1
|
Zhang J, Takeda K, Takeuchi M, Komatsu K, Zhu J, Yamaguchi Y. A Model-Based Trial Design With a Randomization Scheme Considering Pharmacokinetics Exposure for Dose Optimization in Oncology. Pharm Stat 2025; 24:e2454. [PMID: 39551616 DOI: 10.1002/pst.2454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 10/04/2024] [Accepted: 11/04/2024] [Indexed: 11/19/2024]
Abstract
The primary purpose of an oncology dose-finding trial for novel anticancer agents has been shifting from determining the maximum tolerated dose to identifying an optimal dose (OD) that is tolerable and therapeutically beneficial for subjects in subsequent clinical trials. In 2022, the FDA Oncology Center of Excellence initiated Project Optimus to reform the paradigm of dose optimization and dose selection in oncology drug development and issued a draft guidance. The guidance suggests that dose-finding trials include randomized dose-response cohorts of multiple doses and incorporate information on pharmacokinetics (PK) in addition to safety and efficacy data to select the OD. Furthermore, PK information could be a quick alternative to efficacy data to predict the minimum efficacious dose and decide the dose assignment. This article proposes a model-based trial design for dose optimization with a randomization scheme based on PK outcomes in oncology. A simulation study shows that the proposed design has advantages compared to the other designs in the percentage of correct OD selection and the average number of patients assigned to OD in various realistic settings.
Collapse
Affiliation(s)
- Jun Zhang
- Data Science, Astellas Pharma China, Beijing, China
| | - Kentaro Takeda
- Data Science, Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| | - Masato Takeuchi
- Early Development New Technologies, Astellas Pharma Inc, Tokyo, Japan
| | - Kanji Komatsu
- Early Development New Technologies, Astellas Pharma Inc, Tokyo, Japan
| | - Jing Zhu
- Data Science, Astellas Pharma China, Beijing, China
| | - Yusuke Yamaguchi
- Data Science, Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| |
Collapse
|
2
|
Zhou Y, Gong J, Deng X, Shen L, Ge A, Fan H, Ling J, Wu S, Liu L. A comprehensive exploration of adverse reactions to lapatinib: a disproportionate analysis based on the FAERS database. Expert Opin Drug Saf 2025:1-10. [PMID: 39985750 DOI: 10.1080/14740338.2025.2471515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Lapatinib, an FDA-approved tyrosine kinase inhibitor, treats HER2+ advanced/metastatic breast cancer. This study comprehensively analyzed its adverse reaction profile using FDA Adverse Event Reporting System (FAERS) to guide clinical use. RESEARCH DESIGN AND METHODS Adverse event (AE) reports for lapatinib from the second quarter of 2007 to the second quarter of 2024 in FAERS were analyzed using Reporting Odds Ratio (ROR), Proportional Reporting Ratio (PRR), Multi-item Gamma Poisson Shrinkage (MGPS) and Bayesian Confidence Propagation Neural Network (BCPNN) to identify AE signals. RESULTS Among 8300 AE reports, females (91.47%) and ages 40-59.9 (33.71%) were predominant. 20 system organ classifications (SOCs) were affected, with gastrointestinal disorders (ROR = 3.46) and skin disorders (ROR = 2.47) most significant. Based on the PT level, a total of 111 PTs were analyzed that met the four algorithms, including typical AEs such as diarrhea (n = 3410), vomiting (n = 856), and rash (n = 856), as well as some rare AEs that were not prompted by the drug inserts, such as neutropenia (n = 252), pericardial effusion (n = 43), lymphedema (n = 20). The majority of lapatinib-associated AEs had onset within 30 days (51%). CONCLUSIONS Lapatinib has a generally favorable safety profile, but gastrointestinal toxicity and dermatotoxicity require close monitoring to prevent serious AEs.
Collapse
Affiliation(s)
- Yao Zhou
- Department of Galactophore, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jie Gong
- Department of Galactophore, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xianguang Deng
- Department of Galactophore, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lele Shen
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Anqi Ge
- Department of Galactophore, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Hongqiao Fan
- Department of Aesthetic Plastic Surgery, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jie Ling
- Hunan Academy of Chinese Medicine, Changsha, Hunan, China
| | - Shiting Wu
- Department of Galactophore, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lifang Liu
- Department of Galactophore, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
3
|
Mukherjee S, Joshi V, Reddy KP, Singh N, Das P, Datta P. Biopharmaceutical and pharmacokinetic attributes to drive nanoformulations of small molecule tyrosine kinase inhibitors. Asian J Pharm Sci 2024; 19:100980. [PMID: 39640056 PMCID: PMC11617995 DOI: 10.1016/j.ajps.2024.100980] [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: 10/16/2023] [Revised: 06/13/2024] [Accepted: 06/29/2024] [Indexed: 12/07/2024] Open
Abstract
Buoyed by the discovery of small-molecule tyrosine kinase inhibitors (smTKIs), significant impact has been made in cancer chemotherapeutics. However, some of these agents still encounter off-target toxicities and suboptimal efficacies due to their inferior biopharmaceutical and/or pharmacokinetic properties. Almost all of these molecules exhibit significant inter- and intra-patient variations in plasma concentration-time profiles. Thus, therapeutic drug monitoring, dose adjustments and precision medicine are being contemplated by clinicians. Complex formulations or nanoformulation-based drug delivery systems offer promising approaches to provide drug encapsulation or spatiotemporal control over the release, overcoming the biopharmaceutical and pharmacokinetic limitations and improving the therapeutic outcomes. In this context, the present review comprehensively tabulates and critically analyzes all the relevant properties (T1/2, solubility, pKa, therapeutic index, IC50, metabolism etc.) of the approved smTKIs. A detailed appraisal is conducted on the advancements made in complex formulations of smTKIs, with a focus on strategies to enhance their pharmacokinetic profile, tumor targeting ability, and therapeutic efficacy. Various nanocarrier platforms, have been discussed, highlighting their unique features and potential applications in cancer therapy. Nanoformulations have been shown to improve area under the curve and peak plasma concentration, and reduce dosing frequency for several smTKIs in animal models. It is inferred that extensive efforts will be made in developing complex formulations of smTKIs in near future. There, the review concludes with key recommendations for the developing of smTKIs to facilitate early clinical translation.
Collapse
Affiliation(s)
| | | | - Kolimi Prashanth Reddy
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kolkata 700054, West Bengal, India
| | - Nidhi Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kolkata 700054, West Bengal, India
| | - Priyanka Das
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kolkata 700054, West Bengal, India
| | - Pallab Datta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kolkata 700054, West Bengal, India
| |
Collapse
|
4
|
Bi X, He Z, Luo Z, Huang W, Diao X, Ye J. Digital colloid-enhanced Raman spectroscopy for the pharmacokinetic detection of bioorthogonal drugs. Chem Sci 2024:d4sc02553a. [PMID: 39144465 PMCID: PMC11320124 DOI: 10.1039/d4sc02553a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/24/2024] [Indexed: 08/16/2024] Open
Abstract
Bioorthogonal drug molecules are currently gaining prominence for their excellent efficacy, safety and metabolic stability. Pharmacokinetic study is critical for understanding their mechanisms and guiding pharmacotherapy, which is primarily performed with liquid chromatography-mass spectrometry as the gold standard. For broader and more efficient applications in clinics and fundamental research, further advancements are especially desired in cheap and portable instrumentation as well as rapid and tractable pretreatment procedures. Surface-enhanced Raman spectroscopy (SERS) is capable of label-free detection of various molecules based on the spectral signatures with high sensitivity even down to a single-molecule level. But limited by irreproducibility at low concentrations and spectral interference in complex biofluids, SERS hasn't been widely applied for pharmacokinetics, especially in live animals. In this work, we propose a new method to quantify bioorthogonal drug molecules with signatures at the spectral silent region (SR) by the digital colloid-enhanced Raman spectroscopy (dCERS) technique. This method was first validated using 4-mercaptobenzonitrile in a mixture of analogous molecules, exhibiting reliable and specific identification capability based on the unique SR signature and Poisson-determined quantification accuracy. We further developed a single-step serum pretreatment method and successfully profiled the pharmacokinetic behavior of an anticancer drug, erlotinib, from animal studies. In a word, this method, superior in sensitivity, controllable accuracy, minimal background interference and facile pretreatment and measurement, promises diverse applications in fundamental studies and clinical tests of bioorthogonal drug molecules.
Collapse
Affiliation(s)
- Xinyuan Bi
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Shanghai Jiao Tong University Shanghai 200030 P. R. China
| | - Zhicheng He
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Shanghai Jiao Tong University Shanghai 200030 P. R. China
| | - Zhewen Luo
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Shanghai Jiao Tong University Shanghai 200030 P. R. China
| | - Wensi Huang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Shanghai Jiao Tong University Shanghai 200030 P. R. China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201210 P. R. China
| | - Xingxing Diao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201210 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Jian Ye
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Shanghai Jiao Tong University Shanghai 200030 P. R. China
- Institute of Medical Robotics, Shanghai Jiao Tong University Shanghai 200240 P. R. China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127 P. R. China
- Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200233 P. R. China
| |
Collapse
|
5
|
Abbass EM, Al-Karmalawy AA, Sharaky M, Khattab M, Alzahrani AYA, Hassaballah AI. Rational design and eco-friendly one-pot multicomponent synthesis of novel ethylidenehydrazineylthiazol-4(5H)-ones as potential apoptotic inducers targeting wild and mutant EGFR-TK in triple negative breast cancer. Bioorg Chem 2024; 142:106936. [PMID: 37890211 DOI: 10.1016/j.bioorg.2023.106936] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/15/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023]
Abstract
A novel series of ethylidenehydrazineylthiazol-4(5H)-ones were synthesized using various eco-friendly one-pot multicomponent synthetic techniques. The anticancer activity of compounds (4a-m) was tested against 11 cancer cell lines. While the IC50 of all compounds was evaluated against the most sensitive cell lines (MDA-MB-468 and FaDu). Our SAR study pinpointed that compound 4a, having a phenyl substituent, exhibited a significant growth inhibition % against all cancer cell lines. The frontier anticancer candidates against the MDA-MB-468 were also examined against the wild EGFR (EGFR-WT) and mutant EGFR (EGFR-T790M) receptors. Most of the synthesized compounds exhibited a higher inhibitory potential against EGFR-T790M than the wild type of EGFR. Remarkably, compound 4k exhibited the highest inhibitory activity against both EGFR-WT and EGFR-T790M with IC50 values (0.051 and 0.021 µM), respectively. The pro-apoptotic protein markers (p53, BAX, caspase 3, caspase 6, caspase 8, and caspase 9) and the anti-apoptotic key marker (BCL-2) were also measured to propose a mechanism of action for the compound 4k as an apoptotic inducer for MDA-MB-468. Investigation of the cell cycle arrest potential of compound 4k was also conducted on MDA-MB-468 cancer cells. We also evaluated the inhibitory activities of compounds (4a-m) against both EGFR-WT and EGFR-T790M using two different molecular docking processes.
Collapse
Affiliation(s)
- Eslam M Abbass
- Department of Chemistry, Faculty of Science, Ain Shams University, Abbassiya 11566, Cairo, Egypt
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza 12566, Egypt.
| | - Marwa Sharaky
- Cancer Biology Department, Pharmacology Unit, National Cancer Institute (NCI), Cairo University, Cairo, Egypt
| | - Muhammad Khattab
- Office of Research, University of Western Australia, Perth, Australia; Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries, National Research Centre, Cairo, Egypt
| | | | - Aya I Hassaballah
- Department of Chemistry, Faculty of Science, Ain Shams University, Abbassiya 11566, Cairo, Egypt
| |
Collapse
|
6
|
Sherif AY, Harisa GI, Alanazi FK. The Chimera of TPGS and Nanoscale Lipid Carriers as Lymphatic Drug Delivery Vehicles to Fight Metastatic Cancers. Curr Drug Deliv 2024; 21:525-543. [PMID: 37183467 DOI: 10.2174/1567201820666230512122825] [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: 10/07/2022] [Revised: 01/24/2023] [Accepted: 02/13/2023] [Indexed: 05/16/2023]
Abstract
The lymphatic system (LS) plays a crucial role in fluid balance, transportation of macromolecules, and immune response. Moreover, LS is a channel for microbial invasion and cancer metastasis. Particularly, solid tumors, including lung, breast, melanoma, and prostate cancers, are metastasized across highways of LS. Subsequently, the fabrication of chimeric lymphatic drug delivery systems (LDDS) is a promising strategy to fight cancer metastasis and control microbial pandemics. In this regard, LDDS, in terms of PEG-nanoscaled lipid carriers, elicited a revolution during the COVID-19 pandemic as cargoes for mRNA vaccines. The drug delivered by the lymphatic pathway escapes first-pass metabolism and enhances the drug's bioavailability. Ample approaches, including synthesis of prodrugs, trigging of chylomicron biosynthesis, and fabrication of nanocarriers, facilitate lymphatic drug delivery. Specifically, nanoscales lipid cargoes have the propensity to lymphatic trafficking. Interestingly, TPGSengineered nanoscale lipid cargoes enhance lymphatic trafficking, increase tissue permeation, and, specifically, uptake. Moreover, they overcome biological barriers, control biodistribution, and enhance organelles localization. Most anticancer agents are non-specific, have low bioavailability, and induced drug resistance. Therefore, TPGS-engineered nanoscale lipid chimeras improve the therapeutic impact of anticancer agents. This review highlights lymphatic cancer metastasis, nanoscales lipid cargoes as LDDS, and their influence on lymphatic trafficking, besides the methods of LDD studies.
Collapse
Affiliation(s)
- Abdelrahman Y Sherif
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Gamaleldin I Harisa
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Biochemistry and Molecular Biology, College of Pharmacy, Al-Azhar University, Nasr City, Cairo, Egypt
| | - Fars K Alanazi
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
7
|
Takeda K, Zhu J, Li R, Yamaguchi Y. A Bayesian optimal interval design for dose optimization with a randomization scheme based on pharmacokinetics outcomes in oncology. Pharm Stat 2023; 22:1104-1115. [PMID: 37545018 DOI: 10.1002/pst.2332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/15/2023] [Accepted: 07/26/2023] [Indexed: 08/08/2023]
Abstract
The primary objective of an oncology dose-finding trial for novel therapies, such as molecularly targeted agents and immune-oncology therapies, is to identify the optimal dose (OD) that is tolerable and therapeutically beneficial for subjects in subsequent clinical trials. Pharmacokinetic (PK) information is considered an appropriate indicator for evaluating the level of drug intervention in humans from a pharmacological perspective. Several novel anticancer agents have been shown to have significant exposure-efficacy relationships, and some PK information has been considered an important predictor of efficacy. This paper proposes a Bayesian optimal interval design for dose optimization with a randomization scheme based on PK outcomes in oncology. A simulation study shows that the proposed design has advantages compared to the other designs in the percentage of correct OD selection and the average number of patients allocated to OD in various realistic settings.
