1
|
Li T, Zhou S, Wang L, Zhao T, Wang J, Shao F. Docetaxel, cyclophosphamide, and epirubicin: application of PBPK modeling to gain new insights for drug-drug interactions. J Pharmacokinet Pharmacodyn 2024; 51:367-384. [PMID: 38554227 DOI: 10.1007/s10928-024-09912-z] [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: 12/25/2023] [Accepted: 02/20/2024] [Indexed: 04/01/2024]
Abstract
The new adjuvant chemotherapy of docetaxel, epirubicin, and cyclophosphamide has been recommended for treating breast cancer. It is necessary to investigate the potential drug-drug Interactions (DDIs) since they have a narrow therapeutic window in which slight differences in exposure might result in significant differences in treatment efficacy and tolerability. To guide clinical rational drug use, this study aimed to evaluate the DDI potentials of docetaxel, cyclophosphamide, and epirubicin in cancer patients using physiologically based pharmacokinetic (PBPK) models. The GastroPlus™ was used to develop the PBPK models, which were refined and validated with observed data. The established PBPK models accurately described the pharmacokinetics (PKs) of three drugs in cancer patients, and the predicted-to-observed ratios of all the PK parameters met the acceptance criterion. The PBPK model predicted no significant changes in plasma concentrations of these drugs during co-administration, which was consistent with the observed clinical phenomenon. Besides, the verified PBPK models were then used to predict the effect of other Cytochrome P450 3A4 (CYP3A4) inhibitors/inducers on these drug exposures. In the DDI simulation, strong CYP3A4 modulators changed the exposure of three drugs by 0.71-1.61 fold. Therefore, patients receiving these drugs in combination with strong CYP3A4 inhibitors should be monitored regularly to prevent adverse reactions. Furthermore, co-administration of docetaxel, cyclophosphamide, or epirubicin with strong CYP3A4 inducers should be avoided. In conclusion, the PBPK models can be used to further investigate the DDI potential of each drug and to develop dosage recommendations for concurrent usage by additional perpetrators or victims.
Collapse
Affiliation(s)
- Tongtong Li
- Phase I Clinical Trial Unit, The First Affiliated Hospital With Nanjing Medical University, Nanjing, 210029, China
- Department of Clinical Pharmacology, School of Pharmacy College, Nanjing Medical University, Nanjing, 211166, China
| | - Sufeng Zhou
- Phase I Clinical Trial Unit, The First Affiliated Hospital With Nanjing Medical University, Nanjing, 210029, China
| | - Lu Wang
- Phase I Clinical Trial Unit, The First Affiliated Hospital With Nanjing Medical University, Nanjing, 210029, China
| | - Tangping Zhao
- Phase I Clinical Trial Unit, The First Affiliated Hospital With Nanjing Medical University, Nanjing, 210029, China
- Department of Clinical Pharmacology, School of Pharmacy College, Nanjing Medical University, Nanjing, 211166, China
| | - Jue Wang
- Division of Breast Surgery, The First Affiliated Hospital With Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, Jiangsu Province, China
| | - Feng Shao
- Phase I Clinical Trial Unit, The First Affiliated Hospital With Nanjing Medical University, Nanjing, 210029, China.
- Department of Clinical Pharmacology, School of Pharmacy College, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
2
|
Sun Y, Cheng Y, Hertz DL. Using maximum plasma concentration (C max) to personalize taxane treatment and reduce toxicity. Cancer Chemother Pharmacol 2024; 93:525-539. [PMID: 38734836 DOI: 10.1007/s00280-024-04677-1] [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: 02/01/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Taxanes are a widely used class of anticancer agents that play a vital role in the treatment of a variety of cancers. However, toxicity remains a major concern of using taxane drugs as some toxicities are highly prevalent, they can not only adversely affect patient prognosis but also compromise the overall treatment plan. Among all kinds of factors that associated with taxane toxicity, taxane exposure has been extensively studied, with different pharmacokinetic (PK) parameters being used as toxicity predictors. Compared to other widely used predictors such as the area under the drug plasma concentration curve versus time (AUC) and time above threshold plasma drug concentration, maximum plasma concentration (Cmax) is easier to collect and shows promise for use in clinical practice. In this article, we review the previous research on using Cmax to predict taxane treatment outcomes. While Cmax and toxicity have been extensively studied, research on the relationship between Cmax and efficacy is lacking. Most of the articles find a positive relationship between Cmax and toxicity but several articles have contradictory findings. Future clinical trials are needed to validate the relationship between Cmax and treatment outcome and determine whether Cmax can serve as a useful surrogate endpoint of taxane treatment efficacy.
