe C Ornstein
- Department of Hematology and Medical Oncology, Cleveland Clinic-Taussig Cancer Institute, Cleveland, OH, USA.
| | - Sumanta K Pal
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Laura S Wood
- Department of Hematology and Medical Oncology, Cleveland Clinic-Taussig Cancer Institute, Cleveland, OH, USA
| | - Jackie M Tomer
- Department of Hematology and Medical Oncology, Cleveland Clinic-Taussig Cancer Institute, Cleveland, OH, USA
| | - Brian P Hobbs
- Department of Hematology and Medical Oncology, Cleveland Clinic-Taussig Cancer Institute, Cleveland, OH, USA
| | - Xuefei S Jia
- Department of Hematology and Medical Oncology, Cleveland Clinic-Taussig Cancer Institute, Cleveland, OH, USA
| | - Kimberly D Allman
- Department of Hematology and Medical Oncology, Cleveland Clinic-Taussig Cancer Institute, Cleveland, OH, USA
| | - Allison Martin
- Department of Hematology and Medical Oncology, Cleveland Clinic-Taussig Cancer Institute, Cleveland, OH, USA
| | - Thomas Olencki
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Nancy B Davis
- Division of Hematology and Oncology, Vanderbilt Ingram Cancer Center, Nashville, TN, USA
| | - Timothy D Gilligan
- Department of Hematology and Medical Oncology, Cleveland Clinic-Taussig Cancer Institute, Cleveland, OH, USA
| | - Amir Mortazavi
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - W Kimryn Rathmell
- Division of Hematology and Oncology, Vanderbilt Ingram Cancer Center, Nashville, TN, USA
| | - Jorge A Garcia
- Department of Hematology and Medical Oncology, Cleveland Clinic-Taussig Cancer Institute, Cleveland, OH, USA
| | - Brian I Rini
- Department of Hematology and Medical Oncology, Cleveland Clinic-Taussig Cancer Institute, Cleveland, OH, USA
| |
Collapse
|
33
|
Gougis P, Palmieri LJ, Funck-Brentano C, Paci A, Flippot R, Mir O, Coriat R. Major pitfalls of protein kinase inhibitors prescription: A review of their clinical pharmacology for daily use. Crit Rev Oncol Hematol 2019; 141:112-124. [PMID: 31276964 DOI: 10.1016/j.critrevonc.2019.06.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 12/13/2022] Open
Abstract
Protein kinase inhibitors (PKI) are a growing class of anticancer agents. They are prescribed with flat doses, and their oral administration is associated with interindividual variability in exposure. Patients can be over- or underexposed, due to numerous factors. We reviewed key pharmacokinetic concepts and mechanisms by which PKIs prescription could be altered. Challenging situations that could lead to increased toxicity or to therapeutic failure are described and recommendation for clinicians are proposed. Finally, the interest of therapeutic drug monitoring and indications for its use in daily practice is discussed.
Collapse
Affiliation(s)
- Paul Gougis
- Sorbonne Université, Faculty of Medicine, AP-HP, Pitié-Salpêtrière Hospital, Department of Pharmacology and Clinical Investigation Center, F-75013, Paris, France; CLIP² Galilée, Pitié-Salpêtrière Hospital, F-75013, Paris, France.
| | - Lola-Jade Palmieri
- AP-HP, Cochin Hospital, Department of gastroenterology and gastrointestinal oncology, Université Paris Descartes, Paris, France
| | - Christian Funck-Brentano
- Sorbonne Université, Faculty of Medicine, AP-HP, Pitié-Salpêtrière Hospital, Department of Pharmacology and Clinical Investigation Center, F-75013, Paris, France; INSERM, CIC-1421 and UMR ICAN 1166, Institute of Cardiometabolism and Nutrition (ICAN), F-75013, Paris, France
| | - Angelo Paci
- Service de Pharmacologie, Département de Biologie et Pathologie Médicales, Gustave Roussy et Université Paris Saclay, Villejuif, France
| | - Ronan Flippot
- Department of Medical Oncology, Gustave Roussy, 114 rue Edouard Vaillant, 94800, Villejuif, France
| | - Olivier Mir
- Department of Medical Oncology, Gustave Roussy, 114 rue Edouard Vaillant, 94800, Villejuif, France; Department of Ambulatory Care, Gustave Roussy Cancer Campus, Villejuif, France
| | - Romain Coriat
- AP-HP, Cochin Hospital, Department of gastroenterology and gastrointestinal oncology, Université Paris Descartes, Paris, France
| |
Collapse
|
34
|
Miao E, Seetharamu N, Sullivan K, Eng S, Lee CS. Impact of Tyrosine Kinase Inhibitor Starting Dose on Outcomes in Patients With Non-Small Cell Lung Cancer. J Pharm Pract 2019; 34:11-16. [PMID: 31167592 DOI: 10.1177/0897190019840596] [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] [Indexed: 12/22/2022]
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors (TKIs) can cause intolerable adverse events in patients with non-small cell lung cancer (NSCLC) and may be prescribed at a lower dose. OBJECTIVE Our objective was to analyze the starting doses of oral EGFR and ALK TKIs in patients diagnosed with NSCLC at our institution. METHODS We conducted a retrospective chart review with patients on EGFR and ALK TKIs for NSCLC. Patients were categorized into 2 groups: patients initiated on Food and Drug Administration (FDA) standard dose (SD) and patients initiated on a reduced dose (RD). Progression-free survival (PFS), overall survival (OS) and other treatment outcomes were compared between both groups. RESULTS Ninety patients were included for analysis. The median time-to-progression for the SD group (n = 67) and RD group (n = 23) were 13.4 months (95% confidence interval [CI]:8.9-15.6) and 15.1 months (95%CI: 5.6-21.5), respectively. Median time-to-death was not estimable for OS. The predicted OS probability at approximately 15 months post treatment initiation for the SD group and RD group was 81.8% and 80.5%, respectively. CONCLUSION Patients who initiated TKI therapy at a RD did not have different PFS and 15-month survival outcomes than patients who initiated TKI therapy at the FDA SD.
