1
|
Cook BE, Nag S, Arakawa R, Lin EYS, Stratman N, Guckian K, Hering H, Lulla M, Choi J, Salinas C, Genung NE, Morén AF, Bolin M, Boscutti G, Plisson C, Martarello L, Halldin C, Kaliszczak MA. Development of a PET Tracer for OGA with Improved Kinetics in the Living Brain. J Nucl Med 2023; 64:1588-1593. [PMID: 37934021 PMCID: PMC10586483 DOI: 10.2967/jnumed.122.265225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/31/2023] [Indexed: 07/08/2023] Open
Abstract
O-GlcNAcylation is thought to play a role in the development of tau pathology in Alzheimer's disease because of its ability to modulate tau's aggregation propensity. O-GlcNAcylation is regulated by 2 enzymes: O-GlcNAc transferase and O-GlcNAcase (OGA). Development of a PET tracer would therefore be an essential tool for developing therapeutic small-molecule inhibitors of OGA, enabling clinical testing of target engagement and dose selection. Methods: A collection of small-molecule compounds was screened for inhibitory activity and high-affinity binding to OGA, as well as favorable PET tracer attributes (multidrug resistance protein 1 efflux, central nervous system PET multiparameter optimization, etc.). Two lead compounds with high affinity and selectivity for OGA were selected for further profiling, including OGA binding to tissue homogenate using a radioligand competition binding assay. In vivo pharmacokinetics were established using a microdosing approach with unlabeled compounds in rats. In vivo imaging studies were performed in rodents and nonhuman primates (NHPs) with 11C-labeled compounds. Results: Two selected candidates, BIO-735 and BIO-578, displayed promising attributes in vitro. After radiolabeling with tritium, [3H]BIO-735 and [3H]BIO-578 binding in rodent brain homogenates demonstrated dissociation constants of 0.6 and 2.3 nM, respectively. Binding was inhibited, concentration-dependently, by homologous compounds and thiamet G, a well-characterized and structurally diverse OGA inhibitor. Imaging studies in rats and NHPs showed both tracers had high uptake in the brain and inhibition of binding to OGA in the presence of a nonradioactive compound. However, only BIO-578 demonstrated reversible binding kinetics within the time frame of a PET study with a 11C-labeled molecule to enable quantification using kinetic modeling. Specificity of tracer uptake was confirmed with a 10 mg/kg blocking dose of thiamet G. Conclusion: We describe the development and testing of 2 11C PET tracers targeting the protein OGA. The lead compound BIO-578 demonstrated high affinity and selectivity for OGA in rodent and human postmortem brain tissue, leading to its further testing in NHPs. NHP PET imaging studies showed that the tracer had excellent brain kinetics, with full inhibition of specific binding by thiamet G. These results suggest that the tracer [11C]BIO-578 is well suited for further characterization in humans.
Collapse
Affiliation(s)
| | - Sangram Nag
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
- Stockholm County Council, Stockholm, Sweden; and
| | - Ryosuke Arakawa
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
- Stockholm County Council, Stockholm, Sweden; and
| | | | | | | | | | | | | | | | | | - Anton Forsberg Morén
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
- Stockholm County Council, Stockholm, Sweden; and
| | - Martin Bolin
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
- Stockholm County Council, Stockholm, Sweden; and
| | | | | | | | - Christer Halldin
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
- Stockholm County Council, Stockholm, Sweden; and
| | | |
Collapse
|
2
|
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: 55] [Impact Index Per Article: 11.0] [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
|
3
|
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: 1.1] [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
|
4
|
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: 2.2] [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
|
5
|
Use of microdosing and accelerator mass spectrometry to evaluate the pharmacokinetic linearity of a novel tricyclic GyrB/ParE inhibitor in rats. Antimicrob Agents Chemother 2014; 58:6477-83. [PMID: 25136019 DOI: 10.1128/aac.03300-14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Determining the pharmacokinetics (PKs) of drug candidates is essential for understanding their biological fate. The ability to obtain human PK information early in the drug development process can help determine if future development is warranted. Microdosing was developed to assess human PKs, at ultra-low doses, early in the drug development process. Microdosing has also been used in animals to confirm PK linearity across subpharmacological and pharmacological dose ranges. The current study assessed the PKs of a novel antimicrobial preclinical drug candidate (GP-4) in rats as a step toward human microdosing studies. Dose proportionality was determined at 3 proposed therapeutic doses (3, 10, and 30 mg/kg of body weight), and PK linearity between a microdose and a pharmacological dose was assessed in Sprague-Dawley rats. Plasma PKs over the 3 pharmacological doses were proportional. Over the 10-fold dose range, the maximum concentration in plasma and area under the curve (AUC) increased 9.5- and 15.8-fold, respectively. PKs from rats dosed with a (14)C-labeled microdose versus a (14)C-labeled pharmacological dose displayed dose linearity. In the animals receiving a microdose and the therapeutically dosed animals, the AUCs from time zero to infinity were 2.6 ng · h/ml and 1,336 ng · h/ml, respectively, and the terminal half-lives were 5.6 h and 1.4 h, respectively. When the AUC values were normalized to a dose of 1.0 mg/kg, the AUC values were 277.5 ng · h/ml for the microdose and 418.2 ng · h/ml for the pharmacological dose. This 1.5-fold difference in AUC following a 300-fold difference in dose is considered linear across the dose range. On the basis of the results, the PKs from the microdosed animals were considered to be predictive of the PKs from the therapeutically dosed animals.
