1
|
Pure shift NMR and DFT methods for revealing long-range heteronuclear couplings. Chem Phys Lett 2022. [DOI: 10.1016/j.cplett.2022.140079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
2
|
Wang Y, Lin Q, Shi H, Cheng D. Fluorine-18: Radiochemistry and Target-Specific PET Molecular Probes Design. Front Chem 2022; 10:884517. [PMID: 35844642 PMCID: PMC9277085 DOI: 10.3389/fchem.2022.884517] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 05/19/2022] [Indexed: 12/02/2022] Open
Abstract
The positron emission tomography (PET) molecular imaging technology has gained universal value as a critical tool for assessing biological and biochemical processes in living subjects. The favorable chemical, physical, and nuclear characteristics of fluorine-18 (97% β+ decay, 109.8 min half-life, 635 keV positron energy) make it an attractive nuclide for labeling and molecular imaging. It stands that 2-[18F]fluoro-2-deoxy-D-glucose ([18F]FDG) is the most popular PET tracer. Besides that, a significantly abundant proportion of PET probes in clinical use or under development contain a fluorine or fluoroalkyl substituent group. For the reasons given above, 18F-labeled radiotracer design has become a hot topic in radiochemistry and radiopharmaceutics. Over the past decades, we have witnessed a rapid growth in 18F-labeling methods owing to the development of new reagents and catalysts. This review aims to provide an overview of strategies in radiosynthesis of [18F]fluorine-containing moieties with nucleophilic [18F]fluorides since 2015.
Collapse
Affiliation(s)
- Yunze Wang
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Institute of Nuclear Medicine, Fudan University, Shanghai, China
- Shanghai Institute of Medical Imaging, Shanghai, China
| | - Qingyu Lin
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Institute of Nuclear Medicine, Fudan University, Shanghai, China
- Shanghai Institute of Medical Imaging, Shanghai, China
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Institute of Nuclear Medicine, Fudan University, Shanghai, China
- Shanghai Institute of Medical Imaging, Shanghai, China
| | - Dengfeng Cheng
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Institute of Nuclear Medicine, Fudan University, Shanghai, China
- Shanghai Institute of Medical Imaging, Shanghai, China
| |
Collapse
|
3
|
The Unique Pharmacometrics of Small Molecule Therapeutic Drug Tracer Imaging for Clinical Oncology. Cancers (Basel) 2020; 12:cancers12092712. [PMID: 32971780 PMCID: PMC7563483 DOI: 10.3390/cancers12092712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/11/2020] [Accepted: 09/17/2020] [Indexed: 12/30/2022] Open
Abstract
Simple Summary New clinical radiology scans using trace amounts of therapeutic cancer drugs labeled with radioisotope injected into patients can provide oncologists with fundamentally unique insights about drug delivery to tumors. This new application of radiology aims to improve how cancer drugs are used, towards improving patient outcomes. The article reviews published clinical research in this important new field. Abstract Translational development of radiolabeled analogues or isotopologues of small molecule therapeutic drugs as clinical imaging biomarkers for optimizing patient outcomes in targeted cancer therapy aims to address an urgent and recurring clinical need in therapeutic cancer drug development: drug- and target-specific biomarker assays that can optimize patient selection, dosing strategy, and response assessment. Imaging the in vivo tumor pharmacokinetics and biomolecular pharmacodynamics of small molecule cancer drugs offers patient- and tumor-specific data which are not available from other pharmacometric modalities. This review article examines clinical research with a growing pharmacopoeia of investigational small molecule cancer drug tracers.
Collapse
|
4
|
Tay NES, Chen W, Levens A, Pistritto VA, Huang Z, Wu Z, Li Z, Nicewicz DA. 19F- and 18F-Arene Deoxyfluorination via Organic Photoredox-Catalysed Polarity-Reversed Nucleophilic Aromatic Substitution. Nat Catal 2020; 3:734-742. [PMID: 33791591 PMCID: PMC8009013 DOI: 10.1038/s41929-020-0495-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 07/08/2020] [Indexed: 11/09/2022]
Abstract
Nucleophilic aromatic substitution (SNAr) is routinely used to install 19F- and 18F- in aromatic molecules, but is typically limited to electron-deficient arenes due to kinetic barriers associated with C-F bond formation. Here we demonstrate that a polarity-reversed photoredox-catalysed arene deoxyfluorination operating via cation radical-accelerated nucleophilic aromatic substitution (CRA-SNAr) enables the fluorination of electron-rich arenes with 19F- and 18F- under mild conditions, thus complementing the traditional arene polarity requirements necessary for SNAr-based fluorination. The utility of our radiofluorination strategy is highlighted by short reaction times, compatibility with multiple nucleofuges, and high radiofluorination yields, especially that of an important cancer positron emission tomography (PET) agent [18F]5-fluorouracil ([18F]FU). Taken together, our fluorination approach enables the development of fluorinated and radiofluorinated compounds that can be difficult to access by classical SNAr strategies, with the potential for use in the synthesis and discovery of PET radiopharmaceuticals.
Collapse
Affiliation(s)
- Nicholas E S Tay
- Department of Chemistry, University of North Carolina at Chapel Hill, Venable Laboratories, Chapel Hill, North Carolina, 27599-3290, USA
| | - Wei Chen
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina- Chapel Hill, Chapel Hill, NC 27514, USA
| | - Alison Levens
- Department of Chemistry, University of North Carolina at Chapel Hill, Venable Laboratories, Chapel Hill, North Carolina, 27599-3290, USA
| | - Vincent A Pistritto
- Department of Chemistry, University of North Carolina at Chapel Hill, Venable Laboratories, Chapel Hill, North Carolina, 27599-3290, USA
| | - Zeng Huang
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina- Chapel Hill, Chapel Hill, NC 27514, USA
| | - Zhanhong Wu
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina- Chapel Hill, Chapel Hill, NC 27514, USA
| | - Zibo Li
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina- Chapel Hill, Chapel Hill, NC 27514, USA
| | - David A Nicewicz
- Department of Chemistry, University of North Carolina at Chapel Hill, Venable Laboratories, Chapel Hill, North Carolina, 27599-3290, USA
| |
Collapse
|
5
|
Impact of regional differences along the gastrointestinal tract of healthy adults on oral drug absorption: An UNGAP review. Eur J Pharm Sci 2019; 134:153-175. [DOI: 10.1016/j.ejps.2019.04.013] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/03/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023]
|
6
|
Heinzmann K, Carter LM, Lewis JS, Aboagye EO. Multiplexed imaging for diagnosis and therapy. Nat Biomed Eng 2017; 1:697-713. [PMID: 31015673 DOI: 10.1038/s41551-017-0131-8] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 08/02/2017] [Indexed: 12/12/2022]
Abstract
Complex molecular and metabolic phenotypes depict cancers as a constellation of different diseases with common themes. Precision imaging of such phenotypes requires flexible and tunable modalities capable of identifying phenotypic fingerprints by using a restricted number of parameters while ensuring sensitivity to dynamic biological regulation. Common phenotypes can be detected by in vivo imaging technologies, and effectively define the emerging standards for disease classification and patient stratification in radiology. However, for the imaging data to accurately represent a complex fingerprint, the individual imaging parameters need to be measured and analysed in relation to their wider spatial and molecular context. In this respect, targeted palettes of molecular imaging probes facilitate the detection of heterogeneity in oncogene-driven alterations and their response to treatment, and lead to the expansion of rational-design elements for the combination of imaging experiments. In this Review, we evaluate criteria for conducting multiplexed imaging, and discuss its opportunities for improving patient diagnosis and the monitoring of therapy.