Collapse
Affiliation(s)
- Emily Miao
- College of Pharmacy and Health Sciences, 4131St John's University, Queens, NY, USA
| | - Nagashree Seetharamu
- Division of Medical Oncology and Hematology, Northwell Health Cancer Institute, Donald & Barbara Zucker School of Medicine at Hofstra/Northwell, Lake Success, NY, USA
| | - Kevin Sullivan
- Division of Medical Oncology and Hematology, Northwell Health Cancer Institute, Donald & Barbara Zucker School of Medicine at Hofstra/Northwell, Lake Success, NY, USA
| | - Stephen Eng
- College of Pharmacy and Health Sciences, 4131St John's University, Queens, NY, USA
| | - Chung-Shien Lee
- Division of Medical Oncology and Hematology, Northwell Health Cancer Institute, Donald & Barbara Zucker School of Medicine at Hofstra/Northwell, Lake Success, NY, USA.,Department of Clinical Health Professions, College of Pharmacy and Health Sciences, St John's University, Queens, NY, USA
| |
Collapse
|
35
|
Cheng W, Kletas V, Kollmannsberger C, de Lemos M. Survival outcomes associated with different sunitinib dosing regimens in metastatic renal cell carcinoma. J Oncol Pharm Pract 2019; 26:67-73. [DOI: 10.1177/1078155219837333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Standard dosing regimen of sunitinib for metastatic renal cell carcinoma consists of four weeks treatment followed by two weeks rest (intermittent dosing). Alternative regimens have been suggested, including continuous daily dosing (continuous dosing) and non-conventional dosing (non-conventional dosing: e.g. two weeks on/one week off, non-conventional dosing), to provide more individualized therapy with less toxicities. It is unclear whether non-standard sunitinib dosing affects survival outcomes. Patients Metastatic renal cell carcinoma patients treated with sunitinib between 1 July 2007 and 1 July 2011 at our institution. Methods Medical records and dispensing data were reviewed retrospectively to categorize sunitinib dosing as intermittent dosing, continuous dosing, or non-conventional dosing. Primary outcome was to compare overall survival associated with varying regimens, with secondary outcomes of progression-free survival and incidence of treatment discontinuation due to adverse effects. Results A total of 180 patients were identified. Most patients received intermittent dosing ( n = 120, 67%), followed by continuous dosing ( n = 32, 18%) and non-conventional dosing ( n = 28, 16%). Compared to intermittent dosing, continuous dosing was associated with similar overall survival (median 9 vs. 13 months, HR 0.67, 95% CI: 0.43–1.06, p = 0.088) while non-conventional dosing was associated with significantly longer overall survival (median 9 vs. 23 months, HR 0.55, 95% CI: 0.34–0.90, p = 0.016). Progression-free survival was significantly better for continuous dosing (median 4 vs. 9 months, HR 0.61, 95% CI: 0.40–0.94, p = 0.025) and non-conventional dosing (median 4 vs. 10 months, HR 0.61, 95% CI: 0.39–0.95, p = 0.03) when compared to intermittent dosing. Similar to prior sunitinib trials, a significant proportion of patients (20%) discontinued sunitinib therapy due to adverse effects. Conclusions Based on retrospective, real-world data, alternative sunitinib dosing regimens appear to be viable options for patients with metastatic renal cell carcinoma.
