1
|
Nakatsugawa E, Naito T, Shibata K, Kitajima R, Kawakami J. Impacts of genetic polymorphisms and cancer cachexia on naldemedine pharmacokinetics and bowel movements in patients receiving opioid analgesics. Fundam Clin Pharmacol 2024; 38:596-605. [PMID: 38192190 DOI: 10.1111/fcp.12976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND/OBJECTIVES Clinical responses to naldemedine vary between individuals with advanced cancer. This is a prospective, single-center, observational study aimed to evaluate the influence of genetic polymorphisms and cachexia status on plasma naldemedine and clinical responses. METHODS Forty-eight patients being treated with naldemedine for opioid-induced constipation under treatment of cancer pain were enrolled. Plasma naldemedine concentrations were determined on the fourth day or later after administration of naldemedine, and the associations with genotypes, cachexia status, and clinical responses were assessed. RESULTS Cancer patients exhibited a large variation in the plasma naldemedine concentrations, and it was correlated with serum total protein level. Patients who were homozygous CYP3A5*3 had a higher plasma concentration of naldemedine than those with the *1 allele. ABCB1 genotypes tested in this study were not associated with plasma naldemedine. A negative correlation was observed between the plasma naldemedine concentration and 4β-hydroxycholesterol level. The plasma naldemedine concentration was lower in patients with refractory cachexia than in those with precachexia and cachexia. While serum levels of interleukin-6 (IL-6) and acute-phase proteins were higher in patients with refractory cachexia, they were not associated with plasma naldemedine. A higher plasma concentration of naldemedine, CYP3A5*3/*3, and an earlier naldemedine administration after starting opioid analgesics were related to improvement of bowel movements. CONCLUSION Plasma naldemedine increased under deficient activity of CYP3A5 in cancer patients. Cachectic patients with a higher serum IL-6 had a lower plasma naldemedine. Plasma naldemedine, related to CYP3A5 genotype, and the initiation timing of naldemedine were associated with improved bowel movements.
Collapse
Affiliation(s)
- Emi Nakatsugawa
- Department of Hospital Pharmacy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takafumi Naito
- Department of Hospital Pharmacy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Pharmacy, Shinshu University Hospital, Matsumoto, Japan
| | - Kaito Shibata
- Department of Hospital Pharmacy, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Pharmacy, Shinshu University Hospital, Matsumoto, Japan
| | - Ryo Kitajima
- Division of Palliative Care Center, Hamamatsu University Hospital, Hamamatsu, Japan
| | - Junichi Kawakami
- Department of Hospital Pharmacy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
2
|
Cheng F, Wang H, Li W, Zhang Y. Clinical pharmacokinetics and drug-drug interactions of tyrosine-kinase inhibitors in chronic myeloid leukemia: A clinical perspective. Crit Rev Oncol Hematol 2024; 195:104258. [PMID: 38307392 DOI: 10.1016/j.critrevonc.2024.104258] [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: 09/27/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 02/04/2024] Open
Abstract
In the past decade, numerous tyrosine kinase inhibitors (TKIs) have been introduced in the treatment of chronic myeloid leukemia. Given the significant interpatient variability in TKIs pharmacokinetics, potential drug-drug interactions (DDIs) can greatly impact patient therapy. This review aims to discuss the pharmacokinetic characteristics of TKIs, specifically focusing on their absorption, distribution, metabolism, and excretion profiles. Additionally, it provides a comprehensive overview of the utilization of TKIs in special populations such as the elderly, children, and patients with liver or kidney dysfunction. We also highlight known or suspected DDIs between TKIs and other drugs, highlighting various clinically relevant interactions. Moreover, specific recommendations are provided to guide haemato-oncologists, oncologists, and clinical pharmacists in managing DDIs during TKI treatment in daily clinical practice.
