1
|
Quinn TM, Bruce AM, Burt T, Dhaliwal K. Phase 0 trials/ Intra-Target-Microdosing (ITM) and the lung: a review. BMC Pulm Med 2024; 24:425. [PMID: 39210357 PMCID: PMC11363577 DOI: 10.1186/s12890-024-03193-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
The COVID-19 pandemic has highlighted the importance of efficient drug discovery in respiratory disease. The traditional set up of clinical trials is expensive and allows for significant attrition of new drugs, many of which undergo extensive safety testing before being abandoned for lack of efficacy. Phase 0 trials, named as they sit between pre-clinical research and phase I, allow for the testing of sub-clinical microdoses in humans to gather early pharmacokinetic (PK), pharmacodynamic (PD) and mechanistic data, before deciding on which drugs to advance further. This early data can improve the efficiency and cost effectiveness of drug development and reduce the extent of animal testing. Phase 0 trials traditionally have utilised sub-therapeutic microdoses of compounds administered intravenously with readouts focusing on PK - measured using highly sensitive methods such as accelerator mass spectrometry (AMS) and liquid chromatography tandem mass spectrometry (LC-MS/MS) of peripheral blood, as well as whole-body positron emission tomography (PET). Mathematical models allow for extrapolation of this PK data to support the further testing of larger, systemically effective doses. However, this extrapolation method is limited at providing robust PD or target engagement/ mode of action data. Using an Intra-Target Microdosing (ITM) approach, a small compartment of the body (about 1% or less) is exposed to potentially clinically active local concentrations. This allows for the collection of PD data, evidence of target cell engagement, as well as the opportunity to extrapolate systemic PK and PD data. This approach has the potential within the pulmonary system for the study and rapid and cost-effective development of new and repurposed drugs.
Collapse
Affiliation(s)
- Tom M Quinn
- Baillie Gifford Pandemic Science Hub, Centre for Inflammation Research, Institute for Regeneration & Repair, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK.
- Department of Respiratory Medicine, Western General Hospital, Edinburgh, UK.
| | - Annya M Bruce
- Baillie Gifford Pandemic Science Hub, Centre for Inflammation Research, Institute for Regeneration & Repair, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK
| | - Tal Burt
- Burt Consultancy, LLC, New York, NY, USA
| | - Kevin Dhaliwal
- Baillie Gifford Pandemic Science Hub, Centre for Inflammation Research, Institute for Regeneration & Repair, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK.
- Department of Respiratory Medicine, New Royal Infirmary of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK.
| |
Collapse
|
2
|
Weiss HM, Sugiyama Y, van Duijn E. Editorial: Incorporating Phase 0 microdosing as a powerful tool into a new vision of drug development. Front Pharmacol 2024; 15:1455643. [PMID: 39081956 PMCID: PMC11287074 DOI: 10.3389/fphar.2024.1455643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024] Open
Affiliation(s)
| | - Yuichi Sugiyama
- iHuman Institute, Shanghai Tech University, Shanghai, China
- Laboratory of Quantitative System Pharmacokinetics, Pharmacodynamics, Josai International University (JIU), Tokyo, Japan
| | - Esther van Duijn
- The Netherlands Organization for Applied Scientific Research, The Hague, Netherlands
| |
Collapse
|
3
|
Ioannidis I, Lefkaritis G, Georgiades SN, Pashalidis I, Kontoghiorghes GJ. Towards Clinical Development of Scandium Radioisotope Complexes for Use in Nuclear Medicine: Encouraging Prospects with the Chelator 1,4,7,10-Tetraazacyclododecane-1,4,7,10-tetraacetic Acid (DOTA) and Its Analogues. Int J Mol Sci 2024; 25:5954. [PMID: 38892142 PMCID: PMC11173192 DOI: 10.3390/ijms25115954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/15/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
Scandium (Sc) isotopes have recently attracted significant attention in the search for new radionuclides with potential uses in personalized medicine, especially in the treatment of specific cancer patient categories. In particular, Sc-43 and Sc-44, as positron emitters with a satisfactory half-life (3.9 and 4.0 h, respectively), are ideal for cancer diagnosis via Positron Emission Tomography (PET). On the other hand, Sc-47, as an emitter of beta particles and low gamma radiation, may be used as a therapeutic radionuclide, which also allows Single-Photon Emission Computed Tomography (SPECT) imaging. As these scandium isotopes follow the same biological pathway and chemical reactivity, they appear to fit perfectly into the "theranostic pair" concept. A step-by-step description, initiating from the moment of scandium isotope production and leading up to their preclinical and clinical trial applications, is presented. Recent developments related to the nuclear reactions selected and employed to produce the radionuclides Sc-43, Sc-44, and Sc-47, the chemical processing of these isotopes and the main target recovery methods are also included. Furthermore, the radiolabeling of the leading chelator, 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA), and its structural analogues with scandium is also discussed and the advantages and disadvantages of scandium complexation are evaluated. Finally, a review of the preclinical studies and clinical trials involving scandium, as well as future challenges for its clinical uses and applications, are presented.
Collapse
Affiliation(s)
- Ioannis Ioannidis
- Department of Chemistry, University of Cyprus, 2109 Nicosia, Cyprus; (I.I.); (G.L.); (S.N.G.); (I.P.)
| | - George Lefkaritis
- Department of Chemistry, University of Cyprus, 2109 Nicosia, Cyprus; (I.I.); (G.L.); (S.N.G.); (I.P.)
| | - Savvas N. Georgiades
- Department of Chemistry, University of Cyprus, 2109 Nicosia, Cyprus; (I.I.); (G.L.); (S.N.G.); (I.P.)
| | - Ioannis Pashalidis
- Department of Chemistry, University of Cyprus, 2109 Nicosia, Cyprus; (I.I.); (G.L.); (S.N.G.); (I.P.)
| | - George J. Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, 3021 Limassol, Cyprus
| |
Collapse
|
4
|
Hu C, Yang W. Alternatives to animal models to study bacterial infections. Folia Microbiol (Praha) 2023; 68:703-739. [PMID: 37632640 DOI: 10.1007/s12223-023-01084-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 08/02/2023] [Indexed: 08/28/2023]
Abstract
Animal testing has made a significant and unequalled contribution to important discoveries and advancements in the fields of research, medicine, vaccine development, and drug discovery. Each year, millions of animals are sacrificed for various experiments, and this is an ongoing process. However, the debate on the ethical and sensible usage of animals in in vivo experimentation is equally important. The need to explore and adopt newer alternatives to animals so as to comply with the goal of reduce, refine, and replace needs attention. Besides the ever-increasing debate on ethical issues, animal research has additional drawbacks (need of trained labour, requirement of breeding area, lengthy protocols, high expenses, transport barriers, difficulty to extrapolate data from animals to humans, etc.). With this scenario, the present review has been framed to give a comprehensive insight into the possible alternative options worth exploring in this direction especially targeting replacements for animal models of bacterial infections. There have been some excellent reviews discussing on the alternate methods for replacing and reducing animals in drug research. However, reviews that discuss the replacements in the field of medical bacteriology with emphasis on animal bacterial infection models are purely limited. The present review discusses on the use of (a) non-mammalian models and (b) alternative systems such as microfluidic chip-based models and microdosing aiming to give a detailed insight into the prospects of these alternative platforms to reduce the number of animals being used in infection studies. This would enlighten the scientific community working in this direction to be well acquainted with the available new approaches and alternatives so that the 3R strategy can be successfully implemented in the field of antibacterial drug research and testing.