Collapse
Affiliation(s)
- Yuchen Sun
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Yue Cheng
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Daniel L Hertz
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI, USA.
| |
Collapse
|
3
|
Zhao J, Yan D, Li Y, Xu X, Li F, Zhang S, Jin J, Qiu F. Simultaneous determination of 11 oral targeted antineoplastic drugs and 2 active metabolites by LC-MS/MS in human plasma and its application to therapeutic drug monitoring in cancer patients. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1237:124100. [PMID: 38547701 DOI: 10.1016/j.jchromb.2024.124100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/11/2024] [Accepted: 03/17/2024] [Indexed: 04/13/2024]
Abstract
Interindividual exposure differences have been identified in oral targeted antineoplastic drugs (OADs) owing to the pharmacogenetic background of the patients and their susceptibility to multiple factors, resulting in insufficient efficacy or adverse effects. Therapeutic drug monitoring (TDM) can prevent sub-optimal concentrations of OADs and improve their clinical treatment. This study aimed to develop and validate an LC-MS/MS method for the simultaneous quantification of 11 OADs (gefitinib, imatinib, lenvatinib, regorafenib, everolimus, osimertinib, sunitinib, tamoxifen, lapatinib, fruquintinib and sorafenib) and 2 active metabolites (N-desethyl sunitinib and Z-endoxifen) in human plasma. Protein precipitation was used to extract OADs from the plasma samples. Chromatographic separation was performed using an Eclipse XDB-C18 (4.6 × 150 mm, 5 μm) column with a gradient elution of the mobile phase composed of 2 mM ammonium acetate with 0.1 % formic acid in water (solvent A) and methanol (solvent B) at a flow rate of 0.8 mL/min. Mass analysis was performed using positive ion mode electrospray ionization in multiple-reaction monitoring mode. The developed method was validated following FDA bioanalytical guidelines. The calibration curves were linear over the range of 2-400 ng/mL for gefitinib, imatinib, lenvatinib, regorafenib, and everolimus; 1-200 ng/mL for osimertinib, sunitinib, N-desethyl sunitinib, tamoxifen, and Z-endoxifen; and 5-1000 ng/mL for lapatinib, fruquintinib, and sorafenib, with all coefficients of correlation above 0.99. The intra- and inter-day imprecision was below 12.81 %. This method was successfully applied to the routine TDM of gefitinib, lenvatinib, regorafenib, osimertinib, fruquintinib, and sorafenib to optimize the dosage regimens.
Collapse
Affiliation(s)
- Jing Zhao
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China
| | - Dongming Yan
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China
| | - Yue Li
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China
| | - Xiaoqing Xu
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China
| | - Fengling Li
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China
| | - Shuang Zhang
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China
| | - Jingyi Jin
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China.
| | - Furong Qiu
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201213, China.