Collapse
|
6
|
Takahashi R, Ma S, Yue Q, Kim-Kang H, Yi Y, Lyssikatos JP, Regal K, Hunt KW, Kallan NC, Siu M, Hop CECA, Liu X, Khojasteh SC. Dose-dependent exposure and metabolism of GNE-892, a β-secretase inhibitor, in monkeys: contributions by P450, AO, and P-gp. Eur J Drug Metab Pharmacokinet 2014; 40:171-85. [DOI: 10.1007/s13318-014-0198-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 03/26/2014] [Indexed: 02/04/2023]
|
7
|
Lappin G, Noveck R, Burt T. Microdosing and drug development: past, present and future. Expert Opin Drug Metab Toxicol 2013; 9:817-34. [PMID: 23550938 DOI: 10.1517/17425255.2013.786042] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Microdosing is an approach to early drug development where exploratory pharmacokinetic data are acquired in humans using inherently safe sub-pharmacologic doses of drug. The first publication of microdose data was 10 years ago and this review comprehensively explores the microdose concept from conception, over the past decade, up until the current date. AREAS COVERED The authors define and distinguish the concept of microdosing from similar approaches. The authors review the ability of microdosing to provide exploratory pharmacokinetics (concentration-time data) but exclude microdosing using positron emission tomography. The article provides a comprehensive review of data within the peer-reviewed literature as well as the latest applications and a look into the future, towards where microdosing may be headed. EXPERT OPINION Evidence so far suggests that microdosing may be a better predictive tool of human pharmacokinetics than alternative methods and combination with physiologically based modelling may lead to much more reliable predictions in the future. The concept has also been applied to drug-drug interactions, polymorphism and assessing drug concentrations over time at its site of action. Microdosing may yet have more to offer in unanticipated directions and provide benefits that have not been fully realised to date.
Collapse
Affiliation(s)
- Graham Lappin
- University of Lincoln, Brayford Pool, Lincoln, LN6 7TS, UK.
| | | | | |
Collapse
|
8
|
Thomas AT, Stewart BJ, Ognibene TJ, Turteltaub KW, Bench G. Directly coupled high-performance liquid chromatography-accelerator mass spectrometry measurement of chemically modified protein and peptides. Anal Chem 2013; 85:3644-50. [PMID: 23413773 DOI: 10.1021/ac303609n] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Quantitation of low-abundance protein modifications involves significant analytical challenges, especially in biologically important applications, such as studying the role of post-translational modifications in biology and measurement of the effects of reactive drug metabolites. (14)C labeling combined with accelerator mass spectrometry (AMS) provides exquisite sensitivity for such experiments. Here, we demonstrate real-time (14)C quantitation of high-performance liquid chromatography (HPLC) separations by liquid sample accelerator mass spectrometry (LS-AMS). By enabling direct HPLC-AMS coupling, LS-AMS overcomes several major limitations of conventional HPLC-AMS, where individual HPLC fractions must be collected and converted to graphite before measurement. To demonstrate LS-AMS and compare the new technology to traditional solid sample AMS (SS-AMS), reduced and native bovine serum albumin (BSA) was modified by (14)C-iodoacetamide, with and without glutathione present, producing adducts on the order of 1 modification in every 10(6) to 10(8) proteins. (14)C incorporated into modified BSA was measured by solid carbon AMS and LS-AMS. BSA peptides were generated by tryptic digestion. Analysis of HPLC-separated peptides was performed in parallel by LS-AMS, fraction collection combined with SS-AMS, and (for peptide identification) electrospray ionization and tandem mass spectrometry (ESI-MS/MS). LS-AMS enabled (14)C quantitation from ng sample sizes and was 100 times more sensitive to (14)C incorporated in HPLC-separated peptides than SS-AMS, resulting in a lower limit of quantitation of 50 zmol (14)C/peak. Additionally, LS-AMS turnaround times were minutes instead of days, and HPLC trace analyses required 1/6th the AMS instrument time required for analysis of graphite fractions by SS-AMS.
Collapse
Affiliation(s)
- Avi T Thomas
- Center for Accelerator Mass Spectrometry, Lawrence Livermore National Laboratory, Livermore, California 94551, United States.
| | | | | | | | | |
Collapse
|
9
|
|
10
|
Lim MS, Seong SJ, Park J, Seo JJ, Lee J, Yu KS, Lee HW, Yoon YR. Assessment of pharmacokinetic proportionality of levofloxacin and cyclosporine over a 100-fold dose range in healthy human volunteers. Expert Opin Drug Metab Toxicol 2012; 8:399-405. [DOI: 10.1517/17425255.2012.666237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
11
|
Penner N, Xu L, Prakash C. Radiolabeled Absorption, Distribution, Metabolism, and Excretion Studies in Drug Development: Why, When, and How? Chem Res Toxicol 2012; 25:513-31. [DOI: 10.1021/tx300050f] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Natalia Penner
- Department
of Drug Metabolism and Preclinical Safety, Biogen Idec, Cambridge, Massachusetts 02142
| | - Lin Xu
- Department
of Drug Metabolism and Preclinical Safety, Biogen Idec, Cambridge, Massachusetts 02142
| | - Chandra Prakash
- Department
of Drug Metabolism and Preclinical Safety, Biogen Idec, Cambridge, Massachusetts 02142
| |
Collapse
|
12
|
Harrison A, Gardner I, Hay T, Dickins M, Beaumont K, Phipps A, Purkins L, Allan G, Christian R, Duckworth J, Gurrell I, Kempshall S, Savage M, Seymour M, Simpson M, Taylor L, Turnpenny P. Case studies addressing human pharmacokinetic uncertainty using a combination of pharmacokinetic simulation and alternative first in human paradigms. Xenobiotica 2011; 42:57-74. [PMID: 21992032 DOI: 10.3109/00498254.2011.622418] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PF-184298 ((S)-2,3-dichloro-N-isobutyl-N-pyrrolidin-3-ylbenzamide) and PF-4776548 ((3-(4-fluoro-2-methoxy-benzyl)-7-hydroxy-8,9-dihydro-3H,7H-pyrrolo[2,3-c][1,7]naphthyridin-6-one)) are novel compounds which were selected to progress to human studies. Discordant human pharmacokinetic predictions arose from pre-clinical in vivo studies in rat and dog, and from human in vitro studies, resulting in a clearance prediction range of 3 to >20 mL min⁻¹ kg⁻¹ for PF-184298, and 5 to >20 mL min⁻¹ kg⁻¹ for PF-4776548. A package of work to investigate the discordance for PF-184298 is described. Although ultimately complementary to the human pharmacokinetic data in characterising the disposition of PF-184298 in humans, these data did not provide any further confidence in pharmacokinetic prediction. A fit for purpose human pharmacokinetic study was conducted for each compound, with an oral pharmacologically active dose for PF-184298, and an intravenous and oral microdose for PF-4776548. This provided a relatively low cost, clear decision making approach, resulting in the termination of PF-4776548 and further progression of PF-184298. A retrospective analysis of the data showed that, if the tools had been available at the time, the pharmacokinetics of PF-184298 in human could have been predicted from a population based simulation tool in combination with physicochemical properties and in vitro human intrinsic clearance.