Collapse
Affiliation(s)
- Chengming Hu
- Queen Mary College, Nanchang University, Nanchang, China
| | - Wenlong Yang
- Department of Infectious Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
5
|
Bhattacharya C, Sandinge AS, Bragg RA, Heijer M, Yan J, Andersson LC, Jurva U, Pelay-Gimeno M, Vaes WHJ, de Ligt RAF, Gränfors M, Amilon C, Lindstedt EL, Menakuru SR, Garkaviy P, Weidolf L, Gopaul VS. Application of Accelerator Mass Spectrometry to Characterize the Mass Balance Recovery and Disposition of AZD4831, a Novel Myeloperoxidase Inhibitor, following Administration of an Oral Radiolabeled Microtracer Dose in Humans. Drug Metab Dispos 2023; 51:451-463. [PMID: 36639243 DOI: 10.1124/dmd.122.001100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/15/2022] [Accepted: 12/15/2022] [Indexed: 01/15/2023] Open
Abstract
This study evaluated the mass balance and disposition of AZD4831, a novel myeloperoxidase inhibitor, in six healthy participants using a 14C-labeled microtracer coupled with analysis by accelerator mass spectrometry (AMS). A single oral dose of 10 mg 14C-AZD4831 (14.8 kBq) was administered as a solution, and 14C levels were quantified by AMS in blood, urine, and feces over 336 hours postdose. AZD4831 was rapidly absorbed, and AZD4831 plasma concentrations declined in a biphasic manner, with a long half-life of 52 hours. AZD4831 was eliminated via metabolism and renal excretion. An N-carbamoyl glucuronide metabolite of AZD4831 (M7), formed primarily via UGT1A1, was the predominant circulating metabolite. Presumably, M7 contributed to the long half-life of AZD4831 via biliary elimination and hydrolysis/enterohepatic recirculation of AZD4831. On average, ∼84% of administered 14C-AZD4831 was recovered by 336 hours postdose (urine, 51.2%; feces, 32.4%). Between 32%-44% of the dose was excreted as unchanged AZD4831 in urine, indicating renal elimination as the major excretory route. Only 9.7% of overall fecal recovery was recorded in the first 48 hours, with the remainder excreted over 48%-336 hours, suggesting that most fecal recovery was due to biliary elimination. Furthermore, only 6% of unchanged AZD4831 was recovered in feces. Overall, the fraction of the administered AZD4831 dose absorbed was high. 14C-AZD4831 was well tolerated. These findings contribute to increasing evidence that human absorption, distribution, metabolism, and excretion studies can be performed with acceptable mass balance recovery at therapeutically relevant doses and low radiolabel-specific activity using an AMS-14C microtracer approach. SIGNIFICANCE STATEMENT: In this study, the human absorption, distribution, metabolism, and excretion (hADME) of the novel myeloperoxidase inhibitor AZD4831 was assessed following oral administration. This included investigation of the disposition of M7, the N-carbamoyl glucuronide metabolite. Resolution of challenges highlighted in this study contributes to increasing evidence that hADME objectives can be achieved in a single study for compounds with therapeutically relevant doses and low radiolabel-specific activity by using an AMS-14C microtracer approach, thus reducing the need for preclinical radiolabeled studies.
Collapse
Affiliation(s)
- Chandrali Bhattacharya
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland (C.B.); DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism (A.-S.S., J.Y., U.J., L.C.A., V.S.G.); Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences (M.H.); and Early Product Development, Pharmaceutical Sciences (M.G.), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Early Chemical Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (R.A.B.); TNO, Leiden, The Netherlands (M.P.-G., W.H.J.V., R.A.F.d.L.); Quotient Sciences, Nottingham, United Kingdom (S.R.M.); Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (P.G.); and Formerly BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (L.W., C.A., E.-L.L.)
| | - Ann-Sofie Sandinge
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland (C.B.); DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism (A.-S.S., J.Y., U.J., L.C.A., V.S.G.); Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences (M.H.); and Early Product Development, Pharmaceutical Sciences (M.G.), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Early Chemical Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (R.A.B.); TNO, Leiden, The Netherlands (M.P.-G., W.H.J.V., R.A.F.d.L.); Quotient Sciences, Nottingham, United Kingdom (S.R.M.); Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (P.G.); and Formerly BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (L.W., C.A., E.-L.L.)
| | - Ryan A Bragg
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland (C.B.); DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism (A.-S.S., J.Y., U.J., L.C.A., V.S.G.); Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences (M.H.); and Early Product Development, Pharmaceutical Sciences (M.G.), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Early Chemical Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (R.A.B.); TNO, Leiden, The Netherlands (M.P.-G., W.H.J.V., R.A.F.d.L.); Quotient Sciences, Nottingham, United Kingdom (S.R.M.); Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (P.G.); and Formerly BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (L.W., C.A., E.-L.L.)
| | - Maria Heijer
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland (C.B.); DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism (A.-S.S., J.Y., U.J., L.C.A., V.S.G.); Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences (M.H.); and Early Product Development, Pharmaceutical Sciences (M.G.), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Early Chemical Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (R.A.B.); TNO, Leiden, The Netherlands (M.P.-G., W.H.J.V., R.A.F.d.L.); Quotient Sciences, Nottingham, United Kingdom (S.R.M.); Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (P.G.); and Formerly BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (L.W., C.A., E.-L.L.)
| | - Jingjing Yan
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland (C.B.); DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism (A.-S.S., J.Y., U.J., L.C.A., V.S.G.); Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences (M.H.); and Early Product Development, Pharmaceutical Sciences (M.G.), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Early Chemical Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (R.A.B.); TNO, Leiden, The Netherlands (M.P.-G., W.H.J.V., R.A.F.d.L.); Quotient Sciences, Nottingham, United Kingdom (S.R.M.); Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (P.G.); and Formerly BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (L.W., C.A., E.-L.L.)
| | - Linda C Andersson
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland (C.B.); DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism (A.-S.S., J.Y., U.J., L.C.A., V.S.G.); Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences (M.H.); and Early Product Development, Pharmaceutical Sciences (M.G.), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Early Chemical Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (R.A.B.); TNO, Leiden, The Netherlands (M.P.-G., W.H.J.V., R.A.F.d.L.); Quotient Sciences, Nottingham, United Kingdom (S.R.M.); Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (P.G.); and Formerly BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (L.W., C.A., E.-L.L.)
| | - Ulrik Jurva
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland (C.B.); DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism (A.-S.S., J.Y., U.J., L.C.A., V.S.G.); Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences (M.H.); and Early Product Development, Pharmaceutical Sciences (M.G.), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Early Chemical Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (R.A.B.); TNO, Leiden, The Netherlands (M.P.-G., W.H.J.V., R.A.F.d.L.); Quotient Sciences, Nottingham, United Kingdom (S.R.M.); Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (P.G.); and Formerly BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (L.W., C.A., E.-L.L.)