Collapse
Affiliation(s)
- Fang Cheng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430022, China
| | - Hongxiang Wang
- Department of Hematology, the Central Hospital of Wuhan, 430014, China
| | - Weiming Li
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430022, China.
| |
Collapse
|
3
|
Wang F, Zhang X, Wang Y, Chen Y, Lu H, Meng X, Ye X, Chen W. Activation/Inactivation of Anticancer Drugs by CYP3A4: Influencing Factors for Personalized Cancer Therapy. Drug Metab Dispos 2023; 51:543-559. [PMID: 36732076 DOI: 10.1124/dmd.122.001131] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 02/04/2023] Open
Abstract
Cytochrome P450 3A4 (CYP3A4), one of the most important members of the cytochrome P450 subfamily, is a crucial catalyst in the metabolism of numerous drugs. As it catalyzes numerous processes for drug activation or inactivation, the pharmacological activities and clinical outcomes of anticancer drugs metabolized by CYP3A4 are highly dependent on the enzyme's activity and expression. Due to the complexity of tumor microenvironments and various influencing factors observed in human in vitro models and clinical studies, the pharmacokinetics of most anticancer drugs are influenced by the extent of induction or inhibition of CYP3A4-mediated metabolism, and these details are not fully recognized and highlighted. Therefore, this interindividual variability due to genetic and nongenetic factors, together with the narrow therapeutic index of most anticancer drugs, contributes to their unique set of exposures and responses, which have important implications for achieving the expected efficacy and minimizing adverse events of chemotherapy for cancer in individuals. To elucidate the mechanisms of CYP3A4-mediated activation/inactivation of anticancer drugs associated with personalized therapy, this review focuses on the underlying determinants that contribute to differences in CYP3A4 metabolic activity and provides a comprehensive and valuable overview of the significance of these factors, which differs from current considerations for dosing regimens in cancer therapy. We also discuss knowledge gaps, challenges, and opportunities to explore optimal dosing regimens for drug metabolic activation/inactivation in individual patients, with particular emphasis on pooling and analyzing clinical information that affects CYP3A4 activity. SIGNIFICANCE STATEMENT: This review focuses on anticancer drugs that are activated/deactivated by CYP3A4 and highlights outstanding factors affecting the interindividual variability of CYP3A4 activity in order to gain a detailed understanding of CYP3A4-mediated drug metabolism mechanisms. A systematic analysis of available information on the underlying genetic and nongenetic determinants leading to variation in CYP3A4 metabolic activity to predict therapeutic response to drug exposure, maximize efficacy, and avoid unpredictable adverse events has clinical implications for the identification and development of CYP3A4-targeted cancer therapeutics.
Collapse
Affiliation(s)
- Fengling Wang
- Department of Pharmacy, Hefei Hospital, Affiliated to Anhui Medical University (The Second People's Hospital of Hefei), Hefei, Anhui, China (F.W., X.M., X.Y.); School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (F.W.); School of Pharmacy (F.W., X.Z., Y.W., Y.C., H.L., W.C.) and Institute of Pharmaceutics, School of Pharmaceutical Sciences (X.Z., H.L., W.C.), Anhui University of Chinese Medicine, Hefei, Anhui, China; The Second People's Hospital of Hefei, Affiliated to Bengbu Medical College, Hefei, Anhui, China (F.W., X.M., X.Y.); and MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, Anhui, China (W.C.)
| | - Xue Zhang
- Department of Pharmacy, Hefei Hospital, Affiliated to Anhui Medical University (The Second People's Hospital of Hefei), Hefei, Anhui, China (F.W., X.M., X.Y.); School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (F.W.); School of Pharmacy (F.W., X.Z., Y.W., Y.C., H.L., W.C.) and Institute of Pharmaceutics, School of Pharmaceutical Sciences (X.Z., H.L., W.C.), Anhui University of Chinese Medicine, Hefei, Anhui, China; The Second People's Hospital of Hefei, Affiliated to Bengbu Medical College, Hefei, Anhui, China (F.W., X.M., X.Y.); and MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, Anhui, China (W.C.)