Collapse
Affiliation(s)
- Kathrin Heinzmann
- Department of Surgery and Cancer, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Lukas M Carter
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Eric O Aboagye
- Department of Surgery and Cancer, Imperial College London, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
7
|
Hoover AJ, Lazari M, Ren H, Narayanam MK, Murphy JM, van Dam RM, Hooker JM, Ritter T. A Transmetalation Reaction Enables the Synthesis of [ 18F]5-Fluorouracil from [ 18F]Fluoride for Human PET Imaging. Organometallics 2016; 35:1008-1014. [PMID: 27087736 PMCID: PMC4829938 DOI: 10.1021/acs.organomet.6b00059] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Indexed: 01/06/2023]
Abstract
Translation of new 18F-fluorination reactions to produce radiotracers for human positron emission tomography (PET) imaging is rare because the chemistry must have useful scope and the process for 18F-labeled tracer production must be robust and simple to execute. The application of transition metal mediators has enabled impactful 18F-fluorination methods, but to date none of these reactions have been applied to produce a human-injectable PET tracer. In this article we present chemistry and process innovations that culminate in the first production from [18F]fluoride of human doses of [18F]5-fluorouracil, a PET tracer for cancer imaging in humans. The first preparation of nickel σ-aryl complexes by transmetalation from arylboronic acids or esters was developed and enabled the synthesis of the [18F]5-fluorouracil precursor. Routine production of >10 mCi doses of [18F]5-fluorouracil was accomplished with a new instrument for azeotrope-free [18F]fluoride concentration in a process that leverages the tolerance of water in nickel-mediated 18F-fluorination.
Collapse
Affiliation(s)
- Andrew J Hoover
- Department of Chemistry and Chemical Biology, Harvard University , 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| | - Mark Lazari
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine at University of California, Los Angeles , 570 Westwood Plaza, Los Angeles, California 90095, United States
| | - Hong Ren
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, United States; Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Maruthi Kumar Narayanam
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine at University of California, Los Angeles , 570 Westwood Plaza, Los Angeles, California 90095, United States
| | - Jennifer M Murphy
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine at University of California, Los Angeles , 570 Westwood Plaza, Los Angeles, California 90095, United States
| | - R Michael van Dam
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine at University of California, Los Angeles , 570 Westwood Plaza, Los Angeles, California 90095, United States
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, United States; Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Tobias Ritter
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States; Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States; Max-Planck-Institut für Kohlenforschung, Kaiser-Wilhelm-Platz 1, D-45470 Mülheim an der Ruhr, Germany
| |
Collapse
|
8
|
Mahajan A, Goh V, Basu S, Vaish R, Weeks AJ, Thakur MH, Cook GJ. Bench to bedside molecular functional imaging in translational cancer medicine: to image or to imagine? Clin Radiol 2015; 70:1060-82. [PMID: 26187890 DOI: 10.1016/j.crad.2015.06.082] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 06/03/2015] [Accepted: 06/08/2015] [Indexed: 02/05/2023]
Abstract
Ongoing research on malignant and normal cell biology has substantially enhanced the understanding of the biology of cancer and carcinogenesis. This has led to the development of methods to image the evolution of cancer, target specific biological molecules, and study the anti-tumour effects of novel therapeutic agents. At the same time, there has been a paradigm shift in the field of oncological imaging from purely structural or functional imaging to combined multimodal structure-function approaches that enable the assessment of malignancy from all aspects (including molecular and functional level) in a single examination. The evolving molecular functional imaging using specific molecular targets (especially with combined positron-emission tomography [PET] computed tomography [CT] using 2- [(18)F]-fluoro-2-deoxy-D-glucose [FDG] and other novel PET tracers) has great potential in translational research, giving specific quantitative information with regard to tumour activity, and has been of pivotal importance in diagnoses and therapy tailoring. Furthermore, molecular functional imaging has taken a key place in the present era of translational cancer research, producing an important tool to study and evolve newer receptor-targeted therapies, gene therapies, and in cancer stem cell research, which could form the basis to translate these agents into clinical practice, popularly termed "theranostics". Targeted molecular imaging needs to be developed in close association with biotechnology, information technology, and basic translational scientists for its best utility. This article reviews the current role of molecular functional imaging as one of the main pillars of translational research.
Collapse
Affiliation(s)
- A Mahajan
- Division of Imaging Sciences and Biomedical Engineering, King's College London, UK; Department of Radiodiagnosis, Tata Memorial Centre, Mumbai, 400012, India.
| | - V Goh
- Division of Imaging Sciences and Biomedical Engineering, King's College London, UK
| | - S Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Mumbai, 400 012, India
| | - R Vaish
- Department of Head and Neck Surgical Oncology, Tata Memorial Centre, Mumbai, 400012, India
| | - A J Weeks
- Division of Imaging Sciences and Biomedical Engineering, King's College London, UK
| | - M H Thakur
- Department of Radiodiagnosis, Tata Memorial Centre, Mumbai, 400012, India
| | - G J Cook
- Division of Imaging Sciences and Biomedical Engineering, King's College London, UK; Department of Nuclear Medicine, Guy's and St Thomas NHS Foundation Trust Hospital, London, UK
| |
Collapse
|
9
|
Saleem A, Murphy P, Plisson C, Lahn M. Why are we failing to implement imaging studies with radiolabelled new molecular entities in early oncology drug development? ScientificWorldJournal 2014; 2014:269605. [PMID: 25202719 PMCID: PMC4151371 DOI: 10.1155/2014/269605] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 07/18/2014] [Indexed: 11/18/2022] Open
Abstract
In early drug development advanced imaging techniques can help with progressing new molecular entities (NME) to subsequent phases of drug development and thus reduce attrition. However, several organizational, operational, and regulatory hurdles pose a significant barrier, potentially limiting the impact these techniques can have on modern drug development. Positron emission tomography (PET) of radiolabelled NME is arguably the best example of a complex technique with a potential to deliver unique decision-making data in small cohorts of subjects. However, to realise this potential the impediments to timely inclusion of PET into the drug development process must be overcome. In the present paper, we discuss the value of PET imaging with radiolabelled NME during early anticancer drug development, as exemplified with one such NME. We outline the multiple hurdles and propose options on how to streamline the organizational steps for future studies.
Collapse
Affiliation(s)
- Azeem Saleem
- Imanova Ltd., Centre for Imaging Sciences, Imperial College Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Philip Murphy
- GlaxoSmithKline Global Imaging Unit, Stockley Park West, 1-3 Ironbridge Road, Uxbridge, Middlesex UB11 1BT, UK
| | - Christophe Plisson
- Imanova Ltd., Centre for Imaging Sciences, Imperial College Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Michael Lahn
- Early Phase Oncology Clinical Investigation, Eli Lilly Corporate Center, Building 31/4, 893 S. Delaware Street, Indianapolis, IN 46285, USA
| |
Collapse
|
10
|
Van der Veldt AAM, Lammertsma AA, Smit EF. Scheduling of anticancer drugs: timing may be everything. Cell Cycle 2012; 11:4339-43. [PMID: 23032365 DOI: 10.4161/cc.22187] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Many cancer patients are treated with a combination of anticancer drugs. Here, we discuss the importance of drug scheduling and the need for studies that investigate the optimal timing of the various anticancer drugs. Positron emission tomography (PET) using radiolabeled anticancer drugs could be an important tool for those studies.