| | - Marta Pelay-Gimeno
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland (C.B.); DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism (A.-S.S., J.Y., U.J., L.C.A., V.S.G.); Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences (M.H.); and Early Product Development, Pharmaceutical Sciences (M.G.), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Early Chemical Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (R.A.B.); TNO, Leiden, The Netherlands (M.P.-G., W.H.J.V., R.A.F.d.L.); Quotient Sciences, Nottingham, United Kingdom (S.R.M.); Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (P.G.); and Formerly BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (L.W., C.A., E.-L.L.)
| | - Wouter H J Vaes
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland (C.B.); DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism (A.-S.S., J.Y., U.J., L.C.A., V.S.G.); Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences (M.H.); and Early Product Development, Pharmaceutical Sciences (M.G.), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Early Chemical Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (R.A.B.); TNO, Leiden, The Netherlands (M.P.-G., W.H.J.V., R.A.F.d.L.); Quotient Sciences, Nottingham, United Kingdom (S.R.M.); Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (P.G.); and Formerly BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (L.W., C.A., E.-L.L.)
| | - Rianne A F de Ligt
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland (C.B.); DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism (A.-S.S., J.Y., U.J., L.C.A., V.S.G.); Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences (M.H.); and Early Product Development, Pharmaceutical Sciences (M.G.), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Early Chemical Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (R.A.B.); TNO, Leiden, The Netherlands (M.P.-G., W.H.J.V., R.A.F.d.L.); Quotient Sciences, Nottingham, United Kingdom (S.R.M.); Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (P.G.); and Formerly BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (L.W., C.A., E.-L.L.)
| | - Malin Gränfors
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland (C.B.); DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism (A.-S.S., J.Y., U.J., L.C.A., V.S.G.); Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences (M.H.); and Early Product Development, Pharmaceutical Sciences (M.G.), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Early Chemical Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (R.A.B.); TNO, Leiden, The Netherlands (M.P.-G., W.H.J.V., R.A.F.d.L.); Quotient Sciences, Nottingham, United Kingdom (S.R.M.); Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (P.G.); and Formerly BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (L.W., C.A., E.-L.L.)
| | - Carl Amilon
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland (C.B.); DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism (A.-S.S., J.Y., U.J., L.C.A., V.S.G.); Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences (M.H.); and Early Product Development, Pharmaceutical Sciences (M.G.), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Early Chemical Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (R.A.B.); TNO, Leiden, The Netherlands (M.P.-G., W.H.J.V., R.A.F.d.L.); Quotient Sciences, Nottingham, United Kingdom (S.R.M.); Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (P.G.); and Formerly BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (L.W., C.A., E.-L.L.)
| | - Eva-Lotte Lindstedt
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland (C.B.); DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism (A.-S.S., J.Y., U.J., L.C.A., V.S.G.); Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences (M.H.); and Early Product Development, Pharmaceutical Sciences (M.G.), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Early Chemical Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (R.A.B.); TNO, Leiden, The Netherlands (M.P.-G., W.H.J.V., R.A.F.d.L.); Quotient Sciences, Nottingham, United Kingdom (S.R.M.); Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (P.G.); and Formerly BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (L.W., C.A., E.-L.L.)
| | - Somasekhara R Menakuru
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland (C.B.); DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism (A.-S.S., J.Y., U.J., L.C.A., V.S.G.); Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences (M.H.); and Early Product Development, Pharmaceutical Sciences (M.G.), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Early Chemical Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (R.A.B.); TNO, Leiden, The Netherlands (M.P.-G., W.H.J.V., R.A.F.d.L.); Quotient Sciences, Nottingham, United Kingdom (S.R.M.); Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (P.G.); and Formerly BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (L.W., C.A., E.-L.L.)
| | - Pavlo Garkaviy
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland (C.B.); DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism (A.-S.S., J.Y., U.J., L.C.A., V.S.G.); Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences (M.H.); and Early Product Development, Pharmaceutical Sciences (M.G.), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Early Chemical Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (R.A.B.); TNO, Leiden, The Netherlands (M.P.-G., W.H.J.V., R.A.F.d.L.); Quotient Sciences, Nottingham, United Kingdom (S.R.M.); Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (P.G.); and Formerly BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (L.W., C.A., E.-L.L.)
| | - Lars Weidolf
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland (C.B.); DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism (A.-S.S., J.Y., U.J., L.C.A., V.S.G.); Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences (M.H.); and Early Product Development, Pharmaceutical Sciences (M.G.), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Early Chemical Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (R.A.B.); TNO, Leiden, The Netherlands (M.P.-G., W.H.J.V., R.A.F.d.L.); Quotient Sciences, Nottingham, United Kingdom (S.R.M.); Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (P.G.); and Formerly BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (L.W., C.A., E.-L.L.)
| | - V Sashi Gopaul
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland (C.B.); DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism (A.-S.S., J.Y., U.J., L.C.A., V.S.G.); Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences (M.H.); and Early Product Development, Pharmaceutical Sciences (M.G.), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Early Chemical Development, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom (R.A.B.); TNO, Leiden, The Netherlands (M.P.-G., W.H.J.V., R.A.F.d.L.); Quotient Sciences, Nottingham, United Kingdom (S.R.M.); Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (P.G.); and Formerly BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (L.W., C.A., E.-L.L.)
| |
Collapse
|
6
|
Carmichael O. The Role of fMRI in Drug Development: An Update. ADVANCES IN NEUROBIOLOGY 2023; 30:299-333. [PMID: 36928856 DOI: 10.1007/978-3-031-21054-9_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Functional magnetic resonance imaging (fMRI) of the brain is a technology that holds great potential for increasing the efficiency of drug development for the central nervous system (CNS). In preclinical studies and both early- and late-phase human trials, fMRI has the potential to improve cross-species translation of drug effects, help to de-risk compounds early in development, and contribute to the portfolio of evidence for a compound's efficacy and mechanism of action. However, to date, the utilization of fMRI in the CNS drug development process has been limited. The purpose of this chapter is to explore this mismatch between potential and utilization. This chapter provides introductory material related to fMRI and drug development, describes what is required of fMRI measurements for them to be useful in a drug development setting, lists current capabilities of fMRI in this setting and challenges faced in its utilization, and ends with directions for future development of capabilities in this arena. This chapter is the 5-year update of material from a previously published workshop summary (Carmichael et al., Drug DiscovToday 23(2):333-348, 2018).