| | - Yanyan Wang
- Department of Pharmacy, Hefei Hospital, Affiliated to Anhui Medical University (The Second People's Hospital of Hefei), Hefei, Anhui, China (F.W., X.M., X.Y.); School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (F.W.); School of Pharmacy (F.W., X.Z., Y.W., Y.C., H.L., W.C.) and Institute of Pharmaceutics, School of Pharmaceutical Sciences (X.Z., H.L., W.C.), Anhui University of Chinese Medicine, Hefei, Anhui, China; The Second People's Hospital of Hefei, Affiliated to Bengbu Medical College, Hefei, Anhui, China (F.W., X.M., X.Y.); and MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, Anhui, China (W.C.)
| | - Yunna Chen
- Department of Pharmacy, Hefei Hospital, Affiliated to Anhui Medical University (The Second People's Hospital of Hefei), Hefei, Anhui, China (F.W., X.M., X.Y.); School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (F.W.); School of Pharmacy (F.W., X.Z., Y.W., Y.C., H.L., W.C.) and Institute of Pharmaceutics, School of Pharmaceutical Sciences (X.Z., H.L., W.C.), Anhui University of Chinese Medicine, Hefei, Anhui, China; The Second People's Hospital of Hefei, Affiliated to Bengbu Medical College, Hefei, Anhui, China (F.W., X.M., X.Y.); and MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, Anhui, China (W.C.)
| | - Huiyu Lu
- Department of Pharmacy, Hefei Hospital, Affiliated to Anhui Medical University (The Second People's Hospital of Hefei), Hefei, Anhui, China (F.W., X.M., X.Y.); School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (F.W.); School of Pharmacy (F.W., X.Z., Y.W., Y.C., H.L., W.C.) and Institute of Pharmaceutics, School of Pharmaceutical Sciences (X.Z., H.L., W.C.), Anhui University of Chinese Medicine, Hefei, Anhui, China; The Second People's Hospital of Hefei, Affiliated to Bengbu Medical College, Hefei, Anhui, China (F.W., X.M., X.Y.); and MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, Anhui, China (W.C.)
| | - Xiangyun Meng
- Department of Pharmacy, Hefei Hospital, Affiliated to Anhui Medical University (The Second People's Hospital of Hefei), Hefei, Anhui, China (F.W., X.M., X.Y.); School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (F.W.); School of Pharmacy (F.W., X.Z., Y.W., Y.C., H.L., W.C.) and Institute of Pharmaceutics, School of Pharmaceutical Sciences (X.Z., H.L., W.C.), Anhui University of Chinese Medicine, Hefei, Anhui, China; The Second People's Hospital of Hefei, Affiliated to Bengbu Medical College, Hefei, Anhui, China (F.W., X.M., X.Y.); and MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, Anhui, China (W.C.)
| | - Xi Ye
- Department of Pharmacy, Hefei Hospital, Affiliated to Anhui Medical University (The Second People's Hospital of Hefei), Hefei, Anhui, China (F.W., X.M., X.Y.); School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (F.W.); School of Pharmacy (F.W., X.Z., Y.W., Y.C., H.L., W.C.) and Institute of Pharmaceutics, School of Pharmaceutical Sciences (X.Z., H.L., W.C.), Anhui University of Chinese Medicine, Hefei, Anhui, China; The Second People's Hospital of Hefei, Affiliated to Bengbu Medical College, Hefei, Anhui, China (F.W., X.M., X.Y.); and MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, Anhui, China (W.C.)