Collapse
Affiliation(s)
- Anthony Harrison
- Department of Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Sandwich Laboratories, Sandwich, Kent, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Dueker SR, Vuong LT, Lohstroh PN, Giacomo JA, Vogel JS. Quantifying exploratory low dose compounds in humans with AMS. Adv Drug Deliv Rev 2011; 63:518-31. [PMID: 21047543 PMCID: PMC3062634 DOI: 10.1016/j.addr.2010.10.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Revised: 10/08/2010] [Accepted: 10/18/2010] [Indexed: 11/20/2022]
Abstract
Accelerator Mass Spectrometry is an established technology whose essentiality extends beyond simply a better detector for radiolabeled molecules. Attomole sensitivity reduces radioisotope exposures in clinical subjects to the point that no population need be excluded from clinical study. Insights in human physiochemistry are enabled by the quantitative recovery of simplified AMS processes that provide biological concentrations of all labeled metabolites and total compound related material at non-saturating levels. In this paper, we review some of the exploratory applications of AMS (14)C in toxicological, nutritional, and pharmacological research. This body of research addresses the human physiochemistry of important compounds in their own right, but also serves as examples of the analytical methods and clinical practices that are available for studying low dose physiochemistry of candidate therapeutic compounds, helping to broaden the knowledge base of AMS application in pharmaceutical research.
Collapse
|
14
|
Wagner CC, Langer O. Approaches using molecular imaging technology -- use of PET in clinical microdose studies. Adv Drug Deliv Rev 2011; 63:539-46. [PMID: 20887762 DOI: 10.1016/j.addr.2010.09.011] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Revised: 09/21/2010] [Accepted: 09/22/2010] [Indexed: 12/11/2022]
Abstract
Positron emission tomography (PET) imaging uses minute amounts of radiolabeled drug tracers and thereby meets the criteria for clinical microdose studies. The advantage of PET, when compared to other analytical methods used in microdose studies, is that the pharmacokinetics (PK) of a drug can be determined in the tissue targeted for drug treatment. PET microdosing already offers interesting applications in clinical oncology and in the development of central nervous system pharmaceuticals and is extending its range of application to many other fields of pharmaceutical medicine. Although requirements for preclinical safety testing for microdose studies have been cut down by regulatory authorities, radiopharmaceuticals increasingly need to be produced under good manufacturing practice (GMP) conditions, which increases the costs of PET microdosing studies. Further challenges in PET microdosing include combining PET with other ultrasensitive analytical methods, such as accelerator mass spectrometry (AMS), to gain plasma PK data of drugs, beyond the short PET examination periods. Finally, conducting clinical PET studies with radiolabeled drugs both at micro- and therapeutic doses is encouraged to answer the question of dose linearity in clinical microdosing.
Collapse
Affiliation(s)
- Claudia C Wagner
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger-Gürtel 18-20, A-1090, Vienna, Austria
| | | |
Collapse
|
15
|
New ultrasensitive detection technologies and techniques for use in microdosing studies. Bioanalysis 2011; 1:357-66. [PMID: 21083172 DOI: 10.4155/bio.09.40] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
In a microdosing study, subpharmacologically active doses of drug are given to human volunteers at an early stage of development in order to obtain preliminary pharmacokinetic data. The very low doses of drug administered (≤100 µg) consequently lead to very low concentrations of drug appearing in the body and therefore highly sensitive analytical techniques are required. There are three such analytical technologies currently used in microdosing studies: PET, liquid chromatography (LC)-tandem mass spectrometry (MS/MS) and accelerator mass spectrometry (AMS). Both PET and AMS employ radioisotopic tracers. PET is an imaging technique and AMS is an extremely sensitive isotope ratio method, able to measure drug concentrations in the ag/ml range. LC-MS/MS does not require the presence of an isotopic tracer and its sensitivity is in the pg/ml range. This review examines each of these three analytical modalities in the context of performing microdosing studies.
Collapse
|
16
|
Minamide Y, Osawa Y, Nishida H, Igarashi H, Kudoh S. A highly sensitive LC-MS/MS method capable of simultaneously quantitating celiprolol and atenolol in human plasma for a cassette cold-microdosing study. J Sep Sci 2011; 34:1590-8. [PMID: 21595025 DOI: 10.1002/jssc.201100089] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 03/15/2011] [Accepted: 04/05/2011] [Indexed: 11/11/2022]
Abstract
A highly sensitive simultaneous quantitative method for a cassette cold-microdosing study on celiprolol and atenolol was developed with liquid chromatography-tandem mass spectrometry. The method utilizes a combination of solid-phase extraction (SPE) with strong cation exchange (SCX) cartridge columns and reversed-phase chromatography with an ODS analytical column. SCX-SPE cartridge columns (100 mg sorbent) were used for a selective extraction of celiprolol, atenolol and metoprolol (internal standard) from 500 μL of human plasma samples. Turbo-ion spray at positive mode was employed for the ionization of the drug compounds. Quantitation was performed on a triple quadrupole mass spectrometer by selected reaction monitoring with the transitions of m/z 380 to m/z 251 for celiprolol and m/z 267 to m/z 145 for atenolol. Separation of analytes was achieved on an ODS column (100 mm length × 2.1 mm id, 3 μm) by a gradient elution with 10 mM formic acid and methanol by varying their proportion at a flow rate of 0.2 mL/min. The method was validated in the range of 1-250 pg/mL for celiprolol and 2.5-250 pg/mL for atenolol and was successfully applied to the elucidation of pharmacokinetic profiling in a cold cassette microdosing study of the β-blockers.
Collapse
Affiliation(s)
- Yoshiyuki Minamide
- Bioanalysis Research Center, Shin Nippon Biomedical Laboratories Ltd, Gunma, Japan.
| | | | | | | | | |
Collapse
|
17
|
Coleman RA. A human approach to drug development: opportunities and limitations. Altern Lab Anim 2011; 38 Suppl 1:21-5. [PMID: 21275479 DOI: 10.1177/026119291003801s06] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The pharmaceutical industry is failing in its primary function, with increasing expenditure and decreased output in terms of new medicines brought to market. It cannot carry on as it is, without sliding into a terminal decline. It must, therefore, take some positive steps toward addressing its problems. We do not have to look far to see one very obvious problem, namely, the industry's continuing reliance on nonhuman biology as the basis of its evaluation of potential safety and efficacy. The time has come to focus on the relevant, and to realise that more human-based testing is essential, if the industry is to survive as a source of innovation in drug therapy. This can incorporate earlier clinical testing, in the form of microdosing, and promotion of the development of more-powerful computational approaches based on human information. Fortunately, headway is being made in both approaches. However, a problem remains in the lack of functional evaluation of human tissues, where the lack of commitment, and the inadequacy of the tissue resource itself, are hampering any serious developments. An outline of a collaborative scheme is proposed, that will address this issue, central to which is improved access to research tissues from heart-beating organ donors.