Collapse
Affiliation(s)
- Owen Carmichael
- Pennington Biomedical Research Center, Baton Rouge, LA, USA.
| |
Collapse
|
7
|
de las Heras B, Bouyoucef‐Cherchalli D, Reeve L, Reichl A, Mandarino D, Flach S, Vidal L, van Brummelen EMJ, Steeghs N. Healthy volunteers in first-in-human oncology drug development for small molecules. Br J Clin Pharmacol 2022; 88:1773-1784. [PMID: 34558113 PMCID: PMC10234445 DOI: 10.1111/bcp.15092] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/03/2021] [Accepted: 09/14/2021] [Indexed: 12/20/2022] Open
Abstract
This review provides tools to consider the inclusion of healthy volunteers (HVs) in first-in-human (FIH) oncology clinical trials with small molecules, including targeted and immunomodulatory agents, a strategy that was not envisioned with classic chemotherapy. To enable an FIH oncology trial in HVs compared to cancer patients (CPs), a robust nonclinical package must be generated, which includes toxicokinetic and pharmacokinetic studies, as well as more extensive safety pharmacology, toxicology and genotoxicity studies. This strategy could provide an early clinical characterization of the pharmacokinetic parameters and clinical safety profile in the absence of comorbidities and concomitant medication. It also avoids the ethical issue of administrating subtherapeutic doses to CPs, and could potentially help to accelerate the timelines of clinical drug development for patient care. That being said, stakeholders involved in these studies need to proceed with caution, fully understand the regulatory guidance and thoroughly evaluate the benefits and risks. This paper serves to address the regulatory guidance and other considerations needed when using healthy volunteers in early oncology trials.
Collapse
Affiliation(s)
- Begoña de las Heras
- Labcorp Drug Development Inc., headquarters in BurlingtonNorth CarolinaUSA
- Madrid Medical Doctors AssociationMadridSpain
| | | | - Lesley Reeve
- Labcorp Drug Development Inc., headquarters in BurlingtonNorth CarolinaUSA
| | - Andreas Reichl
- Labcorp Drug Development Inc., headquarters in BurlingtonNorth CarolinaUSA
| | - Debra Mandarino
- Labcorp Drug Development Inc., headquarters in BurlingtonNorth CarolinaUSA
| | - Stephen Flach
- Labcorp Drug Development Inc., headquarters in BurlingtonNorth CarolinaUSA
| | - Laura Vidal
- Labcorp Drug Development Inc., headquarters in BurlingtonNorth CarolinaUSA
| | | | | |
Collapse
|
8
|
Pant AB. The Implementation of the Three Rs in Regulatory Toxicity and Biosafety Assessment: The Indian Perspective. Altern Lab Anim 2021; 48:234-251. [PMID: 33523713 DOI: 10.1177/0261192920986811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Animal models have long served as a basis for scientific experimentation, biomedical research, drug development and testing, disease modelling and toxicity studies, as they are widely thought to provide meaningful, human-relevant predictions. However, many of these systems are resource intensive and time-consuming, have low predictive value and are associated with great social and ethical dilemmas. Often drugs appear to be effective and safe in these classical animal models, but later prove to be ineffective and/or unsafe in clinical trials. These issues have paved the way for a paradigm shift from the use of in vivo approaches, toward the 'science of alternatives'. This has fuelled several research and regulatory initiatives, including the ban on the testing of cosmetics on animals. The new paradigm has been shifted toward increasing the relevance of the models for human predictivity and translational efficacy, and this has resulted in the recent development of many new methodologies, from 3-D bio-organoids to bioengineered 'human-on-a-chip' models. These improvements have the potential to significantly advance medical research globally. This paper offers a stance on the existing strategies and practices that utilise alternatives to animals, and outlines progress on the incorporation of these models into basic and applied research and education, specifically in India. It also seeks to provide a strategic roadmap to streamline the future directions for the country's policy changes and investments. This strategic roadmap could be a useful resource to guide research institutions, industries, regulatory agencies, contract research organisations and other stakeholders in transitioning toward modern approaches to safety and risk assessment that could replace or reduce the use of animals without compromising the safety of humans or the environment.
Collapse
Affiliation(s)
- Aditya B Pant
- System Toxicology and Health Risk Assessment Group, 538266Council of Scientific and Industrial Research-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, Uttar Pradesh, India
| |
Collapse
|
9
|
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
|
10
|
Bhogal N, Combes R. TGN1412: Time to Change the Paradigm for the Testing of New Pharmaceuticals. Altern Lab Anim 2019; 34:225-39. [PMID: 16704293 DOI: 10.1177/026119290603400204] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Clinical studies in human volunteers are an essential part of drug development. These studies are designed to account for possible differences between the effects of pharmaceutical products in pre-clinical studies and in humans. However, the tragic outcome of the recent Phase 1 clinical trial on TGN1412 casts considerable doubt over the relevance of this traditional drug development paradigm to the testing of therapeutic agents for human use. The role of alternatives to animal testing is considered, and a series of recommendations are made, which could ensure that clinical trials are well informed and based on the most relevant scientific information.
Collapse
Affiliation(s)
- Nirmala Bhogal
- FRAME, 96-98 North Sherwood Street, Nottingham, NG1 4EE, UK.
| | | |
Collapse
|
11
|
Bhogal N, Combes R. The UK Food Standards Agency Draft Report on Variability and Uncertainty in Toxicology: A Response by FRAME. Altern Lab Anim 2019; 34:539-44. [PMID: 17121477 DOI: 10.1177/026119290603400501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
At the request of the Food Standards Agency, the Committee on Toxicity of Chemicals in Food, Consumer Products and the Environment (COT) established a Working Group on Variation and Uncertainty in Toxicology (WG VUT). In April 2006, the WG VUT produced a draft report for public consultation. FRAME made a submission in response to this consultation in July 2006. We commend the WG VUT for its comprehensive account of many of the problems associated with risk assessment, and for making recommendations about the problems that need to be addressed. We were particularly encouraged by the WG VUTs recognition of the need for guidelines on how toxicological studies should be conducted and data analysed. However, we believe that the report has not achieved all of its objectives. It does not adequately consider how modern technologies, experimental design, statistical analysis and species extrapolation can be used in practice to address variability and uncertainty. There is a disproportionate focus on the sources of variability and uncertainty in human data, with relatively little consideration of how variation and uncertainty due to animal tests can be addressed. Furthermore, it is clear that, until the advantages and limitations of all toxicological methods are fully appraised and testing strategies and guidelines are agreed, the scope for improving the existing approaches to risk assessment will be severely limited. Hence, the use of alternative methods for hazard identification and characterisation merit more consideration than they were given in the draft report.
Collapse
Affiliation(s)
- Nirmala Bhogal
- FRAME, 96-98 North Sherwood Street, Nottingham, NG1 4EE, UK.
| | | |
Collapse
|
12
|
Carmichael O, Schwarz AJ, Chatham CH, Scott D, Turner JA, Upadhyay J, Coimbra A, Goodman JA, Baumgartner R, English BA, Apolzan JW, Shankapal P, Hawkins KR. The role of fMRI in drug development. Drug Discov Today 2018; 23:333-348. [PMID: 29154758 PMCID: PMC5931333 DOI: 10.1016/j.drudis.2017.11.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/19/2017] [Accepted: 11/13/2017] [Indexed: 12/17/2022]
Abstract
Functional magnetic resonance imaging (fMRI) has been known for over a decade to have the potential to greatly enhance the process of developing novel therapeutic drugs for prevalent health conditions. However, the use of fMRI in drug development continues to be relatively limited because of a variety of technical, biological, and strategic barriers that continue to limit progress. Here, we briefly review the roles that fMRI can have in the drug development process and the requirements it must meet to be useful in this setting. We then provide an update on our current understanding of the strengths and limitations of fMRI as a tool for drug developers and recommend activities to enhance its utility.
