1
|
Aoki Y, Rowland M, Sugiyama Y. When to consider intra-target microdosing: physiologically based pharmacokinetic modeling approach to quantitatively identify key factors for observing target engagement. Front Pharmacol 2024; 15:1366160. [PMID: 39119606 PMCID: PMC11306728 DOI: 10.3389/fphar.2024.1366160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 05/29/2024] [Indexed: 08/10/2024] Open
Abstract
Intra-Target Microdosing (ITM), integral to Phase 0 clinical studies, offers a novel approach in drug development, effectively bridging the gap between preclinical and clinical phases. This methodology is especially relevant in streamlining early drug development stages. Our research utilized a Physiologically Based Pharmacokinetic (PBPK) model and Monte Carlo simulations to examine factors influencing the effectiveness of ITM in achieving target engagement. The study revealed that ITM is capable of engaging targets at levels akin to systemically administered therapeutic doses for specific compounds. However, we also observed a notable decrease in the probability of success when the predicted therapeutic dose exceeds 10 mg. Additionally, our findings identified several critical factors affecting the success of ITM. These encompass both lower dissociation constants, higher systemic clearance and an optimum abundance of receptors in the target organ. Target tissues characterized by relatively low blood flow rates and high drug clearance capacities were deemed more conducive to successful ITM. These insights emphasize the necessity of taking into account each drug's unique pharmacokinetic and pharmacodynamic properties, along with the physiological characteristics of the target tissue, in determining the suitability of ITM.
Collapse
Affiliation(s)
- Yasunori Aoki
- Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, Josai International University, Tokyo, Japan
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Malcom Rowland
- Centre for Applied Pharmacokinetic Research, School of Pharmacy, University of Manchester, Manchester, United Kingdom
| | - Yuichi Sugiyama
- Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, Josai International University, Tokyo, Japan
- iHuman Institute, ShanghaiTech University, Shanghai, China
| |
Collapse
|
2
|
Mukai H, Ogawa K, Kato N, Kawakami S. Recent advances in lipid nanoparticles for delivery of nucleic acid, mRNA, and gene editing-based therapeutics. Drug Metab Pharmacokinet 2022; 44:100450. [PMID: 35381574 PMCID: PMC9363157 DOI: 10.1016/j.dmpk.2022.100450] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/30/2022] [Accepted: 02/01/2022] [Indexed: 12/26/2022]
Abstract
Lipid nanoparticles (LNPs) are becoming popular as a means of delivering therapeutics, including those based on nucleic acids and mRNA. The mRNA-based coronavirus disease 2019 vaccines are perfect examples to highlight the role played by drug delivery systems in advancing human health. The fundamentals of LNPs for the delivery of nucleic acid- and mRNA-based therapeutics, are well established. Thus, future research on LNPs will focus on addressing the following: expanding the scope of drug delivery to different constituents of the human body, expanding the number of diseases that can be targeted, and studying the change in the pharmacokinetics of LNPs under physiological and pathological conditions. This review article provides an overview of recent advances aimed at expanding the application of LNPs, focusing on the pharmacokinetics and advantages of LNPs. In addition, analytical techniques, library construction and screening, rational design, active targeting, and applicability to gene editing therapy have also been discussed.
Collapse
Affiliation(s)
- Hidefumi Mukai
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki, 852-8588, Japan; Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.
| | - Koki Ogawa
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki, 852-8588, Japan
| | - Naoya Kato
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki, 852-8588, Japan
| | - Shigeru Kawakami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki, 852-8588, Japan.
| |
Collapse
|
3
|
Advancements in practical and scientific bioanalytical approaches to metabolism studies in drug development. Bioanalysis 2021; 13:913-930. [PMID: 33961500 DOI: 10.4155/bio-2021-0050] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Advancement in metabolism profiling approaches and bioanalytical techniques has been revolutionized over the last two decades. Different in vitro and in vivo approaches along with advanced bioanalytical techniques are enabling the accurate qualitative and quantitative analysis of metabolites. This review summarizes various modern in vitro and in vivo approaches for executing metabolism studies with special emphasis on the recent advancement in the field. Advanced bioanalytical techniques, which can be employed in metabolism studies, have been discussed suggesting their particular application based on specific study objectives. This article can efficiently guide the researchers to scientifically plan metabolism studies and their bioanalysis during drug development programs taking advantage of a detailed understanding of instances of failure in the past.
Collapse
|
4
|
Mukai H, Watanabe Y. Review: PET imaging with macro- and middle-sized molecular probes. Nucl Med Biol 2021; 92:156-170. [PMID: 32660789 DOI: 10.1016/j.nucmedbio.2020.06.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 12/16/2022]
Abstract
Recent progress in radiolabeling of macro- and middle-sized molecular probes has been extending possibilities to use PET molecular imaging for dynamic application to drug development and therapeutic evaluation. Theranostics concept also accelerated the use of macro- and middle-sized molecular probes for sharpening the contrast of proper target recognition even the cellular types/subtypes and proper selection of the patients who should be treated by the same molecules recognition. Here, brief summary of the present status of immuno-PET, and then further development of advanced technologies related to immuno-PET, peptidic PET probes, and nucleic acids PET probes are described.
Collapse
Affiliation(s)
- Hidefumi Mukai
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| |
Collapse
|
5
|
Nomura S, Takahashi M, Kato AH, Wada Y, Watanabe Y, Yamashita F, Mukai H. Biosorption-based 64Cu-labeling of bacteria for pharmacokinetic positron-emission tomography. Int J Pharm 2020; 590:119950. [PMID: 33027635 DOI: 10.1016/j.ijpharm.2020.119950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/29/2020] [Accepted: 10/02/2020] [Indexed: 02/06/2023]
Abstract
Biosorption-based bacterial 64Cu-labeling and its application in pharmacokinetic positron-emission tomography (PET) were investigated. Both gram-positive and gram-negative bacteria were efficiently labeled with [64Cu]Cu2+ ion in saline at room temperature within 5 min. The labeling ratio for Escherichia coli drastically decreased with trypsin pretreatment and the co-presence of excess Cu2+ ion, indicating the existence of specific Cu2+ binding sites on the E. coli cell surface. Washing with lysogeny broth medium was effective in purifying 64Cu-labeled E. coli for kinetic study; the labeling stability was approximately 90% in serum for 15 min. According to dynamic PET imaging in colon-26 tumor-bearing mice, 64Cu-labeled E. coli immediately disappeared from the blood circulation and primarily accumulated in the liver. In addition, transient pulmonary distribution was observed, being in a dose-dependently accelerated manner. Considering the simplicity and versatility of biosorption-based bacterial 64Cu-labeling without genetic modification, the early-phase pharmacokinetic PET with 64Cu-labeled bacteria is promising for assessing toxicological aspects of bacteria-mediated cancer therapy as well as a variety of bacterial pathogenicities in infectious diseases.
Collapse
Affiliation(s)
- Shoko Nomura
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-shimoadachi cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Maiko Takahashi
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Akari Hashiba Kato
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yasuhiro Wada
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Fumiyoshi Yamashita
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-shimoadachi cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hidefumi Mukai
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Department of Pharmaceutical Informatics, Graduate School of Biomedical Science, Nagasaki University, 1-7-1 Sakamotomachi, Nagasaki 852-8588, Japan.
| |
Collapse
|
6
|
Burt T, Young G, Lee W, Kusuhara H, Langer O, Rowland M, Sugiyama Y. Phase 0/microdosing approaches: time for mainstream application in drug development? Nat Rev Drug Discov 2020; 19:801-818. [PMID: 32901140 DOI: 10.1038/s41573-020-0080-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
Abstract
Phase 0 approaches - which include microdosing - evaluate subtherapeutic exposures of new drugs in first-in-human studies known as exploratory clinical trials. Recent progress extends phase 0 benefits beyond assessment of pharmacokinetics to include understanding of mechanism of action and pharmacodynamics. Phase 0 approaches have the potential to improve preclinical candidate selection and enable safer, cheaper, quicker and more informed developmental decisions. Here, we discuss phase 0 methods and applications, highlight their advantages over traditional strategies and address concerns related to extrapolation and developmental timelines. Although challenges remain, we propose that phase 0 approaches be at least considered for application in most drug development scenarios.
Collapse
Affiliation(s)
- Tal Burt
- Burt Consultancy LLC. talburtmd.com, New York, NY, USA. .,Phase-0/Microdosing Network. Phase-0Microdosing.org, New York, NY, USA.
| | - Graeme Young
- GlaxoSmithKline Research and Development Ltd, Ware, UK
| | - Wooin Lee
- Seoul National University, Seoul, Republic of Korea
| | | | - Oliver Langer
- Medical University of Vienna, Vienna, Austria.,AIT Austrian Institute of Technology GmbH, Vienna, Austria
| | | | | |
Collapse
|
7
|
Burt T, Vuong LT, Baker E, Young GC, McCartt AD, Bergstrom M, Sugiyama Y, Combes R. Phase 0, including microdosing approaches: Applying the Three Rs and increasing the efficiency of human drug development. Altern Lab Anim 2019; 46:335-346. [PMID: 30657329 DOI: 10.1177/026119291804600603] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Phase 0 approaches, including microdosing, involve the use of sub-therapeutic exposures to the tested drugs, thus enabling safer, more-relevant, quicker and cheaper first-in-human (FIH) testing. These approaches also have considerable potential to limit the use of animals in human drug development. Recent years have witnessed progress in applications, methodology, operations, and drug development culture. Advances in applications saw an expansion in therapeutic areas, developmental scenarios and scientific objectives, in, for example, protein drug development and paediatric drug development. In the operational area, the increased sensitivity of Liquid Chromatography Tandem Mass Spectrometry (LC-MS/MS), expansion of the utility of Positron Emission Tomography (PET) imaging, and the introduction of Cavity Ring-Down Spectroscopy (CRDS), have led to the increased accessibility and utility of Phase 0 approaches, while reducing costs and exposure to radioactivity. PET has extended the application of microdosing, from its use as a predominant tool to record pharmacokinetics, to a method for recording target expression and target engagement, as well as cellular and tissue responses. Advances in methodology include adaptive Phase 0/Phase 1 designs, cassette and cocktail microdosing, and Intra-Target Microdosing (ITM), as well as novel modelling opportunities and simulations. Importantly, these methodologies increase the predictive power of extrapolation from microdose to therapeutic level exposures. However, possibly the most challenging domain in which progress has been made, is the culture of drug development. One of the main potential values of Phase 0 approaches is the opportunity to terminate development early, thus not only applying the principle of 'kill-early-kill-cheap' to enhance the efficiency of drug development, but also obviating the need for the full package of animal testing required for therapeutic level Phase 1 studies. Finally, we list developmental scenarios that utilised Phase 0 approaches in novel drug development.