| | - Weidong Chen
- Department of Pharmacy, Hefei Hospital, Affiliated to Anhui Medical University (The Second People's Hospital of Hefei), Hefei, Anhui, China (F.W., X.M., X.Y.); School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (F.W.); School of Pharmacy (F.W., X.Z., Y.W., Y.C., H.L., W.C.) and Institute of Pharmaceutics, School of Pharmaceutical Sciences (X.Z., H.L., W.C.), Anhui University of Chinese Medicine, Hefei, Anhui, China; The Second People's Hospital of Hefei, Affiliated to Bengbu Medical College, Hefei, Anhui, China (F.W., X.M., X.Y.); and MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, Anhui, China (W.C.)
| |
Collapse
|
4
|
Hussaarts KGAM, Veerman GDM, Jansman FGA, van Gelder T, Mathijssen RHJ, van Leeuwen RWF. Clinically relevant drug interactions with multikinase inhibitors: a review. Ther Adv Med Oncol 2019; 11:1758835918818347. [PMID: 30643582 PMCID: PMC6322107 DOI: 10.1177/1758835918818347] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/17/2018] [Indexed: 12/11/2022] Open
Abstract
Multikinase inhibitors (MKIs), including the tyrosine kinase inhibitors (TKIs), have rapidly become an established factor in daily (hemato)-oncology practice. Although the oral route of administration offers improved flexibility and convenience for the patient, challenges arise in the use of MKIs. As MKIs are prescribed extensively, patients are at increased risk for (severe) drug–drug interactions (DDIs). As a result of these DDIs, plasma pharmacokinetics of MKIs may vary significantly, thereby leading to high interpatient variability and subsequent risk for increased toxicity or a diminished therapeutic outcome. Most clinically relevant DDIs with MKIs concern altered absorption and metabolism. The absorption of MKIs may be decreased by concomitant use of gastric acid-suppressive agents (e.g. proton pump inhibitors) as many kinase inhibitors show pH-dependent solubility. In addition, DDIs concerning drug (uptake and efflux) transporters may be of significant clinical relevance during MKI therapy. Furthermore, since many MKIs are substrates for cytochrome P450 isoenzymes (CYPs), induction or inhibition with strong CYP inhibitors or inducers may lead to significant alterations in MKI exposure. In conclusion, DDIs are of major concern during MKI therapy and need to be monitored closely in clinical practice. Based on the current knowledge and available literature, practical recommendations for management of these DDIs in clinical practice are presented in this review.
Collapse
Affiliation(s)
- Koen G A M Hussaarts
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - G D Marijn Veerman
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Frank G A Jansman
- Department of Clinical Pharmacy, Deventer Hospital, Deventer, The Netherlands
| | - Teun van Gelder
- Department of Hospital Pharmacy, Erasmus MC, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | |
Collapse
|
5
|
Yamazaki S, Loi CM, Kimoto E, Costales C, Varma MV. Application of Physiologically Based Pharmacokinetic Modeling in Understanding Bosutinib Drug-Drug Interactions: Importance of Intestinal P-Glycoprotein. Drug Metab Dispos 2018; 46:1200-1211. [PMID: 29739809 DOI: 10.1124/dmd.118.080424] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 05/07/2018] [Indexed: 12/21/2022] Open
Abstract
Bosutinib is an orally available Src/Abl tyrosine kinase inhibitor indicated for the treatment of patients with Ph+ chronic myelogenous leukemia at a clinically recommended dose of 500 mg once daily. Clinical results indicated that increases in bosutinib oral exposures were supraproportional at the lower doses (50-200 mg) and approximately dose-proportional at the higher doses (200-600 mg). Bosutinib is a substrate of CYP3A4 and P-glycoprotein and exhibits pH-dependent solubility with moderate intestinal permeability. These findings led us to investigate the factors influencing the underlying pharmacokinetic mechanisms of bosutinib with physiologically based pharmacokinetic (PBPK) models. Our primary objectives were to: 1) refine the previously developed bosutinib PBPK model on the basis of the latest oral bioavailability data and 2) verify the refined PBPK model with P-glycoprotein kinetics on the basis of the bosutinib drug-drug interaction (DDI) results with ketoconazole and rifampin. Additionally, the verified PBPK model was applied to predict bosutinib DDIs with dual CYP3A/P-glycoprotein inhibitors. The results indicated that 1) the refined PBPK model adequately described the observed plasma concentration-time profiles of bosutinib and 2) the verified PBPK model reasonably predicted the effects of ketoconazole and rifampin on bosutinib exposures by accounting for intestinal P-glycoprotein inhibition/induction. These results suggested that bosutinib DDI mechanism could involve not only CYP3A4-mediated metabolism but also P-glycoprotein-mediated efflux on absorption. In summary, P-glycoprotein kinetics could constitute an element in the PBPK models critical to understanding the pharmacokinetic mechanism of dual CYP3A/P-glycoprotein substrates, such as bosutinib, that exhibit nonlinear pharmacokinetics owing largely to a saturation of intestinal P-glycoprotein-mediated efflux.