Collapse
Affiliation(s)
- Kentaro Takeda
- Data Science, Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| | - Jing Zhu
- Data Science, Astellas Pharma China, Beijing, China
| | - Ran Li
- Data Science, Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| | - Yusuke Yamaguchi
- Data Science, Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| |
Collapse
|
8
|
Ma Y, Chen Q, Zhang Y, Xue J, Liu Q, Zhao Y, Yang Y, Huang Y, Fang W, Hou Z, Li S, Wang J, Zhang L, Zhao H. Pharmacokinetics, safety, tolerability, and feasibility of apatinib in combination with gefitinib in stage IIIB-IV EGFR-mutated non-squamous NSCLC: a drug-drug interaction study. Cancer Chemother Pharmacol 2023; 92:411-418. [PMID: 37518060 DOI: 10.1007/s00280-023-04563-2] [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: 03/15/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023]
Abstract
PURPOSE Apatinib combined with gefitinib was proven to benefit advanced EGFR-mutant NSCLC patients in first-line treatment. This study aimed to evaluate the drug-drug interaction of gefitinib and apatinib when coadministered in EGFR-mutated NSCLC patients. METHODS In this phase 1b, multi-center, open-label, fixed-sequence study, the drug-drug interaction of gefitinib and apatinib was evaluated when coadministered in EGFR-mutated NSCLC patients. Patients received single-agent apatinib 500 mg QD on days 1-4. Gefitinib 250 mg QD was given on days 5-15 and combined with apatinib 500 mg QD on days 12-15. Serial blood samples were drawn on days 4 and 15. The plasma concentrations and other pharmacokinetics parameters were measured for apatinib with and without gefitinib. RESULTS The study enrolled 22 patients and 20 were analyzed for pharmacokinetics. There were no distinct differences in apatinib Cmax and AUC0-τ with versus without gefitinib (geometric LSM ratio, 0.96 [90% CI 0.84-1.10] for Cmax and 1.12 [90% CI 0.96-1.30] for AUC0-τ). Similar PFS and grade of treatment-emergent adverse events (TEAEs) were found between different Cmax and AUC0-τ of apatinib and gefitinib at 500 mg apatinib and 250 mg gefitinib dose levels. CONCLUSIONS Apatinib pharmacokinetics parameters were not significantly changed when coadministered with gefitinib. All TEAEs were manageable, and there was no need to change the dose level when combining apatinib and gefitinib (ClinicalTrials.gov identifier: NCT04390984, May 18, 2020).
Collapse
Affiliation(s)
- Yuxiang Ma
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 651 Dongfeng Road East, Guangdong, 510060, Guangzhou, China
| | - Qun Chen
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 651 Dongfeng Road East, Guangdong, 510060, Guangzhou, China
| | - Yang Zhang
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 651 Dongfeng Road East, Guangdong, 510060, Guangzhou, China
| | - Jinhui Xue
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 651 Dongfeng Road East, Guangdong, 510060, Guangzhou, China
| | - Qianwen Liu
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 651 Dongfeng Road East, Guangdong, 510060, Guangzhou, China
| | - Yuanyuan Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 651 Dongfeng Road East, Guangdong, 510060, Guangzhou, China
| | - Yunpeng Yang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 651 Dongfeng Road East, Guangdong, 510060, Guangzhou, China
| | - Yan Huang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 651 Dongfeng Road East, Guangdong, 510060, Guangzhou, China
| | - Wenfeng Fang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 651 Dongfeng Road East, Guangdong, 510060, Guangzhou, China
| | - Zhiguo Hou
- Department of Medical Affairs, Jiangsu Hengrui Pharmaceuticals Co., Ltd., No.1288 Haike Road, Pudong New Area, Shanghai, 200120, China
| | - Shaorong Li
- Department of Clinical Pharmacology, Clinical Research and Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd., No. 1288 Haike Road, Pudong New Area, Shanghai, China
| | - Jing Wang
- Department of Clinical Pharmacology, Clinical Research and Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd., No. 1288 Haike Road, Pudong New Area, Shanghai, China
| | - Li Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 651 Dongfeng Road East, Guangdong, 510060, Guangzhou, China.
| | - Hongyun Zhao
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 651 Dongfeng Road East, Guangdong, 510060, Guangzhou, China.
| |
Collapse
|
9
|
Chauhan G, Wang X, Yousry C, Gupta V. Scalable Production and In Vitro Efficacy of Inhaled Erlotinib Nanoemulsion for Enhanced Efficacy in Non-Small Cell Lung Cancer (NSCLC). Pharmaceutics 2023; 15:pharmaceutics15030996. [PMID: 36986858 PMCID: PMC10054254 DOI: 10.3390/pharmaceutics15030996] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is a global concern as one of the leading causes of cancer deaths. The treatment options for NSCLC are limited to systemic chemotherapy, administered either orally or intravenously, with no local chemotherapies to target NSCLC. In this study, we have prepared nanoemulsions of tyrosine kinase inhibitor (TKI), erlotinib, using the single step, continuous manufacturing, and easily scalable hot melt extrusion (HME) technique without additional size reduction step. The formulated nanoemulsions were optimized and evaluated for their physiochemical properties, in vitro aerosol deposition behavior, and therapeutic activity against NSCLC cell lines both in vitro and ex vivo. The optimized nanoemulsion showed suitable aerosolization characteristics for deep lung deposition. The in vitro anti-cancer activity was tested against the NSCLC A549 cell line which exhibited 2.8-fold lower IC50 for erlotinib-loaded nanoemulsion, as compared to erlotinib-free solution. Furthermore, ex vivo studies using a 3D spheroid model also revealed higher efficacy of erlotinib-loaded nanoemulsion against NSCLC. Hence, inhalable nanoemulsion can be considered as a potential therapeutic approach for the local lung delivery of erlotinib to NSCLC.
Collapse
Affiliation(s)
- Gautam Chauhan
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Xuechun Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Carol Yousry
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Kasr El-Aini, Cairo 11562, Egypt
| | - Vivek Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| |
Collapse
|
10
|
Diniz F, Azevedo M, Sousa F, Osório H, Campos D, Sampaio P, Gomes J, Sarmento B, Reis CA. Polymeric nanoparticles targeting Sialyl-Tn in gastric cancer: A live tracking under flow conditions. Mater Today Bio 2022; 16:100417. [PMID: 36105678 PMCID: PMC9465339 DOI: 10.1016/j.mtbio.2022.100417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 11/30/2022]
Abstract
Drug delivery using nanoparticles (NPs) represents a potential approach for therapy in cancer, such gastric cancer (GC) due to their targeting ability and controlled release properties. The use of advanced nanosystems that deliver anti-cancer drugs specifically to tumor cells may strongly rely on the expression of cancer-associated targets. Glycans aberrantly expressed by cancer cells are attractive targets for such delivery strategy. Sialylated glycans, such as Sialyl-Tn (STn) are aberrantly expressed in several epithelial tumors, including GC, being a potential target for a delivery nanosystem. The aim of this study was the development of NPs surface-functionalized with a specific antibody targeting the STn glycan and further evaluate this nanosystem effectiveness regarding its specificity and recognition capacity. Our results showed that the NPs surface-functionalized with anti-STn antibody efficiently are recognized by cells displaying the cancer-associated STn antigen under static and live cell monitoring flow conditions. This uncovers the potential use of such NPs for drug delivery in cancer. However, flow exposure was disclosed as an important biomechanical parameter to be taken into consideration. Here we presented an innovative and successful methodology to live track the NPs targeting STn antigen under shear stress, simulating the physiological flow. We demonstrate that unspecific binding of NPs agglomerates did not occur under flow conditions, in contrast with static assays. This robust approach can be applied for in vitro drug studies, giving valuable insights for in vivo studies.
Collapse
Affiliation(s)
- Francisca Diniz
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP -Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- ICBAS- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Maria Azevedo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Flávia Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB- Instituto Nacional de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- CESPU-IUCS, 4585-116 Gandra, Portugal
| | - Hugo Osório
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP -Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Diana Campos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP -Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Paula Sampaio
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Joana Gomes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP -Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB- Instituto Nacional de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- CESPU-IUCS, 4585-116 Gandra, Portugal
| | - Celso A. Reis
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP -Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- ICBAS- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- FMUP- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Corresponding author. i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
| |
Collapse
|
11
|
AlRasheed H, Almomen A, Aljohar HI, Arafah M, Almotawa RY, Alossaimi MA, Alzoman NZ. Aspartames Alter Pharmacokinetics Parameters of Erlotinib and Gefitinib and Elevate Liver Enzymes in Wistar Rats. Pharmaceuticals (Basel) 2022; 15:1400. [PMID: 36422530 PMCID: PMC9699004 DOI: 10.3390/ph15111400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/06/2022] [Accepted: 11/07/2022] [Indexed: 01/26/2025] Open
Abstract
Background: Erlotinib (ERL) and gefitinib (GEF) are extensively metabolized by CYP450 enzymes. Aspartame (ASP), an artificial sweetener, induces CYP2E1 and CYP3A2 enzymes in the brain and could increase liver enzymes. In this work, the influence of ASP on the pharmacokinetics (PK) of ERL and GEF in Wistar rats was evaluated. Methods: The PKs of ERL and GEF were evaluated after receiving 175 mg/kg or 1000 mg/kg of ASP for four weeks using UPLC-MS/MS. Levels of liver enzymes after four weeks of ASP consumption were also evaluated. Results: ASP 175 mg/kg was able to significantly alter levels of Cmax (36% increase for ERL, 38% decrease for GEF), AUC0-72 (205% increase for ERL, 41% increase for GEF), and AUC0-∞ (112% increase for ERL, 14% increase for GEF). Moreover, ASP 175 mg/kg decreased the apparent oral clearance ERL and GEF by 58% and 13%, respectively. ASP 1000 mg/kg increased Cmax of ERL by 159% and decreased GEF's Cmax by and 73%. Both AUC0-72 and AUC0-∞ were increased by ASP 1000 for ERL and decreased for GEF. CL/F decreased by 64% for ERL and increased by 38.8% for GEF. Moreover, data indicated that ASP significantly increased levels of liver enzymes within two weeks of administration. Conclusions: Although ASP 175 and 1000 mg/kg alter ERL and GEF PKs parameters, ASP 1000 mg/kg has the highest impact on most parameters. ASP 1000 mg/kg also can significantly increase activities of liver enzymes indicating the possibility of inducing liver injury. Therefore, it might be of clinical importance to avoid the administration of aspartame containing products while on ERL or GEF therapy.
Collapse
Affiliation(s)
- Hajer AlRasheed
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia
| | - Aliyah Almomen
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia
| | - Haya I. Aljohar
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia
| | - Maria Arafah
- Department of Pathology, College of Medicine, King Saud University, P.O. Box 2925, Riyadh 11421, Saudi Arabia
| | - Rana Y. Almotawa
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia
| | - Manal A. Alossaimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdelaziz University, P.O. Box 173, Alkharj 11942, Saudi Arabia
| | - Nourah Z. Alzoman
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia
| |
Collapse
|
12
|
Cranberry Ingestion Modulated Drug Transporters and Metabolizing Enzymes: Gefitinib Used as a Probe Substrate in Rats. Molecules 2022; 27:molecules27185772. [PMID: 36144507 PMCID: PMC9501900 DOI: 10.3390/molecules27185772] [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: 08/08/2022] [Revised: 09/03/2022] [Accepted: 09/06/2022] [Indexed: 12/03/2022] Open
Abstract
Cranberry, a polyphenol-rich functional food, is commonly used for the prophylaxis of urinary tract infections. Gefitinib, an anticancer agent clinically prescribed to treat non-small-cell lung cancer, is a substrate of P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP), and metabolized mainly by cytochrome P450 (CYP) 3A4 and CYP2D6. This study used gefitinib as a probe substrate to investigate the modulation of cranberry on P-gp, BCRP, CYP3A4 and CYP2D6. Rats were administered gefitinib with and without 5.0 g/kg of cranberry as juice (CJ). The concentration of gefitinib in serum was determined by LC-MS/MS. The results showed that CJ significantly increased the Cmax and AUC0-t of gefitinib by 28% and 55%, respectively. Mechanism studies indicated that CJ activated P-gp, and cranberry metabolites (CM) inhibited CYP2D6. Moreover, the protein level of P-gp in rat enterocytes was decreased, whereas that in hepatocytes was increased. In addition, the protein levels of BCRP, CYP3A4 and CYP2D6 in enterocytes and hepatocytes were decreased. In conclusion, CJ ingestion affected the activities and protein levels of P-gp, BCRP, CYP3A4 and CYP2D6.
Collapse
|
13
|
Zhao T, Li X, Chen Y, Du J, Chen X, Wang D, Wang L, Zhao S, Wang C, Meng Q, Sun H, Liu K, Wu J. Risk assessment and molecular mechanism study of drug-drug interactions between rivaroxaban and tyrosine kinase inhibitors mediated by CYP2J2/3A4 and BCRP/P-gp. Front Pharmacol 2022; 13:914842. [PMID: 36071847 PMCID: PMC9441481 DOI: 10.3389/fphar.2022.914842] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/14/2022] [Indexed: 11/21/2022] Open
Abstract
Cancer patients generally has a high risk of thrombotic diseases. However, anticoagulant therapy always aggravates bleeding risks. Rivaroxaban is one of the most widely used direct oral anticoagulants, which is used as anticoagulant treatment or prophylaxis in clinical practice. The present study aimed to systemically estimate the combination safety of rivaroxaban with tyrosine kinase inhibitors (TKIs) based on human cytochrome P450 (CYPs) and efflux transporters and to explore the drug–drug interaction (DDI) mechanisms in vivo and in vitro. In vivo pharmacokinetic experiments and in vitro enzyme incubation assays and bidirectional transport studies were conducted. Imatinib significantly increased the rivaroxaban Cmax value by 90.43% (p < 0.05) and the area under the curve value by 119.96% (p < 0.01) by inhibiting CYP2J2- and CYP3A4-mediated metabolism and breast cancer resistance protein (BCRP)- and P-glycoprotein (P-gp)-mediated efflux transportation in the absorption phase. In contrast, the combination of sunitinib with rivaroxaban reduced the exposure in vivo by 62.32% (p < 0.05) and the Cmax value by 72.56% (p < 0.05). In addition, gefitinib potently inhibited CYP2J2- and CYP3A4-mediated rivaroxaban metabolism with Ki values of 2.99 μΜ and 4.91 μΜ, respectively; however, it almost did not affect the pharmacokinetics of rivaroxaban in vivo. Taken together, clinically significant DDIs were observed in the combinations of rivaroxaban with imatinib and sunitinib. Imatinib increased the bleeding risks of rivaroxaban, while sunitinib had a risk of reducing therapy efficiency. Therefore, more attention should be paid to aviod harmful DDIs in the combinations of rivaroxaban with TKIs.