Collapse
|
36
|
Schuller Y, Gispen-de Wied C, Hollak CEM, Leufkens HGM, Stoyanova-Beninska V. Dose-Finding Studies Among Orphan Drugs Approved in the EU: A Retrospective Analysis. J Clin Pharmacol 2018; 59:229-244. [PMID: 30192386 PMCID: PMC6585723 DOI: 10.1002/jcph.1304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/22/2018] [Indexed: 11/21/2022]
Abstract
In the development process for new drugs, dose‐finding studies are of major importance. Absence of these studies may lead to failed phase 3 trials and delayed marketing authorization. In our study we investigated to what extent dose‐finding studies are performed in the case of orphan drugs for metabolic and oncologic indications. We identified all orphan drugs that were authorized until August 1, 2017. European Public Assessment Reports were used to extract the final dose used in the summary of product characteristics, involvement of healthy volunteers, study type, end points used, number of patients, number of doses, studies in special populations, and dose used for phase 3 studies. Each drug was checked for major objections and dose changes postmarketing. We included 49 orphan drugs, of which 28 were indicated for metabolic disorders and 21 for oncologic indications. Dose‐finding studies were performed in 32 orphan drugs, and studies in healthy volunteers in 26. The absence of dose‐finding studies was mostly due to the rarity of the disease. In this case the dose was determined based on factors such as animal studies or clinical experience. Dose‐related major objections were raised for 9 orphan drugs. Postmarketing dose‐finding studies were conducted in 18 orphan drugs, but dose changes were applied in only 2 drugs. In conclusion, dose‐finding studies in the case of metabolic and oncologic orphan drugs were conducted in the development programs of two thirds of orphan drugs. Dose‐finding studies performed postmarketing suggest that registered doses are not always optimal. It is thus important to perform more robust dose‐finding studies both pre‐ and postmarketing.
Collapse
Affiliation(s)
- Yvonne Schuller
- Department of Endocrinology and Metabolism, Academic Medical Center, Meibergdreef, Amsterdam, The Netherlands
| | - Christine Gispen-de Wied
- College ter Beoordeling Geneesmiddelen/Medicines Evaluation Board, Graadt van Roggenweg, Utrecht, The Netherlands
| | - Carla E M Hollak
- Department of Endocrinology and Metabolism, Academic Medical Center, Meibergdreef, Amsterdam, The Netherlands
| | - Hubertus G M Leufkens
- Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg, Utrecht, The Netherlands
| | - Violeta Stoyanova-Beninska
- College ter Beoordeling Geneesmiddelen/Medicines Evaluation Board, Graadt van Roggenweg, Utrecht, The Netherlands
| |
Collapse
|
37
|
Koldenhof JJ, Lankheet NAG, Steeghs N, Teunissen SCCM, Witteveen PO. Patient-reported outcome measures in a pharmacokinetic study with sunitinib, a prospective cohort study. Support Care Cancer 2018; 26:2641-2650. [PMID: 29464389 PMCID: PMC6018583 DOI: 10.1007/s00520-018-4075-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 01/29/2018] [Indexed: 01/02/2023]
Abstract
PURPOSE During treatment with tyrosine kinase inhibitors, such as sunitinib, patients experience treatment and/or disease-related symptoms. Although application of patient-reported outcome measures (PROMs) enhances early recognition of symptoms, early clinical trials are focused on symptom severity objectified by the Common Terminology Criteria for Adverse Events (CTCAE) in order to evaluate drug safety and to determine a personalized and/or safe dosage range. To gain insight into patient-reported symptoms in addition to healthcare professional-reported adverse events (AEs), a substudy was conducted in an ongoing pharmacokinetic-guided sunitinib dosing study. METHODS In patients for whom sunitinib was considered standard therapy or patients with advanced/metastatic tumors for whom no standard therapy was available, patient-reported symptoms and well-being besides healthcare professional-reported AEs were assessed. RESULTS Twenty-nine patients were included for analysis. Over 50% of them experienced a decreased well-being, caused by symptoms of mild and moderate intensity. Compared to healthcare professionals, all measured symptoms, with the exception of fatigue and vomiting, were reported statistically significantly more often by patients. CONCLUSIONS Application of PROMs in early clinical trials on personalized or individualized oral targeted anticancer agents is feasible and enhances early recognition of symptom burden due to multiple CTCAE grade 1-2 AEs, just as pro-active symptom management and effect evaluation of interventions performed. Application of PROMs in these trials might be clinically relevant in obtaining dose-limiting toxicities.