| |
Collapse
|
4
|
Maliszewska O, Roszkowska A, Lipiński M, Treder N, Olędzka I, Kowalski P, Bączek T, Bień E, Krawczyk MA, Plenis A. Profiling Docetaxel in Plasma and Urine Samples from a Pediatric Cancer Patient Using Ultrasound-Assisted Dispersive Liquid-Liquid Microextraction Combined with LC-MS/MS. Pharmaceutics 2023; 15:pharmaceutics15041255. [PMID: 37111740 PMCID: PMC10143245 DOI: 10.3390/pharmaceutics15041255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
In recent years, therapeutic drug monitoring (TDM) has been applied in docetaxel (DOC)-based anticancer therapy to precisely control various pharmacokinetic parameters, including the concentration of DOC in biofluids (e.g., plasma or urine), its clearance, and its area under the curve (AUC). The ability to determine these values and to monitor DOC levels in biological samples depends on the availability of precise and accurate analytical methods that both enable fast and sensitive analysis and can be implemented in routine clinical practice. This paper presents a new method for isolating DOC from plasma and urine samples based on the coupling of microextraction and advanced liquid chromatography with tandem mass spectrometry (LC-MS/MS). In the proposed method, biological samples are prepared via ultrasound-assisted dispersive liquid-liquid microextraction (UA-DLLME) using ethanol (EtOH) and chloroform (Chl) as the desorption and extraction solvents, respectively. The proposed protocol was fully validated according to the Food and Drug Administration (FDA) and the International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use (ICH) requirements. The developed method was then applied to monitor the DOC profile in plasma and urine samples collected from a pediatric patient suffering from cardiac angiosarcoma (AS) with metastasis to lungs and mediastinal lymph nodes, who was receiving treatment with DOC at a dose of 30 mg/m2 body surface area. Due to the rarity of this disease, TDM was carried out to determine the exact levels of DOC at particular time points to ascertain which levels were conducive to maximizing the treatment's effectiveness while minimizing the drug's toxicity. To this end, the concentration-time profiles of DOC in the plasma and urine samples were determined, and the levels of DOC at specific time intervals up to 3 days after administration were measured. The results showed that DOC was present at higher concentrations in the plasma than in the urine samples, which is due to the fact that this drug is primarily metabolized in the liver and then eliminated with the bile. The obtained data provided information about the pharmacokinetic profile of DOC in pediatric patients with cardiac AS, which enabled the dose to be adjusted to achieve the optimal therapeutic regimen. The findings of this work demonstrate that the optimized method can be applied for the routine monitoring of DOC levels in plasma and urine samples as a part of pharmacotherapy in oncological patients.
Collapse
Affiliation(s)
- Olga Maliszewska
- Department of Analytical Chemistry, Medical University of Gdansk, 80-416 Gdańsk, Poland
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416 Gdańsk, Poland
| | - Anna Roszkowska
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416 Gdańsk, Poland
| | - Marcin Lipiński
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, 80-211 Gdańsk, Poland
| | - Natalia Treder
- Department of Analytical Chemistry, Medical University of Gdansk, 80-416 Gdańsk, Poland
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416 Gdańsk, Poland
| | - Ilona Olędzka
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416 Gdańsk, Poland
| | - Piotr Kowalski
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416 Gdańsk, Poland
| | - Tomasz Bączek
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, 80-416 Gdańsk, Poland
| | - Ewa Bień
- Department of Pediatrics, Hematology and Oncology, Medical University of Gdansk, 80-211 Gdańsk, Poland
| | - Małgorzata Anna Krawczyk
- Department of Pediatrics, Hematology and Oncology, Medical University of Gdansk, 80-211 Gdańsk, Poland
| | - Alina Plenis
- Department of Analytical Chemistry, Medical University of Gdansk, 80-416 Gdańsk, Poland
| |
Collapse
|
5
|