Collapse
Affiliation(s)
- Tal Burt
- Burt Consultancy, LLC, Durham, NC, USA
| | | | - Elizabeth Baker
- Physicians Committee for Responsible Medicine, Washington, DC, USA
| | - Graeme C Young
- Translational Medicine, Research, GSK, David Jack Centre for R&D, Ware, Hertfordshire, UK
| | | | - Mats Bergstrom
- Department of Pharmacology and PET-centre, Uppsala University, Uppsala, Sweden
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN (The Institute of Physical and Chemical Research(, Yokohama, Kanagawa, Japan
| | | |
Collapse
|
8
|
Liu Y, Li L, Yu J, Fan YX, Lu XB. Carbon nanoparticle lymph node tracer improves the outcomes of surgical treatment in papillary thyroid cancer. Cancer Biomark 2019; 23:227-233. [PMID: 30198867 DOI: 10.3233/cbm-181386] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Papillary thyroid carcinoma (PTC) is the most common thyroid malignancy which is generally accompanied by lymph node metastasis. OBJECTIVE Our study evaluated whether carbon nanoparticle lymph node tracer can improve the outcomes of surgical treatment in papillary thyroid cancer (PTC). METHODS Ninety-two patients were selected and underwent total thyroidectomy and lymph node resection. Our study placed 45 individuals into the treatment group (carbon nanoparticle group) and 47 cohorts in the control group (no carbon nanoparticle group). RESULTS Carbon nanoparticle application remarkably improved lymph nodes detection rate in patients (4.7 ± 3.0; P< 0.05) compared to those in control groups (3.5 ± 2.3). The rate of parathyroid accidental resection in the carbon nanoparticle group significantly decreased (6.67%) compared to the control group (21.28%). Incidents of hypoparathyroidism and hypocalcaemia significantly decreased among the carbon nanoparticle cohorts. CONCLUSIONS Our study definitively showed that carbon nanoparticles can be used to effectively treat lymphatic carcinoma. Our study presented clinical evidences for potential application of carbon nanoparticle in improving the management of PTC patients.
Collapse
Affiliation(s)
- Yang Liu
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan, China.,Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan, China
| | - Lin Li
- Department of Dermatology, The Affiliated Children's Hospital of Zhengzhou University, Zhengzhou 450002, Henan, China.,Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan, China
| | - Jie Yu
- Department of Ultrasonagraphy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan, China
| | - Yu-Xia Fan
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan, China
| | - Xiu-Bo Lu
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, Henan, China
| |
Collapse
|
9
|
Mukai H, Hatanaka K, Yagi N, Warashina S, Zouda M, Takahashi M, Narushima K, Yabuuchi H, Iwano J, Kuboyama T, Enokizono J, Wada Y, Watanabe Y. Pharmacokinetic evaluation of liposomal nanoparticle-encapsulated nucleic acid drug: A combined study of dynamic PET imaging and LC/MS/MS analysis. J Control Release 2018; 294:185-194. [PMID: 30529725 DOI: 10.1016/j.jconrel.2018.12.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 12/04/2018] [Accepted: 12/06/2018] [Indexed: 12/13/2022]
Abstract
In vivo biodistribution analyses, especially in tumors, of nucleic acids delivered with nanoparticles are important to develop drug delivery technologies for medical use. We previously developed wrapsome® (WS), an ~100 nm liposomal nanoparticle that can encapsulate siRNA, and reported that WS accumulates in tumors in vivo and inhibits their growth by an enhanced permeability and retention effect. In the present study, we evaluated the pharmacokinetics of nucleic acid-containing nanoparticles by combining dynamic positron emission tomography (PET) imaging and liquid chromatography-tandem mass spectrometry (LC/MS/MS) analysis. An 18-mer phosphorothioate oligodeoxynucleotide (ODN), trabedersen, was used as a model drug and was encapsulated in WS. Dynamic PET imaging and time-activity curve analysis of WS-encapsulated 64Cu-labeled ODNs administered to mice with MIA PaCa-2 subcutaneous xenograft tumors showed tumor accumulation (~3% injected dose per gram (%ID/g)) and liver accumulation (~30 %ID/g) at 24 h. Under these conditions, LC/MS/MS analysis showed that the level of intact ODNs was 1.62 %ID/g in the tumor and 1.70 %ID/g in the liver. From these pharmacokinetic data, the intact/accumulated ODN ratios were calculated using the following equation: intact/accumulated ODN ratio (%) = %ID/g LC/MS/MS, tissue, mean/%ID/g PET, tissue, mean × 100. Interestingly, the ratios for the tumor and kidney were maintained at 20-50% over 48 h after administration of the WS-encapsulated form. In contrast, the ratio for the liver rapidly decreased at 24 h, showing the same pattern as that for naked ODN. These different patterns indicate that WS effectively protected the ODN in the tumor and kidney, but protected it less efficiently in the liver. A combined approach of dynamic PET imaging and LC/MS/MS analysis will assist the development of nanoparticle-encapsulated nucleic acid drugs, such as those using WSs, to determine their detailed pharmacokinetics.
Collapse
Affiliation(s)
- Hidefumi Mukai
- Molecular Network Control Imaging Unit, Molecular Network Control Research Project, Center Director's Strategic Program, RIKEN Center for Life Science Technologies, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| | - Kentaro Hatanaka
- Research Core Function Laboratories, Research Function Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Nobuhiro Yagi
- Research Core Function Laboratories, Research Function Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Shota Warashina
- Molecular Network Control Imaging Unit, Molecular Network Control Research Project, Center Director's Strategic Program, RIKEN Center for Life Science Technologies, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Maki Zouda
- Molecular Network Control Imaging Unit, Molecular Network Control Research Project, Center Director's Strategic Program, RIKEN Center for Life Science Technologies, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Maiko Takahashi
- Molecular Network Control Imaging Unit, Molecular Network Control Research Project, Center Director's Strategic Program, RIKEN Center for Life Science Technologies, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Kazuya Narushima
- Research Core Function Laboratories, Research Function Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Hayato Yabuuchi
- Research Core Function Laboratories, Research Function Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Junko Iwano
- Research Core Function Laboratories, Research Function Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Takeshi Kuboyama
- Research Core Function Laboratories, Research Function Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan.
| | - Junichi Enokizono
- Research Core Function Laboratories, Research Function Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Yasuhiro Wada
- Pathophysiological and Health Science Team, Imaging Platform and Innovation Group, Division of Bio-Function Dynamics Imaging, RIKEN Center for Life Science Technologies, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yasuyoshi Watanabe
- Pathophysiological and Health Science Team, Imaging Platform and Innovation Group, Division of Bio-Function Dynamics Imaging, RIKEN Center for Life Science Technologies, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| |
Collapse
|
10
|
Battista C, Howell BA, Siler SQ, Watkins PB. An Introduction to DILIsym® Software, a Mechanistic Mathematical Representation of Drug-Induced Liver Injury. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/978-1-4939-7677-5_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
11
|
Atzrodt J, Derdau V, Kerr WJ, Reid M. Deuterium- und tritiummarkierte Verbindungen: Anwendungen in den modernen Biowissenschaften. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201704146] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jens Atzrodt
- Isotope Chemistry and Metabolite Synthesis, Integrated Drug Discovery, Medicinal Chemistry; Industriepark Höchst, G876 65926 Frankfurt Deutschland
| | - Volker Derdau
- Isotope Chemistry and Metabolite Synthesis, Integrated Drug Discovery, Medicinal Chemistry; Industriepark Höchst, G876 65926 Frankfurt Deutschland
| | - William J. Kerr
- Department of Pure and Applied Chemistry, WestCHEM; University of Strathclyde; 295 Cathedral Street Glasgow Scotland G1 1XL Großbritannien
| | - Marc Reid
- Department of Pure and Applied Chemistry, WestCHEM; University of Strathclyde; 295 Cathedral Street Glasgow Scotland G1 1XL Großbritannien
| |
Collapse
|
12
|
Atzrodt J, Derdau V, Kerr WJ, Reid M. Deuterium- and Tritium-Labelled Compounds: Applications in the Life Sciences. Angew Chem Int Ed Engl 2018; 57:1758-1784. [PMID: 28815899 DOI: 10.1002/anie.201704146] [Citation(s) in RCA: 421] [Impact Index Per Article: 70.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/27/2017] [Indexed: 12/19/2022]
Abstract
Hydrogen isotopes are unique tools for identifying and understanding biological and chemical processes. Hydrogen isotope labelling allows for the traceless and direct incorporation of an additional mass or radioactive tag into an organic molecule with almost no changes in its chemical structure, physical properties, or biological activity. Using deuterium-labelled isotopologues to study the unique mass-spectrometric patterns generated from mixtures of biologically relevant molecules drastically simplifies analysis. Such methods are now providing unprecedented levels of insight in a wide and continuously growing range of applications in the life sciences and beyond. Tritium (3 H), in particular, has seen an increase in utilization, especially in pharmaceutical drug discovery. The efforts and costs associated with the synthesis of labelled compounds are more than compensated for by the enhanced molecular sensitivity during analysis and the high reliability of the data obtained. In this Review, advances in the application of hydrogen isotopes in the life sciences are described.