Collapse
Affiliation(s)
- J J Koldenhof
- Department of Medical Oncology, HP B02.225, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, Netherlands.
| | - N A G Lankheet
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, Netherlands
| | - N Steeghs
- Department of Medical Oncology and Clinical Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, Netherlands
| | - S C C M Teunissen
- Department of Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - P O Witteveen
- Department of Medical Oncology, HP B02.225, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, Netherlands
| |
Collapse
|
38
|
Correlation of plasma erlotinib trough concentration with skin rash in Chinese NSCLC patients harboring exon 19 deletion mutation. Cancer Chemother Pharmacol 2018; 82:551-559. [DOI: 10.1007/s00280-018-3642-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 07/04/2018] [Indexed: 02/06/2023]
|
39
|
Grassi P, Verzoni E, Ratta R, Porcu L, Prisciandaro M, Mennitto A, Calareso G, de Braud F, Procopio G. Does Dose Modification Affect Efficacy of First-Line Pazopanib in Metastatic Renal Cell Carcinoma? Drugs R D 2018; 17:461-467. [PMID: 28801802 PMCID: PMC5629143 DOI: 10.1007/s40268-017-0203-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background Pazopanib is a standard treatment for metastatic renal cell carcinoma (mRCC), and 800 mg/daily is considered the optimal dose. However, some patients require dose modification because of toxicity. Whether a reduced dose of pazopanib is as effective as the standard dose in achieving clinical benefit remains unclear. Objectives Our objective was to conduct a retrospective analysis to investigate the clinical effect of different therapeutic doses of first-line pazopanib in patients with mRCC. Methods Consecutive patients with mRCC treated with first-line pazopanib between 2011 and 2016 at the Istituto Nazionale Tumori of Milan were retrospectively analysed for demographics, response, outcomes, and toxicity. Three patient groups were compared: group 1 received the standard dose of 800 mg/day; group 2 started with 800 mg/day and then reduced the dose to 400 or 600 mg/day because of toxicity; and group 3 received a reduced starting dose of 400 or 600 mg/day because they had an Eastern Cooperative Oncology Group (ECOG) performance status (PS) of 2 and/or comorbidities. Results In total, 69 patients were evaluated: 34 in group 1, 19 in group 2, and 16 in group 3. After a median follow-up of 13.9 months (range 0.3–43.8), 27 (39.1%) patients had progressive disease (PD) and three (4.3%) patients had died. The incidence rate of PD or death per 100 person-months was 2.5 [95% confidence interval (CI) 0.6–4.4; hazard ratio (HR) 1] in group 1 and 3.9 (95% CI 0–14.3; HR 1.43) in the combined group (2 + 3). The discontinuation rate due to PD was 28% in group 1, 42% in group 2, and 44% in group 3. The objective response rate was 44, 11, and 19% in groups 1, 2, and 3, respectively. Conclusions Our results may suggest that patients with mRCC receiving a lower dose of first-line pazopanib might not have a meaningful progression-free survival advantage compared with those receiving a standard dose. These data highlight that proper management of treatment-related side effects may lead to optimal drug exposure.
Collapse
Affiliation(s)
- Paolo Grassi
- Oncologia Medica 1, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133, Milan, Italy.
| | - Elena Verzoni
- Oncologia Medica 1, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Raffaele Ratta
- Oncologia Medica 1, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Luca Porcu
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Michele Prisciandaro
- Oncologia Medica 1, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Alessia Mennitto
- Oncologia Medica 1, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Giuseppina Calareso
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo de Braud
- Oncologia Medica 1, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133, Milan, Italy
| | - Giuseppe Procopio
- Oncologia Medica 1, Fondazione IRCCS Istituto Nazionale Tumori, Via Venezian 1, 20133, Milan, Italy
| |
Collapse
|
40
|
Gao S, Tao Z, Zhou J, Wang Z, Yun Y, Li M, Zhang F, Chen W, Miao Y. One-Step Solid Extraction for Simultaneous Determination of Eleven Commonly Used Anticancer Drugs and One Active Metabolite in Human Plasma by HPLC-MS/MS. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2018; 2018:7967694. [PMID: 30046507 PMCID: PMC6036832 DOI: 10.1155/2018/7967694] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/07/2018] [Indexed: 06/08/2023]
Abstract
Therapeutic drug monitoring for anticancer drugs could timely reflect in vivo drug exposure, and it was a powerful tool for adjusting and maintaining drug concentration into a reasonable range, so that an enhanced efficacy and declined adverse reactions could be achieved. A liquid chromatography-tandem mass spectrometry method had been developed and fully validated for simultaneous determination of paclitaxel, docetaxel, vinblastine, vinorelbine, pemetrexed, carboplatin, etoposide, cyclophosphamide, ifosfamide, gemcitabine, irinotecan, and SN-38 (an active metabolite of irinotecan) in human plasma from cancer patients after intravenous drip of chemotherapy drugs. One-step solid-phase extraction was successfully applied using an Ostro sample preparation 96-well plate for plasma samples pretreated with acetonitrile containing 0.1% formic acid. Chromatographic separation was achieved on an Atlantis T3-C18 column (2.1 × 100 mm, 3.0 μm) with gradient elution using a mobile phase consisting of acetonitrile and 10 mM ammonium acetate plus 0.1% formic acid in water, and the flow rate was 0.25 mL/min. The Agilent G6410A triple quadrupole liquid chromatography-mass spectrometry system was operated under the multiple reaction monitoring mode with an electrospray ionization in the positive mode. Linear range was 25.0-2500.0 ng for paclitaxel, 10.0-1000.0 ng for docetaxel and SN-38, 100.0-10000.0 ng for vinorelbine and pemetrexed, 10.0-10000.0 ng for vinblastine and irinotecan, 1.0-1000.0 ng for cyclophosphamide and ifosfamide, 50.0-5000.0 ng for carboplatin, etoposide, and gemcitabine. Linearity coefficients of correlation were >0.99 for all analytes. The intraday and interday accuracy and precision of the method were within ±15.0% and less than 15%. The mean recovery and matrix effect as well as stability of all the analytes ranged from 56.2% to 98.9% and 85.2% to 101.3% as well as within ±15.0%. This robust and efficient method was successfully applied to implement therapeutic drug monitoring for cancer patients in clinical application.