Collapse
|
18
|
|
19
|
Abstract
The concept of microdosing has been around for approximately 10 years. In this time there have been an increasing number of drugs reported in the literature where the pharmacokinetics at a microdose have been compared with those observed at a therapeutic dose. Currently, approximately 80% of the microdose pharmacokinetics available in the public domain have been shown to scale to those observed at a therapeutic dose, within a twofold difference. Microdosing is now being extended into areas of drug development other than purely pharmacokinetic prediction. Microdosing has been applied to the study of drug-drug interactions by giving human volunteers a microdose of the candidate drug before and after the administration of a drug known to inhibit or induce certain enzymes, such as the cytochrome P450s. Early data on the metabolism of a drug candidate can be obtained by administering a (14)C-drug to human volunteers and comparing the plasma concentration-time curves for total (14)C and unchanged parent compound. Full metabolic profiles can be generated as an early indication of the drug's metabolism in humans, prior to Phase 1 clinical studies. Microdosing is also being applied to situations where the concentration of a drug in cell or tissue types is key to its efficacy. The application of microdosing as a tool in drug development is therefore widening into new and previously unforeseen fields.
Collapse
|
20
|
Accelerator mass spectrometry-enabled studies: current status and future prospects. Bioanalysis 2011; 2:519-41. [PMID: 20440378 DOI: 10.4155/bio.09.188] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Accelerator mass spectrometry is a detection platform with exceptional sensitivity compared with other bioanalytical platforms. Accelerator mass spectrometry (AMS) is widely used in archeology for radiocarbon dating applications. Early exploration of the biological and pharmaceutical applications of AMS began in the early 1990s. AMS has since demonstrated unique problem-solving ability in nutrition science, toxicology and pharmacology. AMS has also enabled the development of new applications, such as Phase 0 microdosing. Recent development of AMS-enabled applications has transformed this novelty research instrument to a valuable tool within the pharmaceutical industry. Although there is now greater awareness of AMS technology, recognition and appreciation of the range of AMS-enabled applications is still lacking, including study-design strategies. This review aims to provide further insight into the wide range of AMS-enabled applications. Examples of studies conducted over the past two decades will be presented, as well as prospects for the future of AMS.
Collapse
|
21
|
Analytical validation of accelerator mass spectrometry for pharmaceutical development. Bioanalysis 2011; 2:469-85. [PMID: 21083256 DOI: 10.4155/bio.10.14] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The validation parameters for pharmaceutical analyses were examined for the accelerator mass spectrometry measurement of (14)C/C ratio, independent of chemical separation procedures. The isotope ratio measurement was specific (owing to the (14)C label), stable across samples storage conditions for at least 1 year, linear over four orders of magnitude with an analytical range from 0.1 Modern to at least 2000 Modern (instrument specific). Furthermore, accuracy was excellent (between 1 and 3%), while precision expressed as coefficient of variation was between 1 and 6% determined primarily by radiocarbon content and the time spent analyzing a sample. Sensitivity, expressed as LOD and LLOQ was 1 and 10 attomoles of (14)C, respectively (which can be expressed as compound equivalents) and for a typical small molecule labeled at 10% incorporated with (14)C corresponds to 30 fg equivalents. Accelerator mass spectrometry provides a sensitive, accurate and precise method of measuring drug compounds in biological matrices.
Collapse
|
22
|
Sensitivity and proportionality assessment of metabolites from microdose to high dose in rats using LC-MS/MS. Bioanalysis 2011; 2:407-19. [PMID: 21083251 DOI: 10.4155/bio.10.10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The objective of this study was to evaluate the sensitivity requirement for LC-MS/MS as an analytical tool to characterize metabolites in plasma and urine at microdoses in rats and to investigate proportionality of metabolite exposure from a microdose of 1.67 µg/kg to a high dose of 5000 µg/kg for atorvastatin, ofloxacin, omeprazole and tamoxifen. RESULTS Only the glucuronide metabolite of ofloxacin, the hydroxylation metabolite of omeprazole and the hydration metabolite of tamoxifen were characterized in rat plasma at microdose by LC-MS/MS. The exposure of detected metabolites of omeprazole and tamoxifen appeared to increase in a nonproportional manner with increasing doses. Exposure of ortho- and para-hydroxyatorvastatin, but not atorvastatin and lactone, increased proportionally with increasing doses. CONCLUSION LC-MS/MS has demonstrated its usefulness for detecting and characterizing the major metabolites in plasma and urine at microdosing levels in rats. The exposure of metabolites at microdose could not simply be used to predict their exposure at higher doses.
Collapse
|
23
|
Henderson PT, Li T, He M, Zhang H, Malfatti M, Gandara D, Grimminger PP, Danenberg KD, Beckett L, de Vere White RW, Turteltaub KW, Pan CX. A microdosing approach for characterizing formation and repair of carboplatin-DNA monoadducts and chemoresistance. Int J Cancer 2011; 129:1425-34. [PMID: 21128223 DOI: 10.1002/ijc.25814] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Accepted: 11/04/2010] [Indexed: 12/13/2022]
Abstract
Formation and repair of platinum (Pt)-induced DNA adducts is a critical step in Pt drug-mediated cytotoxicity. Measurement of Pt-DNA adduct kinetics in tumors may be useful for better understanding chemoresistance and therapeutic response. However, this concept has yet to be rigorously tested because of technical challenges in measuring the adducts at low concentrations and consistent access to sufficient tumor biopsy material. Ultrasensitive accelerator mass spectrometry was used to detect [(14)C]carboplatin-DNA monoadducts at the attomole level, which are the precursors to Pt-DNA crosslink formation, in six cancer cell lines as a proof-of-concept. The most resistant cells had the lowest monoadduct levels at all time points over 24 hr. [(14)C]Carboplatin "microdoses" (1/100th the pharmacologically effective concentration) had nearly identical adduct formation and repair kinetics compared to therapeutically relevant doses, suggesting that the microdosing approach can potentially be used to determine the pharmacological effects of therapeutic treatment. Some of the possible chemoresistance mechanisms were also studied, such as drug uptake/efflux, intracellular inactivation and DNA repair in selected cell lines. Intracellular inactivation and efficient DNA repair each contributed significantly to the suppression of DNA monoadduct formation in the most resistant cell line compared to the most sensitive cell line studied (p < 0.001). Nucleotide excision repair (NER)-deficient and -proficient cells showed substantial differences in carboplatin monoadduct concentrations over 24 hr that likely contributed to chemoresistance. The data support the utility of carboplatin microdosing as a translatable approach for defining carboplatin-DNA monoadduct formation and repair, possibly by NER, which may be useful for characterizing chemoresistance in vivo.