Collapse
Affiliation(s)
- Tingting Zhao
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Xuening Li
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Yanwei Chen
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jie Du
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Xiaodong Chen
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Dalong Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Liyan Wang
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shan Zhao
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Changyuan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, China
| | - Qiang Meng
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, China
| | - Huijun Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, China
| | - Kexin Liu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, China
| | - Jingjing Wu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, China
- *Correspondence: Jingjing Wu,
| |
Collapse
|
14
|
pH-responsive albumin-coated biopolymeric nanoparticles with lapatinab for targeted breast cancer therapy. BIOMATERIALS ADVANCES 2022; 139:213039. [PMID: 35908475 DOI: 10.1016/j.bioadv.2022.213039] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/14/2022] [Accepted: 07/17/2022] [Indexed: 12/17/2022]
Abstract
One can enhance the therapeutic index of anti-cancer drugs using albumin as a tumor homing agent for targeted cancer therapy. Herein, we sought to load lapatinib (LAPA) into small albumin-coated biopolymeric (poly-lactic co-glycolic acid (PLGA)) nanoparticles (APL NPs) by an emulsification method to improve the anti-tumor efficacy of lapatinib. The prepared APL NPs exhibited a small spherical core with an average diameter of 120.5 ± 10.2 nm with a narrow particle size distribution, high drug loading capacity (LC of 9.65 ± 1.53 %), good entrapment efficiency (EE of 75.55 ± 3.25 %), enhanced colloidal stability and a pH-responsive controlled drug release profile. Their cell-uptake and cancer cell growth inhibition were significantly higher compared to free LAPA and uncoated PLGA-LAPA (UPL) NPs, most likely because aggressive breast tumor cells over-express albumin receptors and utilize albumin as nutrient source for their growth. In addition, APL NPs possessed enhanced tumor accumulation and prolonged blood residence time compared to free LAPA and UPL NPs, allowing for potent tumor growth inhibition while exhibiting excellent biosafety. In short, the current study exploited a new and simple strategy to concurrently improve the safety and efficacy of LAPA for breast cancer treatment.
Collapse
|
15
|
Brain Metastasis Treatment: The Place of Tyrosine Kinase Inhibitors and How to Facilitate Their Diffusion across the Blood-Brain Barrier. Pharmaceutics 2021; 13:pharmaceutics13091446. [PMID: 34575525 PMCID: PMC8468523 DOI: 10.3390/pharmaceutics13091446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/28/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022] Open
Abstract
The incidence of brain metastases has been increasing constantly for the last 20 years, because of better control of metastases outside the brain, and the failure of most drugs to cross the blood–brain barrier at relevant pharmacological concentrations. Recent advances in the molecular biology of cancer have led to the identification of numerous molecular alterations, some of them targetable with the development of specific targeted therapies, including tyrosine kinase inhibitors. In this narrative review, we set out to describe the state-of-the-art in the use of tyrosine kinase inhibitors for the treatment of melanoma, lung cancer, and breast cancer brain metastases. We also report preclinical and clinical pharmacological data on brain exposure to tyrosine kinase inhibitors after oral administration and describe the most recent advances liable to facilitate their penetration of the blood–brain barrier at relevant concentrations and limit their physiological efflux.
Collapse
|
16
|
Microwave-assisted solid-phase synthesis of nitrogen-doping carbon dot with good solvent compatibility and its sensing of sunitinib. Anal Bioanal Chem 2021; 413:6435-6447. [PMID: 34401928 DOI: 10.1007/s00216-021-03609-9] [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: 06/09/2021] [Revised: 07/19/2021] [Accepted: 08/06/2021] [Indexed: 10/20/2022]
Abstract
Microwave-assisted solid-phase synthesis method was simple, convenient, and fast, and herein adopted to produce nitrogen-doping carbon dots (N-CDs) in only 3 min. The N-CDs possessed high fluorescence quantum yield up to 15.9% with satisfactory stability to the environmental pH, ionic strength, and ultraviolet radiation. Particularly, the N-CDs had excellent dispersibility in both water and water-compatible organic solvents with similar fluorescence properties. Sunitinib, a small-molecule tyrosine inhibitor effective for some solid tumors, was found to quench the fluorescence of N-CDs in these media via the inner-filter effect. Hence, it was convenient to combine the proper sample pretreatment with the N-CD probe for sensing sunitinib avoiding the medium incompatibility problem. For rat plasma sample, salting-out liquid-liquid extraction was employed to minimize the sample matrix and concentrate the target sunitinib from aqueous to acetonitrile. The fluorescence detection of sunitinib was then achieved in acetonitrile by the addition of the proper amount of N-CDs. The method provided a good linearity of 0.1 μg/mL to 7 μg/mL with a limit of detection of 30 ng/mL, which met the requirement of the therapeutic drug monitoring of sunitinib. The developed method was potential for on-site detection of sunitinib.
Collapse
|
17
|
Erdoğar N, Akkın S, Varan G, Bilensoy E. Erlotinib complexation with randomly methylated β-cyclodextrin improves drug solubility, intestinal permeability, and therapeutic efficacy in non-small cell lung cancer. Pharm Dev Technol 2021; 26:797-806. [PMID: 34219578 DOI: 10.1080/10837450.2021.1946695] [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: 10/20/2022]
Abstract
The purpose of this study was to investigate the impact of anticancer drug erlotinib-randomly methylated-β-cyclodextrin complex (ERL-RAMEB CD) on drug solubility and dissolution rate. Phase solubility study showed erlotinib displayed maximum solubility in RAMEB CD solution and the stability constant (Kc) was calculated to be 370 ± 16 M-1. The optimal formulation was obtained with ERL-RAMEB CD in a 1:1 molar ratio using the co-lyophilization technique. Differential scanning calorimetry (DSC) and Scanning electron microscopy (SEM) verified the inclusion of complex formation. In vitro dissolution study confirmed ERL-RAMEB CD as a favorable approach to increase drug dissolution with a 1.5-fold increase than free ERL at 1 h. An improved dissolution with ∼88.4% drug release at 1 h was observed, in comparison with Erlotinib with ∼58.7% release in 45 min. The in vitro cytotoxicity results indicated that the ERL-RAMEB CD inclusion complex reduced cell viability than free erlotinib. Caco-2 cell uptake that is indicative of drug intestinal permeability resulted in a 5-fold higher uptake of ERL-RAMEB CD inclusion complex than the ERL solution. Hence, ERL-RAMEB CD complexation displays a strong potential to increase dissolution and permeability of erlotinib leading eventually to enhanced oral bioavailability.
Collapse
Affiliation(s)
- Nazlı Erdoğar
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Safiye Akkın
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Gamze Varan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Erem Bilensoy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
18
|
Kaehler M, Cascorbi I. Pharmacogenomics of Impaired Tyrosine Kinase Inhibitor Response: Lessons Learned From Chronic Myelogenous Leukemia. Front Pharmacol 2021; 12:696960. [PMID: 34262462 PMCID: PMC8273252 DOI: 10.3389/fphar.2021.696960] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 06/17/2021] [Indexed: 12/25/2022] Open
Abstract
The use of small molecules became one key cornerstone of targeted anti-cancer therapy. Among them, tyrosine kinase inhibitors (TKIs) are especially important, as they were the first molecules to proof the concept of targeted anti-cancer treatment. Since 2001, TKIs can be successfully used to treat chronic myelogenous leukemia (CML). CML is a hematologic neoplasm, predominantly caused by reciprocal translocation t(9;22)(q34;q11) leading to formation of the so-called BCR-ABL1 fusion gene. By binding to the BCR-ABL1 kinase and inhibition of downstream target phosphorylation, TKIs, such as imatinib or nilotinib, can be used as single agents to treat CML patients resulting in 80 % 10-year survival rates. However, treatment failure can be observed in 20-25 % of CML patients occurring either dependent or independent from the BCR-ABL1 kinase. Here, we review approved TKIs that are indicated for the treatment of CML, their side effects and limitations. We point out mechanisms of TKI resistance focusing either on BCR-ABL1-dependent mechanisms by summarizing the clinically observed BCR-ABL1-mutations and their implications on TKI binding, as well as on BCR-ABL1-independent mechanisms of resistances. For the latter, we discuss potential mechanisms, among them cytochrome P450 implications, drug efflux transporter variants and expression, microRNA deregulation, as well as the role of alternative signaling pathways. Further, we give insights on how TKI resistance could be analyzed and what could be learned from studying TKI resistance in CML in vitro.
Collapse
Affiliation(s)
| | - Ingolf Cascorbi
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
19
|
Ezhilarasan D. Advantages and challenges in nanomedicines for chronic liver diseases: A hepatologist's perspectives. Eur J Pharmacol 2021; 893:173832. [PMID: 33359144 DOI: 10.1016/j.ejphar.2020.173832] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 12/01/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022]
Abstract
Chronic liver diseases (CLD) are responsible for significant morbidity and mortality worldwide. CLD patients are at a high risk of developing progressive liver fibrosis, cirrhosis, hepatocellular carcinoma (HCC), and subsequent liver failure. To date, there is no specific and effective therapies exist for patients with various forms of CLD. The application of nanotechnology has emerged as a rapidly developing area of interest for the safe and target-specific delivery of poorly aqueous soluble hepatoprotective agents and nucleic acids (siRNA/miRNAs) in CLD. The nanoparticle combination improves bioavailability and plasma stability of drugs with poor aqueous solubility. However, the extent of successful functional delivery of nanoparticles into hepatocytes is often surprisingly low. High Kupffer cells interaction reduces the nanomedicine efficacy. During fibrosis, the extracellular matrix accumulation in the perisinusoidal space restricts nanoparticle delivery to hepatocytes. The availability and uptake of nanoparticles exposure to different cells in the liver microenvironment is as Kupffer cells > sinusoidal endothelial cells > HSCs > hepatocytes. The most widely used strategy to reduce nanoparticles and macrophages interaction is to coat the particle surface with polyethylene glycol. The cationic charged nanoparticles have increased hepatocyte delivery by increased cellular interaction by disrupting the endosomal system via their pH buffering capacity. The immune clearance and toxicity of nanoparticles are mainly unpredictable. Therefore, more elaborate knowledge on exact cellular uptake and intracellular accumulation, trafficking, and endosomal sorting of nanoparticle is the need of the hour to improve the rational carrier design.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Drug and Molecular Medicine Laboratory (The Blue Lab), Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), No.162, PH Road, Chennai, Tamil Nadu, 600 077, India.
| |
Collapse
|
20
|
Wang M, Du Q, Zuo L, Xue P, Lan C, Sun Z. Metabolism and Distribution of Novel Tumor Targeting Drugs In Vivo. Curr Drug Metab 2020; 21:996-1008. [PMID: 33183197 DOI: 10.2174/1389200221666201112110638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/30/2020] [Accepted: 09/22/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND As a new tumor therapy, targeted therapy is becoming a hot topic due to its high efficiency and low toxicity. Drug effects of targeted tumor drugs are closely related to pharmacokinetics, so it is important to understand their distribution and metabolism in vivo. METHODS A systematic review of the literature on the metabolism and distribution of targeted drugs over the past 20 years was conducted, and the pharmacokinetic parameters of approved targeted drugs were summarized in combination with the FDA's drug instructions. Targeting drugs are divided into two categories: small molecule inhibitors and monoclonal antibodies. Novel targeting drugs and their mechanisms of action, which have been developed in recent years, are summarized. The distribution and metabolic processes of each drug in the human body are reviewed. RESULTS In this review, we found that the distribution and metabolism of small molecule kinase inhibitors (TKI) and monoclonal antibodies (mAb) showed different characteristics based on the differences of action mechanism and molecular characteristics. TKI absorbed rapidly (Tmax ≈ 1-4 h) and distributed in large amounts (Vd > 100 L). It was mainly oxidized and reduced by cytochrome P450 CYP3A4. However, due to the large molecular diameter, mAb was distributed to tissues slowly, and the volume of distribution was usually very low (Vd < 10 L). It was mainly hydrolyzed and metabolized into peptides and amino acids by protease hydrolysis. In addition, some of the latest drugs are still in clinical trials, and the in vivo process still needs further study. CONCLUSION According to the summary of the research progress of the existing targeting drugs, it is found that they have high specificity, but there are still deficiencies in drug resistance and safety. Therefore, the development of safer and more effective targeted drugs is the future research direction. Meanwhile, this study also provides a theoretical basis for clinical accurate drug delivery.
Collapse
Affiliation(s)
- Mengli Wang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qiuzheng Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lihua Zuo
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Xue
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chao Lan
- Department of Emergency Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhi Sun
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
21
|
Honeywell RJ, Kathmann I, Giovannetti E, Tibaldi C, Smit EF, Rovithi MN, Verheul HM, Peters GJ. Epithelial Transfer of the Tyrosine Kinase Inhibitors Erlotinib, Gefitinib, Afatinib, Crizotinib, Sorafenib, Sunitinib, and Dasatinib: Implications for Clinical Resistance. Cancers (Basel) 2020; 12:cancers12113322. [PMID: 33182766 PMCID: PMC7696666 DOI: 10.3390/cancers12113322] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/07/2020] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Tyrosine kinase inhibitors (TKIs) specifically inhibit phosphorylation of signaling pathways of cancer cells, thereby inhibiting their growth. They are characterized by a poor solubility and high protein binding, leading to a large variability in gut uptake after oral administration and variation in the clinical efficacy. We used the CaCo2 gut epithelial model to characterize the gut absorption of 7 TKIs and observed a large variation in apical/basolateral (mimicking gut/blood) transfer, with 4 TKIs showing a negative and 3 a neutral transfer. A highly negative transfer may lead to pharmacokinetic resistance. Intracellular uptake of TKIs was high for sunitinib and crizotinib, intermediate for gefitinib, dasatinib and sorafenib, low for afatinib and not detectable for erlotinib. These properties may explain a high red blood cell to plasma ratio for most TKIs investigated. Although TKIs are poorly absorbed the latter property may compensate for this. Abstract Background: tyrosine kinase inhibitors (TKIs) inhibit phosphorylation of signaling proteins. TKIs often show large variations in the clinic due to poor pharmacology, possibly leading to resistance. We compared gut absorption of inhibitors of epidermal growth factor receptor (erlotinib, gefitinib, and afatinib), ALK-cMET (crizotinib), PDGFR/BCR-Abl (dasatinib), and multikinase inhibitors (sunitinib and sorafenib). In clinical samples, we measured the disposition of each compound within various blood compartments. Methods: we used an optimized CaCo2 gut epithelial model to characterize 20 µM TKI absorption. The apical/basolateral transfer is considered to represent the gut/blood transfer. Drugs were measured using LC-MS/MS. Results: sorafenib and sunitinib showed the highest apical/basolateral transfer (Papp 14.1 and 7.7 × 10−6 cm/s, respectively), followed by dasatinib (3.4), afatinib (1.5), gefitinib (0.38), erlotinib (0.13), and crizotinib (n.d.). However, the net absorptions for dasatinib, afatinib, crizotinib, and erlotinib were highly negative (efflux ratios >5) or neutral/negative, sorafenib (0.86), gefitinib (1.0), and sunitinib (1.6). A high negative absorption may result in resistance because of a poor exposure of tissues to the drug. Accumulation of the TKIs at the end of the transfer period (A->B) was not detectable for erlotinib, very low for afatinib 0.45 pmol/μg protein), followed by gefitinib (0.79), dasatinib (1.1), sorafenib (1.65), and crizotinib (2.11), being highest for sunitinib (11.9). A similar pattern was found for accumulation of these drugs in other colon cell lines, WiDr and HT29. In clinical samples, drugs accumulated consistently in red blood cells; blood to plasma ratios were all >3 (sorafenib) or over 30 for erlotinib. Conclusions: TKIs are consistently poorly absorbed, but accumulation in red blood cells seems to compensate for this.