Collapse
Affiliation(s)
- Owen Carmichael
- Pennington Biomedical Research Center, Baton Rouge, LA, USA.
| | | | - Christopher H Chatham
- Translational Medicine Neuroscience and Biomarkers, Roche Innovation Center, Basel, Switzerland
| | | | - Jessica A Turner
- Psychology Department & Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | | | | | | | - Richard Baumgartner
- Biostatistics and Research Decision Sciences (BARDS), Merck & Co., Inc., Kenilworth, NJ, USA
| | | | - John W Apolzan
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | | | | |
Collapse
|
13
|
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
|
14
|
Flach S, Croft M, Ding J, Budhram R, Pankratz T, Pennick M, Scarfe G, Troy S, Getsy J. Pharmacokinetics, absorption, and excretion of radiolabeled revexepride: a Phase I clinical trial using a microtracer and accelerator mass spectrometry-based approach. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:3125-3132. [PMID: 27729771 PMCID: PMC5045912 DOI: 10.2147/dddt.s107843] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Purpose Gastroesophageal reflux disease involves the reflux of gastric and/or duodenal content into the esophagus. Prokinetic therapies, such as the selective 5-hydroxytryptamine receptor 4 agonist revexepride, may aid gastric emptying. This Phase I study evaluated the pharmacokinetics and excretion pathways of [14C]revexepride in healthy individuals using a microtracer approach with accelerator mass spectrometry. Participants and methods Six healthy men received a single oral dose of 2 mg [14C]revexepride containing ~200 nCi of radioactivity; blood, urine, and fecal samples were collected over a 10-day period. Results Almost 100% of 14C was recovered: 38.2%±10.3% (mean ± standard deviation) was recovered in urine, and 57.3%±0.4% was recovered in feces. Blood cell uptake was low, based on the blood plasma total radioactivity ratio of 0.8. The mean revexepride renal clearance was 8.6 L/h, which was slightly higher than the typical glomerular filtration rate in healthy individuals. Time to reach maximal concentration was 1.75±1.17 hours (mean ± standard deviation). No safety signals were identified. Conclusion This study demonstrated that revexepride had rapid and moderate-to-good oral absorption. Excretion of radioactivity was completed with significant amounts in feces and urine. Renal clearance slightly exceeded the typical glomerular filtration rate, suggesting the involvement of active transportation in the renal tubules.
Collapse
Affiliation(s)
| | | | - Jie Ding
- Covance Laboratories Inc., Madison, WI, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Flach S, Scarfe G, Dragone J, Ding J, Seymour M, Pennick M, Pankratz T, Troy S, Getsy J. A Phase I Study to Investigate the Absorption, Pharmacokinetics, and Excretion of [(14)C]Prucalopride After a Single Oral Dose in Healthy Volunteers. Clin Ther 2016; 38:2106-15. [PMID: 27614912 DOI: 10.1016/j.clinthera.2016.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 08/05/2016] [Accepted: 08/05/2016] [Indexed: 12/20/2022]
Abstract
PURPOSE Chronic constipation is a prevalent gastrointestinal disorder globally. It is often treated with medications such as laxatives. Newer therapies to improve gastric motility include the selective 5-hydroxytryptamine receptor-4 agonist prucalopride, which is licensed for the treatment of chronic constipation in adults. The aim of this study was to investigate the pharmacokinetic properties and excretion of prucalopride in healthy individuals, using a microtracer approach with (14)C radioactivity detection using liquid scintillation counting and accelerator mass spectrometry. METHODS This was a single-period, open-label, nonrandomized absorption, metabolism, and excretion study of [(14)C]prucalopride. Participants were 6 healthy men aged 18 to 50 years. After screening, participants were administered a single dose of [(14)C]prucalopride succinate 2 mg (~200 nCi). Postadministration, urine, feces, and blood samples were collected over a 10-day period. Safety and adverse event data were also collected. FINDINGS Almost 100% of the administered dose of radioactivity was recovered, with a mean (SD) of 84.2% (8.88%) recovered in urine and 13.3% (1.73%) recovered in feces. The mean blood-to-plasma concentration ratio of 1.9 indicated uptake of prucalopride into blood cells. The renal clearance of prucalopride was 17.0 (2.5) L/h, which is higher than the glomerular filtration rate in healthy individuals, suggesting active renal transport of prucalopride. Prucalopride was well tolerated, with no serious adverse events reported. IMPLICATIONS Prucalopride was well absorbed and excreted mainly by the kidneys, including both passive and active transporter mechanisms. Quantitative recovery of the radioactive dose was achieved. Consistent with previous studies, prucalopride was generally well tolerated. ClinicalTrials.gov identifier: NCT01807000.
Collapse
Affiliation(s)
| | - Graeme Scarfe
- Shire Pharmaceuticals Ltd, Chineham, Basingstoke, Hampshire, United Kingdom
| | | | - Jie Ding
- Covance Laboratories, Madison, Wisconsin
| | | | - Mike Pennick
- Shire Pharmaceuticals Ltd, Chineham, Basingstoke, Hampshire, United Kingdom
| | | | - Steven Troy
- Shire Pharmaceuticals Inc, Lexington, Massachusetts
| | - Jay Getsy
- Shire Pharmaceuticals Inc, Lexington, Massachusetts
| |
Collapse
|
16
|
Romero L, Vela JM. Alternative Models in Drug Discovery and Development Part II:In VivoNonmammalian and Exploratory/Experimental Human Models. ACTA ACUST UNITED AC 2014. [DOI: 10.1002/9783527679348.ch03] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
17
|
Perloff M, Steele VE. Early-phase development of cancer prevention agents: challenges and opportunities. Cancer Prev Res (Phila) 2013; 6:379-83. [PMID: 23466485 PMCID: PMC3657502 DOI: 10.1158/1940-6207.capr-12-0463] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Chemoprevention is the administration of agents (drugs, biologics, dietary supplements, or nutrients) to reduce the risk of developing cancer or prevent the recurrence of cancer. The National Cancer Institute, Division of Cancer Prevention (NCI, DCP), is a major sponsor of cancer preventive preclinical and clinical research. As such, it has developed a comprehensive drug development program specifically designed to meet the requirements needed for cancer preventive drugs to achieve initial regulatory approval. Clinical development of cancer prevention agents presents unique challenges that are not encountered with most cancer therapeutic agents. To meet these challenges, NCI, DCP has implemented new approaches and programs, including phase 0 clinical trial designs and microdose studies. In addition, the PREVENT Cancer Program was recently implemented by NCI, DCP to offer a formalized structure for moving drugs forward in the prevention pipeline using a continue/not continue decision process. Likewise, DCP has implemented a Clinical Trials Consortium to further develop these agents. These and other approaches will be discussed in this commentary.