Collapse
Affiliation(s)
- Paul T Henderson
- Division of Hematology and Oncology, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Jubert C, Mata J, Bench G, Dashwood R, Pereira C, Tracewell W, Turteltaub K, Williams D, Bailey G. Effects of chlorophyll and chlorophyllin on low-dose aflatoxin B(1) pharmacokinetics in human volunteers. Cancer Prev Res (Phila) 2009; 2:1015-22. [PMID: 19952359 PMCID: PMC5314947 DOI: 10.1158/1940-6207.capr-09-0099] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Chlorophyll (Chla) and chlorophyllin (CHL) were shown previously to reduce carcinogen bioavailability, biomarker damage, and tumorigenicity in trout and rats. These findings were partially extended to humans, where CHL reduced excretion of aflatoxin B(1) (AFB(1))-DNA repair products in Chinese unavoidably exposed to dietary AFB(1). However, neither AFB(1) pharmacokinetics nor Chla effects were examined. We conducted an unblinded crossover study to establish AFB(1) pharmacokinetic parameters among four human volunteers, and to explore possible effects of CHL or Chla cotreatment in three of those volunteers. For protocol 1, fasted subjects received an Institutional Review Board-approved dose of 14C-AFB(1) (30 ng, 5 nCi) by capsule with 100 mL water, followed by normal eating and drinking after 2 hours. Blood and cumulative urine samples were collected over 72 hours, and 14C- AFB(1) equivalents were determined by accelerator mass spectrometry. Protocols 2 and 3 were similar except capsules also contained 150 mg of purified Chla or CHL, respectively. Protocols were repeated thrice for each volunteer. The study revealed rapid human AFB(1) uptake (plasma k(a), 5.05 + or - 1.10 h(-1); T(max), 1.0 hour) and urinary elimination (95% complete by 24 hours) kinetics. Chla and CHL treatment each significantly impeded AFB(1) absorption and reduced Cmax and AUCs (plasma and urine) in one or more subjects. These initial results provide AFB(1) pharmacokinetic parameters previously unavailable for humans, and suggest that Chla or CHL co-consumption may limit the bioavailability of ingested aflatoxin in humans, as they do in animal models.
Collapse
Affiliation(s)
- Carole Jubert
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Cooke MS, Henderson PT, Evans MD. Sources of extracellular, oxidatively-modified DNA lesions: implications for their measurement in urine. J Clin Biochem Nutr 2009; 45:255-70. [PMID: 19902015 PMCID: PMC2771246 DOI: 10.3164/jcbn.sr09-41] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Accepted: 04/29/2009] [Indexed: 12/14/2022] Open
Abstract
There is a robust mechanistic basis for the role of oxidation damage to DNA in the aetiology of various major diseases (cardiovascular, neurodegenerative, cancer). Robust, validated biomarkers are needed to measure oxidative damage in the context of molecular epidemiology, to clarify risks associated with oxidative stress, to improve our understanding of its role in health and disease and to test intervention strategies to ameliorate it. Of the urinary biomarkers for DNA oxidation, 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG) is the most studied. However, there are a number of factors which hamper our complete understanding of what meausrement of this lesion in urine actually represents. DNA repair is thought to be a major contributor to urinary 8-oxodG levels, although the precise pathway(s) has not been proven, plus possible contribution from cell turnover and diet are possible confounders. Most recently, evidence has arisen which suggests that nucleotide salvage of 8-oxodG and 8-oxoGua can contribute substantially to 8-oxoG levels in DNA and RNA, at least in rapidly dividing cells. This new observation may add an further confounder to the conclusion that 8-oxoGua or 8-oxodG, and its nucleobase equivalent 8-oxoguanine, concentrations in urine are simply a consequence of DNA repair. Further studies are required to define the relative contributions of metabolism, disease and diet to oxidised nucleic acids and their metabolites in urine in order to develop urinalyis as a better tool for understanding human disease.
Collapse
Affiliation(s)
- Marcus S Cooke
- Radiation and Oxidative Stress Section, Department of Cancer Studies and Molecular Medicine, Robert Kilpatrick Clinical Sciences Bilding, University of Leicester, LE2 7LX, UK
| | | | | |
Collapse
|
26
|
Penner N, Klunk LJ, Prakash C. Human radiolabeled mass balance studies: objectives, utilities and limitations. Biopharm Drug Dispos 2009; 30:185-203. [PMID: 19544285 DOI: 10.1002/bdd.661] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The determination of metabolic pathways of a drug candidate through the identification of circulating and excreted metabolites is vitally important to understanding its physical and biological effects. Knowledge of metabolite profiles of a drug candidate in animals and humans is essential to ensure that animal species used in toxicological evaluations of new drug candidates are appropriate models of humans. The recent FDA final guidance recommends that human oxidative metabolites whose exposure exceeds 10% of the parent AUC at steady-state should be assessed in at least one of the preclinical animal species used in toxicological assessment. Additional toxicological testing on metabolites that have higher exposure in humans than in preclinical species may be required. The metabolite profiles in laboratory animals and humans are generally accomplished by mass balance and excretion studies in which radiolabeled drugs are administered to these species. The biological fluids are collected, analysed for total radioactivity and evaluated for a quantitative profile of metabolites. Thus, these studies not only determine the rates and routes of excretion but also provide very critical information on the metabolic pathways of drugs in preclinical species and humans. In addition, these studies are required by regulatory agencies for the new drug approval process. Despite the usefulness of these radiolabeled mass balance studies, there is little concrete guidance on how to perform or assess these complex studies. This article examines the objectives, utilities and limitations of these studies and how these studies could be used for the determination of the metabolite exposure in animals and humans.