Collapse
Affiliation(s)
- Astrid A M Van der Veldt
- Department of Nuclear Medicine & PET Research, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
11
|
Aboagye EO, Gilbert FJ, Fleming IN, Beer AJ, Cunningham VJ, Marsden PK, Visvikis D, Gee AD, Groves AM, Kenny LM, Cook GJ, Kinahan PE, Myers M, Clarke L. Recommendations for measurement of tumour vascularity with positron emission tomography in early phase clinical trials. Eur Radiol 2012; 22:1465-78. [DOI: 10.1007/s00330-011-2311-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 09/08/2011] [Accepted: 09/27/2011] [Indexed: 12/22/2022]
|
12
|
Tomasi G, Kimberley S, Rosso L, Aboagye E, Turkheimer F. Double-input compartmental modeling and spectral analysis for the quantification of positron emission tomography data in oncology. Phys Med Biol 2012; 57:1889-906. [DOI: 10.1088/0031-9155/57/7/1889] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
13
|
Sharma R, Aboagye E. Development of radiotracers for oncology--the interface with pharmacology. Br J Pharmacol 2012; 163:1565-85. [PMID: 21175573 DOI: 10.1111/j.1476-5381.2010.01160.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
There is an increasing role for positron emission tomography (PET) in oncology, particularly as a component of early phase clinical trials. As a non-invasive functional imaging modality, PET can be used to assess both pharmacokinetics and pharmacodynamics of novel therapeutics by utilizing radiolabelled compounds. These studies can provide crucial information early in the drug development process that may influence the further development of novel therapeutics. PET imaging probes can also be used as early biomarkers of clinical response and to predict clinical outcome prior to the administration of therapeutic agents. We discuss the role of PET imaging particularly as applied to phase 0 studies and discuss the regulations involved in the development and synthesis of novel radioligands. The review also discusses currently available tracers and their role in the assessment of pharmacokinetics and pharmacodynamics as applied to oncology.
Collapse
Affiliation(s)
- Rohini Sharma
- Comprehensive Cancer Imaging Centre, Imperial College London Hammersmith Campus, Du Cane Road, London, UK
| | | |
Collapse
|
14
|
Nguyen QD, Challapalli A, Smith G, Fortt R, Aboagye EO. Imaging apoptosis with positron emission tomography: 'bench to bedside' development of the caspase-3/7 specific radiotracer [(18)F]ICMT-11. Eur J Cancer 2012; 48:432-40. [PMID: 22226480 DOI: 10.1016/j.ejca.2011.11.033] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 11/21/2011] [Indexed: 12/20/2022]
Abstract
The capacity to evade apoptosis has been defined as one of the hallmarks of cancer and, thus, effective anti-cancer therapy often induces apoptosis. A biomarker for imaging apoptosis could assist in monitoring the efficacy of a wide range of current and future therapeutics. Despite the potential, there are limited clinical examples of the use of positron emission tomography for imaging of apoptosis. [(18)F]ICMT-11 is a novel reagent designed to non-invasively image caspase-3 activation and, hence, drug-induced apoptosis. Radiochemistry development of [(18)F]ICMT-11 has been undertaken to improve specific radioactivity, reduce content of stable impurities, reduce synthesis time and enable automation for manufacture of multi-patient dose. Due to the promising mechanistic and safety profile of [(18)F]ICMT-11, the radiotracer is transitioning to clinical development and has been selected as a candidate radiotracer by the QuIC-ConCePT consortium for further evaluation in preclinical models and humans. A successful outcome will allow use of the radiotracer as qualified method for evaluating the pharmaceutical industry's next generation therapeutics.
Collapse
Affiliation(s)
- Quang-Dé Nguyen
- Department of Surgery and Cancer, Imperial College, London, UK
| | | | | | | | | |
Collapse
|
15
|
Wagner CC, Langer O. Approaches using molecular imaging technology -- use of PET in clinical microdose studies. Adv Drug Deliv Rev 2011; 63:539-46. [PMID: 20887762 DOI: 10.1016/j.addr.2010.09.011] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Revised: 09/21/2010] [Accepted: 09/22/2010] [Indexed: 12/11/2022]
Abstract
Positron emission tomography (PET) imaging uses minute amounts of radiolabeled drug tracers and thereby meets the criteria for clinical microdose studies. The advantage of PET, when compared to other analytical methods used in microdose studies, is that the pharmacokinetics (PK) of a drug can be determined in the tissue targeted for drug treatment. PET microdosing already offers interesting applications in clinical oncology and in the development of central nervous system pharmaceuticals and is extending its range of application to many other fields of pharmaceutical medicine. Although requirements for preclinical safety testing for microdose studies have been cut down by regulatory authorities, radiopharmaceuticals increasingly need to be produced under good manufacturing practice (GMP) conditions, which increases the costs of PET microdosing studies. Further challenges in PET microdosing include combining PET with other ultrasensitive analytical methods, such as accelerator mass spectrometry (AMS), to gain plasma PK data of drugs, beyond the short PET examination periods. Finally, conducting clinical PET studies with radiolabeled drugs both at micro- and therapeutic doses is encouraged to answer the question of dose linearity in clinical microdosing.
Collapse
Affiliation(s)
- Claudia C Wagner
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger-Gürtel 18-20, A-1090, Vienna, Austria
| | | |
Collapse
|
16
|
New ultrasensitive detection technologies and techniques for use in microdosing studies. Bioanalysis 2011; 1:357-66. [PMID: 21083172 DOI: 10.4155/bio.09.40] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
In a microdosing study, subpharmacologically active doses of drug are given to human volunteers at an early stage of development in order to obtain preliminary pharmacokinetic data. The very low doses of drug administered (≤100 µg) consequently lead to very low concentrations of drug appearing in the body and therefore highly sensitive analytical techniques are required. There are three such analytical technologies currently used in microdosing studies: PET, liquid chromatography (LC)-tandem mass spectrometry (MS/MS) and accelerator mass spectrometry (AMS). Both PET and AMS employ radioisotopic tracers. PET is an imaging technique and AMS is an extremely sensitive isotope ratio method, able to measure drug concentrations in the ag/ml range. LC-MS/MS does not require the presence of an isotopic tracer and its sensitivity is in the pg/ml range. This review examines each of these three analytical modalities in the context of performing microdosing studies.
