1
|
Jiang D, Song Z, Ma Y, Zhang X, Bing H, Xiong X, Hu Y, Dong F, Zhao R. Development, validation, and clinical application of LC-MS/MS method for simultaneous determination of ibrutinib, zanubrutinib, orelabrutinib, acalabrutinib, and their active metabolites in patients with B-cell lymphoma. Anal Bioanal Chem 2025; 417:821-834. [PMID: 39702674 DOI: 10.1007/s00216-024-05701-2] [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: 10/16/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Bruton's tyrosine kinase inhibitors (BTKis) exhibit significant interindividual pharmacokinetics, making therapeutic drug monitoring (TDM) a promising approach for personalized therapy. However, simultaneous quantification of multiple BTKis poses technical challenges. A unified protocol for BTKis detection would be clinically desirable. Herein, we developed and validated a novel LC-MS/MS method for the simultaneous analysis of four BTKis including ibrutinib (IBR), zanubrutinib (ZAN), orelabrutinib (ORE), and acalabrutinib (ACB) and active metabolite of IBR and ACB (DIH and ACBM, respectively) in human plasma. The samples were prepared by liquid-liquid extraction using tert-butyl methyl ether. Ibrutinb-d4 (IS) was used as an internal standard. Chromatographic separation was obtained on an XBridge C18 column and connected to an LC-30AD system coupled to an API 4000+ mass spectrometer. The mobile phase comprised 10 mM ammonium acetate containing 0.1% formic acid and acetonitrile containing 0.1% formic acid. The optimized multiple reaction monitoring transitions of m/z 441.4 → 138.3, 475.4 → 304.2, 472.5 → 455.5, 428.3 → 411.5, 466.1 → 372.2, 482.2 → 388.4, and 445.5 → 142.5 were selected to inspect IBR, DIH, ZAN, ORE, ACB, ACBM, and IS, respectively. The method exhibited linearity from 1 to 1000 ng/mL (r > 0.99) for all analytes, with intra-day and inter-day precision of 1.8 to 9.7% and accuracy below 15%. Recovery ranged from 90.4 to 113.6%, and matrix effect varied from 89.3 to 111.0%. All compounds demonstrated stability under relevant conditions. Application of the method to 57 blood samples from 18 patients demonstrated high interpatient variability, with ORE plasma concentrations ranging from 25.6 to 89.9%. The validated LC-MS/MS method provides a feasible, specific, and rapid approach for quantification of BTKis in clinical settings. Simultaneous determination of four BTKis and their metabolites in a single extraction process and chromatographic run reduces analysis time, cost, and resources. The observed variability among individuals highlights the value of TDM for personalized treatment.
Collapse
Affiliation(s)
- Dan Jiang
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China
- Therapeutic Drug Monitoring and Clinical Toxicology Center, Peking University, Beijing, 100191, China
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Zaiwei Song
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China
- Therapeutic Drug Monitoring and Clinical Toxicology Center, Peking University, Beijing, 100191, China
| | - Yi Ma
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China
- Therapeutic Drug Monitoring and Clinical Toxicology Center, Peking University, Beijing, 100191, China
| | - Xu Zhang
- Department of Hematology, Peking University Third Hospital, Beijing, 100191, China
| | - Hao Bing
- Clinical Research Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
| | - Xin Xiong
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China
- Therapeutic Drug Monitoring and Clinical Toxicology Center, Peking University, Beijing, 100191, China
| | - Yang Hu
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China
- Therapeutic Drug Monitoring and Clinical Toxicology Center, Peking University, Beijing, 100191, China
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Fei Dong
- Department of Hematology, Peking University Third Hospital, Beijing, 100191, China.
| | - Rongsheng Zhao
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China.
- Therapeutic Drug Monitoring and Clinical Toxicology Center, Peking University, Beijing, 100191, China.
| |
Collapse
|
2
|
Rousseau A, Géraud A, Geiss R, Farcet A, Spano JP, Hamy AS, Gougis P. Safety of solid oncology drugs in older patients: a narrative review. ESMO Open 2024; 9:103965. [PMID: 39481329 PMCID: PMC11567126 DOI: 10.1016/j.esmoop.2024.103965] [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: 07/09/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
The older population represents ∼50%-60% of the population of newly diagnosed patients with cancer. Due to physiological and pathological aging and the increased presence of comorbidities and frailty factors, this population is at higher risk of serious toxicity from anticancer drugs and, consequently, often under-treated. Despite the complexity of these treatments, a good knowledge of the pharmacology of anticancer drugs and potentially risky situations can limit the emergence of potentially lethal toxicities in this population. This review focuses on optimizing systemic oncology treatments for older patients, emphasizing the unique characteristics of each therapeutic class and the necessity for a precautionary approach for this vulnerable population.