Collapse
Affiliation(s)
- Natalia Penner
- Department of Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, MA 02142, USA
| | | | | |
Collapse
|
27
|
Microdosing: a valuable tool for accelerating drug development and the role of bioanalytical methods in meeting the challenge. Bioanalysis 2009; 1:1293-305. [DOI: 10.4155/bio.09.107] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The concept of specifically determining the clinical pharmacokinetics of a compound using a very low nonpharmacologically active dose (microdose) with an abridged safety and chemistry, manufacturing and control package is relatively new. It is not without its controversy and it is still a subject of discussion. Here, the rationale and application of this approach are examined, together with the regulatory and bioanalytical framework. There are two bioanalytical methods commonly used for human microdosing studies: LC–MS/MS and accelerator MS (AMS). Each method has advantages and disadvantages with the choice of instrumentation being closely tied to the primary objective(s) of the study. If a rapid decision is required on the appropriateness of a pharmacokinetic profile or if a choice is needed from a series of compounds, especially before radiolabeled material is available, LC–MS/MS may be preferable. However, if extreme sensitivity is required, data are required on all drug-related material and metabolites, or a simultaneous intravenous microdose is used to determine absolute bioavailability (sometimes referred to as microtracing), AMS becomes the analytical method of choice. Examples are provided of microdosing studies utilizing both of these bioanalytical techniques. It is emphasized that microdosing is only one tool in the drug developer’s tool box and it should be used in the context of all available data. However, when used appropriately, microdosing is a valuable tool, bridging between lead optimization and early clinical development.
Collapse
|
28
|
Hah SS, Henderson PT, Turteltaub KW. Recent advances in biomedical applications of accelerator mass spectrometry. J Biomed Sci 2009; 16:54. [PMID: 19534792 PMCID: PMC2712465 DOI: 10.1186/1423-0127-16-54] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2008] [Accepted: 06/17/2009] [Indexed: 11/13/2022] Open
Abstract
The use of radioisotopes has a long history in biomedical science, and the technique of accelerator mass spectrometry (AMS), an extremely sensitive nuclear physics technique for detection of very low-abundant, stable and long-lived isotopes, has now revolutionized high-sensitivity isotope detection in biomedical research, because it allows the direct determination of the amount of isotope in a sample rather than measuring its decay, and thus the quantitative analysis of the fate of the radiolabeled probes under the given conditions. Since AMS was first used in the early 90's for the analysis of biological samples containing enriched 14C for toxicology and cancer research, the biomedical applications of AMS to date range from in vitro to in vivo studies, including the studies of 1) toxicant and drug metabolism, 2) neuroscience, 3) pharmacokinetics, and 4) nutrition and metabolism of endogenous molecules such as vitamins. In addition, a new drug development concept that relies on the ultrasensitivity of AMS, known as human microdosing, is being used to obtain early human metabolism information of candidate drugs. These various aspects of AMS are reviewed and a perspective on future applications of AMS to biomedical research is provided.
Collapse
Affiliation(s)
- Sang Soo Hah
- Department of Chemistry and Research Institute for Basic Sciences, Kyung Hee University 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Korea.
| | | | | |
Collapse
|
29
|
Lappin G, Garner RC. The utility of microdosing over the past 5 years. Expert Opin Drug Metab Toxicol 2009; 4:1499-506. [PMID: 19040326 DOI: 10.1517/17425250802531767] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Microdosing studies (human Phase 0) are used to select drug candidates for Phase I clinical trials on the basis of their pharmacokinetic properties, using subpharmacologic doses (maximum 100 microg). There are questions as to whether pharmacokinetic data obtained at these low doses will predict those at the clinically relevant dose. OBJECTIVE To review the current literature on microdosing and assess how well microdose data have predicted the pharmacokinetics obtained at a therapeutic dose. METHODS All data published in the peer reviewed literature comparing pharmacokinetics at a microdose with a therapeutic dose were reviewed, excluding those studies aimed at imaging. CONCLUSIONS Of the 18 drugs reported, 15 demonstrated linear pharmacokinetics within a factor of 2 between a microdose and a therapeutic dose. Therefore, data that support the utility of microdosing are beginning to emerge.
Collapse
Affiliation(s)
- Graham Lappin
- Xceleron Ltd, The Biocentre, Innovation Way, York, YO10 5NY, UK.
| | | |
Collapse
|
30
|
Miyaji Y, Ishizuka T, Kawai K, Hamabe Y, Miyaoka T, Oh-hara T, Ikeda T, Kurihara A. Use of An Intravenous Microdose of 14C-labeled drug and Accelerator Mass Spectrometry to measure Absolute Oral Bioavailability in Dogs; Cross-comparison of Assay Methods by Accelerator Mass Spectrometry and Liquid Chromatography-Tandem Mass Spectrometry. Drug Metab Pharmacokinet 2009; 24:130-8. [DOI: 10.2133/dmpk.24.130] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
31
|
Vuong LT, Ruckle JL, Blood AB, Reid MJ, Wasnich RD, Synal HA, Dueker SR. Use of Accelerator Mass Spectrometry to Measure the Pharmacokinetics and Peripheral Blood Mononuclear Cell Concentrations of Zidovudine. J Pharm Sci 2008; 97:2833-43. [PMID: 17854048 DOI: 10.1002/jps.21160] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The remarkable sensitivity of accelerator mass spectrometry (AMS) is finding many new applications in pharmacology. In this study AMS was used to measure [(14)C]-Zidovudine (ZDV) concentrations at the drug's site of action (peripheral blood mononuclear cells, PBMCs) following a dose of 520 ng (less than one-millionth of the standard daily dose) to a healthy volunteer. In addition, the pharmacokinetics of this microdose were determined and compared to previously published parameters for therapeutic doses. Microdose ZDV pharmacokinetic parameters fell within reported 95% confidence intervals or standard deviations of most previously published values for therapeutic doses. Blood, urine, stool, saliva, and isolated PBMCs were collected periodically through 96 h postdose and analyzed for ZDV and metabolite concentrations. The results showed that ZDV is rapidly absorbed and eliminated, has one major metabolite, and is sequestered in PBMCs. (14)C mass balance assessments indicated a significant portion of ZDV remained after 96 h with a much prolonged elimination half-life. Results of this study demonstrate the usefulness of microdosing and AMS as a tool for studying the pharmacokinetic characteristics, including PBMC concentrations, of ZDV and underscore the value of AMS as a tool with which to perform pharmacokinetic and mass balance studies using trace amounts of radiolabeled compound.