Collapse
|
17
|
Deckers R, Moonen CT. Ultrasound triggered, image guided, local drug delivery. J Control Release 2010; 148:25-33. [DOI: 10.1016/j.jconrel.2010.07.117] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Accepted: 07/18/2010] [Indexed: 10/19/2022]
|
18
|
Aboagye EO. The future of imaging: developing the tools for monitoring response to therapy in oncology: the 2009 Sir James MacKenzie Davidson Memorial lecture. Br J Radiol 2010; 83:814-22. [PMID: 20716650 DOI: 10.1259/bjr/77317821] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Since the days of Sir James MacKenzie Davidson, radiology discoveries have been shaping the way patients are managed. The lecture on which this review is based focused not on anatomical imaging, which already has a great impact on patient management, but on the molecular imaging of cancer targets and pathways. In this post-genomic era, we have several tools at our disposal to enable the discovery of new probes for stratifying patients for therapy and for monitoring response to therapy sooner than is possible using conventional cross-sectional imaging methods. I describe a chemical library approach to discovering new imaging agents, as well as novel methods for improving the metabolic stability of existing probes. Finally, I describe the evaluation of these probes for clinical use in both pre-clinical and clinical validation. The chemical library approach is exemplified by the discovery of isatin sulfonamide probes for imaging apoptosis in tumours. This approach allowed important desirable features of radiopharmaceuticals to be incorporated into the design strategy. A lead compound, [(18)F]ICMT11, is selectively taken up in vitro in cancer cells and in vivo in tumours undergoing apoptosis. Improvement of the metabolic stability of a probe is exemplified by work on [(18)F]fluoro-[1,2-(2)H(2)]choline ("[(18)F]-D4-choline"), a novel probe for imaging choline metabolism. Deuterium substitution significantly reduced the systemic metabolism of this compound relative to that of non-deuteriated analogues, supporting its future clinical use. In order for probes to be useful for monitoring response a number of validation and/or qualification studies need to be performed, including assessments of whether the probe measures the target or pathway of interest in a specific and reproducible manner, whether the probe is stable to metabolism in vivo, what is the best time to assess response with these probes and finally whether changes in radiotracer uptake are associated with clinical outcome. [(18)F]Fluorothymidine, a probe for proliferation imaging has been validated and qualified for use in breast cancer. In summary, the ability to create new molecules that can report on specific targets and pathways provides a strategy for studying response to treatment in cancer earlier than it is currently possible. This could fundamentally change the way medicine is practised in the next 5-10 years.
Collapse
Affiliation(s)
- E O Aboagye
- Comprehensive Cancer Imaging Centre at Imperial College, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London W12 0NN, UK.
| |
Collapse
|
19
|
|
20
|
Kötz B, West C, Saleem A, Jones T, Price P. Blood flow and Vd (water): both biomarkers required for interpreting the effects of vascular targeting agents on tumor and normal tissue. Mol Cancer Ther 2009; 8:303-9. [DOI: 10.1158/1535-7163.mct-08-1016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
21
|
Saleem A, Price PM. Early tumor drug pharmacokinetics is influenced by tumor perfusion but not plasma drug exposure. Clin Cancer Res 2009; 14:8184-90. [PMID: 19088034 DOI: 10.1158/1078-0432.ccr-08-1324] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE Pharmacokinetic parameters derived from plasma sampling are used as a surrogate of tumor pharmacokinetics. However, pharmacokinetics-modulating strategies do not always result in increased therapeutic efficacy. Nonsurrogacy of plasma kinetics may be due to tissue-specific factors such as tumor perfusion. EXPERIMENTAL DESIGN To assess the impact of tumor perfusion and plasma drug exposure on tumor pharmacokinetics, positron emission tomography studies were done with oxygen-15 radiolabeled water in 12 patients, with 6 patients undergoing positron emission tomography studies with carbon-11 radiolabeled N-[2-(dimethylamino)ethyl]acridine-4-carboxamide and the other 6 with fluorine-18 radiolabeled 5-fluorouracil. RESULTS We found that tumor blood flow (mL blood/mL tissue/minute) was significantly correlated to early tumor radiotracer uptake between 4 and 6 minutes [standard uptake value (SUV)4-6; rho = 0.79; P = 0.002], tumor radiotracer exposure over 10 minutes [area under the time-activity curve (AUC)0-10; predominantly parent drug; rho = 0.86; P < 0.001], and tumor radiotracer exposure over 60 minutes (AUC0-60; predominantly radiolabeled metabolites; rho = 0.80; P = 0.002). Similarly, fractional volume of distribution of radiolabeled water in tumor (Vd) was significantly correlated with SUV4-6 (rho = 0.80; P = 0.002), AUC0-10 (rho = 0.85; P < 0.001), and AUC0-60 (rho = 0.66; P = 0.02). In contrast, no correlation was observed between plasma drug or total radiotracer exposure over 60 minutes and tumor drug uptake or exposure. Tumor blood flow was significantly correlated to Vd (rho = 0.69; P = 0.014), underlying the interdependence of tumor perfusion and Vd. CONCLUSIONS Tumor perfusion is a key factor that influences tumor drug uptake/exposure. Tumor vasculature-targeting strategies may thus result in improved tumor drug exposure and therefore drug efficacy.
Collapse
Affiliation(s)
- Azeem Saleem
- Academic Department of Radiation Oncology, The Christie Hospital NHS Foundation Trust, Manchester.
| | | |
Collapse
|
22
|
Lodge MA, Jacene HA, Pili R, Wahl RL. Reproducibility of tumor blood flow quantification with 15O-water PET. J Nucl Med 2008; 49:1620-7. [PMID: 18832120 PMCID: PMC2587033 DOI: 10.2967/jnumed.108.052076] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Noninvasive methods for quantifying tumor blood flow (TBF) have a potentially important role in the field of drug development. (15)O-water PET has been used in several studies aimed at monitoring response to novel treatments. Assessing the significance of changes in TBF requires knowledge of the reproducibility of the technique. This article quantifies the reproducibility of the (15)O-water technique for TBF applications. METHODS A total of 43 pairs of replicate (15)O-water studies were performed on 23 different patients with cancer. TBF was estimated using a standard, single-compartment model, and the replicate data were used to assess the reproducibility of the method. RESULTS The magnitude of the differences between replicate flow measurements was found to be proportional to their means. TBF was measured with a within-subject coefficient of variation of 13.4% and a repeatability of 37.1%. The volume of distribution was measured with a within-subject coefficient of variation of 8.6% and a repeatability of 24.0%. CONCLUSION (15)O-water PET can be used to measure TBF with a reproducibility that is consistent with other applications of the technique. The short half-life of the isotope permits multiple replicate studies to be performed during the same imaging session, allowing the reproducibility of the average flow estimate to be adapted to the required task. (15)O-water PET is a powerful and robust tool for TBF quantification.