Collapse
Affiliation(s)
- Richard J. Honeywell
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands; (R.J.H.); (I.K.); (E.G.); (M.N.R.)
- Department of Pharmacy, Amsterdam UMC, VU University Medical Center, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands
| | - Ietje Kathmann
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands; (R.J.H.); (I.K.); (E.G.); (M.N.R.)
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands; (R.J.H.); (I.K.); (E.G.); (M.N.R.)
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per la Scienza, 56017 Pisa, Italy
| | - Carmelo Tibaldi
- Division of Oncology, Department of Oncology, S. Luca Hospital, 55100 Lucca, Italy;
| | - Egbert F. Smit
- Department of Thoracic Oncology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands;
| | - Maria N. Rovithi
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands; (R.J.H.); (I.K.); (E.G.); (M.N.R.)
| | - Henk M.W. Verheul
- Department of Medical Oncology, Radboud University Medical Center, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, The Netherlands;
| | - Godefridus J. Peters
- Department of Medical Oncology, Amsterdam UMC, VU University Medical Center, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands; (R.J.H.); (I.K.); (E.G.); (M.N.R.)
- Department of Biochemistry, Medical University of Gdansk, 80-211 Gdansk, Poland
- Correspondence: ; Tel.: +31-20-444-2633
| |
Collapse
|
22
|
Eliesen GAM, van Hove H, Meijer MH, van den Broek PHH, Pertijs J, Roeleveld N, van Drongelen J, Russel FGM, Greupink R. Toxicity of anticancer drugs in human placental tissue explants and trophoblast cell lines. Arch Toxicol 2020; 95:557-571. [PMID: 33083868 PMCID: PMC7870638 DOI: 10.1007/s00204-020-02925-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 10/05/2020] [Indexed: 12/28/2022]
Abstract
The application of anticancer drugs during pregnancy is associated with placenta-related adverse pregnancy outcomes. Therefore, it is important to study placental toxicity of anticancer drugs. The aim of this study was to compare effects on viability and steroidogenesis in placental tissue explants and trophoblast cell lines. Third trimester placental tissue explants were exposed for 72 h (culture day 4–7) to a concentration range of doxorubicin, paclitaxel, cisplatin, carboplatin, crizotinib, gefitinib, imatinib, or sunitinib. JEG-3, undifferentiated BeWo, and syncytialised BeWo cells were exposed for 48 h to the same drugs and concentrations. After exposure, tissue and cell viability were assessed and progesterone and estrone levels were quantified in culture medium. Apart from paclitaxel, all compounds affected both cell and tissue viability at clinically relevant concentrations. Paclitaxel affected explant viability moderately, while it reduced cell viability by 50% or more in all cell lines, at 3–10 nM. Doxorubicin (1 µM) reduced viability in explants to 83 ± 7% of control values, whereas it fully inhibited viability in all cell types. Interference with steroid release in explants was difficult to study due to large variability in measurements, but syncytialised BeWo cells proved suitable for this purpose. We found that 1 µM sunitinib reduced progesterone release to 76 ± 6% of control values, without affecting cell viability. While we observed differences between the models for paclitaxel and doxorubicin, most anticancer drugs affected viability significantly in both placental explants and trophoblast cell lines. Taken together, the placenta should be recognized as a potential target organ for toxicity of anticancer drugs.
Collapse
Affiliation(s)
- Gaby A M Eliesen
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, (Route 137), PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Hedwig van Hove
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, (Route 137), PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Maartje H Meijer
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, (Route 137), PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Petra H H van den Broek
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, (Route 137), PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Jeanne Pertijs
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, (Route 137), PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Nel Roeleveld
- Department for Health Evidence, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Joris van Drongelen
- Department of Obstetrics and Gynecology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frans G M Russel
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, (Route 137), PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Rick Greupink
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, (Route 137), PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| |
Collapse
|
23
|
Impact of histamine type-2 receptor antagonists on the anticancer efficacy of gefitinib in patients with non-small cell lung cancer. Eur J Clin Pharmacol 2020; 77:381-388. [PMID: 33029650 DOI: 10.1007/s00228-020-03013-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 10/01/2020] [Indexed: 01/23/2023]
Abstract
PURPOSE Gefitinib is one of the standard treatments for non-small cell lung cancer (NSCLC) with epidermal growth factor receptor mutations. It has been reported that acid suppressants (AS) decrease the anti-tumor effect of gefitinib by reducing its solubility. AS is sometimes necessary in cancer patients; however, previous reports have not shown the most compatible AS with gefitinib administration in cancer patients. This study was conducted to determine if histamine type 2 receptor antagonists (H2RAs) can affect the anti-tumor efficacy of gefitinib. METHODS Eighty-seven patients with NSCLC who were administered gefitinib were retrospectively investigated. Patients who were co-administered H2RA were compared with non-AS control patients. H2RA was administered once a day at about 3-5 or 8-12 h after gefitinib intake. The primary endpoint of this study was progression-free survival (PFS), and secondary endpoints were overall survival (OS), overall response rate (ORR), and adverse effects. RESULTS Median PFS in H2RA group and control group was 8.0 months and 9.0 months, respectively, with no significant difference (p = 0.82). The incidence of liver dysfunction was significantly less in patients administered H2RA, whereas there were no differences between the two groups with regard to skin toxicity and diarrhea. Multivariate analysis suggested that H2RA co-administration is not a risk factor for worse PFS and OS (hazard ratio of 0.95, 0.86; 95% confidence interval of 0.60-1.48, 0.52-1.43; p = 0.82 and 0.60, respectively). CONCLUSION This study demonstrated that concomitant administration of H2RA with gefitinib does not affect the efficacy of gefitinib.
Collapse
|
24
|
Almomen A, Maher HM, Alzoman NZ, Shehata SM, Alsubaie A. Flavoured water consumption alters pharmacokinetic parameters and increases exposure of erlotinib and gefitinib in a preclinical study using Wistar rats. PeerJ 2020; 8:e9881. [PMID: 33024629 PMCID: PMC7518156 DOI: 10.7717/peerj.9881] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 08/15/2020] [Indexed: 12/17/2022] Open
Abstract
Background Erlotinib (ERL) and Gefitinib (GEF) are considered first line therapy for the management of non-small cell lung carcinoma (NSCLC). Like other tyrosine kinase inhibitors (TKIs), ERL and GEF are mainly metabolized by the cytochrome P450 (CYP450) CYP3A4 isoform and are substrates for transporter proteins with marked inter-/intra-individual pharmacokinetic (PK) variability. Therefore, ERL and GEF are candidates for drug-drug and food-drug interactions with a consequent effect on drug exposure and/or drug-related toxicities. In recent years, the consumption of flavoured water (FW) has gained in popularity. Among multiple ingredients, fruit extracts, which might constitute bioactive flavonoids, can possess an inhibitory effect on the CYP450 enzymes or transporter proteins. Therefore, in this study we investigated the effects of different types of FW on the PK parameters of ERL and GEF in Wistar rats. Methods ERL and GEF PK parameters in different groups of rats after four weeks consumption of different flavours of FW, namely berry, peach, lime, and pineapple, were determined from plasma drug concentrations using ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). Results Data indicated that tested FWs altered the PK parameters of both ERL and GEF differently. Lime water had the highest impact on most of ERL and GEF PK parameters, with a significant increase in Cmax (95% for ERL, 58% for GEF), AUC0-48 (111% for ERL, 203% for GEF), and AUC0-∞ (200% for ERL, 203% for GEF), along with a significant decrease in the apparent oral clearance of both drugs (65% for ERL, 67% for GEF). The order by which FW affected the PK parameters for ERL and GEF was as follows: lime > pineapple > berry > peach. Conclusion The present study indicates that drinking FW could be of significance in rats receiving ERL or GEF. Our results indicate that the alteration in PKs was mostly recorded with lime, resulting in an enhanced bioavailability, and reduced apparent oral clearance of the drugs. Peach FW had a minimum effect on the PK parameters of ERL and no significant effect on GEF PKs. Accordingly, it might be of clinical importance to evaluate the PK parameters of ERL and GEF in human subjects who consume FW while receiving therapy.
Collapse
Affiliation(s)
- Aliyah Almomen
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hadir M Maher
- Faculty of Pharmacy, Department of Pharmaceutical Analytical Chemistry, Alexandria University, Alexandria, Egypt
| | - Nourah Z Alzoman
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Shereen M Shehata
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Amal Alsubaie
- Biological Products Evaluation Directorate, Saudi Food and Drug Authority, Riyadh, Saudi Arabia
| |
Collapse
|
25
|
Wang MN, Kuang Y, Gong LY, Hua Y, Pei Q, Guo CX, Cao Y, Huang J, Yang GP. First-in-human, phase I single-ascending-dose study of the safety, pharmacokinetics, and relative bioavailability of selatinib, a dual EGFR-ErbB2 inhibitor in healthy subjects. Invest New Drugs 2020; 38:1826-1835. [PMID: 32535812 PMCID: PMC7575490 DOI: 10.1007/s10637-020-00959-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 05/28/2020] [Indexed: 01/09/2023]
Abstract
We assessed the pharmacokinetics and safety of a single oral administration of selatinib to healthy Chinese subjects and evaluated the potential bioavailability advantage of selatinib relative to lapatinib. Healthy subjects aged 18-40 years were enrolled in this two-part study: Part 1, a single ascending dose (50-500 mg), randomized, double-blind, placebo-control study with 64 subjects; and Part 2, an open-label, positive control, randomized, three-treatment, three-period, three-sequence crossover design study, with 6 subjects administered a single 500-mg dose of selatinib tablets (A), selatinib suspension (B), or lapatinib tablets C) per cycle. In part 1, selatinib was well-tolerated up to the planned maximum dose of 500 mg; thus the maximum tolerated dose was not attained. Twenty-two adverse events were observed in 19 (36.5%) of the 52 subjects administered the test drug. The most common drug-related adverse event was diarrhea. The mean selatinib peak plasma concentration was 69.4-494 ng/mL, which was achieved in a median peak time of 3.5-4.5 h, with a mean elimination half-life between 13.8 and 15.8 h. In Part 2, A and B showed similar bioavailability. Plasma exposure to the active drug (selatinib plus the metabolite, lapatinib) after A intake was more than two-fold higher than that of the same dose of C. In the dose range of 50-500 mg, selatinib was safe and well-tolerated by healthy Chinese subjects, and it conformed with linear pharmacokinetics. Active exposure to selatinib was much greater than that to lapatinib, supporting its development as an adjuvant for anticancer treatment.
Collapse
Affiliation(s)
- Meng-Na Wang
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Yun Kuang
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Li-Ying Gong
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China.,Research Center of Drug Clinical Evaluation of Central South University, Changsha, Hunan, 410013, People's Republic of China.,Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Ye Hua
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Qi Pei
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Cheng-Xian Guo
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China.,Research Center of Drug Clinical Evaluation of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Yu Cao
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Jie Huang
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China. .,Research Center of Drug Clinical Evaluation of Central South University, Changsha, Hunan, 410013, People's Republic of China.
| | - Guo-Ping Yang
- Center for Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China. .,Research Center of Drug Clinical Evaluation of Central South University, Changsha, Hunan, 410013, People's Republic of China. .,Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China.
| |
Collapse
|
26
|
Wang L, Ruan Z, Yang D, Hu Y, Liang J, Chen J, Shao R, Xu Y, Guan Y, Jiang B. Pharmacokinetics and Bioequivalence Evaluation of Erlotinib Hydrochloride Tablets: Randomized, Open-Label, 2-Period Crossover Study in Healthy Chinese Subjects. Clin Pharmacol Drug Dev 2020; 10:166-172. [PMID: 32416055 DOI: 10.1002/cpdd.811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/06/2020] [Indexed: 12/24/2022]
Abstract
A randomized, open-label, 2-period crossover study was performed to evaluate the pharmacokinetic properties and bioequivalence of 2 erlotinib hydrochloride tablets (a test formulation and a reference formulation) in healthy Chinese subjects. Subjects were randomized to receive a single oral dose of the erlotinib hydrochloride test or reference formulation (150 mg) under fasting conditions. The washout period was 12 days. Blood samples were collected at scheduled time points, and plasma concentrations were determined using a high-performance liquid chromatography-tandem mass spectrometry method. A noncompartmental method was used to calculate pharmacokinetic parameters and to evaluate the bioequivalence of the 2 formulations. Safety assessments were performed during the whole study period. The results suggest that the pharmacokinetic parameter values of the test formulation were similar to those of the reference formulation. The 90% confidence intervals of the geometric least-squares mean ratios of the test to reference formulation were 94.06% to 105.43% for maximum concentration, 88.21% to 97.57% for area under the concentration-time curve to last measurement, and 87.37% to 97.14% for area under the curve extrapolated to infinity, which are all within the accepted bioequivalence range of 80% to 125%. No serious adverse events occurred during the study. These findings suggest that the 2 erlotinib hydrochloride tablets were bioequivalent in accordance with predetermined regulatory criteria.