Collapse
Affiliation(s)
- Le T Vuong
- Vitalea Science, Inc, Woodland, California 95776, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Bauer M, Wagner CC, Langer O. Microdosing studies in humans: the role of positron emission tomography. Drugs R D 2008; 9:73-81. [PMID: 18298126 DOI: 10.2165/00126839-200809020-00002] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Positron emission tomography (PET)-microdosing comprises the administration of a carbon-11- or fluorine-18-labelled drug candidate to human subjects in order to describe the drug's concentration-time profile in body tissues targeted for treatment. As PET microdosing involves the administration of only microgram amounts of unlabelled drug, the potential toxicological risk to human subjects is very limited. Consequently, regulatory authorities require reduced preclinical safety testing as compared with conventional phase 1 studies. Microdose studies are gaining increasing importance in clinical drug research as they have the potential to shorten time-lines and cut costs along the critical path of drug development. Current applications of PET in anticancer, anti-infective and CNS system drug research are reviewed.
Collapse
Affiliation(s)
- Martin Bauer
- Department of Clinical Pharmacology, Medical University Vienna, Vienna, Austria
| | | | | |
Collapse
|
33
|
Microdosing assessment to evaluate pharmacokinetics and drug metabolism in rats using liquid chromatography-tandem mass spectrometry. Pharm Res 2008; 25:1572-82. [PMID: 18299965 DOI: 10.1007/s11095-008-9555-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Accepted: 02/04/2008] [Indexed: 10/22/2022]
Abstract
PURPOSE To evaluate the sensitivity requirement for LC-MS/MS as an analytical tool to support human microdosing study with sub-pharmacological dose, investigate proportionality of pharmacokinetics from the microdose to therapeutic human equivalent doses in rats and characterize circulating metabolites in rats administered with the microdose. MATERIALS AND METHODS Five drugs of antipyrine, metoprolol, carbamazepine, digoxin and atenolol were administered orally to male Sprague-Dawley rats at 0.167, 1.67, 16.7, 167 and 1,670 microg/kg doses. Plasma samples were extracted using either solid phase extraction or liquid-liquid extraction, and analyzed using LC-MS/MS. RESULTS Using 100 microl of plasma sample, the lower limit of quantitation for antipyrine (10 pg/ml), carbamazepine (1 pg/ml), metoprolol (5 pg/ml), atenolol (20 pg/ml), and digoxin (5 pg/ml) were achieved using an API 5000. Proportional pharmacokinetics were observed from 0.167 microg/kg to 1,670 microg/kg for antipyrine and carbamazepine and from 1.67 to 1,670 microg/kg for atenolol and digoxin, while metoprolol exhibited a non-proportional pharmacokinetics relationship. Several metabolites of carbamazepine were characterized in plasma from rats dosed at 1.67 mug/kg using LC-MS/MS. CONCLUSIONS This study has shown the promise of sensitive LC-MS/MS method to support microdose pharmacokinetics and drug metabolism studies in human.
Collapse
|
34
|
Bertino JS, Greenberg HE, Reed MD. American College of Clinical Pharmacology position statement on the use of microdosing in the drug development process. J Clin Pharmacol 2007; 47:418-22. [PMID: 17389550 DOI: 10.1177/0091270006299092] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Joseph S Bertino
- Ordway Research Institute Drug Development Center, 1365 Washington Avenue, Suite 201, Albany, NY 12206-1066, USA.
| | | | | |
Collapse
|
35
|
Abstract
Drug metabolism information is a necessary component of drug discovery and development. The key issues in drug metabolism include identifying: the enzyme(s) involved, the site(s) of metabolism, the resulting metabolite(s), and the rate of metabolism. Methods for predicting human drug metabolism from in vitro and computational methodologies and determining relationships between the structure and metabolic activity of molecules are also critically important for understanding potential drug interactions and toxicity. There are numerous experimental and computational approaches that have been developed in order to predict human metabolism which have their own limitations. It is apparent that few of the computational tools for metabolism prediction alone provide the major integrated functions needed to assist in drug discovery. Similarly the different in vitro methods for human drug metabolism themselves have implicit limitations. The utilization of these methods for pharmaceutical and other applications as well as their integration is discussed as it is likely that hybrid methods will provide the most success.
Collapse
Affiliation(s)
- Larry J Jolivette
- Preclinical Drug Discovery, Cardiovascular and Urogenital Centre of Excellence in Drug Discovery, GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| | | |
Collapse
|
36
|
McLean MA, Tam CYJ, Baratta MT, Holliman CL, Ings RM, Galluppi GR. Accelerating drug development: methodology to support first-in-man pharmacokinetic studies by the use of drug candidate microdosing. Drug Dev Res 2007. [DOI: 10.1002/ddr.20160] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
37
|
Abstract
Accelerator mass spectrometry (AMS) counts individual rare, usually radio-, isotopes such as radiocarbon at high efficiency and specificity in milligram-sized samples. AMS traces very low chemical doses (micrograms) and radiative doses (100 Bq) of isotope-labeled compounds in animal models and directly in humans for pharmaceutical, nutritional, or toxicological research. Absorption, metabolism, distribution, binding, and elimination are all quantifiable with high precision after appropriate sample definition.