Collapse
Affiliation(s)
- Jens Atzrodt
- Isotope Chemistry and Metabolite Synthesis, Integrated Drug Discovery, Medicinal Chemistry, Industriepark Höchst, G876, 65926, Frankfurt, Germany
| | - Volker Derdau
- Isotope Chemistry and Metabolite Synthesis, Integrated Drug Discovery, Medicinal Chemistry, Industriepark Höchst, G876, 65926, Frankfurt, Germany
| | - William J Kerr
- Department of Pure and Applied Chemistry, WestCHEM, University of Strathclyde, 295 Cathedral Street, Glasgow, Scotland, G1 1XL, UK
| | - Marc Reid
- Department of Pure and Applied Chemistry, WestCHEM, University of Strathclyde, 295 Cathedral Street, Glasgow, Scotland, G1 1XL, UK
| |
Collapse
|
13
|
Quantitative Method for Simultaneous Analysis of a 5-Probe Cocktail for Cytochrome P450 Enzymes. Ther Drug Monit 2017; 38:761-768. [PMID: 27764027 DOI: 10.1097/ftd.0000000000000338] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The metabolic activity of P450 enzymes in vivo can be determined using selective probe drugs. The simultaneous administration of multiple CYP-specific probe drugs is commonly known as the "cocktail approach." Disadvantages of a cocktail are large volumes of samples required for analysis and time-consuming analyses. The aim of this study was to develop and validate a simplified but sensitive method for the simultaneous quantification of 5 probe drugs [caffeine (CYP1A2), metoprolol (CYP2D6), midazolam (CYP3A4), omeprazole (CYP2C19), and S-warfarin (CYP2C9)] in a previously validated cocktail using a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method. METHODS The method entailed a single method for sample preparation that enables quick processing of the samples containing all 5 probe drugs in a small volume of blood (≥10 μL) followed by a chiral and nonchiral LC-MS/MS method. The method was validated for selectivity, specificity, resolution of racemic warfarin, linearity, accuracy, imprecision, recovery, process efficiency, ionization efficiency, and carryover effect. RESULTS The method showed good selectivity without matrix interferences and differentiated S- and R-warfarin enantiomers with adequate resolution (Rs = 1.55). For all analytes, the mean process efficiency was >95%, and the mean ionization efficiency was >97%. Furthermore, the accuracy was between 94.9% and 108% for all analytes, and the within- and between-run imprecision were <11.7% for the lower limit of quantification and <12.6% for the middle level and upper limit of quantification. CONCLUSIONS The method presented here enables the simultaneous quantification of the 5 probes in a very small blood volume (≥10 μL). Furthermore, it is less time consuming than previously reported methods because it requires only 1 simple method for sample preparation followed by a nonchiral and chiral LC-MS/MS method that can be performed sequentially.
Collapse
|
14
|
Berrospe-Rodriguez C, Visser CW, Schlautmann S, Rivas DF, Ramos-Garcia R. Toward jet injection by continuous-wave laser cavitation. JOURNAL OF BIOMEDICAL OPTICS 2017; 22:1-9. [PMID: 29030942 DOI: 10.1117/1.jbo.22.10.105003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/15/2017] [Indexed: 05/06/2023]
Abstract
This is a study motivated by the need to develop a needle-free device for eliminating major global healthcare problems caused by needles. The generation of liquid jets by means of a continuous-wave laser, focused into a light absorbing solution, was studied with the aim of developing a portable and affordable jet injector. We designed and fabricated glass microfluidic devices, which consist of a chamber where thermocavitation is created and a tapered channel. The growth of a vapor bubble displaces and expels the liquid through the channel as a fast traveling jet. Different parameters were varied with the purpose of increasing the jet velocity. The velocity increases with smaller channel diameters and taper ratios, whereas larger chambers significantly reduce the jet speed. It was found that the initial position of the liquid-air meniscus interface and its dynamics contribute to increased jet velocities. A maximum velocity of 94±3 m/s for a channel diameter of D=120 μm, taper ratio n=0.25, and chamber length E=200 μm was achieved. Finally, agarose gel-based skin phantoms were used to demonstrate the potential of our devices to penetrate the skin. The maximum penetration depth achieved was ∼1 mm, which is sufficient to penetrate the stratum corneum and for most medical applications. A meta-analysis shows that larger injection volumes will be required as a next step to medical relevance for laser-induced jet injection techniques in general.
Collapse
Affiliation(s)
- Carla Berrospe-Rodriguez
- Instituto Nacional de Astrofísica, Óptica y Electrónica, Departamento de Óptica, Puebla, Pue., México
| | - Claas Willem Visser
- Harvard University, Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts, United States
- University of Twente, Physics of Fluids Group, MESA+ Institute and Faculty of Science and Technology, The Netherlands
| | - Stefan Schlautmann
- University of Twente, Mesoscale Chemical Systems Group, MESA+ Institute and Faculty of Science and T, The Netherlands
| | - David Fernandez Rivas
- University of Twente, Mesoscale Chemical Systems Group, MESA+ Institute and Faculty of Science and T, The Netherlands
| | - Ruben Ramos-Garcia
- Instituto Nacional de Astrofísica, Óptica y Electrónica, Departamento de Óptica, Puebla, Pue., México
| |
Collapse
|
15
|
Kusuhara H, Takashima T, Fujii H, Takashima T, Tanaka M, Ishii A, Tazawa S, Takahashi K, Takahashi K, Tokai H, Yano T, Kataoka M, Inano A, Yoshida S, Hosoya T, Sugiyama Y, Yamashita S, Hojo T, Watanabe Y. Comparison of pharmacokinetics of newly discovered aromatase inhibitors by a cassette microdosing approach in healthy Japanese subjects. Drug Metab Pharmacokinet 2017; 32:293-300. [PMID: 29137842 DOI: 10.1016/j.dmpk.2017.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/25/2017] [Accepted: 09/13/2017] [Indexed: 01/10/2023]
Abstract
The aim of the present study is to investigate the pharmacokinetics of our newly developed aromatase inhibitors (cetrozole and TMD-322) in healthy subjects by a cassette microdose strategy. A cocktail of cetrozole and TMD-322 was administered intravenously or orally (1.98 μg for each drug) to six healthy volunteers in a crossover fashion. Anastrozole (1.98 μg) was also included in the oral cocktail. Total body clearance and bioavailability were 12.1 ± 7.1 mL/min/kg and 34.9 ± 32.3% for cetrozole, and 16.8 ± 3.5 mL/min/kg and 18.4 ± 12.2% for TMD-322, respectively. The area under the plasma concentration-time curves of cetrozole and TMD-322 after oral administration was markedly lower than that of anastrozole because of their high hepatic clearance. Two subjects out of six exhibited 4- and 17-fold larger exposure of cetrozole than the others following intravenous and oral administration, respectively. Such variation was not observed for TMD-322 and anastrozole. Extensive metabolism of cetrozole and TMD-322 was observed in the CYP2C19 expression system among the test CYP isoforms (CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4). We report the first clinical investigation of our aromatase inhibitors by a cassette microdose strategy in healthy Japanese subjects. This strategy offers an optional approach for candidate selection as a phase zero study in drug development.
Collapse
Affiliation(s)
- Hiroyuki Kusuhara
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tadayuki Takashima
- RIKEN Center for Molecular Imaging Science, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Hisako Fujii
- Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan; Osaka City University Hospital, Center for Drug & Food Clinical Evaluation, 1-2-7 Asahimachi, Abeno-ku, Osaka 545-0051, Japan
| | - Tsutomu Takashima
- Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan; Osaka City University Hospital, Center for Drug & Food Clinical Evaluation, 1-2-7 Asahimachi, Abeno-ku, Osaka 545-0051, Japan
| | - Masaaki Tanaka
- Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Akira Ishii
- Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Shusaku Tazawa
- Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan; RIKEN Center for Life Science Technologies, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Kazuhiro Takahashi
- Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan; RIKEN Center for Life Science Technologies, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Kayo Takahashi
- Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan; RIKEN Center for Life Science Technologies, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hidekichi Tokai
- Osaka City University Hospital, Center for Drug & Food Clinical Evaluation, 1-2-7 Asahimachi, Abeno-ku, Osaka 545-0051, Japan
| | - Tsuneo Yano
- RIKEN Center for Molecular Imaging Science, 6-7-3 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Makoto Kataoka
- Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan
| | - Akihiro Inano
- Clinical Research Center, Fukushima Medical University Hospital, 1 Hikarigaoka, Fukushima City, Fukushima 960-1295, Japan
| | - Suguru Yoshida
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Takamitsu Hosoya
- RIKEN Center for Life Science Technologies, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Research Cluster for Innovation, Yokohama Bio Industry Center, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Shinji Yamashita
- Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan
| | - Taisuke Hojo
- Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan; Osaka City University Hospital, Center for Drug & Food Clinical Evaluation, 1-2-7 Asahimachi, Abeno-ku, Osaka 545-0051, Japan
| | - Yasuyoshi Watanabe
- Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan; RIKEN Center for Life Science Technologies, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| |
Collapse
|
16
|
Burt T, MacLeod D, Lee K, Santoro A, DeMasi DK, Hawk T, Feinglos M, Rowland M, Noveck RJ. Intra-Target Microdosing - A Novel Drug Development Approach: Proof of Concept, Safety, and Feasibility Study in Humans. Clin Transl Sci 2017; 10:351-359. [PMID: 28689370 PMCID: PMC5593161 DOI: 10.1111/cts.12477] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/25/2017] [Indexed: 12/21/2022] Open
Abstract
Intra‐Target Microdosing (ITM) is a novel drug development approach aimed at increasing the efficiency of first‐in‐human (FIH) testing of new molecular entities (NMEs). ITM combines intra‐target drug delivery and “microdosing,” the subpharmacological systemic exposure. We hypothesized that when the target tissue is small (about 1/100th of total body mass), ITM can lead to target therapeutic‐level exposure with minimal (microdose) systemic exposure. Each of five healthy male volunteers received insulin microdose into the radial artery or full therapeutic dose intravenously in separate visits. Insulin and glucose levels were similar between systemic administration and ITM administration in the ipsilateral hand, and glucose levels demonstrated a reduction in the ipsilateral hand but not in the contralateral hand. Positron emission tomography (PET) imaging of 18F‐fluorodeoxyglucose (FDG) uptake demonstrated differences between the ipsilateral and contralateral arms. The procedures were safe and well‐tolerated. Results are consistent with ITM proof‐of‐concept (POC) and demonstrate the ethical, regulatory, and logistical feasibility of the approach.