Collapse
Affiliation(s)
- Shouhong Gao
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Zhengbo Tao
- Department of Orthopaedics, First Affiliated Hospital, China Medical University, 155 Nan Jing Bei Street, Shenyang, Liaoning 110001, China
| | - Jingya Zhou
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Zhipeng Wang
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Yunlei Yun
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Mingming Li
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Feng Zhang
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Wansheng Chen
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Yejun Miao
- Department of Psychiatry, Ankang Hospital, Ningbo, Zhejiang 315000, China
| |
Collapse
|
41
|
Morotti M, Dass PH, Harris AL, Lord S. Pharmacodynamic and Pharmacokinetic Markers For Anti-angiogenic Cancer Therapy: Implications for Dosing and Selection of Patients. Eur J Drug Metab Pharmacokinet 2018; 43:137-153. [PMID: 29019020 DOI: 10.1007/s13318-017-0442-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Angiogenesis is integral to tumour growth and invasion, and is a key target for cancer therapeutics. However, for many of the licensed indications, only a modest clinical benefit has been observed for both monoclonal antibody and small-molecule tyrosine kinase inhibitor anti-angiogenic therapy. Pre-clinical and clinical studies have attempted to evaluate circulating, imaging, genomic, pharmacokinetic, and pharmacodynamic markers that may aid both the selection of patients for treatment and define dosing. Correct dosing is likely to be critical in the context of vascular normalization to allow better delivery of concomitant anti-cancer therapy and novel imaging techniques hold much promise in the early evaluation of pharmacodynamic response to improve efficacy.
Collapse
Affiliation(s)
- Matteo Morotti
- Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, UK.
- Department of Gynaecology Oncology, University of Oxford, Oxford, UK.
- Department of Oncology, Churchill Hospital, University of Oxford, Oxford, OX3 9DU, UK.
| | - Prashanth Hari Dass
- Department of Oncology, Churchill Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Adrian L Harris
- Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, UK
- Department of Oncology, Churchill Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Simon Lord
- Hypoxia and Angiogenesis Group, Cancer Research UK Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, UK
- Department of Oncology, Churchill Hospital, University of Oxford, Oxford, OX3 9DU, UK
| |
Collapse
|
42
|
Maráz A, Cserháti A, Uhercsák G, Szilágyi É, Varga Z, Révész J, Kószó R, Varga L, Kahán Z. Dose escalation can maximize therapeutic potential of sunitinib in patients with metastatic renal cell carcinoma. BMC Cancer 2018; 18:296. [PMID: 29544452 PMCID: PMC5856318 DOI: 10.1186/s12885-018-4209-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 03/09/2018] [Indexed: 12/24/2022] Open
Abstract
Background In patients with metastatic renal cell cancer, based on limited evidence, increased sunitinib exposure is associated with better outcome. The survival and toxicity data of patients receiving individualized dose escalated sunitinib therapy as compared to standard management were analyzed in this study. Methods From July 2013, the data of metastatic renal cell cancer patients with slight progression but still a stable disease according to RECIST 1.1 criteria treated with an escalated dose of sunitinib (first level: 62.5 mg/day in 4/2 or 2 × 2/1 scheme, second level: 75 mg/day in 4/2 or 2 × 2/1 scheme) were collected prospectively. Regarding characteristics, outcome, and toxicity data, an explorative retrospective analysis of the register was carried out, comparing treatments after and before July 1, 2013 in the study (selected patients for escalated dose) and control (standard dose) groups, respectively. Results The study involved 103 patients receiving sunitinib therapy with a median overall and progression free survival of 25.36 ± 2.62 and 14.2 ± 3.22 months, respectively. Slight progression was detected in 48.5% of them. First and second-level dose escalation were indicated in 18.2% and 4.1% of patients, respectively. The dosing scheme was modified in 22.2%. The median progression free survival (39.7 ± 5.1 vs 14.2 ± 1.3 months (p = 0.037)) and the overall survival (57.5 ± 10.7 vs 27.9 ± 2.5 months (p = 0.044)) were significantly better in the study group (with dose escalation) than in the control group. Patients with nephrectomy and lower Memorial Sloan Kettering Cancer Center (MSKCC) scores showed more favorable outcomes. After dose escalation, the most common adverse events were worsening or development of fatigue, hypertension, stomatitis, and weight loss of over 10%. Conclusions Escalation of sunitinib dosing in selected patients with metastatic renal cell cancer, especially in case of slight progression, based on tolerable toxicity is safe and improves outcome. Dose escalation in 12.5 mg steps may be recommended for properly educated patients.