Collapse
Affiliation(s)
- John S Vogel
- Lawrence Livermore National Laboratory, University of California, Livermore, CA 94551, USA.
| |
Collapse
|
38
|
Balani SK, Nagaraja NV, Qian MG, Costa AO, Daniels JS, Yang H, Shimoga PR, Wu JT, Gan LS, Lee FW, Miwa GT. Evaluation of microdosing to assess pharmacokinetic linearity in rats using liquid chromatography-tandem mass spectrometry. Drug Metab Dispos 2006; 34:384-8. [PMID: 16326814 DOI: 10.1124/dmd.105.007195] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The microdosing strategy allows for early assessment of human pharmacokinetics of new chemical entities using more limited safety assessment requirements than those requisite for a conventional phase I program. The current choice for evaluating microdosing is accelerator mass spectrometry (AMS) due to its ultrasensitivity for detecting radiotracers. However, the AMS technique is still expensive to be used routinely and requires the preparation of radiolabeled compounds. This report describes a feasibility study with conventional liquid chromatography-tandem mass spectrometry (LC-MS/MS) technology for oral microdosing assessment in rats, a commonly used preclinical species. The nonlabeled drugs fluconazole and tolbutamide were studied because of their similar pharmacokinetics characteristics in rats and humans. We demonstrate that pharmacokinetics can be readily characterized by LC-MS/MS at a microdose of 1 microg/kg for these molecules in rats, and, hence, LC-MS/MS should be adequate in human microdosing studies. The studies also exhibit linearity in exposure between the microdose and >or=1000-fold higher doses in rats for these drugs, which are known to show a linear dose-exposure relationship in the clinic, further substantiating the potential utility of LC-MS/MS in defining pharmacokinetics from the microdose of drugs. These data should increase confidence in the use of LC-MS/MS in microdose pharmacokinetics studies of new chemical entities in humans. Application of this approach is also described for an investigational compound, MLNX, in which the pharmacokinetics in rats were determined to be nonlinear, suggesting that MLNX pharmacokinetics at microdoses in humans also might not reflect those at the therapeutic doses. These preclinical studies demonstrate the potential applicability of using traditional LC-MS/MS for microdose pharmacokinetic assessment in humans.
Collapse
Affiliation(s)
- Suresh K Balani
- DMPK, Drug Safety & Disposition, Millennium Pharmaceuticals, Inc., 45 Sidney Street, Cambridge, MA 02139, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Brown K, Tompkins EM, White INH. Applications of accelerator mass spectrometry for pharmacological and toxicological research. MASS SPECTROMETRY REVIEWS 2006; 25:127-45. [PMID: 16059873 DOI: 10.1002/mas.20059] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The technique of accelerator mass spectrometry (AMS), known for radiocarbon dating of archeological specimens, has revolutionized high-sensitivity isotope detection in pharmacology and toxicology by allowing the direct determination of the amount of isotope in a sample rather than measuring its decay. It can quantify many isotopes, including 26Al, 14C, 41Ca, and 3H with detection down to attomole (10(-18)) amounts. Pharmacokinetic data in humans have been achieved with ultra-low levels of radiolabel. One of the most exciting biomedical applications of AMS with 14C-labeled potential carcinogens is the detection of modified proteins or DNA in tissues. The relationship between low-level exposure and covalent binding of genotoxic chemicals has been compared in rodents and humans. Such compounds include heterocyclic amines, benzene, and tamoxifen. Other applications range from measuring the absorption of 26Al to monitoring 41Ca turnover in bone. In epoxy-embedded tissue sections, high-resolution imaging of 14C label in cells is possible. The uses of AMS are becoming more widespread with the availability of instrumentation dedicated to the analysis of biomedical samples.
Collapse
Affiliation(s)
- Karen Brown
- Cancer Biomarkers and Prevention Group, Department of Cancer Studies and Molecular Medicine, The Biocentre, University of Leicester, Leicester LE1 7RH, United Kingdom
| | | | | |
Collapse
|
40
|
Ward KW. Recent advances in pharmacokinetic extrapolation from preclinical data to humans. Expert Opin Drug Metab Toxicol 2005; 1:583-94. [PMID: 16863426 DOI: 10.1517/17425255.1.4.583] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The early characterisation of drug metabolism and pharmacokinetic (DMPK) properties of new chemical entities plays a key role in the pharmaceutical industry's effort to reduce attrition. Specifically, a major goal of early DMPK studies is to accurately predict the behaviour of new chemical entities in humans, thus allowing likely failures to be terminated rapidly and resource to be placed on molecules most likely to succeed. The present review summarises progress over the past several years in the key technologies used in the pharmaceutical industry to achieve these goals: namely, in vivo, in vitro and in silico/computational tools. The limitations of the various assays are discussed, with attention also given to likely future directions in this field.
Collapse
Affiliation(s)
- Keith W Ward
- Bausch & Lomb, Global Preclinical Development, Rochester, NY 14603, USA.
| |
Collapse
|
41
|
Hah SS, Kim HM, Sumbad RA, Henderson PT. Hydantoin derivative formation from oxidation of 7,8-dihydro-8-oxo-2'-deoxyguanosine (8-oxodG) and incorporation of 14C-labeled 8-oxodG into the DNA of human breast cancer cells. Bioorg Med Chem Lett 2005; 15:3627-31. [PMID: 15982874 DOI: 10.1016/j.bmcl.2005.05.113] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2005] [Revised: 05/04/2005] [Accepted: 05/10/2005] [Indexed: 11/22/2022]
Abstract
One-electron oxidation of 7,8-dihydro-8-oxo-2'-deoxyguanosine (8-oxodG) yielded a guanidinohydantoin derivative (dGh) and a spiroiminodihydantoin derivative (dSp), both putatively mutagenic products that may be formed in vivo. The nucleoside dGh was the major product at room temperature, regardless of pH. The results are contrary to previously published model studies using 2',3',5'-triacetoxy-8-oxo-7,8-dihydroguanosine (Luo, W.; Miller, J. G.; Rachlin, E. M.; Burrows, C. J. Org. Lett. 2000, 2, 613; Luo, W.; Miller, J.G.; Rachlin, E.M.; Burrows, C.J. Chem. Res. Toxicol. 2001, 14, 927), who observed a spiroiminodihydantoin derivative as the major product at neutral pH. Clearly, the functional groups attached to the ribose moiety of 8-oxodG influence the oxidation chemistry of the nucleobase derivative. To explore this chemistry in vivo, (14)C-labeled 8-oxodG was synthesized and incubated with growing MCF-7 human breast cancer cells, resulting in the incorporation of the compound into cellular DNA as measured by a novel accelerator mass spectrometry assay.
Collapse
Affiliation(s)
- Sang Soo Hah
- Biosciences Directorate and Center for Accelerator Mass Spectrometry, Lawrence Livermore National Laboratory, 7000 E. Avenue, L-441, Livermore, CA 94551, USA
| | | | | | | |
Collapse
|
42
|
Affiliation(s)
- Ian R Wilding
- Pharmaceutical Profiles, Mere Way, Ruddington Fields, Nottingham NG11 6JS, UK.
| | | |
Collapse
|