Collapse
Affiliation(s)
- A Rousseau
- Department of Medical Oncology, Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | - A Géraud
- Department of Medical Oncology, Institut Paoli-Calmette, Marseille, France
| | - R Geiss
- Department of Medical Oncology, Institut Curie, Université Paris Cité, Paris, France
| | - A Farcet
- Department of Medical Oncology, Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | - J-P Spano
- Department of Medical Oncology, Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | - A-S Hamy
- Department of Medical Oncology, Institut Curie, Université Paris Cité, Paris, France; Residual Tumor and Response to Treatment, RT2Lab, INSERM, U932 Cancer & Immunity, Institut Curie, Université Paris Sciences Lettres, Paris, France
| | - P Gougis
- Department of Medical Oncology, Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France; Residual Tumor and Response to Treatment, RT2Lab, INSERM, U932 Cancer & Immunity, Institut Curie, Université Paris Sciences Lettres, Paris, France; Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Assistance Publique - Hôpitaux de Paris (AP-HP), Centre d'Investigation Clinique (CIC-1901), Pharmacology Department, Pitié-Salpêtrière Hospital, Paris, France.
| |
Collapse
|
3
|
Géraud A, Combarel D, Funck-Brentano C, Beaulieu Q, Zahr N, Broutin S, Spano JP, Massard C, Besse B, Gougis P. A Score to Predict the Clinical Usefulness of Therapeutic Drug Monitoring: Application to Oral Molecular Targeted Therapies in Cancer. Clin Pharmacol Ther 2024; 116:678-689. [PMID: 38389482 DOI: 10.1002/cpt.3193] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 01/22/2024] [Indexed: 02/24/2024]
Abstract
Therapeutic drug monitoring (TDM) involves measuring and interpreting drug concentrations in biological fluids to adjust drug dosages. In onco-hematology, TDM guidelines for oral molecular targeted therapies (oMTTs) are varied. This study evaluates a quantitative approach with a score to predict the clinical usefulness of TDM for oMTTs. We identified key parameters for an oMTT's suitability for TDM from standard TDM recommendations. We gathered oMTT pharmacological data, which covered exposure variability (considering pharmacokinetic (PK) impact of food and proton pump inhibitors), technical intricacy (PK linearity and active metabolites), efficacy (exposure-response relationship), and safety (maximum tolerated dose, and exposure-safety relationship). To assess the validity and the relevance of the score and define relevant thresholds, we evaluated molecules with prospective validation or strong recommendations for TDM, both in oncology and in other fields. By September 1, 2021, the US Food and Drug Administration (FDA) approved 67 oMTTs for onco-hematological indications. Scores ranged from 15 (acalabrutinib) to 80 (sunitinib) with an average of 48.3 and a standard deviation of 15.6. Top scorers included sunitinib, sorafenib, cabozantinib, nilotinib, and abemaciclib. Based on scores, drugs were categorized into low (< 40), intermediate (≥ 40 and < 60), and high (≥ 60) relevance for TDM. Notably, negative controls generally scored around or under 40, whereas positive controls had a high score across different indications. In this work, we propose a quantitative and reproducible score to compare the potential usefulness of TDM for oMTTs. Future guidelines should prioritize the TDM for molecules with the highest score.
Collapse
Affiliation(s)
- Arthur Géraud
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Early Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
- Paris-Saclay University, Orsay, France
| | - David Combarel
- Pharmacology Department, Gustave Roussy, Villejuif, France
- Faculty of Pharmacy, Paris-Saclay University, Chatenay-Malabry, France
| | - Christian Funck-Brentano
- Department of Pharmacology and Clinical Investigation Center (CIC-1901), AP-HP.Sorbonne Université, Pitié-Salpêtrière Hospital, INSERM, CIC-1901 and UMR-S 1166, Sorbonne Université Médecine, Paris, France
| | - Quentin Beaulieu
- Department of Pharmacology and Clinical Investigation Center (CIC-1901), AP-HP.Sorbonne Université, Pitié-Salpêtrière Hospital, INSERM, CIC-1901 and UMR-S 1166, Sorbonne Université Médecine, Paris, France
| | - Noël Zahr
- Department of Pharmacology and Clinical Investigation Center (CIC-1901), AP-HP.Sorbonne Université, Pitié-Salpêtrière Hospital, INSERM, CIC-1901 and UMR-S 1166, Sorbonne Université Médecine, Paris, France
| | - Sophie Broutin
- Pharmacology Department, Gustave Roussy, Villejuif, France
| | - Jean-Philippe Spano
- Oncology Department, APHP-Sorbonne Université, Cancer Institute (IUC), Paris, France
- INSERM, UMRS 1136, Paris, France
| | - Christophe Massard
- Early Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
- Centre Eugène Marquis, Rennes, France
| | - Benjamin Besse
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Paris-Saclay University, Orsay, France
| | - Paul Gougis
- Department of Pharmacology and Clinical Investigation Center (CIC-1901), AP-HP.Sorbonne Université, Pitié-Salpêtrière Hospital, INSERM, CIC-1901 and UMR-S 1166, Sorbonne Université Médecine, Paris, France
- Oncology Department, APHP-Sorbonne Université, Cancer Institute (IUC), Paris, France
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, INSERM, U932 Immunity and Cancer, Curie Institute, Université Paris, Paris, France
| |
Collapse
|
4
|
Combarel D, Geraud A, Gougis P. Reply to: "Therapeutic Drug Monitoring of Oral Oncology Drugs: Another Example of Maslow's Hammer" and "Therapeutic Drug Monitoring of Oral Oncology Drugs: Finding the Right Nails". Clin Pharmacol Ther 2024; 116:506-507. [PMID: 38970295 DOI: 10.1002/cpt.3367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 06/23/2024] [Indexed: 07/08/2024]
Affiliation(s)
- David Combarel
- Pharmacology Department, Gustave Roussy, Villejuif, France
- Faculty of Pharmacy, Paris-Saclay University, Chatenay-Malabry, France
| | - Arthur Geraud
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Paul Gougis
- Department of Pharmacology and Clinical Investigation Center (CIC-1901), Pitié-Salpêtrière Hospital, AP-HP Sorbonne Université, Paris, France
- INSERM, CIC-1901 and UMR-S 1166, Sorbonne Université Médecine, Paris, France