Collapse
Affiliation(s)
- T Burt
- Burt Consultancy, LLC, Durham, North Carolina, USA
| | - D MacLeod
- Department of Anesthesiology, Duke University, Durham, North Carolina, USA
| | - K Lee
- MI, CCC-TDI, OPS, SV, Siemens Medical Solutions USA, Inc., Knoxville, Tennessee, USA
| | - A Santoro
- Department of Anesthesiology, Duke University, Durham, North Carolina, USA
| | - D K DeMasi
- Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - T Hawk
- Department of Radiology, Duke University, Durham, North Carolina, USA
| | - M Feinglos
- Department of Endocrinology, Duke University, Durham, North Carolina, USA
| | - M Rowland
- Manchester Pharmacy School, University of Manchester, Manchester, UK
| | - R J Noveck
- Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| |
Collapse
|
17
|
Professor Yuichi Sugiyama: A Brilliant, Creative, Amicable, Charming, and Humorous Pharmaceutical Scientist. J Pharm Sci 2017; 106:2188-2194. [PMID: 28479350 DOI: 10.1016/j.xphs.2017.04.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 04/27/2017] [Indexed: 11/23/2022]
|
18
|
Oku N. Innovations in Liposomal DDS Technology and Its Application for the Treatment of Various Diseases. Biol Pharm Bull 2017; 40:119-127. [PMID: 28154249 DOI: 10.1248/bpb.b16-00857] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Liposomes have been widely used as drug carriers in the field of drug delivery systems (DDS), and they are thought to be ideal nano-capsules for targeting DDS after being injected into the bloodstream. In general, DDS drugs meet the needs of aged and super-aged societies, since the administration route of drugs can be changed, the medication frequency reduced, the adverse effects of drugs suppressed, and so on. In fact, a number of liposomal drugs have been launched and used worldwide including liposomal anticancer drugs, and these drugs have appeared on the market owing to various innovations in liposomal DDS technologies. The accumulation of long-circulating liposomes in cancer tissue is driven by the enhanced permeability and retention (EPR) effect. In this review, liposome-based targeting DDS for cancer therapy is briefly discussed. Since cancer angiogenic vessels are the ideal target of drug carriers after their injection and are critical for cancer growth, damaging of these neovessels has been an approach for eradicating cancer cells. Also, the usage of liposomal DDS for the treatment of ischemic stroke is possible, since we observed that PEGylated liposomes accumulate in the site of cerebral ischemia in transient middle cerebral artery occlusion (t-MCAO) model rats. Interestingly, liposomes carrying neuroprotectants partly suppress ischemia/reperfusion injury of these model rats, suggesting that the EPR effect also works in ischemic diseases by causing an increase in the permeability of the blood vessel endothelium. The potential of liposomal DDS against life-threatening diseases might thus be attractive for supporting long-lived societies.
Collapse
Affiliation(s)
- Naoto Oku
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
19
|
Enright HA, Malfatti MA, Zimmermann M, Ognibene T, Henderson P, Turteltaub KW. Use of Accelerator Mass Spectrometry in Human Health and Molecular Toxicology. Chem Res Toxicol 2016; 29:1976-1986. [PMID: 27726383 PMCID: PMC5203773 DOI: 10.1021/acs.chemrestox.6b00234] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Accelerator mass spectrometry (AMS) has been adopted as a powerful bioanalytical method for human studies in the areas of pharmacology and toxicology. The exquisite sensitivity (10-18 mol) of AMS has facilitated studies of toxins and drugs at environmentally and physiologically relevant concentrations in humans. Such studies include risk assessment of environmental toxicants, drug candidate selection, absolute bioavailability determination, and more recently, assessment of drug-target binding as a biomarker of response to chemotherapy. Combining AMS with complementary capabilities such as high performance liquid chromatography (HPLC) can maximize data within a single experiment and provide additional insight when assessing drugs and toxins, such as metabolic profiling. Recent advances in the AMS technology at Lawrence Livermore National Laboratory have allowed for direct coupling of AMS with complementary capabilities such as HPLC via a liquid sample moving wire interface, offering greater sensitivity compared to that of graphite-based analysis, therefore enabling the use of lower 14C and chemical doses, which are imperative for clinical testing. The aim of this review is to highlight the recent efforts in human studies using AMS, including technological advancements and discussion of the continued promise of AMS for innovative clinical based research.
Collapse
Affiliation(s)
- Heather A. Enright
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA USA
| | - Michael A. Malfatti
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA USA
| | - Maike Zimmermann
- Department of Internal Medicine, Division of Hematology and Oncology, UC Davis Medical Center, Sacramento, CA USA
- Accelerated Medical Diagnostics Incorporated, Berkeley, CA USA
| | - Ted Ognibene
- Center for Accelerator Mass Spectrometry, Lawrence Livermore National Laboratory, Livermore, CA USA
| | - Paul Henderson
- Department of Internal Medicine, Division of Hematology and Oncology, UC Davis Medical Center, Sacramento, CA USA
- Accelerated Medical Diagnostics Incorporated, Berkeley, CA USA
| | - Kenneth W. Turteltaub
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA USA
| |
Collapse
|
20
|
Zimmermann M, Wang SS, Zhang H, Lin TY, Malfatti M, Haack K, Ognibene T, Yang H, Airhart S, Turteltaub KW, Cimino GD, Tepper CG, Drakaki A, Chamie K, de Vere White R, Pan CX, Henderson PT. Microdose-Induced Drug-DNA Adducts as Biomarkers of Chemotherapy Resistance in Humans and Mice. Mol Cancer Ther 2016; 16:376-387. [PMID: 27903751 DOI: 10.1158/1535-7163.mct-16-0381] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 10/05/2016] [Accepted: 11/02/2016] [Indexed: 12/15/2022]
Abstract
We report progress on predicting tumor response to platinum-based chemotherapy with a novel mass spectrometry approach. Fourteen bladder cancer patients were administered one diagnostic microdose each of [14C]carboplatin (1% of the therapeutic dose). Carboplatin-DNA adducts were quantified by accelerator mass spectrometry in blood and tumor samples collected within 24 hours, and compared with subsequent chemotherapy response. Patients with the highest adduct levels were responders, but not all responders had high adduct levels. Four patient-derived bladder cancer xenograft mouse models were used to test the possibility that another drug in the regimen could cause a response. The mice were dosed with [14C]carboplatin or [14C]gemcitabine and the resulting drug-DNA adduct levels were compared with tumor response to chemotherapy. At least one of the drugs had to induce high drug-DNA adduct levels or create a synergistic increase in overall adducts to prompt a corresponding therapeutic response, demonstrating proof-of-principle for drug-DNA adducts as predictive biomarkers. Mol Cancer Ther; 16(2); 376-87. ©2016 AACR.
Collapse
Affiliation(s)
- Maike Zimmermann
- Department of Internal Medicine, Division of Hematology and Oncology and UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, California.,Accelerated Medical Diagnostics Incorporated, Berkeley, California
| | - Si-Si Wang
- Department of Internal Medicine, Division of Hematology and Oncology and UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, California
| | - Hongyong Zhang
- Department of Internal Medicine, Division of Hematology and Oncology and UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, California
| | - Tzu-Yin Lin
- Department of Internal Medicine, Division of Hematology and Oncology and UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, California
| | | | - Kurt Haack
- Lawrence Livermore National Laboratory, Livermore, California
| | - Ted Ognibene
- Lawrence Livermore National Laboratory, Livermore, California
| | | | | | | | - George D Cimino
- Accelerated Medical Diagnostics Incorporated, Berkeley, California
| | - Clifford G Tepper
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, California
| | - Alexandra Drakaki
- Division of Hematology and Oncology, UCLA Medical Center, Los Angeles, California
| | - Karim Chamie
- Department of Urology, UCLA Medical Center, Los Angeles, California
| | - Ralph de Vere White
- Department of Urology, University of California Davis, Sacramento, California
| | - Chong-Xian Pan
- Department of Internal Medicine, Division of Hematology and Oncology and UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, California. .,Department of Urology, University of California Davis, Sacramento, California.,VA Northern California Health Care System, Mather, California
| | - Paul T Henderson
- Department of Internal Medicine, Division of Hematology and Oncology and UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, California. .,Accelerated Medical Diagnostics Incorporated, Berkeley, California
| |
Collapse
|
21
|
Burt T, John CS, Ruckle JL, Vuong LT. Phase-0/microdosing studies using PET, AMS, and LC-MS/MS: a range of study methodologies and conduct considerations. Accelerating development of novel pharmaceuticals through safe testing in humans – a practical guide. Expert Opin Drug Deliv 2016; 14:657-672. [DOI: 10.1080/17425247.2016.1227786] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
22
|
Abstract
INTRODUCTION About 10,000 compounds will be tested for an individual drug to eventually reach the market. It might be helpful recapitulating previous failures and identifying the main factors of the disappointments. AREAS COVERED In this review, the author(s) detailed the 7 cardiovascular compounds discontinued after reaching animal studies or Phase I-III clinical trials during 2015. Meanwhile, the reasons for these discontinuations were reported. Among these drugs, most discontinuations (6 drugs) were attributed to lack of efficacy. In general, failures due to lack of efficacy and safety demonstrate the need for the development of more predictive animal models. However, recent related studies showed that the absence of toxicity in animals provided little or virtually no evidential weight that adverse drug reactions would also be absent in humans. In this case, microdosing and collaborating more closely with biotech companies may be the better choices to improve the success ratio. EXPERT OPINION Future researches may benefit from the seven developments and investigators conducting similar studies may learn from these failures.
Collapse
Affiliation(s)
- Hong-Ping Zhao
- a Center for Instrumental Analysis, Key Laboratory of Drug Quality Control & Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , China
| | - Yan Dai
- a Center for Instrumental Analysis, Key Laboratory of Drug Quality Control & Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , China
| | - Bing-Ren Xiang
- a Center for Instrumental Analysis, Key Laboratory of Drug Quality Control & Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , China
| |
Collapse
|
23
|
The impact of early human data on clinical development: there is time to win. Drug Discov Today 2016; 21:873-9. [PMID: 27046542 DOI: 10.1016/j.drudis.2016.03.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/18/2016] [Accepted: 03/23/2016] [Indexed: 12/16/2022]
Abstract
Modern accelerator mass spectrometry (AMS) methods enable the routine application of this technology in drug development. By the administration of a (14)C-labelled microdose or microtrace, pharmacokinetic (PK) data, such as mass balance, metabolite profiling, and absolute bioavailability (AB) data, can be generated easier, faster, and at lower costs. Here, we emphasize the advances and impact of this technology for pharmaceutical companies. The availability of accurate intravenous (iv) PK and human absorption, distribution, metabolism, and excretion (ADME) information, even before or during Phase I trials, can improve the clinical development plan. Moreover, applying the microtrace approach during early clinical development might impact the number of clinical pharmacology and preclinical safety pharmacology studies required, and shorten the overall drug discovery program.