Collapse
Affiliation(s)
- Anikó Maráz
- Department of Oncotherapy, University of Szeged, Korányi fasor 12, Szeged, H-6720, Hungary.
| | - Adrienn Cserháti
- Department of Oncotherapy, University of Szeged, Korányi fasor 12, Szeged, H-6720, Hungary
| | - Gabriella Uhercsák
- Department of Oncotherapy, University of Szeged, Korányi fasor 12, Szeged, H-6720, Hungary
| | - Éva Szilágyi
- Department of Oncotherapy, University of Szeged, Korányi fasor 12, Szeged, H-6720, Hungary
| | - Zoltán Varga
- Department of Oncotherapy, University of Szeged, Korányi fasor 12, Szeged, H-6720, Hungary
| | - János Révész
- Institute of Radiotherapy and Clinical Oncology, Borsod County Hospital and University Academic Hospital, Szentpéteri Kapu 72-76, Miskolc, H-3526, Hungary
| | - Renáta Kószó
- Department of Oncotherapy, University of Szeged, Korányi fasor 12, Szeged, H-6720, Hungary
| | - Linda Varga
- Department of Oncotherapy, University of Szeged, Korányi fasor 12, Szeged, H-6720, Hungary
| | - Zsuzsanna Kahán
- Department of Oncotherapy, University of Szeged, Korányi fasor 12, Szeged, H-6720, Hungary
| |
Collapse
|
43
|
Béchade D, Chakiba C, Desjardin M, Bécouarn Y, Fonck M. [Hepatotoxicity of tyrosine kinase inhibitors: Mechanisms involved and practical implications]. Bull Cancer 2018; 105:290-298. [PMID: 29471963 DOI: 10.1016/j.bulcan.2017.11.015] [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: 09/07/2017] [Revised: 11/27/2017] [Accepted: 11/29/2017] [Indexed: 12/26/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) are used for the targeted treatment of solid cancers. TKIs produce a variable incidence of liver adverse events (5-25%) which can progress to severe liver injury in a minority of patients if treatment is maintained despite ongoing injury. This risk requires careful patient management to maintain treatment benefit without harm. This review highlights the various mechanisms of idiosyncratic hepatotoxicity, the formation of reactive metabolites and how this leads to toxicity. These critical events depend of the drug-specific characteristics of each TKI and the patient risk factors, especially genetic characterization. With improved understanding of the mechanisms leading to hepatotoxicity, several strategies have been adopted to prevent or treat this side effect. Recommendations on liver function liver test monitoring have been proposed according to each TKI.
Collapse
Affiliation(s)
- Dominique Béchade
- Institut Bergonié, département d'oncologie médicale, 229, cours de l'Argonne, 33076 Bordeaux cedex, France.
| | - Camille Chakiba
- Institut Bergonié, département d'oncologie médicale, 229, cours de l'Argonne, 33076 Bordeaux cedex, France
| | - Marie Desjardin
- Institut Bergonié, département d'oncologie médicale, 229, cours de l'Argonne, 33076 Bordeaux cedex, France
| | - Yves Bécouarn
- Institut Bergonié, département d'oncologie médicale, 229, cours de l'Argonne, 33076 Bordeaux cedex, France
| | - Marianne Fonck
- Institut Bergonié, département d'oncologie médicale, 229, cours de l'Argonne, 33076 Bordeaux cedex, France
| |
Collapse
|
44
|
Hassan W, Chitcholtan K, Sykes P, Garrill A. Ascitic fluid from advanced ovarian cancer patients compromises the activity of receptor tyrosine kinase inhibitors in 3D cell clusters of ovarian cancer cells. Cancer Lett 2018; 420:168-181. [PMID: 29432847 DOI: 10.1016/j.canlet.2018.02.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 01/31/2018] [Accepted: 02/06/2018] [Indexed: 12/18/2022]
Abstract
Ovarian cancer patients in the advanced stages of the disease show clinical ascites, which is associated with a poor prognosis. There is limited understanding of the effect of ascitic fluid on ovarian cancer cells and their response to anticancer drugs. We investigated the antitumour effects of EGFR/Her-2 (canertinib) and c-Met (PHA665752) inhibitors in a 3D cell model of three ovarian cancer lines. Single and combined inhibitor treatments affected cell growth of OVCAR-5 and SKOV-3 cell lines but not OV-90 cell line. Growth reduction was correlated with the down expression of PCNA, EGFR, HER-2, c-MET, ERK and AKT and their phosphorylation status in cells in growth factor supplemented media. However, these effects were not re-producible in OVCAR-5 and SKOV-3 cell lines when they were exposed to ascitic fluid obtained from three ovarian cancer patients. Serum albumin and protein components in the ascitic fluids may reduce the cellular uptake of the inhibitors.