Collapse
Affiliation(s)
- Lu Wang
- Center of Clinical Pharmacology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zourong Ruan
- Center of Clinical Pharmacology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Dandan Yang
- Center of Clinical Pharmacology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yin Hu
- Center of Clinical Pharmacology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jian Liang
- Center of Clinical Pharmacology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jinliang Chen
- Center of Clinical Pharmacology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Rong Shao
- Center of Clinical Pharmacology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yichao Xu
- Center of Clinical Pharmacology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yanlu Guan
- Center of Clinical Pharmacology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Bo Jiang
- Center of Clinical Pharmacology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
27
|
Co-targeting EGFR and mTOR with gefitinib and everolimus in triple-negative breast cancer cells. Sci Rep 2020; 10:6367. [PMID: 32286420 PMCID: PMC7156377 DOI: 10.1038/s41598-020-63310-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 03/24/2020] [Indexed: 12/30/2022] Open
Abstract
Triple-negative breast cancers (TNBC) are unlikely to respond to hormonal therapies and anti-HER2-targeted therapies. TNBCs overexpress EGFR and exhibit constitutive activation of the PI3K/AKT/mTOR signalling pathway. We hypothesized that simultaneously blocking EGFR and mTOR could be a potential therapeutic strategy for the treatment of TNBC. We examined the antitumour activity of the mTOR inhibitor everolimus combined with the EGFR tyrosine kinase inhibitor gefitinib in TNBC cell with or without activating mutations in the PI3K/AKT/mTOR signalling pathway. We demonstrated that everolimus and gefitinib induced synergistic growth inhibition in the PI3K and PTEN-mutant CAL-51 cell line but not in the PTEN-null HCC-1937 cell line. The antiproliferative effect was associated with synergistic inhibition of mTOR and P70S6K phosphorylation, as well as a significant reduction in 4E-BP1 activation in the CAL-51 cell line. We also showed that combination therapy significantly inhibited cell cycle progression and increased apoptosis in this cell line. Gene and protein expression analysis revealed significant downregulation of cell cycle regulators after exposure to combined treatment. Collectively, these results suggested that dual inhibition of mTOR and EGFR may be an effective treatment for TNBC with activating mutations of PI3K.
Collapse
|
28
|
Fink C, Sun D, Wagner K, Schneider M, Bauer H, Dolgos H, Mäder K, Peters SA. Evaluating the Role of Solubility in Oral Absorption of Poorly Water-Soluble Drugs Using Physiologically-Based Pharmacokinetic Modeling. Clin Pharmacol Ther 2019; 107:650-661. [PMID: 31608434 PMCID: PMC7158207 DOI: 10.1002/cpt.1672] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/09/2019] [Indexed: 12/19/2022]
Abstract
Poor aqueous solubility and dissolution of drug candidates drive key decisions on lead series optimization during drug discovery, on formulation optimization, and clinical studies planning during drug development. The interpretation of the in vivo relevance of early pharmaceutical profiling is often confounded by the multiple factors affecting oral systemic exposure. There is growing evidence that in vitro drug solubility may underestimate the true in vivo solubility and lead to drug misclassification. Based on 10 poorly water‐soluble tyrosine kinase inhibitors, this paper demonstrates the use of physiologically‐based pharmacokinetic (PK) analysis in combination with early clinical PK data to identify drugs whose absorption is truly limited by solubility in vivo and, therefore, expected to exhibit food effect. Our study supports a totality of evidence approach using early clinical data to guide decisions on conducting drug interaction studies with food and acid‐reducing agents.
Collapse
Affiliation(s)
- Christina Fink
- Chemical Pharmaceutical Development, Merck Healthcare KGaA, Darmstadt, Germany.,Faculty of Biosciences, Institute of Pharmacy, Martin-Luther University Halle-Wittenberg, Halle/Saale, Germany
| | - Dajun Sun
- Site Management - Analytics Healthcare, Merck KGaA, Darmstadt, Germany
| | - Knut Wagner
- Chemical Pharmaceutical Development, Merck Healthcare KGaA, Darmstadt, Germany
| | - Melanie Schneider
- Chemical Pharmaceutical Development, Merck Healthcare KGaA, Darmstadt, Germany
| | - Holger Bauer
- Global Manufacturing and Supply, Merck Healthcare KGaA, Darmstadt, Germany
| | | | - Karsten Mäder
- Faculty of Biosciences, Institute of Pharmacy, Martin-Luther University Halle-Wittenberg, Halle/Saale, Germany
| | | |
Collapse
|
29
|
Gibson AJW, D'Silva A, Elegbede AA, Tudor RA, Dean ML, Bebb DG, Hao D. Impact of Asian ethnicity on outcome in metastatic EGFR-mutant non-small cell lung cancer. Asia Pac J Clin Oncol 2019; 15:343-352. [PMID: 31486229 DOI: 10.1111/ajco.13234] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022]
Abstract
AIM To determine factors associated with survival in de novo stage IV, non-small cell lung cancer (NSCLC) patients possessing epidermal growth factor receptor mutations (EGFRmut+ ) receiving tyrosine kinase inhibitors (TKI) in the first-line setting. METHODS The Glans-Look Lung Cancer Database was used to retrospectively review stage IV EGFRmut+ NSCLC patients diagnosed 2010-2016 receiving first-line TKI. Patients with overall survival times in the upper quartile (≥34 months) were designated "long-term survivors" (LTS), the remaining deemed "average-term survivors" and characteristics between these groups were compared in univariate analysis, and multivariable models constructed to determine predictors of outcome. RESULTS Of 170 eligible patients, median overall survival was 21 months. LTS were significantly more likely to be of Asian ethnicity, be never-smokers and not possess brain or bone metastases at diagnosis. Asian and non-Asian patients were comparable, save for an increased propensity of Asian patients to be never smokers and have normal-range BMI. Multivariable analysis revealed Asian ethnicity [hazard ratio (HR) = 0.65; P = 0.016] and never-smoking history (HR = 0.65; P = 0.034) as indicators of improved outcome, and presence of brain metastasis at diagnosis an indicator of poor outcome (HR = 2.21; P < 0.001). CONCLUSIONS Analysis of this population-based cohort identifies never-smoking history and absence of brain metastasis along with Asian ethnicity as an independent prognosticators of favorable outcome, and reveals Asian patients to be clinicopathologically similar to non-Asian patients. These findings suggest Asian patients represent a unique subpopulation within EGFRmut+ NSCLC who may possess different biological underpinnings of NSCLC.
Collapse
Affiliation(s)
- Amanda J W Gibson
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Adrijana D'Silva
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Anifat A Elegbede
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Roxana A Tudor
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Michelle L Dean
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Dafydd Gwyn Bebb
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Tom Baker Cancer Centre, Alberta Health Services, Calgary, Canada
| | - Desiree Hao
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Tom Baker Cancer Centre, Alberta Health Services, Calgary, Canada
| |
Collapse
|
30
|
Decoster L, Schallier D. Treatment of older patients with advanced non-small cell lung cancer: A challenge. J Geriatr Oncol 2019; 10:528-533. [DOI: 10.1016/j.jgo.2018.09.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/27/2018] [Accepted: 09/19/2018] [Indexed: 01/06/2023]
|
31
|
Pai SM, Chaikin P, Berg JK. Pharmacokinetics of Lapatinib, a Nonrenally Cleared Drug, in Patients With End-Stage Renal Disease on Maintenance Hemodialysis. J Clin Pharmacol 2019; 59:1379-1383. [PMID: 31074516 DOI: 10.1002/jcph.1430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 04/08/2019] [Indexed: 11/05/2022]
Abstract
Lapatinib, a tyrosine kinase inhibitor, is approved for the treatment of breast cancer. The literature shows that it is metabolized by CYP3A4 and eliminated predominantly (>90%) by the fecal route, with minimal (<2%) renal elimination in healthy subjects (dose of 250 mg); in cancer patients, renal elimination is minimal at therapeutic doses. For nonrenally cleared drugs, while there is ample evidence of pharmacokinetic alterations secondary to renal impairment-induced effects on drug metabolizing enzymes and/or transporters, the effect of end-stage renal disease (ESRD) on lapatinib pharmacokinetics has not been determined. Rather, as stated in the drug's label, the expectation is lack of effect of renal impairment on lapatinib pharmacokinetics based on its minimal renal elimination. The current report addresses this gap with pharmacokinetic data (obtained in a 1-way drug interaction study) in ESRD patients (n = 11) on maintenance hemodialysis and compared with published data in 37 healthy subjects in 3 separate studies. Following a 250-mg oral dose in ESRD patients, the median tmax was 3.0 hours, and geometric mean (95%CI) values for Cmax , AUCinf , and t1/2 were 349 ng/mL (245-499 ng/mL), 4410 ng·h/mL (2960-6580 ng·h/mL), and 14.8 hours (9.7-22.5 hours), respectively. These parameters approximated published values in healthy subjects and demonstrated that renal impairment and hemodialysis did not affect lapatinib pharmacokinetics. The results of the present study in this renally impaired population, the only such information available to date, support the drug's label and are valuable in view of the recognized difficulties in enrolling organ-impaired patients in oncology trials.
Collapse
Affiliation(s)
- Sudhakar M Pai
- Department of Clinical Pharmacology, Akros Pharma, Inc., Princeton, NJ, USA
| | | | | |
Collapse
|
32
|
Seebacher NA, Stacy AE, Porter GM, Merlot AM. Clinical development of targeted and immune based anti-cancer therapies. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:156. [PMID: 30975211 PMCID: PMC6460662 DOI: 10.1186/s13046-019-1094-2] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 02/07/2019] [Indexed: 02/08/2023]
Abstract
Cancer is currently the second leading cause of death globally and is expected to be responsible for approximately 9.6 million deaths in 2018. With an unprecedented understanding of the molecular pathways that drive the development and progression of human cancers, novel targeted therapies have become an exciting new development for anti-cancer medicine. These targeted therapies, also known as biologic therapies, have become a major modality of medical treatment, by acting to block the growth of cancer cells by specifically targeting molecules required for cell growth and tumorigenesis. Due to their specificity, these new therapies are expected to have better efficacy and limited adverse side effects when compared with other treatment options, including hormonal and cytotoxic therapies. In this review, we explore the clinical development, successes and challenges facing targeted anti-cancer therapies, including both small molecule inhibitors and antibody targeted therapies. Herein, we introduce targeted therapies to epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), human epidermal growth factor receptor 2 (HER2), anaplastic lymphoma kinase (ALK), BRAF, and the inhibitors of the T-cell mediated immune response, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein-1 (PD-1)/ PD-1 ligand (PD-1 L).
Collapse
Affiliation(s)
- N A Seebacher
- Faculty of Medicine, The University of Sydney, Camperdown, New South Wales, 2006, Australia
| | - A E Stacy
- Faculty of Medicine, The University of Notre Dame, Darlinghurst, New South Wales, 2010, Australia
| | - G M Porter
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Kensington, New South Wales, 2031, Australia
| | - A M Merlot
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Kensington, New South Wales, 2031, Australia. .,School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Kensington, New South Wales, 2031, Australia. .,UNSW Centre for Childhood Cancer Research, Faculty of Medicine, University of New South Wales, Kensington, New South Wales, 2031, Australia.
| |
Collapse
|
33
|
Impact of single nucleotide polymorphisms on the efficacy and toxicity of EGFR tyrosine kinase inhibitors in advanced non-small cell lung cancer patients. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2019; 781:63-70. [PMID: 31416579 DOI: 10.1016/j.mrrev.2019.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 01/11/2023]
Abstract
EGFR tyrosine kinase inhibitors (EGFR-TKIs) are the treatment of choice for advanced-stage (IIIB-IV) NSCLC patients with mutations in EGFR. However, EGFR-TKIs clinical outcomes vary from person to person and these inter-individual differences may be due to genetic factors such as single nucleotide polymorphisms (SNPs). SNPs in genes involved in EGFR-TKIs pharmacodynamics, metabolism and mechanism of action have been demonstrated to be associated with response, survival and toxicity in advanced NSCLC patients treated with EGFR-TKIs. Here we review the influence of gene polymorphisms in the EGFR pathway on clinical outcome and toxicity to EGFR-TKIs in advanced NSCLC patients. The EGFR-216 polymorphism has reported a strong association between response and/or survival to EGFR-TKIs in Caucasian population. Similarly, the effect of EGFR-CA repeats polymorphisms on survival of advanced NSCLC patients treated with EGFR-TKIs have been confirmed both in Caucasian and Asian population. The influence on toxicity of the -216, -191, CA repeats, Arg497Lys and Asp994Asp polymorphisms in EGFR have also been confirmed. Polymorphisms in AKT (rs1130214 and rs1130233) and SMAD3 (rs6494633, rs11071938 and rs11632964) have been associated with survival in advanced NSCLC patients treated with EGFR-TKIs. However, data come from a limited number of studies and need to be confirmed. Finally, polymorphisms in genes coding proteins of the membrane transporters and cytochrome P450 enzymes have been less extensively investigated. There are few studies with small samples, which complicated the generalization of their role in EGFR-TKIs treatment.