Collapse
Affiliation(s)
- Martin A Lodge
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | | | | | | |
Collapse
|
23
|
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
|
24
|
Sugae S, Suzuki A, Takahashi N, Minamimoto R, Cheng C, Theeraladanon C, Endo I, Togo S, Inoue T, Shimada H. Fluorine-18-labeled 5-fluorouracil is a useful radiotracer for differentiation of malignant tumors from inflammatory lesions. Ann Nucl Med 2008; 22:65-72. [DOI: 10.1007/s12149-007-0081-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2007] [Accepted: 10/17/2007] [Indexed: 10/22/2022]
|
25
|
Saleem A, Aboagye EO, Matthews JC, Price PM. Plasma pharmacokinetic evaluation of cytotoxic agents radiolabelled with positron emitting radioisotopes. Cancer Chemother Pharmacol 2007; 61:865-73. [PMID: 17639391 DOI: 10.1007/s00280-007-0552-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2007] [Accepted: 06/18/2007] [Indexed: 11/28/2022]
Abstract
PURPOSE This study aimed to evaluate the utility of plasma pharmacokinetic analyses of anti-cancer agents from data obtained during positron emission tomography (PET) oncology studies of radiolabelled anti-cancer agents. PATIENTS AND METHODS Thirteen patients were administered fluorine-18 radiolabelled 5-FU ([(18)F]5-FU) admixed with 5-FU, corresponding to a total 5-FU dose of 380-407 mg/m2 (eight patients) and 1 mg/m2 (five patients). Nine patients received 2.2-19.2 microg/m2 of carbon-11 radiolabelled N-[2-(dimethylamino)ethyl]acridine-4-carboxamide ([11C]DACA) at 1/1,000th of phase I dose, as part of phase 0 microdosing study. Radioactivity of parent drug obtained from arterial blood samples, the injected activity of the radiolabelled drug, and the total dose of injected drug were used to obtain plasma drug concentrations. Plasma pharmacokinetic parameters were estimated using model-dependent and model-independent methods. RESULTS 5-FU plasma concentrations at therapeutic doses were above the Km and a single compartment kinetic model was best used to fit the kinetics, with a mean half-life of 8.6 min. Clearance and volumes of distribution (Vd) obtained using both model-dependent and model-independent methods were similar. Mean (SE) clearance was 1,421(144), ml min(-1) and 1,319 (119) ml min(-1) and the mean (SE) Vd was 17.3 (1.8) l and 16.3 (1.9) l by the model-independent method and model-dependent methods, respectively. In contrast, with 1 mg/m2, plasma concentrations of 5-FU were less than the Km and a two-compartment model was used to best fit the kinetics, with the mean 5-FU half-life of 6.5 min. The mean (SE) clearances obtained by the model-independent method and model-dependent methods were 3,089 (314) ml min(-1) and 2,225 (200) ml min(-1), respectively and the mean (SE) Vd were 27.9 (7.0) l and 2.3 (0.4) l, by the model independent and dependent methods, respectively. Extrapolation of AUC0-Clast to AUC0-infinity was less than 3% in both these cohort of patients. A two-compartment model with a mean half-life of 42.1 min was used to best fit the kinetics of DACA; considerable extrapolation (mean 26%) was required to obtain AUC0-infinity from AUC0-Clast. Mean (SE) clearance of DACA was 1,920 (269) ml min(-1), with the model-independent method and 1,627 (287) ml min(-1) with the model-dependent method. Similarly, Vd [mean (SE)] of DACA with the model-independent and model-dependent methods were 118 (22) l and 50 (15) l, respectively. CONCLUSIONS Pharmacokinetic parameters can be estimated with confidence from PET studies for agents given at therapeutic doses, whose half-lives are significantly less than the total sampling time during the scan. Tracer studies performed alone, wherein plasma levels below the Km are expected, may also provide valuable information on drug clearance for the entire range of linear kinetics. However, drugs with half-lives longer than the sampling duration are inappropriate for PET plasma pharmacokinetic evaluation.
Collapse
Affiliation(s)
- A Saleem
- Academic Department of Radiation Oncology, Christie Hospital NHS Foundation Trust, Wilmslow Road, and The University of Manchester Wolfson Molecular Imaging Centre, Manchester M20 4BX, UK.
| | | | | | | |
Collapse
|
26
|
Boddy AV. Recent developments in the clinical pharmacology of classical cytotoxic chemotherapy. Br J Clin Pharmacol 2007; 62:27-34. [PMID: 16842376 PMCID: PMC1885069 DOI: 10.1111/j.1365-2125.2006.02714.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Advances in analytical methods, imaging techniques and an increased understanding of the influence of pharmacogenetic factors have added to our knowledge of the pharmacology of many chemotherapeutic agents. Extending the use of these approaches to pharmacodynamic end-points, together with the application of population-based modelling techniques, offers the potential to develop truly individualized therapy in the future.
Collapse
Affiliation(s)
- Alan V Boddy
- Northern Institute for Cancer Research, Medical School, University of Newcastle, Newcastle upon Tyne, UK.
| |
Collapse
|
27
|
Price P. The role of PET scanning in determining pharmacoselective doses in oncology drug development. ERNST SCHERING RESEARCH FOUNDATION WORKSHOP 2006:185-93. [PMID: 17117724 DOI: 10.1007/978-3-540-49529-1_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Molecular imaging is the most sensitive and specific method for measuring in vivo molecular pathways in man. Its use in oncology has developed significantly over the last 5-10 years. Molecules can be labelled with positron emitting isotopes and the emitted radiation is detected using sensitive positron emission tomography (PET) cameras. It is now possible to measure in vivo and normal tissue pharmacokinetics of anti-cancer drugs and investigate their mechanism of action. Radiolabelling of tracers can be used to measure specific pharmacodynamic endpoints and target identification. Increasing evidence shows how these technologies, when added to early drug development, can rapidly reduce the time for entry into man and early identification of mechanisms of action. With the move towards more segmented markets and identification of specific subgroups, PET's use for noninvasive biomarkers will become in- creasingly important. However, much international effort between academia and industry is required with prioritisation of development of this technology.
Collapse
Affiliation(s)
- P Price
- Academic Department of Radiation Oncology, Christie Hospital NHS Trust, Withington, Manchester, UK.
| |
Collapse
|
28
|
Gupta N, Saleem A, Kötz B, Osman S, Aboagye EO, Phillips R, Vernon C, Wasan H, Jones T, Hoskin PJ, Price PM. Carbogen and nicotinamide increase blood flow and 5-fluorouracil delivery but not 5-fluorouracil retention in colorectal cancer metastases in patients. Clin Cancer Res 2006; 12:3115-23. [PMID: 16707610 DOI: 10.1158/1078-0432.ccr-05-0513] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE To examine whether carbogen and nicotinamide increases 5-fluorouracil (5-FU) delivery to colorectal cancer metastases. EXPERIMENTAL DESIGN Six patients were scanned using positron emission tomography. Two scans were done to coincide with the start of separate chemotherapy cycles. At the second positron emission tomography session, 60 mg/kg nicotinamide was given orally 2 to 3 hours before 10-minute carbogen inhalation. In the middle of carbogen treatment, [15O]H2O (to measure regional tissue perfusion) and then [18F]5-FU (to measure 5-FU tissue pharmacokinetics) were administered. RESULTS Regions of interest were drawn in 12 liver metastases, 6 spleens, 6 livers, and 12 kidneys. Nicotinamide and carbogen administration increased mean blood pO2 from 93 mm Hg (95% confidence interval, 79-198) to 278 mm Hg (95% confidence interval, 241-316; P = 0.031). Regional perfusion (mL(blood)/min/mL(tissue)) increased in metastases (mean change = 52%, range -32% to +261%, P = 0.024), but decreased in kidney (mean change = -42%, range -82% to -11%, P = 0.0005) and liver (mean change = -34%, range -43% to -26%, P = 0.031). 5-FU uptake at 3.75 minutes (m(2)/mL) increased in tumor (mean change = 40%, range -39% to +196%, P = 0.06) and decreased in kidney (mean change = -25%, range -71% to 12%, P = 0.043). 5-FU delivery measured as K1 increased in tumor (mean change = 74%, range -23% to +293%, P = 0.0039). No differences were seen in [18F]5-FU tumor exposure (net area under curve) and retention. CONCLUSION Nicotinamide and carbogen administration can increase 5-FU delivery to colorectal cancer liver metastases. Despite an increase in perfusion and 5-FU delivery, the effects were not directly related and did not increase 5-FU retention or tissue exposure.