Collapse
Affiliation(s)
- Marjorie Perloff
- Division of Cancer Prevention, National Cancer Institute, 6130 Executive Blvd. EPN 2112, Rockville, MD 20892, USA.
| | | |
Collapse
|
18
|
Ieiri I, Doi Y, Maeda K, Sasaki T, Kimura M, Hirota T, Chiyoda T, Miyagawa M, Irie S, Iwasaki K, Sugiyama Y. Microdosing Clinical Study: Pharmacokinetic, Pharmacogenomic (SLCO2B1), and Interaction (Grapefruit Juice) Profiles of Celiprolol Following the Oral Microdose and Therapeutic Dose. J Clin Pharmacol 2013; 52:1078-89. [DOI: 10.1177/0091270011408612] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
19
|
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
|
20
|
Balls M, Combes RD, Bhogal N. The use of integrated and intelligent testing strategies in the prediction of toxic hazard and in risk assessment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 745:221-53. [PMID: 22437821 PMCID: PMC7122993 DOI: 10.1007/978-1-4614-3055-1_13] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
There is increasing concern that insurmountable differences between humans and laboratory animals limit the relevance and reliability for hazard identification and risk assessment purposes of animal data produced by traditional toxicity test procedures. A way forward is offered by the emerging new technologies, which can be directly applied to human material or even to human beings themselves. This promises to revolutionise the evaluation of the safety of chemicals and chemical products of various kinds and, in particular, pharmaceuticals. The available and developing technologies are summarised and it is emphasised that they will need to be used selectively, in integrated and intelligent testing strategies, which, in addition to being scientifically sound, must be manageable and affordable. Examples are given of proposed testing strategies for general chemicals, cosmetic ingredients, candidate pharmaceuticals, inhaled substances, nanoparticles and neurotoxicity.
Collapse
|
21
|
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]
|
22
|
Bae SK, Shon JH. Microdosing studies using accelerated mass spectrometry as exploratory investigational new drug trials. Arch Pharm Res 2011; 34:1789-98. [DOI: 10.1007/s12272-011-1102-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 08/24/2011] [Indexed: 11/24/2022]
|
23
|
Prediction of Nonlinear Intestinal Absorption of CYP3A4 and P-Glycoprotein Substrates from their In Vitro Km Values. Pharm Res 2011; 29:651-68. [DOI: 10.1007/s11095-011-0579-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Accepted: 08/26/2011] [Indexed: 02/05/2023]
|
24
|
Combes RD, Balls M. Integrated testing strategies for toxicity employing new and existing technologies. Altern Lab Anim 2011; 39:213-25. [PMID: 21777036 DOI: 10.1177/026119291103900303] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have developed individual, integrated testing strategies (ITS) for predicting the toxicity of general chemicals, cosmetics, pharmaceuticals, inhaled chemicals, and nanoparticles. These ITS are based on published schemes developed previously for the risk assessment of chemicals to fulfil the requirements of REACH, which have been updated to take account of the latest developments in advanced in chemico modelling and in vitro technologies. In addition, we propose an ITS for neurotoxicity, based on the same principles, for incorporation in the other ITS. The technologies are deployed in a step-wise manner, as a basis for decision-tree approaches, incorporating weight-of-evidence stages. This means that testing can be stopped at the point where a risk assessment and/or classification can be performed, with labelling in accordance with the requirements of the regulatory authority concerned, rather than following a checklist approach to hazard identification. In addition, the strategies are intelligent, in that they are based on the fundamental premise that there is no hazard in the absence of exposure - which is why pharmacokinetic modelling plays a key role in each ITS. The new technologies include the use of complex, three-dimensional human cell tissue culture systems with in vivo-like structural, physiological and biochemical features, as well as dosing conditions. In this way, problems of inter-species extrapolation and in vitro/in vivo extrapolation are minimised. This is reflected in the ITS placing more emphasis on the use of volunteers at the whole organism testing stage, rather than on existing animal testing, which is the current situation.
Collapse
Affiliation(s)
- Robert D Combes
- c/o Fund for the Replacement of Animals in Medical Experiments (FRAME), Nottingham, UK.
| | | |
Collapse
|
25
|
Takashima T, Hashizume Y, Katayama Y, Murai M, Wada Y, Maeda K, Sugiyama Y, Watanabe Y. The Involvement of Organic Anion Transporting Polypeptide in the Hepatic Uptake of Telmisartan in Rats: PET Studies with [11C]Telmisartan. Mol Pharm 2011; 8:1789-98. [DOI: 10.1021/mp200160t] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Tadayuki Takashima
- RIKEN Center for Molecular Imaging Science, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yoshinobu Hashizume
- RIKEN Center for Molecular Imaging Science, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yumiko Katayama
- RIKEN Center for Molecular Imaging Science, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Machiko Murai
- RIKEN Center for Molecular Imaging Science, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yasuhiro Wada
- RIKEN Center for Molecular Imaging Science, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yuichi Sugiyama
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yasuyoshi Watanabe
- RIKEN Center for Molecular Imaging Science, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| |
Collapse
|
26
|
Pharmacokinetic and pharmacogenomic profiles of telmisartan after the oral microdose and therapeutic dose. Pharmacogenet Genomics 2011; 21:495-505. [DOI: 10.1097/fpc.0b013e3283489ce2] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
27
|
Holmes AM, Solari R, Holgate ST. Animal models of asthma: value, limitations and opportunities for alternative approaches. Drug Discov Today 2011; 16:659-70. [PMID: 21723955 DOI: 10.1016/j.drudis.2011.05.014] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 04/15/2011] [Accepted: 05/31/2011] [Indexed: 11/15/2022]
Abstract
Asthma remains an area of considerable unmet medical need. Few new drugs have made it to the clinic during the past 50 years, with many that perform well in preclinical animal models of asthma, failing in humans owing to lack of safety and efficacy. The failure to translate promising drug candidates from animal models to humans has led to questions about the utility of in vivo studies and to demand for more predictive models and tools based on the latest technologies. Following a workshop with experts from academia and the pharmaceutical industry, we suggest here a disease modelling framework designed to better understand human asthma, and accelerate the development of safe and efficacious new asthma drugs that go beyond symptomatic relief.
Collapse
Affiliation(s)
- Anthony M Holmes
- National Centre for the Replacement, Refinement and Reduction of Animals in Research, 20 Park Crescent, London, W1B 1AL, UK.
| | | | | |
Collapse
|
28
|
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
|
29
|
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: 2.1] [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
|
30
|
Bailey J. An assessment of the use of chimpanzees in hepatitis C research past, present and future: 1. Validity of the chimpanzee model. Altern Lab Anim 2011; 38:387-418. [PMID: 21105756 DOI: 10.1177/026119291003800501] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The USA is the only significant user of chimpanzees in biomedical research in the world, since many countries have banned or limited the practice due to substantial ethical, economic and scientific concerns. Advocates of chimpanzee use cite hepatitis C research as a major reason for its necessity and continuation, in spite of supporting evidence that is scant and often anecdotal. This paper examines the scientific and ethical issues surrounding chimpanzee hepatitis C research, and concludes that claims of the necessity of chimpanzees in historical and future hepatitis C research are exaggerated and unjustifiable, respectively. The chimpanzee model has several major scientific, ethical, economic and practical caveats. It has made a relatively negligible contribution to knowledge of, and tangible progress against, the hepatitis C virus compared to non-chimpanzee research, and must be considered scientifically redundant, given the array of alternative methods of inquiry now available. The continuation of chimpanzee use in hepatitis C research adversely affects scientific progress, as well as chimpanzees and humans in need of treatment. Unfounded claims of its necessity should not discourage changes in public policy regarding the use of chimpanzees in US laboratories.