Fan W, Yin W, Zhou F, Wang Y, Fan J, Zang F, Lin B. The correlation between paclitaxel chemotoxicity and the plasma albumin level in cancer patients. J Clin Pharm Ther 2022; 47:2237-2244. [PMID: 36325658 DOI: 10.1111/jcpt.13798] [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: 05/23/2022] [Revised: 09/08/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE The aim of this study was to evaluate the pharmacokinetics of paclitaxel in cancer patients with hypoalbuminemia following paclitaxel-containing chemotherapy and to provide a reference for the prevention of adverse events (AEs) after paclitaxel administration. METHODS Peripheral blood was collected from cancer patients treated with paclitaxel. The plasma concentration of paclitaxel was determined by ultra-high performance liquid chromatography after 24 ± 8 h of chemotherapy, and individual paclitaxel time above a threshold concentration of 0.05 μmol/L (Tc>0.05 ) was calculated using the population pharmacokinetic model. Haematological and non-haematological toxicities were monitored after chemotherapy, and the correlation between different chemotherapy toxicities and Tc>0.05 was evaluated using the Prism software. RESULTS AND DISCUSSION The enrolled patients were divided into the hypoalbuminemia group and normal albumin level group. The mean Tc>0.05 values in the normal albumin level and hypoalbuminemia groups were 36.89 and 24.93 h, respectively (P < 0.001). The risk of myelosuppression was positively correlated with Tc>0.05 . Due to the lower Tc>0.05 , the incidences of immediate AEs such as gastrointestinal reactions and rashes were higher in the hypoalbuminemia group than in the normal albumin level group, and the incidences of delayed AEs such as myelosuppression and neurotoxicity were lower in the hypoalbuminemia group. WHAT IS NEW AND CONCLUSIONS Plasma albumin level has a conclusive effect on Tc>0.05 , which can predict the potential clinical toxicity of paclitaxel. The study provides a theoretical basis for administration of paclitaxel.
Collapse
Affiliation(s)
- Weibin Fan
- Department of Pharmacy, Changxing People's Hospital, Huzhou, China.,Key Laboratory of Intelligent Pharmacy and Individualized Therapy of Huzhou, Huzhou, China
| | - Weiming Yin
- Department of Pharmacy, Changxing People's Hospital, Huzhou, China.,Key Laboratory of Intelligent Pharmacy and Individualized Therapy of Huzhou, Huzhou, China
| | - Feng Zhou
- Department of Respiratory, Zhejiang University School of Medicine Second Affiliated Hospital - Changxing Branch, Huzhou, China
| | - Yinhui Wang
- Department of Pharmacy, Changxing People's Hospital, Huzhou, China.,Key Laboratory of Intelligent Pharmacy and Individualized Therapy of Huzhou, Huzhou, China
| | - Jing Fan
- Department of Pharmacy, Changxing People's Hospital, Huzhou, China.,Key Laboratory of Intelligent Pharmacy and Individualized Therapy of Huzhou, Huzhou, China
| | - Farong Zang
- Department of Respiratory, Zhejiang University School of Medicine Second Affiliated Hospital - Changxing Branch, Huzhou, China
| | - Bin Lin
- Department of Pharmacy, Changxing People's Hospital, Huzhou, China.,Key Laboratory of Intelligent Pharmacy and Individualized Therapy of Huzhou, Huzhou, China
| |
Collapse
|
6
|
Stojanova J, Carland JE, Murnion B, Seah V, Siderov J, Lemaitre F. Therapeutic drug monitoring in oncology - What’s out there: A bibliometric evaluation on the topic. Front Oncol 2022; 12:959741. [PMID: 36439413 PMCID: PMC9685987 DOI: 10.3389/fonc.2022.959741] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 10/07/2022] [Indexed: 11/12/2022] Open
Abstract
Pharmacological therapy is the mainstay of treatment for cancer patients. Despite wide interpatient variability in systemic drug concentrations for numerous antineoplastics, dosing based on body size remains the predominant approach. Therapeutic drug monitoring (TDM) is used for few antineoplastics in specific scenarios. We conducted a rapid bibliometric evaluation of TDM in oncology to capture a snapshot of research in this area over time and explore topics that reflect development in the field. Reports with the composite, indexed term ‘therapeutic drug monitoring’ in the title and abstract were extracted from MEDLINE (inception to August 2021). Reports related to applications in cancer were selected for inclusion and were tagged by study design, antineoplastic drugs and concepts related to TDM. We present a timeline from 1980 to the present indicating the year of first report of antineoplastic agents and key terms. The reports in our sample primarily reflected development and validation of analytical methods with few relating to clinical outcomes to support implementation. Our work emphasises evidence gaps that may contribute to poor uptake of TDM in oncology.