Collapse
Affiliation(s)
- Nishi Gupta
- Cancer Research UK PET Oncology Group and Hammersmith Imanet, Hammersmith Hospital NHS Trust, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Positron emission tomography (PET) allows noninvasive, quantitative studies of various biologic processes in the tumor tissue. By using PET, investigators can study the pharmacokinetics of anticancer drugs, identify various therapeutic targets and monitor the inhibition of these targets during therapy. Furthermore, PET provides various markers to assess tumor response early in the course of therapy. A significant number of studies have now shown that changes in tumor glucose utilization during the first weeks of chemotherapy are significantly correlated with patient outcome. These data suggest that PET may be used as a sensitive test to assess the activity of new cytotoxic agents in phase II studies. Furthermore, early identification of nonresponding tumors provides the opportunity to adjust treatment regimens according to the individual chemosensitivity of the tumor tissue. However, further prospective and randomized validation of PET is still required before PET controlled chemotherapy can be used in clinical practice.
Collapse
Affiliation(s)
- Wolfgang A Weber
- Department of Molecular Medicine and Pharmacology, David Geffen School of Medicine, UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095, USA.
| |
Collapse
|
30
|
Saleem A, Charnley N, Price P. Clinical molecular imaging with positron emission tomography. Eur J Cancer 2006; 42:1720-7. [PMID: 16797972 DOI: 10.1016/j.ejca.2006.02.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Accepted: 02/10/2006] [Indexed: 11/30/2022]
Abstract
Molecular imaging allows for the in vivo evaluation of targeted molecules and biological processes in man. Positron emission tomography (PET) is a highly sensitive and quantitative molecular imaging modality, whose utility in clinical and experimental medicine is increasing by the day. In this article, the principles of PET and its currently accepted applications in oncology, such as cancer staging, treatment response assessment and as a prognostic marker are reviewed. Further, the evolving role of PET in areas of oncology such as radiotherapy treatment planning, anti-cancer drug development and the evaluation of patho-physiological processes which drive a cell into neoplastic activity is discussed.
Collapse
Affiliation(s)
- Azeem Saleem
- The University of Manchester Wolfson Molecular Imaging Centre, 27 Palatine Road, Withington, Manchester, M20 3JL, UK.
| | | | | |
Collapse
|
31
|
Workman P, Aboagye EO, Chung YL, Griffiths JR, Hart R, Leach MO, Maxwell RJ, McSheehy PMJ, Price PM, Zweit J. Minimally invasive pharmacokinetic and pharmacodynamic technologies in hypothesis-testing clinical trials of innovative therapies. J Natl Cancer Inst 2006; 98:580-98. [PMID: 16670384 DOI: 10.1093/jnci/djj162] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Clinical trials of new cancer drugs should ideally include measurements of parameters such as molecular target expression, pharmacokinetic (PK) behavior, and pharmacodynamic (PD) endpoints that can be linked to measures of clinical effect. Appropriate PK/PD biomarkers facilitate proof-of-concept demonstrations for target modulation; enhance the rational selection of an optimal drug dose and schedule; aid decision-making, such as whether to continue or close a drug development project; and may explain or predict clinical outcomes. In addition, measurement of PK/PD biomarkers can minimize uncertainty associated with predicting drug safety and efficacy, reduce the high levels of drug attrition during development, accelerate drug approval, and decrease the overall costs of drug development. However, there are many challenges in the development and implementation of biomarkers that probably explain their disappointingly low implementation in phase I trials. The Pharmacodynamic/Pharmacokinetic Technologies Advisory committee of Cancer Research UK has found that submissions for phase I trials of new cancer drugs in the United Kingdom often lack detailed information about PK and/or PD endpoints, which leads to suboptimal information being obtained in those trials or to delays in starting the trials while PK/PD methods are developed and validated. Minimally invasive PK/PD technologies have logistic and ethical advantages over more invasive technologies. Here we review these technologies, emphasizing magnetic resonance spectroscopy and positron emission tomography, which provide detailed functional and metabolic information. Assays that measure effects of drugs on important biologic pathways and processes are likely to be more cost-effective than those that measure specific molecular targets. Development, validation, and implementation of minimally invasive PK/PD methods are encouraged.
Collapse
Affiliation(s)
- Paul Workman
- Cancer Research UK Centre for Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Laking GR, West C, Buckley DL, Matthews J, Price PM. Imaging vascular physiology to monitor cancer treatment. Crit Rev Oncol Hematol 2006; 58:95-113. [PMID: 16387510 DOI: 10.1016/j.critrevonc.2005.10.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2004] [Revised: 09/30/2004] [Accepted: 10/14/2005] [Indexed: 11/27/2022] Open
Abstract
The primary physiological function of the vasculature is to support perfusion, the nutritive flow of blood through the tissues. Vascular physiology can be studied non-invasively in human subjects using imaging methods such as positron emission tomography (PET), magnetic resonance imaging (MRI), X-ray computed tomography (CT), and Doppler ultrasound (DU). We describe the physiological rationale for imaging vascular physiology with these methods. We review the published data on repeatability. We review the literature on 'before-and-after' studies using these methods to monitor response to treatment in human subjects, in five broad clinical settings: (1) antiangiogenic agents, (2) vascular disruptive agents, (3) conventional cytotoxic drugs, (4) radiation treatment, and (5) agents affecting drug delivery. We argue that imaging of vascular physiology offers an attractive 'functional endpoint' for clinical trials of anticancer treatment. More conventional measures of tumour response, such as size criteria and the uptake of fluorodeoxyglucose, may be insensitive to therapeutically important changes in vascular function.
Collapse
Affiliation(s)
- George R Laking
- Cancer Research UK PET Oncology Group, University of Manchester, Wolfson Molecular Imaging Centre, Manchester M203LJ, UK.
| | | | | | | | | |
Collapse
|
33
|
Abstract
Although most clinical diagnostic imaging studies employ anatomic techniques such as computed tomography (CT) and magnetic resonance (MR) imaging, much of radiology research currently focuses on adapting these conventional methods to physiologic imaging as well as on introducing new techniques and probes for studying processes at the cellular and molecular levels in vivo, i.e. molecular imaging. Molecular imaging promises to provide new methods for the early detection of cancer and support for personalized cancer therapy. Although molecular imaging has been practiced in various incarnations for over 20 years in the context of nuclear medicine, other imaging modalities have only recently been applied to the noninvasive assessment of physiology and molecular events. Nevertheless, there has been sufficient experience with specifically targeted contrast agents and high-resolution techniques for MR imaging and other modalities that we must begin moving these new technologies from the laboratory to the clinic. This brief review outlines several of the more promising areas of pursuit in molecular imaging for oncology with an emphasis on those that show the most immediate likelihood for clinical translation.