Collapse
Affiliation(s)
- Jarrod Bailey
- New England Anti-Vivisection Society, Boston, MA 02108-5100, USA.
| |
Collapse
|
31
|
Stenström K, Sydoff M, Mattsson S. Microdosing for early biokinetic studies in humans. RADIATION PROTECTION DOSIMETRY 2010; 139:348-352. [PMID: 20167793 DOI: 10.1093/rpd/ncq029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Microdosing is a new concept in drug development that--if implemented in the pharmaceutical industry--would mean that new drugs can be tested earlier in humans than done today. The human microdosing concept--or 'Phase 0'--may offer improved candidate selection, reduced failure rates in the drug development line and a reduction in the use of laboratory animals in early drug development, factors which will help to speed up drug development and also reduce the costs. Microdosing utilises sub-pharmacological amounts of the substance to open opportunities for early studies in man. Three technologies are used for microdosing: accelerator mass spectrometry (AMS), positron emission tomography and liquid chromatography-tandem mass spectrometry. This paper focuses on the principle of AMS and discusses the current status of microdosing with AMS.
Collapse
Affiliation(s)
- K Stenström
- Department of Physics, Division of Nuclear Physics, Lund University, PO Box 118, SE-221 00 Lund, Sweden.
| | | | | |
Collapse
|
32
|
|
33
|
Hartung T. Lessons learned from alternative methods and their validation for a new toxicology in the 21st century. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2010; 13:277-90. [PMID: 20574902 DOI: 10.1080/10937404.2010.483945] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Europe and the United States further the development of new toxicological tools in different ways. While the replacement of animal tests has been promoted strongly in Europe over the last decades (following the 3Rs principles--reduce, replace, refine), in the United States the vision for a toxicology in the 21st century (Tox-21c), which was prompted by the National Research Council document only 3 years ago, dominates the discussion. In both cases, there is significant political support. However, while in Europe the horizontal animal welfare legislation from 1986 (which urges the use of 3Rs methods wherever possible) currently under revision and cosmetics and chemical legislation are the primary drivers, in the United States it is mainly federal agencies, most prominently the U.S. Environmental Protection Agency (EPA), that made the implementation of the NRC report their toxicity testing strategy only in 2009. This preempts such likely legislative measures as the reauthorization of the Toxic Substances Control Act (TSCA) in the United States. The European implementation is characterized by substantial broad funding programs to develop 3Rs methods and can be termed a "bottom-up" approach; in contrast, the Tox-21 program represents a "top-down" approach, where programmed research is carried out and commissioned. It is postulated that the two approaches are two sides of the same coin, and instruct and complement each other. However, more importantly, if brought together they can result in a Human Toxicology Project and a real revolution in regulatory toxicology.
Collapse
Affiliation(s)
- Thomas Hartung
- Environmental Health Sciences, Center for Alternatives to Animal Testing, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA.
| |
Collapse
|
34
|
Yamazaki A, Kumagai Y, Yamane N, Tozuka Z, Sugiyama Y, Fujita T, Yokota S, Maeda M. Microdose study of a P-glycoprotein substrate, fexofenadine, using a non-radioisotope-labelled drug and LC/MS/MS. J Clin Pharm Ther 2010; 35:169-75. [DOI: 10.1111/j.1365-2710.2009.01159.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
35
|
Positron emission tomography neuroimaging for a better understanding of the biology of ADHD. Neuropharmacology 2009; 57:601-7. [DOI: 10.1016/j.neuropharm.2009.08.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 07/29/2009] [Accepted: 08/04/2009] [Indexed: 11/20/2022]
|
36
|
Suzuki M, Sumi K, Koyama H, Siqin, Hosoya T, Takashima-Hirano M, Doi H. Pd0-Mediated Rapid Coupling between Methyl Iodide and Heteroarylstannanes: An Efficient and General Method for the Incorporation of a Positron-Emitting11C Radionuclide into Heteroaromatic Frameworks. Chemistry 2009; 15:12489-95. [DOI: 10.1002/chem.200901145] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
37
|
Bibliometric study of radiation application on microdose useful for new drug development. Ann Nucl Med 2009; 23:829-41. [DOI: 10.1007/s12149-009-0311-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Accepted: 09/18/2009] [Indexed: 10/20/2022]
|
38
|
Creton S, Billington R, Davies W, Dent MP, Hawksworth GM, Parry S, Travis KZ. Application of toxicokinetics to improve chemical risk assessment: implications for the use of animals. Regul Toxicol Pharmacol 2009; 55:291-9. [PMID: 19665509 DOI: 10.1016/j.yrtph.2009.08.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Revised: 07/31/2009] [Accepted: 08/03/2009] [Indexed: 11/30/2022]
Abstract
While toxicokinetics has become an integral part of pharmaceutical safety assessment over the last two decades, its use in the chemical industry is relatively new. However, it is recognised as a potentially important tool in human health risk assessment and recent initiatives have advocated greater application of toxicokinetics as part of an improved assessment strategy for crop protection chemicals that could offer greater efficiency, use fewer animals and provide better data for risk assessment purposes. To explore the potential scientific and animal welfare benefits of increased use of toxicokinetic data across the chemical industry, an international workshop was held in 2008. Experts from a wide range of chemical industry sectors, including industrial chemicals, agrochemicals and consumer products, participated in the meeting as well as representatives from relevant regulatory authorities. Pharmaceutical industry experts were also invited, in order to share experiences from the extensive use of toxicokinetics in drug development. Given that increased generation of toxicokinetic data could potentially result in an increased number of animals undergoing testing, technologies and strategies to reduce and refine animal use for this purpose were also considered. This paper outlines and expands upon the key themes that emerged from the workshop.
Collapse
Affiliation(s)
- Stuart Creton
- National Centre for the Replacement, Refinement and Reduction of Animals in Research, London W1B 1AL, UK.