- Oncology Department, APHP-Sorbonne Université, Cancer Institute (IUC), Paris, France
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, INSERM, U932 Immunity and Cancer, Curie Institute, Université Paris, Paris, France
| |
Collapse
|
5
|
Posocco B, Zanchetta M, Orleni M, Gagno S, Montico M, Peruzzi E, Roncato R, Gerratana L, Corsetti S, Puglisi F, Toffoli G. Therapeutic Monitoring of Palbociclib, Ribociclib, Abemaciclib, M2, M20, and Letrozole in Human Plasma: A Novel LC-MS/MS Method. Ther Drug Monit 2024; 46:485-493. [PMID: 38366332 PMCID: PMC11232939 DOI: 10.1097/ftd.0000000000001174] [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: 08/29/2023] [Accepted: 11/13/2023] [Indexed: 02/18/2024]
Abstract
BACKGROUND Therapeutic drug monitoring (TDM) using cyclin-dependent kinase inhibitors (CDK4/6is) is a novel approach for optimizing treatment outcomes. Currently, palbociclib, ribociclib, and abemaciclib are the available CDK4/6is and are primarily coadministered with letrozole. This study aimed to develop and validate an LC-MS/MS method for the simultaneous analysis of CDK4/6is, 2 active metabolites of abemaciclib (M2 and M20), and letrozole in human plasma for use in TDM studies. METHODS Sample pretreatment comprised protein precipitation with methanol and dilution of the supernatant with an aqueous mobile phase. Chromatographic separation was achieved using a reversed-phase XBridge BEH C18 column (2.5 μm, 3.0 × 75 mm XP), with methanol serving as the organic mobile phase and pyrrolidine-pyrrolidinium formate (0.005:0.005 mol/L) buffer (pH 11.3) as the aqueous mobile phase. A triple quadrupole mass spectrometer was used for the detection, with the ESI source switched from negative to positive ionization mode and the acquisition performed in multiple reaction monitoring mode. RESULTS The complete validation procedure was successfully performed in accordance with the latest regulatory guidelines. The following analytical ranges (ng/mL) were established for the tested compounds: 6-300, palbociclib and letrozole; 120-6000, ribociclib; 40-800, abemaciclib; and 20-400, M2 and M20. All results met the acceptance criteria for linearity, accuracy, precision, selectivity, sensitivity, matrix effects, and carryover. A total of 85 patient samples were analyzed, and all measured concentrations were within the validated ranges. The percent difference for the reanalyzed samples ranged from -11.2% to 7.0%. CONCLUSIONS A simple and robust LC-MS/MS method was successfully validated for the simultaneous quantification of CDK4/6is, M2, M20, and letrozole in human plasma. The assay was found to be suitable for measuring steady-state trough concentrations of the analytes in patient samples.
Collapse
Affiliation(s)
- Bianca Posocco
- Experimental and Clinical Pharmacology Unit-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Martina Zanchetta
- Experimental and Clinical Pharmacology Unit-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Marco Orleni
- Experimental and Clinical Pharmacology Unit-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Sara Gagno
- Experimental and Clinical Pharmacology Unit-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Marcella Montico
- Clinical Trial Office, Scientific Direction-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Elena Peruzzi
- Experimental and Clinical Pharmacology Unit-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Rossana Roncato
- Experimental and Clinical Pharmacology Unit-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
- Department of Medicine, University of Udine, Udine, Italy; and
| | - Lorenzo Gerratana
- Department of Medical Oncology-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Serena Corsetti
- Department of Medical Oncology-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Fabio Puglisi
- Department of Medicine, University of Udine, Udine, Italy; and
- Department of Medical Oncology-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| |
Collapse
|
6
|
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
|
7
|
Matte P, Bihan K, Isnard-Bagnis C, Zahr N, Thiery-Vuillemin A, Gougis P, Campedel L. Management of cancer treatments in hemodialysis patients. Bull Cancer 2024; 111:701-719. [PMID: 36931910 DOI: 10.1016/j.bulcan.2023.01.018] [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: 10/27/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 03/17/2023]
Abstract
INTRODUCTION The number of cancer patients receiving long-term hemodialysis (HD) is increasing, and HD could jeopardize treatments' safety and efficacy. Therefore, managing anticancer drugs is critical in this frail population. In addition, evidence of HD safety or risk is regularly released both for cytotoxic chemotherapy (CT) or hormone therapy (HT) as well as new therapies with molecularly targeted therapies (MTT), immune checkpoint inhibitors (ICI), and a summary of current knowledge is needed. METHODS We aimed to synthesize available data on cancer treatments in HD patients using PubMed database, FDA labels, summary of product characteristics (SmPC), FDA and EMA approval documents, guidelines and finally case reports for which relevant pharmacokinetic (PK) data is available. RESULTS For CT, recently proposed guidelines were balanced by the publication of particular toxic reports following them. SmPC was helpful in some cases, but no data was found for most CTs. MTT, both oral and monoclonal antibodies, were rarely modified by HD. However, HD patients have particular frailty that could require dose adaptation despite no substantial PK modification. Similarly, exposure to ICIs is unlikely to be modified by HD since immunoglobulins are not dialyzable. For HT, PK characteristics and HD impact were more heterogeneous and were reviewed molecule by molecule. CONCLUSIONS We summarized current knowledge on HD and cancer treatments. Data remains scarce, and the latest guidelines rely on few clinical data. There is a need to collect both retrospective and prospective data to better characterize the safety and relevant dose and schedule adaptations whenever needed in this situation to reinforce future guidelines.