Collapse
|
34
|
Abdulla DSY, Scheffler M, Brandes V, Ruge M, Kunze S, Merkelbach-Bruse S, Nogova L, Michels S, Fischer R, Riedel R, Büttner R, Persigehl T, Grau S, Galldiks N, Drzezga A, Kobe C, Wolf J. Monitoring Treatment Response to Erlotinib in EGFR-mutated Non-small-cell Lung Cancer Brain Metastases Using Serial O-(2-[ 18F]fluoroethyl)-L-tyrosine PET. Clin Lung Cancer 2018; 20:e148-e151. [PMID: 30528316 DOI: 10.1016/j.cllc.2018.10.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/26/2018] [Accepted: 10/27/2018] [Indexed: 11/24/2022]
Affiliation(s)
- Diana S Y Abdulla
- Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln, Bonn, Germany; Network Genomic Medicine (NGM) Lung Cancer, Cologne, Germany
| | - Matthias Scheffler
- Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln, Bonn, Germany; Network Genomic Medicine (NGM) Lung Cancer, Cologne, Germany
| | - Vanessa Brandes
- Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln, Bonn, Germany; Network Genomic Medicine (NGM) Lung Cancer, Cologne, Germany
| | - Maximilian Ruge
- Center for Integrated Oncology Köln, Bonn, Germany; Department of Stereotaxy and Functional Neurosurgery, University Hospital of Cologne, Cologne, Germany
| | - Sabine Kunze
- Center for Integrated Oncology Köln, Bonn, Germany; Department of Radiotherapy, University Hospital of Cologne, Cologne, Germany
| | - Sabine Merkelbach-Bruse
- Center for Integrated Oncology Köln, Bonn, Germany; Network Genomic Medicine (NGM) Lung Cancer, Cologne, Germany; Department of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Lucia Nogova
- Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln, Bonn, Germany; Network Genomic Medicine (NGM) Lung Cancer, Cologne, Germany
| | - Sebastian Michels
- Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln, Bonn, Germany; Network Genomic Medicine (NGM) Lung Cancer, Cologne, Germany
| | - Rieke Fischer
- Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln, Bonn, Germany; Network Genomic Medicine (NGM) Lung Cancer, Cologne, Germany
| | - Richard Riedel
- Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln, Bonn, Germany; Network Genomic Medicine (NGM) Lung Cancer, Cologne, Germany
| | - Reinhard Büttner
- Center for Integrated Oncology Köln, Bonn, Germany; Network Genomic Medicine (NGM) Lung Cancer, Cologne, Germany; Department of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Thorsten Persigehl
- Center for Integrated Oncology Köln, Bonn, Germany; Department of Radiology, University Hospital of Cologne, Cologne, Germany
| | - Stefan Grau
- Center for Integrated Oncology Köln, Bonn, Germany; Department of Neurosurgery, University Hospital of Cologne, Cologne, Germany
| | - Norbert Galldiks
- Center for Integrated Oncology Köln, Bonn, Germany; Department of Neurology, University Hospital of Cologne, Cologne, Germany; Institute of Neuroscience and Medicine, Research Center Juelich, Juelich, Germany
| | - Alexander Drzezga
- Center for Integrated Oncology Köln, Bonn, Germany; Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| | - Carsten Kobe
- Center for Integrated Oncology Köln, Bonn, Germany; Department of Nuclear Medicine, University Hospital Cologne, Cologne, Germany
| | - Jürgen Wolf
- Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany; Center for Integrated Oncology Köln, Bonn, Germany; Network Genomic Medicine (NGM) Lung Cancer, Cologne, Germany.
| |
Collapse
|
35
|
Wu YL, Zhang L, Kim DW, Liu X, Lee DH, Yang JCH, Ahn MJ, Vansteenkiste JF, Su WC, Felip E, Chia V, Glaser S, Pultar P, Zhao S, Peng B, Akimov M, Tan DSW. Phase Ib/II Study of Capmatinib (INC280) Plus Gefitinib After Failure of Epidermal Growth Factor Receptor (EGFR) Inhibitor Therapy in Patients With EGFR-Mutated, MET Factor-Dysregulated Non-Small-Cell Lung Cancer. J Clin Oncol 2018; 36:3101-3109. [PMID: 30156984 DOI: 10.1200/jco.2018.77.7326] [Citation(s) in RCA: 249] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PURPOSE MET dysregulation occurs in up to 26% of non-small-cell lung cancers (NSCLCs) after epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitor (TKI) treatment. Capmatinib (INC280) is a potent and selective MET inhibitor with preclinical activity in combination with gefitinib in EGFR-mutant, MET-amplified/overexpressing models of acquired EGFR-TKI resistance. This phase Ib/II study investigated the safety and efficacy of capmatinib plus gefitinib in patients with EGFR-mutated, MET-dysregulated (amplified/overexpressing) NSCLC who experienced disease progression while receiving EGFR-TKI treatment. METHODS Patients in phase Ib received capmatinib 100- to 800-mg capsules once per day or 200- to 600-mg capsules or tablets twice per day, plus gefitinib 250 mg once per day. Patients in phase II received the recommended phase II dose. The primary end point was the overall response rate (ORR) per Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. RESULTS Sixty-one patients were treated in phase Ib, and 100 were treated in phase II. The recommended phase II dose was capmatinib 400 mg twice per day plus gefitinib 250 mg once per day. Preliminary clinical activity was observed, with an ORR across phase Ib/II of 27%. Increased activity was seen in patients with high MET-amplified tumors, with a phase II ORR of 47% in patients with a MET gene copy number ≥ 6. Across phases Ib and II, the most common drug-related adverse events were nausea (28%), peripheral edema (22%), decreased appetite (21%), and rash (20%); the most common drug-related grade 3/4 adverse events were increased amylase and lipase levels (both 6%). No significant drug-drug interactions between capmatinib and gefitinib were evident. CONCLUSION This study, focused on a predominant EGFR-TKI resistance mechanism in patients with EGFR-mutated NSCLC, shows that the combination of capmatinib with gefitinib is a promising treatment for patients with EGFR-mutated, MET-dysregulated NSCLC, particularly MET-amplified disease.
Collapse
Affiliation(s)
- Yi-Long Wu
- Yi-Long Wu, Guangdong General Hospital and Guangdong Academy of Medical Sciences; Li Zhang, Sun Yat-sen University Cancer Center, Guangdong; Xiaoqing Liu, Affiliated Hospital of the Chinese Academy of Military Medical Sciences, Beijing; Sylvia Zhao and Bin Peng, Novartis Institutes for Biomedical Research, Shanghai, People's Republic of China; Dong-Wan Kim, Seoul National University Hospital; Dae Ho Lee, University of Ulsan College of Medicine; Myung-Ju Ahn, Samsung Medical Center, Seoul, Republic of Korea; James Chih-Hsin Yang, National Taiwan University Hospital, Taipei; Wu-Chou Su, National Cheng Kung University Hospital, Tainan, Taiwan; Johan F. Vansteenkiste, University Hospital KU Leuven, Leuven, Belgium; Enriqueta Felip, Vall d'Hebron University Hospital, Barcelona, Spain; Vincent Chia and Philippe Pultar, Novartis Pharmaceuticals, East Hanover, NJ; Sabine Glaser and Mikhail Akimov, Novartis Pharma AG, Basel, Switzerland; and Daniel S.W. Tan, National Cancer Centre Singapore, Singapore
| | - Li Zhang
- Yi-Long Wu, Guangdong General Hospital and Guangdong Academy of Medical Sciences; Li Zhang, Sun Yat-sen University Cancer Center, Guangdong; Xiaoqing Liu, Affiliated Hospital of the Chinese Academy of Military Medical Sciences, Beijing; Sylvia Zhao and Bin Peng, Novartis Institutes for Biomedical Research, Shanghai, People's Republic of China; Dong-Wan Kim, Seoul National University Hospital; Dae Ho Lee, University of Ulsan College of Medicine; Myung-Ju Ahn, Samsung Medical Center, Seoul, Republic of Korea; James Chih-Hsin Yang, National Taiwan University Hospital, Taipei; Wu-Chou Su, National Cheng Kung University Hospital, Tainan, Taiwan; Johan F. Vansteenkiste, University Hospital KU Leuven, Leuven, Belgium; Enriqueta Felip, Vall d'Hebron University Hospital, Barcelona, Spain; Vincent Chia and Philippe Pultar, Novartis Pharmaceuticals, East Hanover, NJ; Sabine Glaser and Mikhail Akimov, Novartis Pharma AG, Basel, Switzerland; and Daniel S.W. Tan, National Cancer Centre Singapore, Singapore
| | - Dong-Wan Kim
- Yi-Long Wu, Guangdong General Hospital and Guangdong Academy of Medical Sciences; Li Zhang, Sun Yat-sen University Cancer Center, Guangdong; Xiaoqing Liu, Affiliated Hospital of the Chinese Academy of Military Medical Sciences, Beijing; Sylvia Zhao and Bin Peng, Novartis Institutes for Biomedical Research, Shanghai, People's Republic of China; Dong-Wan Kim, Seoul National University Hospital; Dae Ho Lee, University of Ulsan College of Medicine; Myung-Ju Ahn, Samsung Medical Center, Seoul, Republic of Korea; James Chih-Hsin Yang, National Taiwan University Hospital, Taipei; Wu-Chou Su, National Cheng Kung University Hospital, Tainan, Taiwan; Johan F. Vansteenkiste, University Hospital KU Leuven, Leuven, Belgium; Enriqueta Felip, Vall d'Hebron University Hospital, Barcelona, Spain; Vincent Chia and Philippe Pultar, Novartis Pharmaceuticals, East Hanover, NJ; Sabine Glaser and Mikhail Akimov, Novartis Pharma AG, Basel, Switzerland; and Daniel S.W. Tan, National Cancer Centre Singapore, Singapore
| | - Xiaoqing Liu
- Yi-Long Wu, Guangdong General Hospital and Guangdong Academy of Medical Sciences; Li Zhang, Sun Yat-sen University Cancer Center, Guangdong; Xiaoqing Liu, Affiliated Hospital of the Chinese Academy of Military Medical Sciences, Beijing; Sylvia Zhao and Bin Peng, Novartis Institutes for Biomedical Research, Shanghai, People's Republic of China; Dong-Wan Kim, Seoul National University Hospital; Dae Ho Lee, University of Ulsan College of Medicine; Myung-Ju Ahn, Samsung Medical Center, Seoul, Republic of Korea; James Chih-Hsin Yang, National Taiwan University Hospital, Taipei; Wu-Chou Su, National Cheng Kung University Hospital, Tainan, Taiwan; Johan F. Vansteenkiste, University Hospital KU Leuven, Leuven, Belgium; Enriqueta Felip, Vall d'Hebron University Hospital, Barcelona, Spain; Vincent Chia and Philippe Pultar, Novartis Pharmaceuticals, East Hanover, NJ; Sabine Glaser and Mikhail Akimov, Novartis Pharma AG, Basel, Switzerland; and Daniel S.W. Tan, National Cancer Centre Singapore, Singapore
| | - Dae Ho Lee
- Yi-Long Wu, Guangdong General Hospital and Guangdong Academy of Medical Sciences; Li Zhang, Sun Yat-sen University Cancer Center, Guangdong; Xiaoqing Liu, Affiliated Hospital of the Chinese Academy of Military Medical Sciences, Beijing; Sylvia Zhao and Bin Peng, Novartis Institutes for Biomedical Research, Shanghai, People's Republic of China; Dong-Wan Kim, Seoul National University Hospital; Dae Ho Lee, University of Ulsan College of Medicine; Myung-Ju Ahn, Samsung Medical Center, Seoul, Republic of Korea; James Chih-Hsin Yang, National Taiwan University Hospital, Taipei; Wu-Chou Su, National Cheng Kung University Hospital, Tainan, Taiwan; Johan F. Vansteenkiste, University Hospital KU Leuven, Leuven, Belgium; Enriqueta Felip, Vall d'Hebron University Hospital, Barcelona, Spain; Vincent Chia and Philippe Pultar, Novartis Pharmaceuticals, East Hanover, NJ; Sabine Glaser and Mikhail Akimov, Novartis Pharma AG, Basel, Switzerland; and Daniel S.W. Tan, National Cancer Centre Singapore, Singapore
| | - James Chih-Hsin Yang
- Yi-Long Wu, Guangdong General Hospital and Guangdong Academy of Medical Sciences; Li Zhang, Sun Yat-sen University Cancer Center, Guangdong; Xiaoqing Liu, Affiliated Hospital of the Chinese Academy of Military Medical Sciences, Beijing; Sylvia Zhao and Bin Peng, Novartis Institutes for Biomedical Research, Shanghai, People's Republic of China; Dong-Wan Kim, Seoul National University Hospital; Dae Ho Lee, University of Ulsan College of Medicine; Myung-Ju Ahn, Samsung Medical Center, Seoul, Republic of Korea; James Chih-Hsin Yang, National Taiwan University Hospital, Taipei; Wu-Chou Su, National Cheng Kung University Hospital, Tainan, Taiwan; Johan F. Vansteenkiste, University Hospital KU Leuven, Leuven, Belgium; Enriqueta Felip, Vall d'Hebron University Hospital, Barcelona, Spain; Vincent Chia and Philippe Pultar, Novartis Pharmaceuticals, East Hanover, NJ; Sabine Glaser and Mikhail Akimov, Novartis Pharma AG, Basel, Switzerland; and Daniel S.W. Tan, National Cancer Centre Singapore, Singapore
| | - Myung-Ju Ahn
- Yi-Long Wu, Guangdong General Hospital and Guangdong Academy of Medical Sciences; Li Zhang, Sun Yat-sen University Cancer Center, Guangdong; Xiaoqing Liu, Affiliated Hospital of the Chinese Academy of Military Medical Sciences, Beijing; Sylvia Zhao and Bin Peng, Novartis Institutes for Biomedical Research, Shanghai, People's Republic of China; Dong-Wan Kim, Seoul National University Hospital; Dae Ho Lee, University of Ulsan College of Medicine; Myung-Ju Ahn, Samsung Medical Center, Seoul, Republic of Korea; James Chih-Hsin Yang, National Taiwan University Hospital, Taipei; Wu-Chou Su, National Cheng Kung University Hospital, Tainan, Taiwan; Johan F. Vansteenkiste, University Hospital KU Leuven, Leuven, Belgium; Enriqueta Felip, Vall d'Hebron University Hospital, Barcelona, Spain; Vincent Chia and Philippe Pultar, Novartis Pharmaceuticals, East Hanover, NJ; Sabine Glaser and Mikhail Akimov, Novartis Pharma AG, Basel, Switzerland; and Daniel S.W. Tan, National Cancer Centre Singapore, Singapore
| | - Johan F Vansteenkiste
- Yi-Long Wu, Guangdong General Hospital and Guangdong Academy of Medical Sciences; Li Zhang, Sun Yat-sen University Cancer Center, Guangdong; Xiaoqing Liu, Affiliated Hospital of the Chinese Academy of Military Medical Sciences, Beijing; Sylvia Zhao and Bin Peng, Novartis Institutes for Biomedical Research, Shanghai, People's Republic of China; Dong-Wan Kim, Seoul National University Hospital; Dae Ho Lee, University of Ulsan College of Medicine; Myung-Ju Ahn, Samsung Medical Center, Seoul, Republic of Korea; James Chih-Hsin Yang, National Taiwan University Hospital, Taipei; Wu-Chou Su, National Cheng Kung University Hospital, Tainan, Taiwan; Johan F. Vansteenkiste, University Hospital KU Leuven, Leuven, Belgium; Enriqueta Felip, Vall d'Hebron University Hospital, Barcelona, Spain; Vincent Chia and Philippe Pultar, Novartis Pharmaceuticals, East Hanover, NJ; Sabine Glaser and Mikhail Akimov, Novartis Pharma AG, Basel, Switzerland; and Daniel S.W. Tan, National Cancer Centre Singapore, Singapore