| | | | | | | | | | | | | |
Collapse
|
39
|
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: 5.2] [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
|
40
|
Accelerator mass spectrometry analysis of background (14)C-concentrations in human blood: aiming at reference data for further microdosing studies. Ann Nucl Med 2009; 22:883-9. [PMID: 19142707 DOI: 10.1007/s12149-008-0200-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2007] [Accepted: 07/03/2008] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Phase 0 clinical studies, which are known as microdose trials, are expected to promote drug development and reduce development costs. The accelerator mass spectrometry (AMS) system is expected to play an important role in the microdosing tests, as it is a highly sensitive measurement system that can be used to determine the drug concentrations in these tests. Using the AMS system, we measured the background (14)C-concentration in human blood and evaluated the data for use as a reference in microdose studies that administer (14)C-labeled compounds in humans. METHODS Blood samples of five healthy Japanese volunteers (three men, two women, median age 40.4 +/- 9.8 years) were collected around the same time and just prior to when the subjects ate a meal (between 12:00 noon and 2:00 pm). Centrifugal separations of blood that was allowed to clot and the plasma were performed at 503 g for 2 min at 4 degrees C. Background (14)C-concentration for each of the samples was measured using the AMS system. The Institute of Accelerator Analysis, which is the first contract research organization in Japan that is capable of providing AMS analysis services for carbon dating and bioanalysis work, performed the AMS analysis. RESULTS The mean (14)C-concentration in blood was 1.613 +/- 0.125 dpm/ml (men 1.668 +/- 0.114 dpm/ml, women 1.514 +/- 0.076 dpm/ml), in clots 2.373 +/- 0.087 dpm/ml (men 2.381 +/- 0.101 dpm/ml, women 2.357 +/- 0.060 dpm/ ml), and in plasma 0.648 +/- 0.049 dpm/ml (men 0.647 +/- 0.059 dpm/ml, women 0.649 +/- 0.032 dpm/ml). The coefficient variation (CV) for blood was 7.8% (men 6.9%, women 5.0%), for clots 3.7% (men 4.3%, women 2.5%), and for plasma 7.6% (men 9.1%, women 4.9%). The (14)C-concentrations of the clot and blood were higher than those of plasma. The (14)C-concentrations in the blood and plasma were slightly different between individuals when compared with the values for the clot, although the differences were quite small, with a CV value less than 7.8%. CONCLUSIONS Even though the (14)C-concentration differed only slightly between individuals, (14)C-concentrations of the clot and blood were higher than those of the plasma. Therefore, the variation and difference of the background data for blood and plasma might be of use as a reference for microdosing test evaluations.
Collapse
|
41
|
Zimmer L. Can positron emission tomography facilitate paediatric drug development? Fundam Clin Pharmacol 2008; 22:595-8. [DOI: 10.1111/j.1472-8206.2008.00650.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
42
|
Langley C. Better Models of Human Pain. Altern Lab Anim 2008. [DOI: 10.1177/026119290803600506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Chris Langley
- ScienceSources Consultancy Hitchin Hertfordshire, UK
| |
Collapse
|
43
|
Lappin G, Stevens L. Biomedical accelerator mass spectrometry: recent applications in metabolism and pharmacokinetics. Expert Opin Drug Metab Toxicol 2008; 4:1021-33. [DOI: 10.1517/17425255.4.8.1021] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
44
|
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: 2.1] [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
|
45
|
Boddy AV, Sludden J, Griffin MJ, Garner C, Kendrick J, Mistry P, Dutreix C, Newell DR, O'Brien SG. Pharmacokinetic investigation of imatinib using accelerator mass spectrometry in patients with chronic myeloid leukemia. Clin Cancer Res 2007; 13:4164-9. [PMID: 17634544 DOI: 10.1158/1078-0432.ccr-06-2179] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To investigate the potential use of accelerator mass spectrometry (AMS) in the study of the clinical pharmacology of imatinib. EXPERIMENTAL DESIGN Six patients who were receiving imatinib (400 mg/d) as part of their ongoing treatment for chronic myeloid leukemia (CML) received a dose containing a trace quantity (13.6 kBq) of (14)C-imatinib. Blood samples were collected from patients before and at various times up to 72 h after administration of the test dose and were processed to provide samples of plasma and peripheral blood lymphocytes (PBL). Samples were analyzed by AMS, with chromatographic separation of parent compound from metabolites. In addition, plasma samples were analyzed by liquid chromatography/mass spectrometry (LCMS). RESULTS Analysis of the AMS data indicated that imatinib was rapidly absorbed and could be detected in plasma up to 72 h after administration. Imatinib was also detectable in PBL at 24 h after administration of the (14)C-labeled dose. Comparison of plasma concentrations determined by AMS with those derived by LCMS analysis gave similar average estimates of area under plasma concentration time curve (26 +/- 3 versus 27 +/- 11 microg/mL.h), but with some variation within each individual. CONCLUSIONS Using this technique, data were obtained in a small number of patients on the pharmacokinetics of a single dose of imatinib in the context of chronic dosing, which could shed light on possible pharmacologic causes of resistance to imatinib in CML.
Collapse
Affiliation(s)
- Alan V Boddy
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kimmelman J. Ethics at phase 0: clarifying the issues. THE JOURNAL OF LAW, MEDICINE & ETHICS : A JOURNAL OF THE AMERICAN SOCIETY OF LAW, MEDICINE & ETHICS 2007; 35:727-514. [PMID: 18076522 DOI: 10.1111/j.1748-720x.2007.00194.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The Food and Drug Administration (FDA) and the European Agency for the Evaluation of Medicinal Products (EMEA) recently issued documents encouraging sponsors to consider microdose testing before launching Phase I trials, and many commentators predict that such methodologies will be applied more routinely in drug development. However, exploratory testing has provoked several ethical criticisms. Skeptics question the value and validity of microdose trials, and whether they present a reasonable balance of risks and benefits for subjects. Another major criticism is that such studies serve mainly commercial ends. The present article explores these and other ethical concerns for studies conducted in the oncology setting. It concludes that microdosing is not inconsistent with prevailing practices in Phase I research, and that in principle, such studies could strengthen the ethical basis for Phase I trials by providing them better evidentiary justification.
Collapse
|
47
|
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
|
48
|
Kelloff GJ, Krohn KA, Larson SM, Weissleder R, Mankoff DA, Hoffman JM, Link JM, Guyton KZ, Eckelman WC, Scher HI, O'Shaughnessy J, Cheson BD, Sigman CC, Tatum JL, Mills GQ, Sullivan DC, Woodcock J. The progress and promise of molecular imaging probes in oncologic drug development. Clin Cancer Res 2006; 11:7967-85. [PMID: 16299226 DOI: 10.1158/1078-0432.ccr-05-1302] [Citation(s) in RCA: 190] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
As addressed by the recent Food and Drug Administration Critical Path Initiative, tools are urgently needed to increase the speed, efficiency, and cost-effectiveness of drug development for cancer and other diseases. Molecular imaging probes developed based on recent scientific advances have great potential as oncologic drug development tools. Basic science studies using molecular imaging probes can help to identify and characterize disease-specific targets for oncologic drug therapy. Imaging end points, based on these disease-specific biomarkers, hold great promise to better define, stratify, and enrich study groups and to provide direct biological measures of response. Imaging-based biomarkers also have promise for speeding drug evaluation by supplementing or replacing preclinical and clinical pharmacokinetic and pharmacodynamic evaluations, including target interaction and modulation. Such analyses may be particularly valuable in early comparative studies among candidates designed to interact with the same molecular target. Finally, as response biomarkers, imaging end points that characterize tumor vitality, growth, or apoptosis can also serve as early surrogates of therapy success. This article outlines the scientific basis of oncology imaging probes and presents examples of probes that could facilitate progress. The current regulatory opportunities for new and existing probe development and testing are also reviewed, with a focus on recent Food and Drug Administration guidance to facilitate early clinical development of promising probes.
Collapse
Affiliation(s)
- Gary J Kelloff
- Cancer Imaging Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Bergström M, Långström B. Pharmacokinetic studies with PET. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2006; 62:279-317. [PMID: 16329260 DOI: 10.1007/3-7643-7426-8_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Affiliation(s)
- Mats Bergström
- Uppsala Imanet, GE Health Care, Box 967, SE-751 09 Uppsala, Sweden.
| | | |
Collapse
|
50
|
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: 1.9] [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
|