Collapse
Affiliation(s)
- Jana Stojanova
- Department of Clinical Pharmacology and Toxicology, St. Vincent’s Hospital, Sydney, NSW, Australia
- Interdisciplinary Centre for Health Studies (CIESAL), Universidad de Valparaiíso, Valparaiíso, Chile
- *Correspondence: Jana Stojanova,
| | - Jane E. Carland
- Department of Clinical Pharmacology and Toxicology, St. Vincent’s Hospital, Sydney, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, Sydney, NSW, Australia
| | - Bridin Murnion
- Department of Clinical Pharmacology and Toxicology, St. Vincent’s Hospital, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Vincent Seah
- Department of Clinical Pharmacology and Toxicology, St. Vincent’s Hospital, Sydney, NSW, Australia
| | - Jim Siderov
- Pharmacy Department, Austin Health, Heidelberg, VIC, Australia
| | - Florian Lemaitre
- Université de Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail), Rennes, France
- INSERM, Centre d’Investigation Clinique, Rennes, France
| |
Collapse
|
7
|
Ultrasonic Extraction and Separation of Taxanes from Taxus cuspidata Optimized by Response Surface Methodology. SEPARATIONS 2022. [DOI: 10.3390/separations9080193] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Taxanes are natural compounds with strong antitumor activity. In this study, we first extracted taxanes from the needles of Taxus cuspidata using ultrasonic (US) extraction, and then assessed the effects of different extraction conditions on the yields of eight target compounds. Response surface methodology (RSM) was further used to optimize the extraction conditions: when the liquid-to-solid ratio was 20.88 times, ultrasonic power was 140.00 W, ultrasonic time was 47.63 min, and ethanol content in solvent was 83.50%, taxane yields reached the maximum value of 354.28 μg/g. Under these conditions, the actual extraction rate of taxanes from the needles was 342.27 μg/g. The scanning electron microscopy (SEM) results indicated that the morphology of the needles, suspension cells, and callus of Taxus cuspidata extracted by ultrasonic wave had changed, the pores of the sections of the needles extracted by ultrasonic wave had become relatively loose, and the pore diameter had obviously increased. The callus and overall structure of the suspension cells extracted by ultrasonic wave were destroyed, forming cell fragments. The components of Taxus cuspidata are complex; the high-performance liquid chromatography (HPLC) method established in this paper is suitable for the rapid and effective separation of taxanes in Taxus cuspidata. We systematically and comprehensively compared the yields of taxanes in needles, callus, and suspension cells of Taxus cuspidata, and the taxane yields were increased by the suspension cell culture.
Collapse
|
8
|
Agema BC, Veerman GDM, Steendam CMJ, Lanser DAC, Preijers T, van der Leest C, Koch BCP, Dingemans AMC, Mathijssen RHJ, Koolen SLW. Improving the tolerability of osimertinib by identifying its toxic limit. Ther Adv Med Oncol 2022; 14:17588359221103212. [PMID: 35677320 PMCID: PMC9168866 DOI: 10.1177/17588359221103212] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/08/2022] [Indexed: 01/05/2023] Open
Abstract
Background: Osimertinib is the cornerstone in the treatment of epidermal growth factor
receptor-mutated non-small cell lung cancer (NSCLC). Nonetheless, ±25% of
patients experience severe treatment-related toxicities. Currently, it is
impossible to identify patients at risk of severe toxicity beforehand.
Therefore, we aimed to study the relationship between osimertinib exposure
and severe toxicity and to identify a safe toxic limit for a preventive dose
reduction. Methods: In this real-life prospective cohort study, patients with NSCLC treated with
osimertinib were followed for severe toxicity (grade ⩾3 toxicity, dose
reduction or discontinuation, hospital admission, or treatment termination).