Collapse
Affiliation(s)
- Paul Matte
- Groupement hospitalier Pitié-Salpêtrière, Assistance publique-Hôpitaux de Paris, service d'oncologie médicale, Paris, France; Groupe de recherche interdisciplinaire francophone en onco-néphrologie (GRIFON), Paris, France
| | - Kevin Bihan
- Groupement hospitalier Pitié-Salpêtrière, Assistance publique-Hôpitaux de Paris, service de pharmacologie, Paris, France
| | - Corinne Isnard-Bagnis
- Groupe de recherche interdisciplinaire francophone en onco-néphrologie (GRIFON), Paris, France; Groupement hospitalier Pitié-Salpêtrière, Assistance publique-Hôpitaux de Paris, Service de néphrologie, Paris, France
| | - Noël Zahr
- Groupement hospitalier Pitié-Salpêtrière, Assistance publique-Hôpitaux de Paris, service de pharmacologie, Paris, France
| | | | - Paul Gougis
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, Inserm, U932 Immunity and Cancer, Institut Curie, Université Paris, 75005 Paris, France
| | - Luca Campedel
- Groupe de recherche interdisciplinaire francophone en onco-néphrologie (GRIFON), Paris, France; Centre hospitalier universitaire Gabriel-Montpied, service d'oncologie médicale, Clermont-Ferrand, France.
| |
Collapse
|
8
|
Cytochrome P-450-mediated herb and food-drug interactions can be identified in cancer patients through patient self-reporting with a tablet application: results of a prospective observational study. ESMO Open 2022; 7:100650. [PMID: 36493603 PMCID: PMC9808458 DOI: 10.1016/j.esmoop.2022.100650] [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: 07/22/2022] [Revised: 09/21/2022] [Accepted: 10/24/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Consumption of herbs, food used as medicine and dietary supplements (HFDSs) is common in cancer patients. Herbs and food-drug interactions (HFDIs) can lead to serious adverse effects and can be prevented. We previously reviewed cytochrome P-450 (CYP)-mediated HFDI for 261 HFDSs and we classified the risk of CYP inhibition and induction on a level of evidence scale from 1 (high evidence, supported by several clinical studies) to 5 (low evidence, only limited preclinical data). PATIENTS AND METHODS We conducted a prospective, non-interventional study (NCT04128865) to assess whether self-assessment of patients could detect HFDI classified as 'probable' (i.e. level 1, 2 or 3 of the scale) in a population of cancer patients. Patients were invited through a tablet application to report their consumption of herbs, regular CYP-interacting food consumption and dietary supplements, as well as some clinical data and cancer treatments. The patient's completion of the survey could be supervised by a health care professional or not. A prespecified threshold of 5% of HFDIs classified as 'probable' detected with the application was deemed relevant. RESULTS Between 29 March 2018 and 22 June 2018, 143 patients completed the survey. Ninety-five patients (66%) reported at least one current systemic cancer treatment and were included in the analyses. Seventy-four patients reported an intake of at least one HFDS (77.9%), while 21 patients reported no HFDS (22.1%). Twenty-two HFDIs classified as 'probable' were found in 16 patients (16.8%) with the application, which was significantly superior to the prespecified threshold (P = 0.02). The interactions were reported with food (n = 19, 86%) more frequently than with herbs (n = 3, 14%) or with dietary supplements (no interaction reported). CONCLUSIONS Self-assessment of HFDS interaction with cancer treatment with an application is feasible and should be considered in daily routine. Prospective interventional studies should be conducted to better assess the clinical benefits of this approach.
Collapse
|
9
|
Akdeniz GY, Akgün H, Özakpınar ÖB, Duracık M, Öztürk M, İşcan E, Başoğlu F. Synthesis and studies of anticancer and antimicrobial activity of new phenylurenyl chalcone derivatives. LETT DRUG DES DISCOV 2022. [DOI: 10.2174/1570180819666220110153542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Phenylurenyl chalcone structures have the potential to act as a scaffold in anticancer drug discovery.