Collapse
|
24
|
Ruiz-de-Angulo A, Zabaleta A, Gómez-Vallejo V, Llop J, Mareque-Rivas JC. Microdosed Lipid-Coated (67)Ga-Magnetite Enhances Antigen-Specific Immunity by Image Tracked Delivery of Antigen and CpG to Lymph Nodes. ACS NANO 2016; 10:1602-1618. [PMID: 26678549 DOI: 10.1021/acsnano.5b07253] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Development of vaccines to prevent and treat emerging new pathogens and re-emerging infections and cancer remains a major challenge. An attractive approach is to build the vaccine upon a biocompatible NP that simultaneously acts as accurate delivery vehicle and radiotracer for PET/SPECT imaging for ultrasensitive and quantitative in vivo imaging of NP delivery to target tissues/organs. Success in developing these nanovaccines will depend in part on having a "correct" NP size and accommodating and suitably displaying antigen and/or adjuvants (e.g., TLR agonists). Here we develop and evaluate a NP vaccine based on iron oxide-selective radio-gallium labeling suitable for SPECT((67)Ga)/PET((68)Ga) imaging and efficient delivery of antigen (OVA) and TLR 9 agonists (CpGs) using lipid-coated magnetite micelles. OVA, CpGs and rhodamine are easily accommodated in the hybrid micelles, and the average size of the construct can be controlled to be ca. 40 nm in diameter to target direct lymphatic delivery of the vaccine cargo to antigen presenting cells (APCs) in the lymph nodes (LNs). While the OVA/CpG-loaded construct showed effective delivery to endosomal TLR 9 in APCs, SPECT imaging demonstrated migration from the injection site to regional and nonregional LNs. In correlation with the imaging results, a range of in vitro and in vivo studies demonstrate that by using this microdosed nanosystem the cellular and humoral immune responses are greatly enhanced and provide protection against tumor challenge. These results suggest that these nanosystems have considerable potential for image-guided development of targeted vaccines that are more effective and limit toxicity.
Collapse
Affiliation(s)
- Ane Ruiz-de-Angulo
- Theranostic Nanomedicine Laboratory, Cooperative Centre for Research in Biomaterials (CIC biomaGUNE) , Paseo Miramón 182, 20009-San Sebastián, Spain
| | - Aintzane Zabaleta
- Theranostic Nanomedicine Laboratory, Cooperative Centre for Research in Biomaterials (CIC biomaGUNE) , Paseo Miramón 182, 20009-San Sebastián, Spain
| | - Vanessa Gómez-Vallejo
- Radiochemistry Platform, Cooperative Centre for Research in Biomaterials (CIC biomaGUNE) , Paseo Miramón 182, 20009-San Sebastián, Spain
| | - Jordi Llop
- Radiochemistry and Nuclear Imaging Laboratory, Cooperative Centre for Research in Biomaterials (CIC biomaGUNE) , Paseo Miramón 182, 20009-San Sebastián, Spain
| | - Juan C Mareque-Rivas
- Theranostic Nanomedicine Laboratory, Cooperative Centre for Research in Biomaterials (CIC biomaGUNE) , Paseo Miramón 182, 20009-San Sebastián, Spain
- IKERBASQUE, Basque Foundation for Science , 48011-Bilbao, Spain
- School of Engineering, The University of Aberdeen , Aberdeen AB24 3UE, U.K
| |
Collapse
|
25
|
Klinghoffer RA, Bahrami SB, Hatton BA, Frazier JP, Moreno-Gonzalez A, Strand AD, Kerwin WS, Casalini JR, Thirstrup DJ, You S, Morris SM, Watts KL, Veiseh M, Grenley MO, Tretyak I, Dey J, Carleton M, Beirne E, Pedro KD, Ditzler SH, Girard EJ, Deckwerth TL, Bertout JA, Meleo KA, Filvaroff EH, Chopra R, Press OW, Olson JM. A technology platform to assess multiple cancer agents simultaneously within a patient's tumor. Sci Transl Med 2016; 7:284ra58. [PMID: 25904742 DOI: 10.1126/scitranslmed.aaa7489] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A fundamental problem in cancer drug development is that antitumor efficacy in preclinical cancer models does not translate faithfully to patient outcomes. Much of early cancer drug discovery is performed under in vitro conditions in cell-based models that poorly represent actual malignancies. To address this inconsistency, we have developed a technology platform called CIVO, which enables simultaneous assessment of up to eight drugs or drug combinations within a single solid tumor in vivo. The platform is currently designed for use in animal models of cancer and patients with superficial tumors but can be modified for investigation of deeper-seated malignancies. In xenograft lymphoma models, CIVO microinjection of well-characterized anticancer agents (vincristine, doxorubicin, mafosfamide, and prednisolone) induced spatially defined cellular changes around sites of drug exposure, specific to the known mechanisms of action of each drug. The observed localized responses predicted responses to systemically delivered drugs in animals. In pair-matched lymphoma models, CIVO correctly demonstrated tumor resistance to doxorubicin and vincristine and an unexpected enhanced sensitivity to mafosfamide in multidrug-resistant lymphomas compared with chemotherapy-naïve lymphomas. A CIVO-enabled in vivo screen of 97 approved oncology agents revealed a novel mTOR (mammalian target of rapamycin) pathway inhibitor that exhibits significantly increased tumor-killing activity in the drug-resistant setting compared with chemotherapy-naïve tumors. Finally, feasibility studies to assess the use of CIVO in human and canine patients demonstrated that microinjection of drugs is toxicity-sparing while inducing robust, easily tracked, drug-specific responses in autochthonous tumors, setting the stage for further application of this technology in clinical trials.
Collapse
Affiliation(s)
| | - S Bahram Bahrami
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | | | - Andrew D Strand
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | | | - Sheng You
- Presage Biosciences, Seattle, WA 98109, USA
| | | | | | - Mandana Veiseh
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | - Joyoti Dey
- Presage Biosciences, Seattle, WA 98109, USA
| | | | | | - Kyle D Pedro
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | - Emily J Girard
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | - Karri A Meleo
- Oncology Department, BluePearl Veterinary Partners, Seattle, WA 98125, USA
| | | | | | - Oliver W Press
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - James M Olson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Department of Pediatrics, University of Washington, Seattle, WA 98195, USA.,Seattle Children's Hospital, Seattle, WA 98105, USA
| |
Collapse
|
26
|
Burt T, Rouse DC, Lee K, Wu H, Layton AT, Hawk TC, Weitzel DH, Chin BB, Cohen-Wolkowiez M, Chow SC, Noveck RJ. Intraarterial Microdosing: A Novel Drug Development Approach, Proof-of-Concept PET Study in Rats. J Nucl Med 2015; 56:1793-9. [PMID: 26315828 PMCID: PMC4767259 DOI: 10.2967/jnumed.115.160986] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 08/26/2015] [Indexed: 12/24/2022] Open
Abstract
UNLABELLED Intraarterial microdosing (IAM) is a novel drug development approach combining intraarterial drug delivery and microdosing. We aimed to demonstrate that IAM leads to target exposure similar to that of systemic full-dose administration but with minimal systemic exposure. IAM could enable the safe, inexpensive, and early study of novel drugs at the first-in-human stage and the study of established drugs in vulnerable populations. METHODS Insulin was administered intraarterially (ipsilateral femoral artery) or systemically to 8 CD IGS rats just before blood sampling or 60-min (18)F-FDG uptake PET imaging of ipsilateral and contralateral leg muscles (lateral gastrocnemius) and systemic muscles (spinotrapezius). The (18)F-FDG uptake slope analysis was used to compare the interventions. Plasma levels of insulin and glucose were compared using area under the curve calculated by the linear trapezoidal method. A physiologically based computational pharmacokinetics/pharmacodynamics model was constructed to simulate the relationship between the administered dose and response over time. RESULTS (18)F-FDG slope analysis found no difference between IAM and systemic full-dose slopes (0.0066 and 0.0061, respectively; 95% confidence interval [CI], -0.024 to 0.029; P = 0.7895), but IAM slope was statistically significantly greater than systemic microdose (0.0018; 95% CI, -0.045 to -0.007; P = 0.0147) and sham intervention (-0.0015; 95% CI, 0.023-0.058; P = 0.0052). The pharmacokinetics/pharmacodynamics data were used to identify model parameters that describe membrane insulin binding and glucose-insulin dynamics. CONCLUSION Target exposure after IAM was similar to systemic full dose administration but with minimal systemic effects. The computational pharmacokinetics/pharmacodynamics model can be generalized to predict whole-body response. Findings should be validated in larger, controlled studies in animals and humans using a range of targets and classes of drugs.