Collapse
Affiliation(s)
- Martin G Pomper
- The Russell H Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, Baltimore, MD 21287-2182, USA.
| |
Collapse
|
34
|
|
35
|
Sun H, Collins JM, Mangner TJ, Muzik O, Shields AF. Imaging the pharmacokinetics of [F-18]FAU in patients with tumors: PET studies. Cancer Chemother Pharmacol 2005; 57:343-8. [PMID: 16001172 DOI: 10.1007/s00280-005-0037-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2004] [Accepted: 04/26/2005] [Indexed: 11/29/2022]
Abstract
PURPOSE FAU (1-(2'-deoxy-2'-fluoro-beta-D: -arabinofuranosyl) uracil) can be phosphorylated by thymidine kinase, methylated by thymidylate synthase, followed by DNA incorporation and thus functions as a DNA synthesis inhibitor. This first-in-human study of [F-18]FAU was conducted in cancer patients to determine its suitability for imaging and also to understand its pharmacokinetics as a potential antineoplastic agent. METHODS Six patients with colorectal (n = 3) or breast cancer (n = 3) were imaged with [F-18]FAU. Serial blood and urine samples were analyzed using HPLC to determine the clearance and metabolites. RESULTS Imaging showed that [F-18]FAU was concentrated in breast tumors and a lymph node metastasis (tumor-to-normal-breast-tissue-ratio 3.7-4.7). FAU retention in breast tumors was significantly higher than in normal breast tissues at 60 min and retained in tumor over 2.5 h post-injection. FAU was not retained above background in colorectal tumors. Increased activity was seen in the kidney and urinary bladder due to excretion. Decreased activity was seen in the bone marrow with a mean SUV 0.6. Over 95% of activity in the blood and urine was present as intact [F-18]FAU at the end of the study. CONCLUSIONS Increased [F-18]FAU retention was shown in the breast tumors but not in colorectal tumors. The increased retention of FAU in the breast compared to bone marrow indicates that FAU may be useful as an unlabeled antineoplastic agent. The low retention in the marrow indicates that unlabeled FAU might lead to little marrow toxicity; however, the images were not of high contrast to consider FAU for diagnostic clinical imaging.
Collapse
Affiliation(s)
- Haihao Sun
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201-2013, USA
| | | | | | | | | |
Collapse
|
36
|
Abstract
Positron emission tomography (PET) imaging of small animals enables researchers to bridge the gap between in vitro science and in vivo human studies. The imaging paradigm can be established and refined in animals before implementation in humans and image data related to ex vivo assays of biological activity. Small animal PET (saPET) imaging enables assessment of baseline focal pathophysiology, pharmacokinetics, biological target modulation and the efficacy of novel drugs. The potential and challenge of this technology as applied to anticancer drug development is discussed here.
Collapse
Affiliation(s)
- Eric O Aboagye
- Molecular Therapy and PET Oncology Research group, The Clinical Sciences Centre, Faculty of Medicine, Hammersmith Hospital Campus, Imperial College London, Rm. 242 MRC Cyclotron Building, London, W12 0NN, UK.
| |
Collapse
|
37
|
Affiliation(s)
- Martin G Pomper
- Department of Radiology, Johns Hopkins University, Baltimore, MD 21287-2182, USA.
| | | |
Collapse
|
38
|
West CML, Jones T, Price P. The potential of positron-emission tomography to study anticancer-drug resistance. Nat Rev Cancer 2004; 4:457-69. [PMID: 15170448 DOI: 10.1038/nrc1368] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Catharine M L West
- Academic Department of Radiation Oncology and Manchester Molecular Imaging Centre, University of Manchester, Christie NHS Trust Hospital, Wilmslow Road, Manchester, M20 4BX, United Kingdom.
| | | | | |
Collapse
|
39
|
Kumar R, Jana S. Positron emission tomography: an advanced nuclear medicine imaging technique from research to clinical practice. Methods Enzymol 2004; 385:3-19. [PMID: 15130730 DOI: 10.1016/s0076-6879(04)85001-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Rakesh Kumar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, NewDelhi 110029, India
| | | |
Collapse
|
40
|
N/A. N/A. Shijie Huaren Xiaohua Zazhi 2003; 11:1410-1413. [DOI: 10.11569/wcjd.v11.i9.1410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
|
41
|
Wells P, Aboagye E, Gunn RN, Osman S, Boddy AV, Taylor GA, Rafi I, Hughes AN, Calvert AH, Price PM, Newell DR. 2-[11C]thymidine positron emission tomography as an indicator of thymidylate synthase inhibition in patients treated with AG337. J Natl Cancer Inst 2003; 95:675-82. [PMID: 12734319 DOI: 10.1093/jnci/95.9.675] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Some anticancer drugs inhibit thymidylate synthase (TS), a key enzyme for thymidine nucleotide biosynthesis. Cells can compensate for depleted thymidine levels by taking up extracellular thymidine via a salvage pathway. We investigated the use of 2-[11C]thymidine positron emission tomography (PET) to measure thymidine salvage kinetics in vivo in humans. METHODS Five patients with advanced gastrointestinal cancer were PET scanned both before and 1 hour after oral administration of the TS inhibitor AG337 (THYMITAQ [nolatrexed]); seven control patients were scanned twice but not treated with AG337. Thymidine salvage kinetics were measured in vivo using 2-[11C]thymidine PET and spectral analysis to obtain the standardized uptake values (SUV), the area under the time-activity curve (AUC), and the fractional retention of thymidine (FRT). Changes in PET parameters between scans in the AG337-treated and control groups were compared using the Mann-Whitney U test. The relationship between AG337 exposure and AG337-induced changes in tumor FRT and in plasma deoxyuridine levels (a conventional pharmacodynamic systemic measure of TS inhibition) was examined using Spearman's regression analysis. Statistical tests were two-sided. RESULTS The between-scan change in FRT in patients treated with AG337 (38% increase, 95% confidence interval [CI] = 8% to 68%) was higher than that in control patients (3% increase, 95% CI = -11% to 17%) (P =.028). The level of AG337-induced increase in both 2-[11C]thymidine FRT and plasma deoxyuridine levels was statistically significantly correlated with AG337 exposure (r = 1.00, P =.01 for both). CONCLUSIONS AG337 administration was associated with increased tumor tracer retention that was consistent with tumor cell uptake of exogenous 2-[11C]thymidine as a result of TS inhibition. 2-[11C]Thymidine PET can be used to measure thymidine salvage kinetics directly in the tissue of interest.
Collapse
Affiliation(s)
- Paula Wells
- Imperial College School of Medicine, Hammersmith Hospital, London, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Positron emission tomography (PET) is increasingly being used in anticancer drug development. The technique is applicable to studies of drug delivery, and where specific probes are available, to provide pharmacodynamic readouts noninvasively in patients. Mathematical modeling of the imaging data enhances the quality of information that is obtained from such studies. This section provides a review of the PET methodologies that have been used for the development of new cancer therapies. Other than imaging of radiolabeled drugs, PET modeling has found extensive application in studies with 2-[11C]thymidine, [18F]fluorodeoxyglucose, H2(15)O, C15O, and receptor ligands.