Methods:
N-Phenethyl-N'-{4-[(2E)-3-phenylprop-2-enoyl]phenyl}urea, 4/3-[(2E)-3-substitutedphenylprop-2-enoyl]phenyl}-N-phenylurea,4/3-[(2E)-3-substitutedphenyl
prop-2-enoyl]phenyl}-N-methylphenyl urea and {4/3-[(2E)-3-substitutedphenylprop-2-enoyl]phenyl}-N-ethylphenyl urea derivatives(1-35)were prepared and evaluated for their anticancer and antimicrobial activity against A-549 Hep-3B, HT-29, CF-7, PC-3, K-562 NIH-3T3 and Huh-7 cell lines and against Staphylococcus aureus (ATCC 6538), Pseudomonas aeruginosa (ATCC 9027), Escherichia coli (ATCC 8739) and Candida albicans (ATCC 10231), respectively.
Results:
While compounds 2, 26, 29, and 34 showed moderate cytotoxic activity on cell line Huh 7, compounds 14 (IC50: 6.42 µM), 16 (IC50: 5.64 µM), 19 (IC50: 6.95 µM) and 34 (IC50: 6.87 µM) showed good cytotoxic activity on Huh-7 cell line close to Sorafenib (IC50: 4.29 µM) (as reference). MIC values of compounds 4 and 22 against E. coli were 25 μg/ml, of compounds 3, 14 and 29 against P. aeruginosa 25 μg/ml and of compounds 11 and 33 against S. aureus 25 μg/ml. On the other hand, the minimum inhibitory concentration of all tested compounds against C. albicans was 25 μg/ml.
Conclusion:
N-Phenethyl-N'-{4-[(2E)-3-phenylprop-2-enoyl]phenyl}urea may be a new candidate to be developed as an anticancer compound.
Collapse
Affiliation(s)
- Güneş Yıldırım Akdeniz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Yeditepe University, Istanbul, Turkey
| | - Hülya Akgün
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Yeditepe University, Istanbul, Turkey
| | - Özlem Bingöl Özakpınar
- Department of Biochemistry, Faculty of Pharmacy, University of Marmara, Istanbul, Turkey
| | - Merve Duracık
- Department of Biochemistry, Faculty of Pharmacy, University of Marmara, Istanbul, Turkey
| | - Mehmet Öztürk
- zmir Biomedicine and Genome Center, Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey
| | - Evin İşcan
- Faculty of Medicine, Izmir Tınaztepe University, Izmir, Turkey
| | - Faika Başoğlu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, European University of Lefke, Lefke, Northern Cyprus, TR-10 Mersin, Turkey
| |
Collapse
|
10
|
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: 1.3] [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
|
11
|
Late phase 1 studies: concepts and outcomes. Lancet Oncol 2021; 22:e446-e455. [PMID: 34592194 DOI: 10.1016/s1470-2045(21)00467-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/27/2021] [Accepted: 08/04/2021] [Indexed: 12/13/2022]
Abstract
Over the past two decades, targeted therapies have become cornerstone treatments for numerous cancers with oncogene addiction. Unfortunately, their effectiveness reduces over time and most patients who receive targeted therapies relapse within 12 months. The emergence of drug-resistance mechanisms in tumours paved the way for next-generation inhibitors. However, insufficient concentration of targeted therapy is a frequent but poorly explored mechanism of treatment failure. Additionally, the maximum tolerated dose (MTD) is not always reached in phase studies, and the recommended phase 2 dose is mostly based on benefit-risk ratio and pharmacokinetic considerations, which could result in a suboptimal dose. This scenario has led us to propose a new concept in clinical drug development: the late phase 1 study. The primary goal of this type of trial is to define an alternative MTD of a drug in patients who are chronically exposed and had an initial benefit from targeted therapy but subsequently progressed without an identified resistance alteration. Intrapatient dose escalation might increase drug concentration and restore drug activity or efficacy.
Collapse
|
12
|
Llopis B, Robidou P, Tissot N, Pinna B, Gougis P, Aubart FC, Campedel L, Abbar B, Weil DR, Uzunov M, Gligorov J, Salem JE, Funck-Brentano C, Zahr N. Development and clinical validation of a simple and fast UPLC-ESI-MS/MS method for simultaneous quantification of nine kinase inhibitors and two antiandrogen drugs in human plasma: Interest for their therapeutic drug monitoring. J Pharm Biomed Anal 2021; 197:113968. [PMID: 33618135 DOI: 10.1016/j.jpba.2021.113968] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/04/2021] [Accepted: 02/09/2021] [Indexed: 12/15/2022]
Abstract
Kinase inhibitors (KIs) and antiandrogen drugs (AAs) are oral anticancer drugs with narrow therapeutic index that exhibit high inter- and intra-individual variability. We describe here a UPLC-MS/MS method for the simultaneous quantification of nine KIs: cobimetinib, dasatinib, ibrutinib, imatinib, nilotinib, palbociclib, ruxolitinib, sorafenib and vemurafenib; two active metabolites of them: N-desmethyl imatinib, N-oxide sorafenib; and two AAs: abiraterone and enzalutamide; with short pre-treatment and run time in order to be easily used in clinical practice for their therapeutic drug monitoring (TDM) and facilitating pharmacokinetics and pharmacokinetics/pharmacodynamics studies. Plasma samples were prepared by a single-step protein precipitation. Analytes were separated on a Waters Acquity UPLC® T3 HSS C18 column by non-linear gradient elution; with subsequent detection by Xevo® TQD triple quadrupole tandem mass spectrometer in a positive ionization mode. Analysis time was 2.8 min per run, and all analytes eluted within 1.46-1.97 minutes. The analytical performance of the method in terms of specificity, sensitivity, linearity, precision, accuracy, matrix effect, extraction recovery, limit of quantification, dilution integrity and stability of analytes under different conditions met all criteria for a bioanalytical method for the quantification of drugs. The calibration curves were linear over the range of 1-500 ng/mL for abiraterone, dasatinib and ibrutinib; 5-500 ng/mL for cobimetinib and palbociclib; 10-5,000 ng/mL for imatinib, N-desmethyl imatinib, nilotinib, sorafenib, N-oxide sorafenib and ruxolitinib; 100-50,000 ng/mL for enzalutamide and 100-100,000 ng/mL for vemurafenib with coefficient of correlation above 0.995 for all analytes. This novel method was successfully applied to TDM in clinical practice.