Collapse
Affiliation(s)
- Tal Burt
- Duke University, Durham, North Carolina; and
| | | | - Kihak Lee
- Siemens Medical Solutions USA, Knoxville, Tennessee
| | - Huali Wu
- Duke University, Durham, North Carolina; and
| | | | | | | | | | | | | | | |
Collapse
|
27
|
A clinical pharmacokinetic microdosing study of docetaxel with Japanese patients with cancer. Cancer Chemother Pharmacol 2015; 76:793-801. [DOI: 10.1007/s00280-015-2844-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 08/02/2015] [Indexed: 01/31/2023]
|
28
|
Bosgra S, Vlaming MLH, Vaes WHJ. To Apply Microdosing or Not? Recommendations to Single Out Compounds with Non-Linear Pharmacokinetics. Clin Pharmacokinet 2015; 55:1-15. [DOI: 10.1007/s40262-015-0308-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
29
|
Yamashita S, Kataoka M, Suzaki Y, Imai H, Morimoto T, Ohashi K, Inano A, Togashi K, Mutaguchi K, Sugiyama Y. An Assessment of the Oral Bioavailability of Three Ca-Channel Blockers Using a Cassette-Microdose Study: A New Strategy for Streamlining Oral Drug Development. J Pharm Sci 2015; 104:3154-61. [PMID: 26037531 DOI: 10.1002/jps.24499] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 04/25/2015] [Accepted: 04/27/2015] [Indexed: 11/08/2022]
Abstract
A cassette-microdose (MD) clinical study was performed to demonstrate its usefulness for identifying the most promising compound for oral use. Three Ca-channel blockers (nifedipine, nicardipine, and diltiazem) were chosen as model drugs. In the MD clinical study, a cassette-dose method was employed in which three model drugs were administered simultaneously. Both intravenous (i.v.) and oral (p.o.) administration studies were conducted to calculate the oral bioavailability (BA). For comparison, p.o. studies with therapeutic dose (ThD) levels were also performed. In all studies, blood concentrations of each drug were successfully determined using liquid chromatography-mass spectrometry with the lower limit of quantification of 0.2-2.0 pg/mL. Oral BA of nifedipine in the MD study was approximately 50% and in the same range with that obtained in the ThD study, whereas other two drugs showed significantly lower BA in the MD study, indicating a dose-dependent absorption. In addition, compared with the ThD study, absorption of nicardipine was delayed in the MD study. As a result, nifedipine was considered to be most promising for oral use. In conclusion, a cassette-MD clinical study is of advantage for oral drug development that enables to identify the candidate having desired properties for oral use.
Collapse
Affiliation(s)
- Shinji Yamashita
- Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Osaka, 573-0101, Japan
| | - Makoto Kataoka
- Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Osaka, 573-0101, Japan
| | - Yuki Suzaki
- General Clinical Research Center, Oita University Hospital, Oita, Japan
| | - Hiromitsu Imai
- General Clinical Research Center, Oita University Hospital, Oita, Japan
| | - Takuya Morimoto
- General Clinical Research Center, Oita University Hospital, Oita, Japan
| | - Kyoichi Ohashi
- General Clinical Research Center, Oita University Hospital, Oita, Japan
| | - Akihiro Inano
- Clinical Research Center, Fukushima Medical University Hospital, Fukushima City, Fukushima, 960-1295, Japan
| | - Kazutaka Togashi
- Pharmaceutical Business Division, Sumika Chemical Analysis Service, Ltd, Osaka, 554-0022, Japan
| | - Kuninori Mutaguchi
- Pharmaceutical Business Division, Sumika Chemical Analysis Service, Ltd, Osaka, 554-0022, Japan
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Research Cluster for Innovation, Yokohama Bio Industry Center, Tsurumi-ku, Yokohama, 230-0045, Japan
| |
Collapse
|
30
|
Vlaming MLH, van Duijn E, Dillingh MR, Brands R, Windhorst AD, Hendrikse NH, Bosgra S, Burggraaf J, de Koning MC, Fidder A, Mocking JAJ, Sandman H, de Ligt RAF, Fabriek BO, Pasman WJ, Seinen W, Alves T, Carrondo M, Peixoto C, Peeters PAM, Vaes WHJ. Microdosing of a Carbon-14 Labeled Protein in Healthy Volunteers Accurately Predicts Its Pharmacokinetics at Therapeutic Dosages. Clin Pharmacol Ther 2015; 98:196-204. [PMID: 25869840 DOI: 10.1002/cpt.131] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 03/30/2015] [Accepted: 04/06/2015] [Indexed: 11/12/2022]
Abstract
Preclinical development of new biological entities (NBEs), such as human protein therapeutics, requires considerable expenditure of time and costs. Poor prediction of pharmacokinetics in humans further reduces net efficiency. In this study, we show for the first time that pharmacokinetic data of NBEs in humans can be successfully obtained early in the drug development process by the use of microdosing in a small group of healthy subjects combined with ultrasensitive accelerator mass spectrometry (AMS). After only minimal preclinical testing, we performed a first-in-human phase 0/phase 1 trial with a human recombinant therapeutic protein (RESCuing Alkaline Phosphatase, human recombinant placental alkaline phosphatase [hRESCAP]) to assess its safety and kinetics. Pharmacokinetic analysis showed dose linearity from microdose (53 μg) [(14) C]-hRESCAP to therapeutic doses (up to 5.3 mg) of the protein in healthy volunteers. This study demonstrates the value of a microdosing approach in a very small cohort for accelerating the clinical development of NBEs.
Collapse
Affiliation(s)
| | | | - M R Dillingh
- Centre for Human Drug Research, Leiden, The Netherlands, UK
| | - R Brands
- AMRIF BV, Wageningen, The Netherlands, UK
| | - A D Windhorst
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands, UK
| | - N H Hendrikse
- Department of Pharmacy and Clinical Pharmacology, VU University Medical Center, Amsterdam, The Netherlands, UK
| | | | - J Burggraaf
- Centre for Human Drug Research, Leiden, The Netherlands, UK
| | | | | | | | | | | | | | | | - W Seinen
- AMRIF BV, Wageningen, The Netherlands, UK.,Utrecht University, Utrecht, The Netherlands, UK
| | - T Alves
- GenIBET/IBET, Oeiras, Portugal
| | | | | | - P A M Peeters
- Centre for Human Drug Research, Leiden, The Netherlands, UK
| | | |
Collapse
|
31
|
Iguchi Y, Michiue H, Kitamatsu M, Hayashi Y, Takenaka F, Nishiki TI, Matsui H. Tumor-specific delivery of BSH-3R for boron neutron capture therapy and positron emission tomography imaging in a mouse brain tumor model. Biomaterials 2015; 56:10-7. [PMID: 25934274 DOI: 10.1016/j.biomaterials.2015.03.061] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 03/30/2015] [Accepted: 03/31/2015] [Indexed: 12/28/2022]
Abstract
Glioblastoma, a malignant brain tumor with poor disease outcomes, is managed in modern medicine by multimodality therapy. Boron neutron capture therapy (BNCT) is an encouraging treatment under clinical investigation. In malignant cells, BNCT consists of two major factors: neutron radiation and boron uptake. To increase boron uptake in cells, we created a mercapto-closo-undecahydrododecaborate ([B12HnSH](2-)2Na(+), BSH) fused with a short arginine peptide (1R, 2R, 3R) and checked cellular uptake in vitro and in vivo. In a mouse brain tumor model, only BSH with at least three arginine domains could penetrate cell membranes of glioma cells in vitro and in vivo. Furthermore, to monitor the pharmacokinetic properties of these agents in vivo, we fused BSH and BSH-3R with 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA); DOTA is a metal chelating agent for labeling positron emission tomography (PET) probe with (64)Cu. We administered BSH-DOTA-(64)Cu and BSH-3R-DOTA-(64)Cu to the tumor model through a mouse tail vein and determined the drugs' pharmacokinetics by PET imaging. BSH-3R showed a high uptake in the tumor area on PET imaging. We concluded that BSH-3R is the ideal boron compound for clinical use during BNCT and that in developing this compound for clinical use, the BSH-3R PET probe is essential for pharmacokinetic imaging.
Collapse
Affiliation(s)
- Yoshiya Iguchi
- Department of Physiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-Ku, Okayama City, Okayama 700-8558, Japan
| | - Hiroyuki Michiue
- Department of Physiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-Ku, Okayama City, Okayama 700-8558, Japan.
| | - Mizuki Kitamatsu
- Department of Applied Chemistry, Faculty of Science and Engineering, Kinki University, 3-4-1 Kowakae, Higashi-Osaka City, Osaka 577-8502, Japan
| | - Yuri Hayashi
- Department of Physiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-Ku, Okayama City, Okayama 700-8558, Japan
| | - Fumiaki Takenaka
- Collaborative Research Center for OMIC, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-Ku, Okayama City, Okayama 700-8558, Japan
| | - Tei-Ichi Nishiki
- Department of Physiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-Ku, Okayama City, Okayama 700-8558, Japan
| | - Hideki Matsui
- Department of Physiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-Ku, Okayama City, Okayama 700-8558, Japan
| |
Collapse
|
32
|
Abstract
Nonclinical safety pharmacology and toxicology testing of drug candidates assess the potential adverse effects caused by the drug in relation to its intended use in humans. Hazards related to a drug have to be identified and the potential risks at the intended exposure have to be evaluated in comparison to the potential benefit of the drug. Preclinical safety is thus an integral part of drug discovery and drug development. It still causes significant attrition during drug development.Therefore, there is a need for smart selection of drug candidates in drug discovery including screening of important safety endpoints. In the recent years,there was significant progress in computational and in vitro technology allowing in silico assessment as well as high-throughput screening of some endpoints at very early stages of discovery. Despite all this progress, in vivo evaluation of drug candidates is still an important part to safety testing. The chapter provides an overview on the most important areas of nonclinical safety screening during drug discovery of small molecules.
Collapse
|
33
|
Abstract
Organic anion-transporting polypeptides or OATPs are central transporters in the disposition of drugs and other xenobiotics. In addition, they mediate transport of a wide variety of endogenous substrates. The critical role of OATPs in drug disposition has spurred research both in academia and in the pharmaceutical industry. Translational aspects with clinical questions are the focus in academia, while the pharmaceutical industry tries to define and understand the role these transporters play in pharmacotherapy. The present overview summarizes our knowledge on the interaction of food constituents with OATPs and on the OATP transport mechanisms. Further, it gives an update on the available information on the structure-function relationship of the OATPs and, finally, covers the transcriptional and posttranscriptional regulation of OATPs.