Collapse
Affiliation(s)
- Shinji Yamazaki
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, San Diego, California (S.Y., C.-M.L.) and Groton, Connecticut (E.K., C.C., M.V.V.)
| | - Cho-Ming Loi
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, San Diego, California (S.Y., C.-M.L.) and Groton, Connecticut (E.K., C.C., M.V.V.)
| | - Emi Kimoto
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, San Diego, California (S.Y., C.-M.L.) and Groton, Connecticut (E.K., C.C., M.V.V.)
| | - Chester Costales
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, San Diego, California (S.Y., C.-M.L.) and Groton, Connecticut (E.K., C.C., M.V.V.)
| | - Manthena V Varma
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research & Development, San Diego, California (S.Y., C.-M.L.) and Groton, Connecticut (E.K., C.C., M.V.V.)
| |
Collapse
|
6
|
Abstract
During recent years, the therapeutic landscape in chronic myeloid leukemia (CML) has changed significantly. Since the clinical introduction of tyrosine kinase inhibitors (TKIs) approximately 15 years ago, patients' concerns have shifted from reduced life expectancy toward long-term toxicities of TKI, depth of remission, and the probability of successful treatment discontinuation. Patients with newly diagnosed CML in chronic phase (at least with a Sokal score not exceeding intermediate) may now expect an almost normal life expectancy. However, even if almost 30% of all newly diagnosed chronic-phase patients might eventually be facing the prospect of a life without CML-specific treatment, based on current knowledge, most, if not all, patients would have to undergo an expected minimum of 5-8 years of TKI treatment and the majority would face a life-long exposure to the side-effects of TKIs. At present, 5 different TKIs are licensed for the treatment of CML, that is, imatinib, which is a first-generation TKI (including its generic derivatives); nilotinib, dasatinib, and bosutinib, which are second-generation TKIs; as well as ponatinib, which is a so-called third-generation TKI and is supposed to be used for patients harboring the T315I-mutation. One of the important, yet unanswered questions is the choice of the best possible TKI upfront for each individual patient. Bosutinib is currently licensed for patients with CML after failure or intolerance of at least 2 other TKIs. It can also be prescribed according to label if after failure of the first TKI therapy, another option does not seem feasible. This review focuses on the existing data on clinical efficacy, tolerability, and side effects of bosutinib treatment in CML patients with the aim to identify patient characteristics and treatment scenarios most suitable for treatment with bosutinib.