| | - Wu-Chou Su
- Yi-Long Wu, Guangdong General Hospital and Guangdong Academy of Medical Sciences; Li Zhang, Sun Yat-sen University Cancer Center, Guangdong; Xiaoqing Liu, Affiliated Hospital of the Chinese Academy of Military Medical Sciences, Beijing; Sylvia Zhao and Bin Peng, Novartis Institutes for Biomedical Research, Shanghai, People's Republic of China; Dong-Wan Kim, Seoul National University Hospital; Dae Ho Lee, University of Ulsan College of Medicine; Myung-Ju Ahn, Samsung Medical Center, Seoul, Republic of Korea; James Chih-Hsin Yang, National Taiwan University Hospital, Taipei; Wu-Chou Su, National Cheng Kung University Hospital, Tainan, Taiwan; Johan F. Vansteenkiste, University Hospital KU Leuven, Leuven, Belgium; Enriqueta Felip, Vall d'Hebron University Hospital, Barcelona, Spain; Vincent Chia and Philippe Pultar, Novartis Pharmaceuticals, East Hanover, NJ; Sabine Glaser and Mikhail Akimov, Novartis Pharma AG, Basel, Switzerland; and Daniel S.W. Tan, National Cancer Centre Singapore, Singapore
| | - Enriqueta Felip
- Yi-Long Wu, Guangdong General Hospital and Guangdong Academy of Medical Sciences; Li Zhang, Sun Yat-sen University Cancer Center, Guangdong; Xiaoqing Liu, Affiliated Hospital of the Chinese Academy of Military Medical Sciences, Beijing; Sylvia Zhao and Bin Peng, Novartis Institutes for Biomedical Research, Shanghai, People's Republic of China; Dong-Wan Kim, Seoul National University Hospital; Dae Ho Lee, University of Ulsan College of Medicine; Myung-Ju Ahn, Samsung Medical Center, Seoul, Republic of Korea; James Chih-Hsin Yang, National Taiwan University Hospital, Taipei; Wu-Chou Su, National Cheng Kung University Hospital, Tainan, Taiwan; Johan F. Vansteenkiste, University Hospital KU Leuven, Leuven, Belgium; Enriqueta Felip, Vall d'Hebron University Hospital, Barcelona, Spain; Vincent Chia and Philippe Pultar, Novartis Pharmaceuticals, East Hanover, NJ; Sabine Glaser and Mikhail Akimov, Novartis Pharma AG, Basel, Switzerland; and Daniel S.W. Tan, National Cancer Centre Singapore, Singapore
| | - Vincent Chia
- Yi-Long Wu, Guangdong General Hospital and Guangdong Academy of Medical Sciences; Li Zhang, Sun Yat-sen University Cancer Center, Guangdong; Xiaoqing Liu, Affiliated Hospital of the Chinese Academy of Military Medical Sciences, Beijing; Sylvia Zhao and Bin Peng, Novartis Institutes for Biomedical Research, Shanghai, People's Republic of China; Dong-Wan Kim, Seoul National University Hospital; Dae Ho Lee, University of Ulsan College of Medicine; Myung-Ju Ahn, Samsung Medical Center, Seoul, Republic of Korea; James Chih-Hsin Yang, National Taiwan University Hospital, Taipei; Wu-Chou Su, National Cheng Kung University Hospital, Tainan, Taiwan; Johan F. Vansteenkiste, University Hospital KU Leuven, Leuven, Belgium; Enriqueta Felip, Vall d'Hebron University Hospital, Barcelona, Spain; Vincent Chia and Philippe Pultar, Novartis Pharmaceuticals, East Hanover, NJ; Sabine Glaser and Mikhail Akimov, Novartis Pharma AG, Basel, Switzerland; and Daniel S.W. Tan, National Cancer Centre Singapore, Singapore
| | - Sabine Glaser
- Yi-Long Wu, Guangdong General Hospital and Guangdong Academy of Medical Sciences; Li Zhang, Sun Yat-sen University Cancer Center, Guangdong; Xiaoqing Liu, Affiliated Hospital of the Chinese Academy of Military Medical Sciences, Beijing; Sylvia Zhao and Bin Peng, Novartis Institutes for Biomedical Research, Shanghai, People's Republic of China; Dong-Wan Kim, Seoul National University Hospital; Dae Ho Lee, University of Ulsan College of Medicine; Myung-Ju Ahn, Samsung Medical Center, Seoul, Republic of Korea; James Chih-Hsin Yang, National Taiwan University Hospital, Taipei; Wu-Chou Su, National Cheng Kung University Hospital, Tainan, Taiwan; Johan F. Vansteenkiste, University Hospital KU Leuven, Leuven, Belgium; Enriqueta Felip, Vall d'Hebron University Hospital, Barcelona, Spain; Vincent Chia and Philippe Pultar, Novartis Pharmaceuticals, East Hanover, NJ; Sabine Glaser and Mikhail Akimov, Novartis Pharma AG, Basel, Switzerland; and Daniel S.W. Tan, National Cancer Centre Singapore, Singapore
| | - Philippe Pultar
- Yi-Long Wu, Guangdong General Hospital and Guangdong Academy of Medical Sciences; Li Zhang, Sun Yat-sen University Cancer Center, Guangdong; Xiaoqing Liu, Affiliated Hospital of the Chinese Academy of Military Medical Sciences, Beijing; Sylvia Zhao and Bin Peng, Novartis Institutes for Biomedical Research, Shanghai, People's Republic of China; Dong-Wan Kim, Seoul National University Hospital; Dae Ho Lee, University of Ulsan College of Medicine; Myung-Ju Ahn, Samsung Medical Center, Seoul, Republic of Korea; James Chih-Hsin Yang, National Taiwan University Hospital, Taipei; Wu-Chou Su, National Cheng Kung University Hospital, Tainan, Taiwan; Johan F. Vansteenkiste, University Hospital KU Leuven, Leuven, Belgium; Enriqueta Felip, Vall d'Hebron University Hospital, Barcelona, Spain; Vincent Chia and Philippe Pultar, Novartis Pharmaceuticals, East Hanover, NJ; Sabine Glaser and Mikhail Akimov, Novartis Pharma AG, Basel, Switzerland; and Daniel S.W. Tan, National Cancer Centre Singapore, Singapore
| | - Sylvia Zhao
- Yi-Long Wu, Guangdong General Hospital and Guangdong Academy of Medical Sciences; Li Zhang, Sun Yat-sen University Cancer Center, Guangdong; Xiaoqing Liu, Affiliated Hospital of the Chinese Academy of Military Medical Sciences, Beijing; Sylvia Zhao and Bin Peng, Novartis Institutes for Biomedical Research, Shanghai, People's Republic of China; Dong-Wan Kim, Seoul National University Hospital; Dae Ho Lee, University of Ulsan College of Medicine; Myung-Ju Ahn, Samsung Medical Center, Seoul, Republic of Korea; James Chih-Hsin Yang, National Taiwan University Hospital, Taipei; Wu-Chou Su, National Cheng Kung University Hospital, Tainan, Taiwan; Johan F. Vansteenkiste, University Hospital KU Leuven, Leuven, Belgium; Enriqueta Felip, Vall d'Hebron University Hospital, Barcelona, Spain; Vincent Chia and Philippe Pultar, Novartis Pharmaceuticals, East Hanover, NJ; Sabine Glaser and Mikhail Akimov, Novartis Pharma AG, Basel, Switzerland; and Daniel S.W. Tan, National Cancer Centre Singapore, Singapore
| | - Bin Peng
- Yi-Long Wu, Guangdong General Hospital and Guangdong Academy of Medical Sciences; Li Zhang, Sun Yat-sen University Cancer Center, Guangdong; Xiaoqing Liu, Affiliated Hospital of the Chinese Academy of Military Medical Sciences, Beijing; Sylvia Zhao and Bin Peng, Novartis Institutes for Biomedical Research, Shanghai, People's Republic of China; Dong-Wan Kim, Seoul National University Hospital; Dae Ho Lee, University of Ulsan College of Medicine; Myung-Ju Ahn, Samsung Medical Center, Seoul, Republic of Korea; James Chih-Hsin Yang, National Taiwan University Hospital, Taipei; Wu-Chou Su, National Cheng Kung University Hospital, Tainan, Taiwan; Johan F. Vansteenkiste, University Hospital KU Leuven, Leuven, Belgium; Enriqueta Felip, Vall d'Hebron University Hospital, Barcelona, Spain; Vincent Chia and Philippe Pultar, Novartis Pharmaceuticals, East Hanover, NJ; Sabine Glaser and Mikhail Akimov, Novartis Pharma AG, Basel, Switzerland; and Daniel S.W. Tan, National Cancer Centre Singapore, Singapore
| | - Mikhail Akimov
- Yi-Long Wu, Guangdong General Hospital and Guangdong Academy of Medical Sciences; Li Zhang, Sun Yat-sen University Cancer Center, Guangdong; Xiaoqing Liu, Affiliated Hospital of the Chinese Academy of Military Medical Sciences, Beijing; Sylvia Zhao and Bin Peng, Novartis Institutes for Biomedical Research, Shanghai, People's Republic of China; Dong-Wan Kim, Seoul National University Hospital; Dae Ho Lee, University of Ulsan College of Medicine; Myung-Ju Ahn, Samsung Medical Center, Seoul, Republic of Korea; James Chih-Hsin Yang, National Taiwan University Hospital, Taipei; Wu-Chou Su, National Cheng Kung University Hospital, Tainan, Taiwan; Johan F. Vansteenkiste, University Hospital KU Leuven, Leuven, Belgium; Enriqueta Felip, Vall d'Hebron University Hospital, Barcelona, Spain; Vincent Chia and Philippe Pultar, Novartis Pharmaceuticals, East Hanover, NJ; Sabine Glaser and Mikhail Akimov, Novartis Pharma AG, Basel, Switzerland; and Daniel S.W. Tan, National Cancer Centre Singapore, Singapore
| | - Daniel S W Tan
- Yi-Long Wu, Guangdong General Hospital and Guangdong Academy of Medical Sciences; Li Zhang, Sun Yat-sen University Cancer Center, Guangdong; Xiaoqing Liu, Affiliated Hospital of the Chinese Academy of Military Medical Sciences, Beijing; Sylvia Zhao and Bin Peng, Novartis Institutes for Biomedical Research, Shanghai, People's Republic of China; Dong-Wan Kim, Seoul National University Hospital; Dae Ho Lee, University of Ulsan College of Medicine; Myung-Ju Ahn, Samsung Medical Center, Seoul, Republic of Korea; James Chih-Hsin Yang, National Taiwan University Hospital, Taipei; Wu-Chou Su, National Cheng Kung University Hospital, Tainan, Taiwan; Johan F. Vansteenkiste, University Hospital KU Leuven, Leuven, Belgium; Enriqueta Felip, Vall d'Hebron University Hospital, Barcelona, Spain; Vincent Chia and Philippe Pultar, Novartis Pharmaceuticals, East Hanover, NJ; Sabine Glaser and Mikhail Akimov, Novartis Pharma AG, Basel, Switzerland; and Daniel S.W. Tan, National Cancer Centre Singapore, Singapore
| |
Collapse
|
36
|
Takeda K, Komatsu K, Morita S. Bayesian dose-finding phase I trial design incorporating pharmacokinetic assessment in the field of oncology. Pharm Stat 2018; 17:725-733. [DOI: 10.1002/pst.1890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 05/28/2018] [Accepted: 06/25/2018] [Indexed: 11/11/2022]
Affiliation(s)
- Kentaro Takeda
- Data Science, Astellas Pharma Global Development, Inc.; IL USA
| | - Kanji Komatsu
- Clinical Pharmacology, Astellas Pharma Inc.; Tokyo Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics; Kyoto University Graduate School of Medicine; Kyoto Japan
| |
Collapse
|
37
|
Fogli S, Polini B, Del Re M, Petrini I, Passaro A, Crucitta S, Rofi E, Danesi R. EGFR-TKIs in non-small-cell lung cancer: focus on clinical pharmacology and mechanisms of resistance. Pharmacogenomics 2018; 19:727-740. [PMID: 29785875 DOI: 10.2217/pgs-2018-0038] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The clinical introduction of EGFR-TKIs within the oncologic armamentarium has changed the therapeutic landscape of non-small-cell lung cancer (NSCLC) creating widespread expectations both in patients and clinicians. However, several gaps in current understanding leave open important questions regarding the use of these drugs in clinical practice. For instance, there is uncertainty in regard to which EGFR-TKI should be given first in naive patients with EGFR-driven malignancies since different generations of drugs are available with different pharmacological profiles. Furthermore, acquired drug resistance may limit the therapeutic potential of EGFR-TKIs and the choice of the best treatment strategy after first-line treatment failure is still debated. This review article is aimed at describing the pharmacological properties of EGFR-TKIs and the current treatment options for NSCLC patients who develop acquired resistance. This information might be useful to design new rational and more effective pharmacological strategies in patients with EGFR-mutant NSCLC.
Collapse
Affiliation(s)
- Stefano Fogli
- Clinical Pharmacology & Pharmacogenetics Unit, Department of Clinical & Experimental Medicine, University of Pisa, Italy
| | - Beatrice Polini
- Clinical Pharmacology & Pharmacogenetics Unit, Department of Clinical & Experimental Medicine, University of Pisa, Italy
| | - Marzia Del Re
- Clinical Pharmacology & Pharmacogenetics Unit, Department of Clinical & Experimental Medicine, University of Pisa, Italy
| | - Iacopo Petrini
- General Pathology, Department of Translational Research & New Technologies in Surgery & Medicine, University of Pisa, Italy
| | - Antonio Passaro
- Division of Thoracic Oncology, European Institute of Oncology, Milan, Italy
| | - Stefania Crucitta
- Clinical Pharmacology & Pharmacogenetics Unit, Department of Clinical & Experimental Medicine, University of Pisa, Italy
| | - Eleonora Rofi
- Clinical Pharmacology & Pharmacogenetics Unit, Department of Clinical & Experimental Medicine, University of Pisa, Italy
| | - Romano Danesi
- Clinical Pharmacology & Pharmacogenetics Unit, Department of Clinical & Experimental Medicine, University of Pisa, Italy
| |
Collapse
|
38
|
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: 0.9] [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
|
39
|
Anti-EGFR monoclonal antibodies and EGFR tyrosine kinase inhibitors as combination therapy for triple-negative breast cancer. Oncotarget 2018; 7:73618-73637. [PMID: 27655662 PMCID: PMC5342003 DOI: 10.18632/oncotarget.12037] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 08/22/2016] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by overexpression of epidermal growth factor receptor (EGFR) and activation of its downstream signaling pathways. Dual targeting of EGFR using one monoclonal antibody (mAb; cetuximab or panitumumab) and one tyrosine kinase inhibitor (EGFR-TKI; gefitinib or erlotinib) is a potential therapeutic approach. We investigated the effect of these therapies in EGFR-expressing TNBC cell lines that do or do not harbor the main activating mutations of EGFR pathways. Cell lines were sensitive to EGFR-TKIs, whereas mAbs were active only in MDA-MB-468 (EGFR amplification) and SUM-1315 (KRAS and PTEN wild-type) cells. MDA-MB-231 (KRAS mutated) and HCC-1937 (PTEN deletion) cells were resistant to mAbs. The combined treatment resulted in a synergistic effect on cell proliferation and superior inhibition of the RAS/MAPK signaling pathway in mAb-sensitive cells. The anti-proliferative effect was associated with G1 cell cycle arrest followed by apoptosis. Sensitivity to therapies was characterized by induction of positive regulators and inactivation of negative regulators of cell cycle. These results suggest that dual EGFR inhibition might result in an enhanced antitumor effect in a subgroup of TNBC. The status of EGFR, KRAS and PTEN could be used as a molecular marker for predicting the response to this therapeutic strategy.