Collapse
Affiliation(s)
- Wafaa Hassan
- School of Biological Sciences, University of Canterbury, Private Bag 4800, Christchurch, 8041, New Zealand.
| | - Kenny Chitcholtan
- Gynaecological Oncology Research Group, Department of Obstetrics and Gynaecology, University of Otago, Christchurch Women's Hospital, 2 Riccarton Avenue, Christchurch, 8011, New Zealand.
| | - Peter Sykes
- Gynaecological Oncology Research Group, Department of Obstetrics and Gynaecology, University of Otago, Christchurch Women's Hospital, 2 Riccarton Avenue, Christchurch, 8011, New Zealand.
| | - Ashley Garrill
- School of Biological Sciences, University of Canterbury, Private Bag 4800, Christchurch, 8041, New Zealand.
| |
Collapse
|
45
|
Schmidinger M, Danesi R, Jones R, McDermott R, Pyle L, Rini B, Négrier S. Individualized dosing with axitinib: rationale and practical guidance. Future Oncol 2017; 14:861-875. [PMID: 29264944 DOI: 10.2217/fon-2017-0455] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Axitinib is a potent, selective, vascular endothelial growth factor receptor inhibitor with demonstrated efficacy as second-line treatment for metastatic renal cell carcinoma. Analyses of axitinib drug exposures have demonstrated high interpatient variability in patients receiving the 5 mg twice-daily (b.i.d.) starting dose. Clinical criteria can be used to assess whether individual patients may benefit further from dose modifications, based on their safety and tolerability data. This review provides practical guidance on the 'flexible dosing' method, to help physicians identify who would benefit from dose escalations, dose reductions or continuation with manageable toxicity at the 5 mg b.i.d. dose. This flexible approach allows patients to achieve the best possible outcomes without compromising safety.
Collapse
Affiliation(s)
- Manuela Schmidinger
- Clinical Division of Oncology, Department of Medicine I & Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Romano Danesi
- Department of Clinical & Experimental Medicine, University of Pisa, Pisa, Italy
| | - Robert Jones
- Institute of Cancer Sciences, University of Glasgow, The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Ray McDermott
- Department of Medical Oncology, St Vincent's University Hospital & The Adelaide & Meath Hospital, Dublin, Ireland
| | - Lynda Pyle
- Renal Cancer Unit, Department of Medicine, Royal Marsden Hospital, London, UK
| | - Brian Rini
- Department of Hematology & Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA
| | - Sylvie Négrier
- Medical Oncology Department, University of Lyon, Centre Léon Bérard, Lyon, France
| |
Collapse
|
46
|
Development and Validation of a Simultaneous Quantification Method of 14 Tyrosine Kinase Inhibitors in Human Plasma Using LC-MS/MS. Ther Drug Monit 2017; 39:43-54. [PMID: 27861317 DOI: 10.1097/ftd.0000000000000357] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND A sensitive liquid chromatography coupled with tandem mass spectrometry (MS/MS) method for the analysis in a small volume of plasma of 14 tyrosine kinase inhibitors currently used (imatinib, dasatinib, ibrutinib, ponatinib, trametinib, sunitinib, cobimetinib, dabrafenib, erlotinib, lapatinib, nilotinib, bosutinib, sorafenib, and vemurafenib) has been developed and validated. This multianalyte liquid chromatography coupled with MS/MS assay is of interest for anticancer drug combination therapy. METHODS After a simple protein precipitation of plasma samples, the chromatographic separation was performed using an ultra performance liquid chromatography system coupled with MS/MS in a positive ionization mode. The mobile phase consisted of a gradient elution of 10 mmol/L formate ammonium buffer containing 0.1% (vol/vol) formic acid (phase A) and acetonitrile with 0.1% (vol/vol) formic acid (phase B) at a flow rate of 300 μL/min. RESULTS The analysis time was 5.0 minutes per run, and all analytes and internal standard eluted within 1.45-1.79 minutes. The calibration curves were linear over the range from 1 to 500 ng/mL for bosutinib, cobimetinib, dasatinib, ibrutinib, and trametinib, from 5 to 500 ng/mL for ponatinib and sunitinib; from 50 to 2500 ng/mL for lapatinib; from 750 to 100,000 ng/mL for vemurafenib, and from 10 to 2500 ng/mL for dabrafenib, erlotinib, imatinib, nilotinib, and sorafenib, with coefficients of correlation above 0.99 for all analytes. The intra- and interday imprecisions were below 14.36%. CONCLUSIONS This method was successfully applied to therapeutic drug monitoring in clinical practice.