Collapse
Affiliation(s)
- Susanne Isfort
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany.,Center for Translational and Clinical Research Aachen (CTC-A), Medical Faculty at the RWTH Aachen University, Aachen, Germany
| | - Tim H Brümmendorf
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
7
|
Isfort S, Crysandt M, Gezer D, Koschmieder S, Brümmendorf TH, Wolf D. Bosutinib: A Potent Second-Generation Tyrosine Kinase Inhibitor. Recent Results Cancer Res 2018; 212:87-108. [PMID: 30069626 DOI: 10.1007/978-3-319-91439-8_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bosutinib is one of the five tyrosine kinase inhibitors which are currently approved for the treatment of chronic myeloid leukemia. By its dual inhibition of Src and ABL kinase and also targeting further kinases, it creates a unique target portfolio which also explains its unique side effect profile. The approval of bosutinib in 2013 made the drug available for patients previously treated with one or more tyrosine kinase inhibitor(s) and for whom imatinib, nilotinib, and dasatinib are not considered appropriate treatment options. As initially the first-line clinical trial comparing bosutinib with imatinib in CML patients in chronic phase did not reach its primary endpoint and therefore the product was not licensed for first-line therapy, a second first-line trial, the so-called BFORE study, was performed and just recently the promising results have been published predicting a quick expansion of the existing label. In comparison with the other approved TKIs, bosutinib harbors a distinct side effect profile with only very few cardiovascular and thromboembolic events and minimal long-term safety issues with most adverse events happening during the first months of treatment. On the other hand, gastrointestinal side effects are very common (e.g., diarrhea rates in more than 80% of the patients) with bosutinib surprising some of the investigators during the early clinical trials evaluating bosutinib. Until then, several approaches have been used to face this problem resulting in extensive supportive efforts (such as early loperamid treatment) as well as new trials testing alternative dosing strategies with early dose adjustment schedules. This article reports preclinical and clinical data available for bosutinib both in hematologic diseases such as CML or ALL and solid tumours as well as other diseases and envisions future perspectives including additional patient groups in which bosutinib might be of clinical benefit.
Collapse
Affiliation(s)
- Susanne Isfort
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany.
| | - Martina Crysandt
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Deniz Gezer
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Tim H Brümmendorf
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Dominik Wolf
- Department of Oncology, Hematology, Immunoncology and Rheumatology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127, Bonn, Germany
| |
Collapse
|
8
|
Hsyu PH, Pignataro DS, Matschke K. Absolute Bioavailability of Bosutinib in Healthy Subjects From an Open-Label, Randomized, 2-Period Crossover Study. Clin Pharmacol Drug Dev 2017; 7:373-381. [PMID: 29058816 DOI: 10.1002/cpdd.396] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 08/28/2017] [Indexed: 12/31/2022]
Abstract
This study evaluated the absolute bioavailability of bosutinib and assessed its safety and tolerability after single-dose oral and intravenous administration. In this phase 1 open-label, 2-sequence, 2-period crossover study, healthy, fed subjects aged 18-55 years were randomized to 1 of 2 treatment sequences (n = 7/sequence): oral bosutinib (100 mg × 5) followed by intravenous bosutinib (120 mg in approximately 240 mL over 1 hour), with a ≥14-day washout, or intravenous bosutinib and then oral bosutinib. Results of plasma pharmacokinetics analyses demonstrated that exposure to intravenous bosutinib was 3-fold higher than for oral bosutinib (16.2 and 5.5 ng·h/mL/mg, respectively), and mean terminal half-life was similar (35.5 and 31.7 hours). The ratio of adjusted geometric means (90%CI) for the dose-normalized area under the plasma concentration-time profile (AUC0-∞ /D) was 33.85% (30.65%-37.38%). Most treatment-emergent adverse events (AEs) were mild in severity. Gastrointestinal (GI) AEs occurred in 9 of 13 subjects given oral bosutinib, whereas no subjects given intravenous bosutinib experienced GI AEs, suggesting bosutinib present in the GI tract had an effect. Bosutinib exhibited an absolute bioavailability of 33.85% based on the ratio of AUC0-∞ /D. Both oral and intravenous bosutinib were safe and well tolerated in healthy, fed adult subjects.