Collapse
|
40
|
Albiol-Chiva J, Esteve-Romero J, Peris-Vicente J. Development of a method to determine axitinib, lapatinib and afatinib in plasma by micellar liquid chromatography and validation by the European Medicines Agency guidelines. J Chromatogr B Analyt Technol Biomed Life Sci 2018; 1074-1075:61-69. [PMID: 29331859 DOI: 10.1016/j.jchromb.2017.12.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 12/19/2017] [Accepted: 12/28/2017] [Indexed: 12/11/2022]
Abstract
A method based on micellar liquid chromatography to quantify the tyrosine kinase inhibitors axitinib, lapatinib and afatinib in plasma is reported. The sample pretreatment was a simple 1/5-dilution in a pure micellar solution, filtration and direct injection, without requiring extraction or purification steps. The three drugs were resolved from the matrix in 17min, using an aqueous solution of 0.07M sodium dodecyl sulfate - 6.0% 1-pentanol, buffered at pH7 with 0.01M phosphate salt as mobile phase, running under isocratic mode at 1mL/min through a C18 column. The detection was performed by absorbance at 260nm. An accurate mathematical relationship was established between the retention factor of each drug and the surfactant/organic solvent concentration in the mobile phase, achieved with a limited number of experiments, in order to optimize these factors. A binding behavior of the analytes face to the micelles was found out. The method was successfully validated by the guidelines of the European Medicines Agency in terms of: selectivity, linearity (r2>0.9995), calibration range (0.5 to 10mg/L), limit of detection (0.2mg/L), carry-over effect, accuracy (-8.1 to +6.9%), precision (<13.8%), dilution integrity, matrix effect, stability and robustness. The procedure was found reliable, practical, economic, accessible, short-time, easy-to-handle, inexpensive, environmental-friendly, safe, useful for the analysis of many samples per day. Finally, the method was applied to the analysis of incurred, using quality control samples in the same analytical run, with adequate results. Therefore, it can be implementable for routine analysis in clinical laboratories.
Collapse
Affiliation(s)
- Jaume Albiol-Chiva
- Departament de Química Física i Analítica, Universitat Jaume I, 12071 Castelló, Spain
| | - Josep Esteve-Romero
- Departament de Química Física i Analítica, Universitat Jaume I, 12071 Castelló, Spain
| | - Juan Peris-Vicente
- Departament de Química Física i Analítica, Universitat Jaume I, 12071 Castelló, Spain; Departament de Química Analítica, Universitat de València, Av/Doctor Moliner 50, 46100 Burjassot, Valencia, Spain.
| |
Collapse
|
41
|
He Y, Zhou L, Gao S, Yin T, Tu Y, Rayford R, Wang X, Hu M. Development and validation of a sensitive LC–MS/MS method for simultaneous determination of eight tyrosine kinase inhibitors and its application in mice pharmacokinetic studies. J Pharm Biomed Anal 2018; 148:65-72. [DOI: 10.1016/j.jpba.2017.09.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/15/2017] [Accepted: 09/08/2017] [Indexed: 11/26/2022]
|
42
|
|
43
|
The effects of lapatinib on CYP3A metabolism of midazolam in patients with advanced cancer. Cancer Chemother Pharmacol 2017; 80:1141-1146. [PMID: 29098381 DOI: 10.1007/s00280-017-3470-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 10/16/2017] [Indexed: 12/26/2022]
Abstract
PURPOSE The potential inhibition of CYP3A4 by lapatinib was studied using midazolam as a probe substrate in patients with cancer. METHODS This was a partially randomized, 4-period, 4-sequence, 4-treatment, cross-over study in 24 patients with advanced cancer. Single 1-mg IV and 3-mg oral doses of midazolam were given 2 days apart, in a partially random order, on study days 1, 3, 9, and 11. Lapatinib 1500-mg was administered orally once daily on study days 4 through 11. Midazolam plasma concentrations were measured up to 24-h post dosing, and lapatinib plasma concentrations measured prior to each midazolam dose. RESULTS Lapatinib increased the geometric mean (95% CIs) midazolam AUC(o-∞) by 45% (31-60%) after the oral dose and by 14% (0-29%) after the IV dose, and prolonged the midazolam elimination half-life by 48% (22-81%) after the oral dose and by 20% (2-40%) after the IV dose. Lapatinib decreased midazolam total clearance by 13% (1-23%), while total bioavailability was increased 23% (4-46%) without changes in apparent volume of distribution or hepatic bioavailability. CONCLUSION These data show that lapatinib caused weak inhibition of gastrointestinal CYP3A4 in vivo. This suggests that oral CYP3A4 drug substrates with a narrow therapeutic index may need dose reduction if lapatinib is to be co-prescribed.
Collapse
|
44
|
Karbownik A, Szałek E, Sobańska K, Klupczynska A, Plewa S, Grabowski T, Wolc A, Moch M, Kokot ZJ, Grześkowiak E. A pharmacokinetic study on lapatinib in type 2 diabetic rats. Pharmacol Rep 2017; 70:191-195. [PMID: 29471066 DOI: 10.1016/j.pharep.2017.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/31/2017] [Accepted: 09/15/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND Diabetes mellitus (DM) is a complex metabolic disorder which affects the function of numerous tissues and alters the pharmacokinetic parameters of many drugs. As many oncological patients are diabetics, it is important to determine the influence of this chronic disease on the pharmacokinetics (PK) of anticancer drugs. Lapatinib is a tyrosine kinase inhibitor (TKI), approved for the treatment of human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer. The aim of the study was to compare the PK of lapatinib in normal and type 2 diabetes mellitus (T2DM) model rats. Additionally, the effect of lapatinib on blood glucose concentrations was examined. METHODS The PK of lapatinib was studied in healthy rats (n=6, the healthy group) and T2DM model rats (n=6, the diabetic group). The rats received lapatinib orally as a single dose of 50mg. Plasma concentrations of lapatinib were measured with high-performance liquid chromatography method coupled with a tandem mass spectrometry. RESULTS The plasma concentrations of lapatinib were increased in the T2DM model rats. There were statistically significant differences between the groups in Cmax (p=0.0104) and AUC0-t (p=0.0265). The reduction of glycaemia in the range of 1.2-41.5% and in the range of 4.1-36.8% was observed in the diabetic and healthy animals, respectively. CONCLUSIONS Higher concentrations of lapatinib in the diabetic rats may suggest the need for application of lower doses of this TKI in patients with DM.
Collapse
Affiliation(s)
- Agnieszka Karbownik
- Department of Clinical Pharmacy and Biopharmacy, Poznań University of Medical Sciences, Poznań, Poland.
| | - Edyta Szałek
- Department of Clinical Pharmacy and Biopharmacy, Poznań University of Medical Sciences, Poznań, Poland
| | - Katarzyna Sobańska
- Department of Clinical Pharmacy and Biopharmacy, Poznań University of Medical Sciences, Poznań, Poland
| | - Agnieszka Klupczynska
- Department of Inorganic and Analytical Chemistry, Poznań University of Medical Sciences, Poznań, Poland
| | - Szymon Plewa
- Department of Inorganic and Analytical Chemistry, Poznań University of Medical Sciences, Poznań, Poland
| | | | - Anna Wolc
- Department of Animal Science, Iowa State University, Ames, IA, USA; Hy-Line International, Dallas Center, IA USA
| | - Marta Moch
- Department of Clinical Pharmacy and Biopharmacy, Poznań University of Medical Sciences, Poznań, Poland
| | - Zenon J Kokot
- Department of Inorganic and Analytical Chemistry, Poznań University of Medical Sciences, Poznań, Poland
| | - Edmund Grześkowiak
- Department of Clinical Pharmacy and Biopharmacy, Poznań University of Medical Sciences, Poznań, Poland
| |
Collapse
|
45
|
A novel humanized mouse lacking murine P450 oxidoreductase for studying human drug metabolism. Nat Commun 2017; 8:39. [PMID: 28659616 PMCID: PMC5489481 DOI: 10.1038/s41467-017-00049-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 05/02/2017] [Indexed: 12/30/2022] Open
Abstract
Only one out of 10 drugs in development passes clinical trials. Many fail because experimental animal models poorly predict human xenobiotic metabolism. Human liver chimeric mice are a step forward in this regard, as the human hepatocytes in chimeric livers generate human metabolites, but the remaining murine hepatocytes contain an expanded set of P450 cytochromes that form the major class of drug-metabolizing enzymes. We therefore generated a conditional knock-out of the NADPH-P450 oxidoreductase (Por) gene combined with Il2rg− /−/Rag2− /−/Fah− /− (PIRF) mice. Here we show that homozygous PIRF mouse livers are readily repopulated with human hepatocytes, and when the murine Por gene is deleted (<5%), they predominantly use human cytochrome metabolism. When given the anticancer drug gefitinib or the retroviral drug atazanavir, the Por-deleted humanized PIRF mice develop higher levels of the major human metabolites than current models. Humanized, murine Por-deficient PIRF mice can thus predict human drug metabolism and should be useful for preclinical drug development. Human liver chimeric mice are increasingly used for drug testing in preclinical development, but express residual murine p450 cytochromes. Here the authors generate mice lacking the Por gene in the liver, and show that human cytochrome metabolism is used following repopulation with human hepatocytes.
Collapse
|
46
|
Wei D, Zhang H, Peng R, Huang C, Bai R. ABCC2 (1249G > A) polymorphism implicates altered transport activity for sorafenib. Xenobiotica 2016; 47:1008-1014. [PMID: 27855531 DOI: 10.1080/00498254.2016.1262976] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
1. Multidrug resistance-associated protein 2 (MRP2), encoded by the ABCC2 gene, is an efflux transporter of several endogenous substrates and xenobiotics. Here, we investigated whether the 1249G > A (rs2273697) polymorphism in ABCC2 affects the ability of MRP2 to pump the multi-tumor drug sorafenib out of cells. 2. Human embryonic kidney 293 (HEK 293) cell lines transfected with ABCC2-1249G and ABCC2-1249A were used to assess the sensitivity and accumulation to sorafenib. The isolated MRP2 were applied to estimate the ATPase activity. 3. The HEK293 cell line overexpressing the ABCC2 1249A allele showed a significantly higher 50% inhibitory concentration (IC50) than a cell line overexpressing ABCC2-1249G or a non-overexpressing control cell line. Intracellular accumulation of sorafenib was much lower in ABCC2-1249A cells than in ABCC2-1249G cells expressing comparable levels of MRP2. Isolated ABCC2-1249A protein showed higher ATPase activity than ABCC2-1249G protein. 4. Our results suggest that the ABCC2 polymorphism 1249G > A increases the ATPase activity of MRP2, leading to greater efflux of sorafenib.
Collapse
Affiliation(s)
- Danyun Wei
- a Wuhan University Renmin Hospital , Wuhan , China
| | - Hong Zhang
- a Wuhan University Renmin Hospital , Wuhan , China
| | - Rui Peng
- a Wuhan University Renmin Hospital , Wuhan , China
| | | | - Ruidan Bai
- a Wuhan University Renmin Hospital , Wuhan , China
| |
Collapse
|
47
|
A Simple High-Performance Liquid Chromatography for Determining Lapatinib and Erlotinib in Human Plasma. Ther Drug Monit 2016; 38:657-662. [DOI: 10.1097/ftd.00000000000003520] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
48
|
Sawicki E, Schellens JHM, Beijnen JH, Nuijen B. Inventory of oral anticancer agents: Pharmaceutical formulation aspects with focus on the solid dispersion technique. Cancer Treat Rev 2016; 50:247-263. [PMID: 27776286 DOI: 10.1016/j.ctrv.2016.09.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/13/2016] [Indexed: 11/16/2022]
Abstract
Dissolution from the pharmaceutical formulation is a prerequisite for complete and consistent absorption of any orally administered drug, including anticancer agents (oncolytics). Poor dissolution of an oncolytic can result in low oral bioavailability, high variability in blood concentrations and with that suboptimal or even failing therapy. This review discusses pharmaceutical formulation aspects and absorption pharmacokinetics of currently licensed orally administered oncolytics. In nearly half of orally dosed oncolytics poor dissolution is likely to play a major role in low and unpredictable absorption. Dissolution-limited drug absorption can be improved with a solid dispersion which is a formulation method that induces super-saturated drug dissolution and with that it enhances in vivo absorption. This review discusses formulation principles with focus on the solid dispersion technology and how it works to enhance drug absorption. There are currently three licensed orally dosed oncolytics formulated as a solid dispersion (everolimus, vemurafenib and regorafenib) and these formulations result in remarkably improved dissolution and absorption compared to what can be achieved with conventional formulations of the respective oncolytics. Because of the successful implementation of these three solid dispersion formulations, we encourage the application of this formulation method for poorly soluble oral oncolytics.
Collapse
Affiliation(s)
- E Sawicki
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute/MC Slotervaart, Louwesweg 6, 1066 EC Amsterdam, The Netherlands.
| | - J H M Schellens
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; Science Faculty, Utrecht Institute for Pharmaceutical Sciences (UIPS), Division of Pharmaco-epidemiology & Clinical Pharmacology, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - J H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute/MC Slotervaart, Louwesweg 6, 1066 EC Amsterdam, The Netherlands; Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands; Science Faculty, Utrecht Institute for Pharmaceutical Sciences (UIPS), Division of Pharmaco-epidemiology & Clinical Pharmacology, Utrecht University, P.O. Box 80082, 3508 TB Utrecht, The Netherlands
| | - B Nuijen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute/MC Slotervaart, Louwesweg 6, 1066 EC Amsterdam, The Netherlands
| |
Collapse
|
49
|
Eigenmann MJ, Frances N, Hoffmann G, Lavé T, Walz AC. Combining Nonclinical Experiments with Translational PKPD Modeling to Differentiate Erlotinib and Gefitinib. Mol Cancer Ther 2016; 15:3110-3119. [DOI: 10.1158/1535-7163.mct-16-0076] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 08/02/2016] [Accepted: 08/26/2016] [Indexed: 11/16/2022]
|
50
|
Rowland A, van Dyk M, Mangoni AA, Miners JO, McKinnon RA, Wiese MD, Rowland A, Kichenadasse G, Gurney H, Sorich MJ. Kinase inhibitor pharmacokinetics: comprehensive summary and roadmap for addressing inter-individual variability in exposure. Expert Opin Drug Metab Toxicol 2016; 13:31-49. [DOI: 10.1080/17425255.2016.1229303] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|