Collapse
Affiliation(s)
- Benoit Llopis
- AP-HP.Sorbonne Université, Department of Pharmacology and Clinical Investigation Center (CIC-1901), Pitié-Salpêtrière Hospital, INSERM, CIC-1901 and UMR-S 1166, Sorbonne Université, Faculty of Medicine Sorbonne Université, Faculty of Medicine, Paris, France
| | - Pascal Robidou
- AP-HP.Sorbonne Université, Department of Pharmacology and Clinical Investigation Center (CIC-1901), Pitié-Salpêtrière Hospital, INSERM, CIC-1901 and UMR-S 1166, Sorbonne Université, Faculty of Medicine Sorbonne Université, Faculty of Medicine, Paris, France
| | - Nadine Tissot
- AP-HP.Sorbonne Université, Department of Pharmacology and Clinical Investigation Center (CIC-1901), Pitié-Salpêtrière Hospital, INSERM, CIC-1901 and UMR-S 1166, Sorbonne Université, Faculty of Medicine Sorbonne Université, Faculty of Medicine, Paris, France
| | - Bruno Pinna
- AP-HP.Sorbonne Université, Department of Pharmacology and Clinical Investigation Center (CIC-1901), Pitié-Salpêtrière Hospital, INSERM, CIC-1901 and UMR-S 1166, Sorbonne Université, Faculty of Medicine Sorbonne Université, Faculty of Medicine, Paris, France
| | - Paul Gougis
- AP-HP.Sorbonne Université, Department of Pharmacology and Clinical Investigation Center (CIC-1901), Pitié-Salpêtrière Hospital, INSERM, CIC-1901 and UMR-S 1166, Sorbonne Université, Faculty of Medicine Sorbonne Université, Faculty of Medicine, Paris, France; AP-HP Sorbonne Université, Pitié-Salpêtrière Hospital, institut universitaire de cancérologie, département d'oncologie médicale, CLIP2, Galilée Paris, France
| | - Fleur Cohen Aubart
- AP-HP Sorbonne Université, Pitié-Salpêtrière Hospital, Service de Médecine Interne 2, Centre National de Référence Maladies Systémiques Rares et Histiocytoses, Paris, France
| | - Luca Campedel
- AP-HP Sorbonne Université, Pitié-Salpêtrière Hospital, institut universitaire de cancérologie, département d'oncologie médicale, CLIP2, Galilée Paris, France
| | - Baptiste Abbar
- AP-HP.Sorbonne Université, Department of Pharmacology and Clinical Investigation Center (CIC-1901), Pitié-Salpêtrière Hospital, INSERM, CIC-1901 and UMR-S 1166, Sorbonne Université, Faculty of Medicine Sorbonne Université, Faculty of Medicine, Paris, France
| | - Damien Roos Weil
- AP-HP Sorbonne Université, Service d'Hématologie Clinique, Pitié-Salpêtrière Hospital, Paris, France
| | - Madalina Uzunov
- AP-HP Sorbonne Université, Service d'Hématologie Clinique, Pitié-Salpêtrière Hospital, Paris, France
| | - Joseph Gligorov
- Institut Universitaire de Cancérologie, AP-HP Sorbonne Université, INSERM U-938, CLIP(2) Galilée, Tenon Hospital, Medical Oncology Department, Paris, France
| | - Joe-Elie Salem
- AP-HP.Sorbonne Université, Department of Pharmacology and Clinical Investigation Center (CIC-1901), Pitié-Salpêtrière Hospital, INSERM, CIC-1901 and UMR-S 1166, Sorbonne Université, Faculty of Medicine Sorbonne Université, Faculty of Medicine, Paris, France
| | - Christian Funck-Brentano
- AP-HP.Sorbonne Université, Department of Pharmacology and Clinical Investigation Center (CIC-1901), Pitié-Salpêtrière Hospital, INSERM, CIC-1901 and UMR-S 1166, Sorbonne Université, Faculty of Medicine Sorbonne Université, Faculty of Medicine, Paris, France
| | - Noël Zahr
- AP-HP.Sorbonne Université, Department of Pharmacology and Clinical Investigation Center (CIC-1901), Pitié-Salpêtrière Hospital, INSERM, CIC-1901 and UMR-S 1166, Sorbonne Université, Faculty of Medicine Sorbonne Université, Faculty of Medicine, Paris, France.