Collapse
Affiliation(s)
- Bruno Stieger
- Department of Clinical Pharmacology and Toxicology, University Hospital, Zürich, Switzerland.
| | - Bruno Hagenbuch
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
34
|
Affiliation(s)
- Soonih Kim
- Pharmaceutical Technology Laboratories, Ono Pharmaceutical Co. Ltd
| |
Collapse
|
35
|
Kim DK, Lee JW, Shin KH, Kim S, Oh KH, Kim M, Yu KS, Lee JP, Lim CS, Kim YS, Joo KW. Dose selection method for pharmacokinetic study in hemodialysis patients using a subpharmacological dose: oseltamivir as a model drug. BMC Nephrol 2014; 15:46. [PMID: 24636040 PMCID: PMC3995549 DOI: 10.1186/1471-2369-15-46] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 03/11/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Dose selection is an important step in pharmacokinetic (PK) studies of hemodialysis patients. We propose a simulation-based dose-selection method for PK studies of hemodialysis patients using a subpharmacological dose of oseltamivir as a model drug. METHODS The concentrations of oseltamivir and its active metabolite, oseltamivir carboxylate (OC), were measured by liquid chromatography-tandem mass spectrometry. To determine a low oseltamivir dose exhibiting PK linearity, a pilot low dose determination investigation (n = 4) was performed using a single administration dose-escalation study. After the dose was determined, a low dose study (n = 10) was performed, and the optimal dose required to reach the hypothetical target OC exposure (area under the concentration-time curve [AUC] of 60,000 ng · hr/mL) was simulated using a nonparametric superposition method. Finally, observed PKs at the optimal dose were compared to the simulated PKs to verify PK predictability. RESULTS In the pilot low dose determination study, 2.5 mg of oseltamivir was determined to be the low dose. Subsequently, we performed a single-dose PK study with the low oseltamivir dose in an additional group of 10 hemodialysis patients. The predicted AUC last of OC following continuous oseltamivir doses was simulated, and 35 mg of oseltamivir corresponded to the hypothetical target AUC last of OC. The observed PK profiles of OC at a 35-mg oseltamivir dose and the simulated data based on the low dose study were in close alignment. CONCLUSION The results indicate that the proposed method provides a rational approach to determine the proper PK dose in hemodialysis patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Kwon Wook Joo
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
36
|
Mukai H, Ozaki D, Cui Y, Kuboyama T, Yamato-Nagata H, Onoe K, Takahashi M, Wada Y, Imanishi T, Kodama T, Obika S, Suzuki M, Doi H, Watanabe Y. Quantitative evaluation of the improvement in the pharmacokinetics of a nucleic acid drug delivery system by dynamic PET imaging with (18)F-incorporated oligodeoxynucleotides. J Control Release 2014; 180:92-9. [PMID: 24566256 DOI: 10.1016/j.jconrel.2014.02.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Revised: 12/27/2013] [Accepted: 02/16/2014] [Indexed: 11/29/2022]
Abstract
Recently, we demonstrated the utility of positron emission tomography (PET) imaging-based pharmacokinetic evaluation studies for preclinical experiments and microdose clinical trials, mainly focused on low molecular weight compounds. In order to investigate the pharmacokinetics of nucleic acid drugs and their drug delivery systems (DDSs) in vivo by using PET imaging, we developed a novel and efficient method for radiolabeling oligodeoxynucleotides with the positron-emitting radionuclide (18)F (stoichiometry-focused Huisgen-type (18)F labeling). By using this method, we succeeded in synthesizing a variety of (18)F-labeled oligodeoxynucleotides with not only phosphodiesters (PO) in natural forms, but also phosphorothioate (PS) and bridged nucleic acid (BNA) in artificial forms, and then performed PET studies and radioactive metabolite analyses of these (18)F-labeled oligodeoxynucleotides. The tissue-distribution and dynamic changes in radioactivity showed significantly different profiles between these antisense oligodeoxynucleotides. The radioactivity of (18)F-labeled PO-DNA and PO-BNA rapidly accumulated in the kidneys and liver and then moved to the renal medulla, ureter, bladder, and intestine. However, the radioactivity of (18)F-labeled PS-DNA and PS-BNA, possessing PS backbone structures, was retained in the blood for relatively long periods and then gradually accumulated in the liver and kidneys. The metabolite analysis showed that (18)F-labeled PO-DNA rapidly degraded by 5min and (18)F-labeled PO-BNA gradually degraded over time by 60min. Conversely, (18)F-labeled PS-DNA and PS-BNA were shown to be much more stable. To demonstrate the usefulness of the PET imaging technique for evaluating the improved targeting potential of the DDS, we designed and synthesized a cholesterol-modified oligodeoxynucleotide, that we developed as an antisense nucleic acid drug against proprotein convertase subtilisin/kexin type 9 (PCSK9) for hypercholesterolemia therapy, and evaluated its pharmacokinetics using PET imaging. As expected, the (18)F-labeled cholesterol-modified PS-BNA-type oligodeoxynucleotide showed much higher and more rapid accumulation in the delivery target organ, that is, the liver, which encourages us to develop this drug. These results suggest that dynamic PET studies using (18)F-incorporated oligodeoxynucleotide synthesized by stoichiometry-focused Huisgen-type labeling is useful for quantitative pharmacokinetic evaluation of nucleic acid drugs and their delivery systems.
Collapse
Affiliation(s)
- Hidefumi Mukai
- Division of Bio-function Dynamics Imaging, RIKEN Center for Life Science Technologies (CLST), 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Daiki Ozaki
- Division of Bio-function Dynamics Imaging, RIKEN Center for Life Science Technologies (CLST), 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yilong Cui
- Division of Bio-function Dynamics Imaging, RIKEN Center for Life Science Technologies (CLST), 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Takeshi Kuboyama
- Division of Bio-function Dynamics Imaging, RIKEN Center for Life Science Technologies (CLST), 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hiroko Yamato-Nagata
- Division of Bio-function Dynamics Imaging, RIKEN Center for Life Science Technologies (CLST), 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kayo Onoe
- Division of Bio-function Dynamics Imaging, RIKEN Center for Life Science Technologies (CLST), 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Maiko Takahashi
- Division of Bio-function Dynamics Imaging, RIKEN Center for Life Science Technologies (CLST), 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yasuhiro Wada
- Division of Bio-function Dynamics Imaging, RIKEN Center for Life Science Technologies (CLST), 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Takeshi Imanishi
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tetsuya Kodama
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Satoshi Obika
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Masaaki Suzuki
- Division of Bio-function Dynamics Imaging, RIKEN Center for Life Science Technologies (CLST), 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hisashi Doi
- Division of Bio-function Dynamics Imaging, RIKEN Center for Life Science Technologies (CLST), 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yasuyoshi Watanabe
- Division of Bio-function Dynamics Imaging, RIKEN Center for Life Science Technologies (CLST), 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| |
Collapse
|
37
|
Shoda LKM, Woodhead JL, Siler SQ, Watkins PB, Howell BA. Linking physiology to toxicity using DILIsym®, a mechanistic mathematical model of drug-induced liver injury. Biopharm Drug Dispos 2013; 35:33-49. [DOI: 10.1002/bdd.1878] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 10/10/2013] [Accepted: 11/01/2013] [Indexed: 12/22/2022]
Affiliation(s)
- Lisl K. M. Shoda
- The Hamner-UNC Institute for Drug Safety Sciences; The Hamner Institutes; Research Triangle Park NC 27709 USA
| | - Jeffrey L. Woodhead
- The Hamner-UNC Institute for Drug Safety Sciences; The Hamner Institutes; Research Triangle Park NC 27709 USA
| | - Scott Q. Siler
- The Hamner-UNC Institute for Drug Safety Sciences; The Hamner Institutes; Research Triangle Park NC 27709 USA
| | - Paul B. Watkins
- The Hamner-UNC Institute for Drug Safety Sciences; The Hamner Institutes; Research Triangle Park NC 27709 USA
| | - Brett A. Howell
- The Hamner-UNC Institute for Drug Safety Sciences; The Hamner Institutes; Research Triangle Park NC 27709 USA
| |
Collapse
|
38
|
Ahn G, Park DM, Park JW, Cho JY, Rhee SJ, Kim HY, Lee DS, Jang IJ, Kim HK. Development and validation of a microfluidic chip-based nano-liquid chromatography–triple quadrupole tandem mass spectrometry method for a sensitive and reliable quantification of 7-ethyl-10-hydroxycamptothecin (SN38) in mouse plasma. Anal Bioanal Chem 2013; 405:9817-24. [DOI: 10.1007/s00216-013-7411-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 08/18/2013] [Accepted: 10/02/2013] [Indexed: 12/20/2022]
|
39
|
Mukai H, Wada Y, Watanabe Y. The synthesis of 64Cu-chelated porphyrin photosensitizers and their tumor-targeting peptide conjugates for the evaluation of target cell uptake and PET image-based pharmacokinetics of targeted photodynamic therapy agents. Ann Nucl Med 2013; 27:625-39. [DOI: 10.1007/s12149-013-0728-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 04/05/2013] [Indexed: 10/26/2022]
|
40
|
Ieiri I, Tsunemitsu S, Maeda K, Ando Y, Izumi N, Kimura M, Yamane N, Okuzono T, Morishita M, Kotani N, Kanda E, Deguchi M, Matsuguma K, Matsuki S, Hirota T, Irie S, Kusuhara H, Sugiyama Y. Mechanisms of pharmacokinetic enhancement between ritonavir and saquinavir; micro/small dosing tests using midazolam (CYP3A4), fexofenadine (p-glycoprotein), and pravastatin (OATP1B1) as probe drugs. J Clin Pharmacol 2013; 53:654-61. [PMID: 23381882 DOI: 10.1002/jcph.62] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 12/05/2012] [Indexed: 11/08/2022]
Abstract
We investigated the mechanisms of ritonavir-mediated enhancement effect on the pharmacokinetics of saquinavir using in vivo probes for CYP3A4 (midazolam), p-glycoprotein (fexofenadine), and OATP1B1 (pravastatin) following oral micro/small dosing. A cocktail of the drugs (2 mg of saquinavir, 100 µg of each probe) was administered to eight healthy volunteers (phase 1), and then coadministered with 20 mg (phase 2) and 100 mg (phase 3) of ritonavir. Plasma concentrations of the drugs were measured by validated LC-MS/MS methods. The mean plasma AUC0-24 (pg hour/mL) of saquinavir at phases 1, 2, and 3 was 101, 2 540, and 23 900 (P < .01), respectively. The relative area under the plasma concentration-time curve (AUC)0-24 ratios of midazolam and fexofenadine at phases 1, 2, and 3 were 1:5.9:14.7 (P < .01), and 1:1.4:2.2 (P < .01-.05), respectively. In contrast, there was no difference in the pharmacokinetics of pravastatin. Inhibition of intestinal and hepatic CYP3A-mediated metabolism, and intestinal p-glycoprotein-mediated efflux of saquinavir, but not OATP1B1, is involved in the enhancement mechanism. Micro/small dosing is useful for examining the mechanism of drug interactions without safety concern.