Collapse
|
9
|
Ono C, Hsyu PH, Abbas R, Loi CM, Yamazaki S. Application of Physiologically Based Pharmacokinetic Modeling to the Understanding of Bosutinib Pharmacokinetics: Prediction of Drug-Drug and Drug-Disease Interactions. Drug Metab Dispos 2017; 45:390-398. [PMID: 28167538 DOI: 10.1124/dmd.116.074450] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/03/2017] [Indexed: 02/13/2025] Open
Abstract
Bosutinib is an orally available Src/Abl tyrosine kinase inhibitor indicated for the treatment of patients with Philadelphia chromosome-positive chronic myelogenous leukemia. Bosutinib is predominantly metabolized by CYP3A4 as the primary clearance mechanism. The main objectives of this study were to 1) develop physiologically based pharmacokinetic (PBPK) models of bosutinib; 2) verify and refine the PBPK models based on clinical study results of bosutinib single-dose drug-drug interaction (DDI) with ketoconazole and rifampin, as well as single-dose drug-disease interaction (DDZI) in patients with renal and hepatic impairment; 3) apply the PBPK models to predict DDI outcomes in patients with weak and moderate CYP3A inhibitors; and 4) apply the PBPK models to predict DDZI outcomes in renally and hepatically impaired patients after multiple-dose administration. Results showed that the PBPK models adequately predicted bosutinib oral exposures in patients after single- and multiple-dose administrations. The PBPK models also reasonably predicted changes in bosutinib exposures in the single-dose DDI and DDZI results, suggesting that the PBPK models were sufficiently developed and verified based on the currently available data. Finally, the PBPK models predicted 2- to 4-fold increases in bosutinib exposures by moderate CYP3A inhibitors, as well as comparable increases in bosutinib exposures in renally and hepatically impaired patients between single- and multiple-dose administrations. Given the challenges in conducting numerous DDI and DDZI studies of anticancer drugs in patients, we believe that the PBPK models verified in our study would be valuable to reasonably predict bosutinib exposures under various scenarios that have not been tested clinically.
Collapse
Affiliation(s)
- Chiho Ono
- Clinical Pharmacology, Pfizer Japan Inc., Tokyo, Japan (C.O.); Clinical Pharmacology, Pfizer Inc., San Diego, California (P.-H.H.); Clinical Pharmacology, Pfizer Essential Health, Collegeville, Pennsylvania (R.A.); and Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide Research and Development, San Diego, California (C.-M.L., S.Y.)
| | - Poe-Hirr Hsyu
- Clinical Pharmacology, Pfizer Japan Inc., Tokyo, Japan (C.O.); Clinical Pharmacology, Pfizer Inc., San Diego, California (P.-H.H.); Clinical Pharmacology, Pfizer Essential Health, Collegeville, Pennsylvania (R.A.); and Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide Research and Development, San Diego, California (C.-M.L., S.Y.)
| | - Richat Abbas
- Clinical Pharmacology, Pfizer Japan Inc., Tokyo, Japan (C.O.); Clinical Pharmacology, Pfizer Inc., San Diego, California (P.-H.H.); Clinical Pharmacology, Pfizer Essential Health, Collegeville, Pennsylvania (R.A.); and Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide Research and Development, San Diego, California (C.-M.L., S.Y.)
| | - Cho-Ming Loi
- Clinical Pharmacology, Pfizer Japan Inc., Tokyo, Japan (C.O.); Clinical Pharmacology, Pfizer Inc., San Diego, California (P.-H.H.); Clinical Pharmacology, Pfizer Essential Health, Collegeville, Pennsylvania (R.A.); and Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide Research and Development, San Diego, California (C.-M.L., S.Y.)
| | - Shinji Yamazaki
- Clinical Pharmacology, Pfizer Japan Inc., Tokyo, Japan (C.O.); Clinical Pharmacology, Pfizer Inc., San Diego, California (P.-H.H.); Clinical Pharmacology, Pfizer Essential Health, Collegeville, Pennsylvania (R.A.); and Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide Research and Development, San Diego, California (C.-M.L., S.Y.)
| |
Collapse
|