Collapse
|
47
|
Revisiting Dosing Regimen Using Pharmacokinetic/Pharmacodynamic Mathematical Modeling: Densification and Intensification of Combination Cancer Therapy. Clin Pharmacokinet 2017; 55:1015-25. [PMID: 26946136 DOI: 10.1007/s40262-016-0374-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Controlling effects of drugs administered in combination is particularly challenging with a densified regimen because of life-threatening hematological toxicities. We have developed a mathematical model to optimize drug dosing regimens and to redesign the dose intensification-dose escalation process, using densified cycles of combined anticancer drugs. A generic mathematical model was developed to describe the main components of the real process, including pharmacokinetics, safety and efficacy pharmacodynamics, and non-hematological toxicity risk. This model allowed for computing the distribution of the total drug amount of each drug in combination, for each escalation dose level, in order to minimize the average tumor mass for each cycle. This was achieved while complying with absolute neutrophil count clinical constraints and without exceeding a fixed risk of non-hematological dose-limiting toxicity. The innovative part of this work was the development of densifying and intensifying designs in a unified procedure. This model enabled us to determine the appropriate regimen in a pilot phase I/II study in metastatic breast patients for a 2-week-cycle treatment of docetaxel plus epirubicin doublet, and to propose a new dose-ranging process. In addition to the present application, this method can be further used to achieve optimization of any combination therapy, thus improving the efficacy versus toxicity balance of such a regimen.
Collapse
|
48
|
Lucas CJ, Martin JH. Pharmacokinetic-Guided Dosing of New Oral Cancer Agents. J Clin Pharmacol 2017; 57 Suppl 10:S78-S98. [DOI: 10.1002/jcph.937] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 04/10/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Catherine J. Lucas
- Discipline of Clinical Pharmacology, School of Medicine and Public Health; University of Newcastle; New South Wales Australia
| | - Jennifer H. Martin
- Discipline of Clinical Pharmacology, School of Medicine and Public Health; University of Newcastle; New South Wales Australia
| |
Collapse
|
49
|
Allard M, Rousseau B, Cardoso E, Bellesoeur A, Blanchet B. Suivi thérapeutique pharmacologique à la 4 e journée de pharmacologie des anti-tumoraux. Bull Cancer 2017; 104:800-806. [DOI: 10.1016/j.bulcan.2017.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/11/2017] [Accepted: 06/13/2017] [Indexed: 10/19/2022]
|
50
|
Lankheet NAG, Desar IME, Mulder SF, Burger DM, Kweekel DM, van Herpen CML, van der Graaf WTA, van Erp NP. Optimizing the dose in cancer patients treated with imatinib, sunitinib and pazopanib. Br J Clin Pharmacol 2017; 83:2195-2204. [PMID: 28500677 DOI: 10.1111/bcp.13327] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 05/08/2017] [Accepted: 05/10/2017] [Indexed: 01/11/2023] Open
Abstract
AIM Fixed dose oral tyrosine kinase inhibitors imatinib, sunitinib and pazopanib show a high interpatient variability in plasma exposure. A relationship between plasma exposure and treatment outcome has been established, which supports the rationale for dose optimization of these drugs. The aim of this study was to monitor how many patients reached adequate trough levels after therapeutic drug monitoring-based dose optimization in daily practice. METHODS A cohort study was performed in patients treated with imatinib, sunitinib or pazopanib of whom follow-up drug levels were measured between August 2012 and April 2016. Patients' characteristics were collected by reviewing electronic patient records. Drug levels were measured using high-performance liquid chromatography coupled with tandem mass spectrometry and trough levels were estimated using a predefined algorithm. Dose interventions were proposed based on trough levels. RESULTS In total, 396 trough levels were determined in 109 patients. Median sample frequency per patient was 3. During the first measurement only 38% of patients showed trough levels within the predefined target ranges despite standard dosing; 52% of the patients showed drug levels below and 10% above the target range. In 35 out of 41 patients (85%) dose interventions led to adequate trough levels. Eventually, 64% of the total cohort reached adequate trough levels. CONCLUSIONS Dose optimization proved an effective tool to reach adequate trough levels in patients treated with imatinib, sunitinib and pazopanib. The percentage of patients with adequate trough levels increased from 38 to 64%. Therapeutic drug monitoring may add to the improvement of efficacy and reduction of toxicity and costs of these treatments.
Collapse
Affiliation(s)
- Nienke A G Lankheet
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ingrid M E Desar
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Sasja F Mulder
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - David M Burger
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, Netherlands
| | - Dinemarie M Kweekel
- Department of Pharmacy, Leiden University Medical Center, Leiden, Netherlands
| | - Carla M L van Herpen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Winette T A van der Graaf
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, Netherlands.,The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Nielka P van Erp
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|