| |
Collapse
|
13
|
Geraud A, Mezquita L, Auclin E, Combarel D, Delahousse J, Gougis P, Massard C, Jovelet C, Caramella C, Adam J, Naltet C, Lavaud P, Gazzah A, Lacroix L, Rouleau E, Vasseur D, Mir O, Planchard D, Paci A, Besse B. Chronic Plasma Exposure to Kinase Inhibitors in Patients with Oncogene-Addicted Non-Small Cell Lung Cancer. Cancers (Basel) 2020; 12:cancers12123758. [PMID: 33327482 PMCID: PMC7764991 DOI: 10.3390/cancers12123758] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 12/10/2020] [Indexed: 12/27/2022] Open
Abstract
Simple Summary In this study, we measured the plasmatic concentration of Kinase inhibitors (KI) among a population with non-small cell lung cancer (NSCLC) harboring driver genetic alterations. They received erlotinib, gefitinib, osimertinib, crizotinib, or dabrafenib (with or without trametinib) for at least three months. The results were measured by ultra-performance liquid chromatography coupled with tandem mass spectrometry and compared to previously published data. Between November 2013 and February 2019, fifty-one samples were analyzed. The main outcome was the rate of samples with suboptimal KI plasma concentrations. Suboptimal plasma concentrations were observed in 51% (26/51) of cases and might contribute to treatment failure. Abstract Kinase inhibitors (KI) have dramatically improved the outcome of treatment in patients with non-small cell lung cancer (NSCLC), which harbors an oncogene addiction. This study assesses KI plasma levels and their clinical relevance in patients chronically exposed to KIs. Plasma samples were collected in NSCLC patients receiving erlotinib, gefitinib, osimertinib, crizotinib, or dabrafenib (with or without trametinib) for at least three months between November 2013 and February 2019 in a single institution. KI drug concentrations were measured by ultra-performance liquid chromatography coupled with tandem mass spectrometry and compared to published data defining optimal plasma concentration. The main outcome was the rate of samples with suboptimal KI plasma concentrations. Secondary outcomes included its impact on T790M mutation emergence in patients receiving a first-generation epidermal growth factor receptor (EGFR) KI. Fifty-one samples were available from 41 patients with advanced NSCLC harboring driver genetic alterations, including EGFR, v-Raf murine sarcoma viral oncogene homolog B (BRAF), anaplastic lymphoma kinase (ALK) or ROS proto-oncogene 1 (ROS1), and who had an available evaluation of chronic KI plasma exposure. Suboptimal plasma concentrations were observed in 51% (26/51) of cases. In EGFR-mutant cases failing first-generation KIs, EGFR exon 20 p.T790M mutation emergence was detected in 31% (4/13) of samples in optimal vs. none in suboptimal concentration (0/5). Suboptimal plasma concentrations of KIs are frequent in advanced NSCLC patients treated with a KI for at least three months and might contribute to treatment failure.
Collapse
Affiliation(s)
- Arthur Geraud
- Cancer Medicine Department, Gustave Roussy, 94805 Villejuif, France; (A.G.); (L.M.); (C.N.); (P.L.); (D.P.)
- Early Drug Development Department (DITEP), Gustave Roussy, 94805 Villejuif, France; (C.M.); (A.G.)
| | - Laura Mezquita
- Cancer Medicine Department, Gustave Roussy, 94805 Villejuif, France; (A.G.); (L.M.); (C.N.); (P.L.); (D.P.)
- Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Medical Oncology Department, Hospital Clínic, 08036 Barcelona, Spain
| | - Edouard Auclin
- Department of Medical and Digestive Oncology, Hôpital Européen Georges Pompidou, Assistance Publique des Hôpitaux de Paris, 75015 Paris, France;
| | - David Combarel
- Pharmacology Department, Gustave Roussy, 94805 Villejuif, France; (D.C.); (J.D.); (A.P.)
- Faculty of Pharmacy, Paris-Saclay University, 92296 Chatenay-Malabry, France
| | - Julia Delahousse
- Pharmacology Department, Gustave Roussy, 94805 Villejuif, France; (D.C.); (J.D.); (A.P.)
| | - Paul Gougis
- Department of Pharmacology and Clinical Investigation Center, Pitié-Salpêtrière Hospital, INSERM, CIC-1421, Sorbonne University, 75013 Paris, France;
- CLIP2 Galilée, Regional Pharmacovigilance Center, Pitié-Salpêtrière Hospital, INSERM, CIC-1421, Sorbonne University, 75013 Paris, France
| | - Christophe Massard
- Early Drug Development Department (DITEP), Gustave Roussy, 94805 Villejuif, France; (C.M.); (A.G.)