Blood for pharmacokinetic analyses was withdrawn during every out-patient
visit. Primary endpoint was the correlation between osimertinib clearance
(exposure) and severe toxicity. Secondary endpoint was the exposure–efficacy
relationship, defined as progression-free survival (PFS) and overall
survival (OS). Results: In total, 819 samples from 159 patients were included in the analysis.
Multivariate competing risk analysis showed osimertinib clearance
(c.q. exposure) to be significantly correlated with
severe toxicity (hazard ratio 0.93, 95% CI: 0.88–0.99). An relative
operating characteristic curve showed the optimal toxic limit to be
259 ng/mL osimertinib. A 50% dose reduction in the high-exposure group, that
is 25.8% of the total cohort, would reduce the risk of severe toxicity by
53%. Osimertinib exposure was not associated with PFS nor OS. Conclusion: Osimertinib exposure is highly correlated with the occurrence of severe
toxicity. To optimize tolerability, patients above the toxic limit
concentration of 259 ng/mL could benefit from a preventive dose reduction,
without fear for diminished effectiveness.
Collapse
Affiliation(s)
- Bram C. Agema
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, Rotterdam 3015 GD, The Netherlands Department of Clinical Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - G. D. Marijn Veerman
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Pulmonology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Christi M. J. Steendam
- Department of Pulmonology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Pulmonology, Amphia Hospital, Breda, The Netherlands
| | - Daan A. C. Lanser
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Tim Preijers
- Department of Clinical Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Birgit C. P. Koch
- Department of Clinical Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Anne-Marie C. Dingemans
- Department of Pulmonology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ron H. J. Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Stijn L. W. Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands Department of Clinical Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
9
|
Lu L, Xu Q, Wang J, Wu S, Luo Z, Lu W. Drug Nanocrystals for Active Tumor-Targeted Drug Delivery. Pharmaceutics 2022; 14:pharmaceutics14040797. [PMID: 35456631 PMCID: PMC9026472 DOI: 10.3390/pharmaceutics14040797] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/19/2022] [Accepted: 03/25/2022] [Indexed: 12/17/2022] Open
Abstract
Drug nanocrystals, which are comprised of active pharmaceutical ingredients and only a small amount of essential stabilizers, have the ability to improve the solubility, dissolution and bioavailability of poorly water-soluble drugs; in turn, drug nanocrystal technology can be utilized to develop novel formulations of chemotherapeutic drugs. Compared with passive targeting strategy, active tumor-targeted drug delivery, typically enabled by specific targeting ligands or molecules modified onto the surface of nanomedicines, circumvents the weak and heterogeneous enhanced permeability and retention (EPR) effect in human tumors and overcomes the disadvantages of nonspecific drug distribution, high administration dosage and undesired side effects, thereby contributing to improving the efficacy and safety of conventional nanomedicines for chemotherapy. Continuous efforts have been made in the development of active tumor-targeted drug nanocrystals delivery systems in recent years, most of which are encouraging and also enlightening for further investigation and clinical translation.
Collapse
Affiliation(s)
- Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China;
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; (Q.X.); (J.W.); (S.W.); (Z.L.)
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
| | - Qianzhu Xu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; (Q.X.); (J.W.); (S.W.); (Z.L.)
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
| | - Jun Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; (Q.X.); (J.W.); (S.W.); (Z.L.)
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
| | - Sunyi Wu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; (Q.X.); (J.W.); (S.W.); (Z.L.)
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
| | - Zimiao Luo
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; (Q.X.); (J.W.); (S.W.); (Z.L.)
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; (Q.X.); (J.W.); (S.W.); (Z.L.)
- Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China
- Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, and Shanghai Frontiers Science Center for Druggability of Cardiovascular Non-Coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China
- Correspondence:
| |
Collapse
|