Collapse
Affiliation(s)
- Ichiro Ieiri
- Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ostenfeld T, Beaumont C, Bullman J, Beaumont M, Jeffrey P. Human microdose evaluation of the novel EP1 receptor antagonist GSK269984A. Br J Clin Pharmacol 2012; 74:1033-44. [PMID: 22497298 PMCID: PMC3522817 DOI: 10.1111/j.1365-2125.2012.04296.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 04/10/2012] [Indexed: 11/27/2022] Open
Abstract
AIM The primary objective was to evaluate the pharmacokinetics (PK) of the novel EP(1) antagonist GSK269984A in human volunteers after a single oral and intravenous (i.v.) microdose (100 µg). METHOD GSK269984A was administered to two groups of healthy human volunteers as a single oral (n= 5) or i.v. (n= 5) microdose (100 µg). Blood samples were collected for up to 24 h and the parent drug concentrations were measured in separated plasma using a validated high pressure liquid chromatography-tandem mass spectrometry method following solid phase extraction. RESULTS Following the i.v. microdose, the geometric mean values for clearance (CL), steady-state volume of distribution (V(ss) ) and terminal elimination half-life (t(1/2) ) of GSK269984A were 9.8 l h(-1) , 62.8 l and 8.2 h. C(max) and AUC(0,∞) were 3.2 ng ml(-1) and 10.2 ng ml(-1) h, respectively; the corresponding oral parameters were 1.8 ng ml(-1) and 9.8 ng ml(-1) h, respectively. Absolute oral bioavailability was estimated to be 95%. These data were inconsistent with predictions of human PK based on allometric scaling of in vivo PK data from three pre-clinical species (rat, dog and monkey). CONCLUSION For drug development programmes characterized by inconsistencies between pre-clinical in vitro metabolic and in vivo PK data, and where uncertainty exists with respect to allometric predictions of the human PK profile, these data support the early application of a human microdose study to facilitate the selection of compounds for further clinical development.
Collapse
Affiliation(s)
- Thor Ostenfeld
- Neurology Discovery Medicine, GlaxoSmithKline R&D, Harlow, UK.
| | | | | | | | | |
Collapse
|
42
|
Cai HK, He HF, Tian W, Zhou MQ, Hu Y, Deng YC. Colorectal cancer lymph node staining by activated carbon nanoparticles suspension in vivo or methylene blue in vitro. World J Gastroenterol 2012; 18:6148-54. [PMID: 23155345 PMCID: PMC3496893 DOI: 10.3748/wjg.v18.i42.6148] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Revised: 08/02/2012] [Accepted: 08/14/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether activated carbon nanoparticles suspension (ACNS) or methylene blue (MB) can increase the detected number of lymph nodes in colorectal cancer.
METHODS: Sixty-seven of 72 colorectal cancer patients treated at our hospital fulfilled the inclusion criteria of the study which was conducted from December 2010 to February 2012. Seven patients refused to participate. Eventually, 60 patients were included, and randomly assigned to three groups (20 in each group): ACNS group (group A), MB group (group B) and non-stained conventional surgical group (group C). In group A, patients received subserosal injection of 1 mL ACNS in a 4-quadrant region around the mass. In group B, the main artery of specimen was identified and isolated after the specimen was removed, and 2 mL MB was slowly injected into the isolated, stretched and fixed vessel. In group C, no ACNS and MB were injected. All the mesentery lymph nodes were isolated and removed systematically by visually inspecting and palpating the adipose tissue.
RESULTS: No difference was observed among the three groups in age, gender, tumor location, tumor diameter, T-stage, degree of differentiation, postoperative complications and peritoneal drainage retention time. The total number of detected lymph nodes was 535, 476 and 223 in the three groups, respectively. The mean number of detected lymph nodes per patient was significantly higher in group A than in group C (26.8 ± 8.4 vs 12.2 ± 3.2, P < 0.001). Similarly, there were significantly more lymph nodes detected in group B than in group C (23.8 ± 6.9 vs 12.2 ± 3.2, P < 0.001). However, there was no significant difference between group A and group B. There were 50, 46 and 32 metastatic lymph nodes dissected in 13 patients of group A, 10 patients of group B and 11 patients of group C, without significant differences among the three groups. Eleven of the 60 patients had insufficient number of detected lymph nodes (< 12). Only one patient with T4a rectal cancer had 10 lymph nodes detected in group B, the other 10 patients were all from group C. Based on the different diameter categories, the number of detected lymph nodes in groups A and B was significantly higher than in group C. However, there was no statistically significant difference between group A and group B. The metastatic lymph nodes were not significant different among the three groups. Similarly, tumor location, T stage and tumor differentiation did not affect the staining results. Body mass index was a minor influencing factor in the two different staining methods. The stained lymph nodes can easily be identified from the mesenteric adipose tissues, and the staining time for lymph nodes was not significantly different compared with unstained group. None of the patients in groups A and B had drug-related complications.
CONCLUSION: Both activated carbon nanoparticles suspension in vivo and methylene blue in vitro can be used as tracers to increase the detected number of lymph nodes in colorectal cancer.
Collapse
|
43
|
Rowland M. Microdosing: A Critical Assessment of Human Data. J Pharm Sci 2012; 101:4067-74. [DOI: 10.1002/jps.23290] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 07/06/2012] [Accepted: 07/20/2012] [Indexed: 11/09/2022]
|
44
|
Sugiyama Y. Evolutional Drug Development with the Use of Microdosing and PET Imaging. Ann Oncol 2012. [DOI: 10.1016/s0923-7534(20)32075-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
45
|
Yamane N, Igarashi A, Kusama M, Maeda K, Ikeda T, Sugiyama Y. Cost-effectiveness analysis of microdose clinical trials in drug development. Drug Metab Pharmacokinet 2012; 28:187-95. [PMID: 22971640 DOI: 10.2133/dmpk.dmpk-12-rg-044] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Microdose (MD) clinical trials have been introduced to obtain human pharmacokinetic data early in drug development. Here we assessed the cost-effectiveness of microdose integrated drug development in a hypothetical model, as there was no such quantitative research that weighed the additional effectiveness against the additional time and/or cost. First, we calculated the cost and effectiveness (i.e., success rate) of 3 types of MD integrated drug development strategies: liquid chromatography-tandem mass spectrometry, accelerator mass spectrometry, and positron emission tomography. Then, we analyzed the cost-effectiveness of 9 hypothetical scenarios where 100 drug candidates entering into a non-clinical toxicity study were selected by different methods as the conventional scenario without MD. In the base-case, where 70 drug candidates were selected without MD and 30 selected evenly by one of the three MD methods, incremental cost-effectiveness ratio per one additional drug approved was JPY 12.7 billion (US$ 0.159 billion), whereas the average cost-effectiveness ratio of the conventional strategy was JPY 24.4 billion, which we set as a threshold. Integrating MD in the conventional drug development was cost-effective in this model. This quantitative analytical model which allows various modifications according to each company's conditions, would be helpful for guiding decisions early in clinical development.
Collapse
Affiliation(s)
- Naoe Yamane
- Pharmaceutical Regulatory Science, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
46
|
High-sensitivity liquid chromatography–tandem mass spectrometry for the simultaneous determination of five drugs and their cytochrome P450-specific probe metabolites in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2012; 895-896:56-64. [DOI: 10.1016/j.jchromb.2012.03.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 03/09/2012] [Accepted: 03/12/2012] [Indexed: 11/23/2022]
|
47
|
Imaging of Gastrointestinal Absorption and Biodistribution of an Orally Administered Probe Using Positron Emission Tomography in Humans. Clin Pharmacol Ther 2012; 91:653-9. [DOI: 10.1038/clpt.2011.267] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
48
|
Affiliation(s)
- Ruth Duncan
- Polymer Therapeutics Lab., Centro de Investigación Príncipe Felipe, Av. Autopista del Saler 16 E-46012, Valencia, Spain
| | - Rogerio Gaspar
- Nanomedicine & Drug Delivery Systems Group, iMed, Faculty of Pharmacy of the University of Lisbon, Av. Prof Gama Pinto, 1649-003 Lisbon, Portugal
| |
Collapse
|
49
|
Zhao HP, Zhang XS, Xiang BR. Discontinued drugs in 2010: cardiovascular drugs. Expert Opin Investig Drugs 2011; 20:1311-25. [PMID: 21870899 DOI: 10.1517/13543784.2011.611500] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
This perspective is a paper discussing drugs dropped from clinical development in the previous years. Specifically, this paper focuses on 16 cardiovascular drugs discontinued in 2010 after reaching Phase I - III clinical trials. Information for this perspective is mainly derived from a search of Pharmaprojects.
Collapse
Affiliation(s)
- Hong-ping Zhao
- Center for Instrumental Analysis, China Pharmaceutical University, Nanjing, Key Laboratory of Drug Quality Control & Pharmacovigilance, Ministry of Education, Jiangsu 210009, People's Republic of China.
| | | | | |
Collapse
|
50
|
Miyata T, Kikuchi K, Kiyomoto H, van Ypersele de Strihou C. New era for drug discovery and development in renal disease. Nat Rev Nephrol 2011; 7:469-77. [PMID: 21727928 DOI: 10.1038/nrneph.2011.84] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Drug discovery and development is a lengthy and expensive process. Testing new agents in humans at an early stage could reduce the time and costs involved in identifying drugs that are likely to succeed in clinical studies. New guidance has outlined the concept of exploratory clinical trials, which provide important information on a drug's distribution as well as its physiological and pharmacological effects in humans. This strategy reduces the need for preclinical testing by limiting the dose and duration of exposure to a new drug in humans to below those required by the traditional testing of investigational new drugs. Exploratory, first-in-man studies should provide insights into human physiology and pharmacology, identify therapeutic targets relevant to disease and increase our knowledge of a drug's characteristics. Implementation of a new drug also requires the development of useful biomarkers of disease and of the drug's efficacy, as well as sensitive molecular imaging techniques. In this Review, we outline the benefits of exploratory clinical trials, especially in academia, and provide an overview of the experimental tools necessary for rational drug discovery and development.
Collapse
Affiliation(s)
- Toshio Miyata
- United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai 980-8575, Japan
| | | | | | | |
Collapse
|