- Paris-Saclay University, Cancer Campus Gustave Roussy, Gustave Roussy, 94805 Villejuif, France
| | - Cécile Jovelet
- Department of Medical Biology and Pathology, Gustave Roussy, 94805 Villejuif, France; (C.J.); (L.L.); (E.R.); (D.V.)
| | | | - Julien Adam
- Pathology Department, Gustave Roussy, 94805 Villejuif, France;
| | - Charles Naltet
- Cancer Medicine Department, Gustave Roussy, 94805 Villejuif, France; (A.G.); (L.M.); (C.N.); (P.L.); (D.P.)
| | - Pernelle Lavaud
- Cancer Medicine Department, Gustave Roussy, 94805 Villejuif, France; (A.G.); (L.M.); (C.N.); (P.L.); (D.P.)
| | - Anas Gazzah
- Early Drug Development Department (DITEP), Gustave Roussy, 94805 Villejuif, France; (C.M.); (A.G.)
| | - Ludovic Lacroix
- Department of Medical Biology and Pathology, Gustave Roussy, 94805 Villejuif, France; (C.J.); (L.L.); (E.R.); (D.V.)
| | - Etienne Rouleau
- Department of Medical Biology and Pathology, Gustave Roussy, 94805 Villejuif, France; (C.J.); (L.L.); (E.R.); (D.V.)
| | - Damien Vasseur
- Department of Medical Biology and Pathology, Gustave Roussy, 94805 Villejuif, France; (C.J.); (L.L.); (E.R.); (D.V.)
| | - Olivier Mir
- Department of Ambulatory Cancer Care, Gustave Roussy, 94805 Villejuif, France;
| | - David Planchard
- Cancer Medicine Department, Gustave Roussy, 94805 Villejuif, France; (A.G.); (L.M.); (C.N.); (P.L.); (D.P.)
| | - Angelo Paci
- Pharmacology Department, Gustave Roussy, 94805 Villejuif, France; (D.C.); (J.D.); (A.P.)
- Faculty of Pharmacy, Paris-Saclay University, 92296 Chatenay-Malabry, France
| | - Benjamin Besse
- Cancer Medicine Department, Gustave Roussy, 94805 Villejuif, France; (A.G.); (L.M.); (C.N.); (P.L.); (D.P.)
- Paris-Saclay University, Cancer Campus Gustave Roussy, Gustave Roussy, 94805 Villejuif, France
- Correspondence: ; Tel.: +331-42-11-43-22
| |
Collapse
|
14
|
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: 2.2] [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
|
15
|
Chen J, Xu H, Pawlak S, James LP, Peltz G, Lee K, Ginman K, Bergeron M, Pithavala YK. The Effect of Rifampin on the Pharmacokinetics and Safety of Lorlatinib: Results of a Phase One, Open-Label, Crossover Study in Healthy Participants. Adv Ther 2020; 37:745-758. [PMID: 31863284 DOI: 10.1007/s12325-019-01198-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Lorlatinib is a third-generation tyrosine kinase inhibitor approved for the treatment of anaplastic lymphoma kinase (ALK)-positive metastatic non-small cell lung cancer; cytochrome P450 (CYP) 3A plays an important role in the metabolism of lorlatinib. METHODS This phase 1, open-label, two-period, crossover study estimated the effect of oral rifampin (a strong CYP3A inducer) on the pharmacokinetics and safety of oral lorlatinib (NCT02804399). Healthy participants received single-dose lorlatinib 100 mg in period 1 followed by rifampin 600 mg/day (days 1-12) and single-dose lorlatinib 100 mg (day 8) in period 2. Blood samples were collected for 120 h after each dose of lorlatinib. RESULTS When a single dose of lorlatinib was administered during daily dosing with rifampin (period 2), the area under the plasma concentration-time profile extrapolated to infinity (AUCinf) and maximum plasma concentration (Cmax) of lorlatinib were 14.74% [90% confidence interval (CI) 12.78%, 17.01%] and 23.88% (90% CI 21.58%, 26.43%), respectively, of those in period 1 (lorlatinib alone). A single dose of lorlatinib was well tolerated in period 1, but elevations in transaminase values were observed in all participants (grade 2-4 in 11 participants) within 1-3 days after a single dose of lorlatinib was administered with ongoing rifampin in period 2. Rifampin dosing was therefore halted. Transaminase levels subsequently returned to normal (median time to recovery: 15 days). No elevations in bilirubin were observed. CONCLUSIONS The addition of a single dose of lorlatinib to daily dosing with rifampin significantly reduced lorlatinib plasma exposure relative to a single dose of lorlatinib administered alone and was associated with severe but self-limiting transaminase elevations in all healthy participants. These observations support the contraindication in the product label against concomitant use of lorlatinib with all strong CYP3A inducers. TRIAL REGISTRATION ClinicalTrials.gov identifier, NCT02804399.
Collapse
Affiliation(s)
- Joseph Chen
- Global Product Development, Pfizer Oncology, New York, NY, USA.
| | - Huiping Xu
- Global Product Development, Clinical Pharmacology, Pfizer Oncology, La Jolla, CA, USA
| | | | - Leonard P James
- Global Product Development, Pfizer Oncology, New York, NY, USA
| | - Gerson Peltz
- Safety Surveillance and Risk Management, Pfizer Oncology, Groton, CT, USA
| | - Kimberly Lee
- Global Product Development, Pfizer Inc., Groton, CT, USA
| | | | | | - Yazdi K Pithavala
- Global Product Development, Clinical Pharmacology, Pfizer Oncology, La Jolla, CA, USA
| |
Collapse
|