Collapse
Affiliation(s)
- Eric O Aboagye
- PET Oncology Group, Department of Cancer Medicine, Imperial College of Science, Technology and Medicine, Faculty of Medicine, Hammersmith Hospital, London, UK
| | | |
Collapse
|
43
|
Propper DJ, de Bono J, Saleem A, Ellard S, Flanagan E, Paul J, Ganesan TS, Talbot DC, Aboagye EO, Price P, Harris AL, Twelves C. Use of positron emission tomography in pharmacokinetic studies to investigate therapeutic advantage in a phase I study of 120-hour intravenous infusion XR5000. J Clin Oncol 2003; 21:203-10. [PMID: 12525511 DOI: 10.1200/jco.2003.02.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE XR5000 (N-[2-(dimethylamino)ethyl]acridine-4-carboxamide) is a topoisomerase I and II inhibitor. Because the cytotoxicity of XR5000 increases markedly with prolonged exposure, we performed a phase I study of weekly XR5000 by 120-hour continuous infusion over 3 weeks. PATIENTS AND METHODS Twenty-four patients with advanced solid cancer were treated at seven dose levels (700 to 4,060 mg/m2/120 hrs) for a total of 67 cycles. Three patients underwent positron emission tomography (PET) studies at the maximum-tolerated dose (MTD) to evaluate normal tissue and tumor carbon-11 radiolabeled XR5000 ([11C]XR5000) pharmacokinetics. RESULTS The dose-limiting toxicity was National Cancer Institute Common Toxicity Criteria (version 1) grade 4 chest and abdominal pain affecting the single patient receiving 4,060 mg/m2/120 hours, and the MTD was 3,010 mg/m2/120 hours. Other grade 3-4 toxicities, affecting single patients at the MTD, were myelosuppression (grade 4), raised bilirubin, vomiting, and somnolence (all grade 3). There was one partial response (adenocarcinoma of unknown primary); the remainder had progressive disease. [11C]XR5000 distributed well into the three tumors studied by PET. Tumor uptake (maximum concentration or area under the concentration versus time curve [AUC]) was less than in normal tissue in which the tumors were located. Tumor exposure (AUC; mean +/- SD in m2/mL/sec) increased when [(11)C]XR5000 was administered during an infusion of XR5000 (0.242 +/- 0.4), compared with [11C]XR5000 given alone (0.209 +/- 0.04; P <.05), indicating that tumor drug exposure was not saturated [corrected]. CONCLUSION The recommended dose for XR5000 in phase II studies is 3,010 mg/m2/120 hours. PET studies with 11C-labeled drug were feasible and demonstrated in vivo distribution into tumors. Saturation of tumor exposure was not reached at the MTD.
Collapse
Affiliation(s)
- D J Propper
- CRC Department of Medical Oncology, Beatson Oncology Centre, Glasgow, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Solomon B, McArthur G, Cullinane C, Zalcberg J, Hicks R. Applications of Positron Emission Tomography in the Development of Molecular Targeted Cancer Therapeutics. BioDrugs 2003; 17:339-54. [PMID: 14498764 DOI: 10.2165/00063030-200317050-00004] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
For molecular targeted cancer therapies to fulfill their promise in cancer treatment, innovative approaches are required to overcome significant obstacles that exist in the clinical development of these agents. Positron emission tomography (PET) is a functional imaging technology that allows rapid, repeated, noninvasive, in vivo assessment and quantification of many biological processes and in some cases molecular pathways targeted by these therapies. It is highly sensitive, with the capacity to detect subnanomolar concentrations of radiotracer and provides superior image resolution to conventional nuclear medicine imaging with gamma cameras. Novel PET radiotracers have been developed that allow visualisation of a variety of processes including tumour metabolism, cell proliferation, apoptosis, hypoxia and blood flow. Furthermore, specific molecular targets including cellular receptors can be identified using radiolabelled receptor ligands or specific monoclonal antibodies. Improvements in imaging technology leading to the development of small-animal PET scanners, with resolution capable of imaging commonly used mouse models of cancer, will enable PET to play an important role in preclinical proof-of-principle drug studies. Such improvements will also facilitate the validation of imaging protocols that can be readily translated to studies in humans. The greatest utility of PET in the development of molecular targeted therapeutics, however, lies in clinical studies, where PET may play a valuable role in a number of situations. These include selection of patients for therapy through noninvasive identification of the presence of specific molecular targets, pharmacokinetic studies with labelled drugs and pharmacodynamic evaluations of biological parameters to select the optimal biological dose, and assessment of response to therapies.
Collapse
Affiliation(s)
- Benjamin Solomon
- Research Division, Division of Haematology, Peter MacCallum Cancer Center, Melbourne, Australia
| | | | | | | | | |
Collapse
|
45
|
Abstract
Positron emission tomography (PET) scanning is evolving as a unique tool for drug development in oncology for improving both the efficacy of established treatment and in evaluating novel anticancer agents. As a non-invasive functional imaging modality, PET has an unrivalled sensitivity when monitoring the pharmacokinetics and pharmacodynamics of drugs and biochemicals when radiolabelled with short living positron-emitting radioisotopes. This is of particular relevance in assessing newer molecular-targeted therapy where conventional evaluation criteria (maximum tolerated dose and tumour shrinkage for example) may be inappropriate. PET has already been applied to a wide number of drugs to demonstrate activity in vivo from standard chemotherapy such as 5-fluorouracil (5-FU) [J Clin Oncol 17 (1999) 1580], to novel molecular agents such as those involved in tumour angiogenesis [Br J Cancer 83 (2000) P6] and antivascular therapy [Proc Annu Meet Am Soc Clin Oncol 19 (2000) 179a]. This review will evaluate the achievements of PET in the drug development process, an approach that promises to facilitate the rapid translation of scientific research into current clinical practice.
Collapse
Affiliation(s)
- N Gupta
- Cancer Research UK PET Oncology Group, Section of Cancer Therapeutics, Imperial College of Science, Technology and Medicine, MRC Cyclotron Unit, Hammersmith Hospital, Du Cane Road, London W12 ONN, UK
| | | | | |
Collapse
|
46
|
Abstract
Over the next three years a large number of novel, mechanistically targeted drugs will enter clinical trials for cancer. The remarkable progress in understanding the molecular biology of cancer has provided an enormous range of validated targets for drug discovery. Following lead optimisation and suitable pharmaceutical formulation these compounds have undergone rapid screening in preclinical models. Innovative methods of clinical development are now essential to ensure optimal dose determination and scheduling. The discovery of novel surrogates for efficacy is essential in this fast moving area and requires imaginative partnerships between academic groups and the pharmaceutical industry.
Collapse
|
47
|
Oral Fluoropyrimidines in Colorectal Cancer. COLORECTAL CANCER 2002. [DOI: 10.1007/978-1-59259-160-2_27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
48
|
Abstract
During the past ten years, positron emission tomography (PET) has been increasingly developed for imaging and quantifying molecular mechanisms in oncology. The technique uses radionuclides to label molecules, which can then be imaged in man. The inherent sensitivity and specificity of PET is unrivalled because it can image molecular interactions and pathways, providing quantitative kinetic information down to the subpicomolar level. This technology has the potential to answer a large number of important clinical questions in translational research in oncology. However, the challenges in the methodology are substantial. Molecular imaging has the potential to assist in the optimization of molecular-based targeted therapies in cancer and to investigate the function of the genome.
Collapse
Affiliation(s)
- P Price
- Molecular Imaging Centre, Academic Department of Radiation Oncology, University of Manchester, UK.
| |
Collapse
|
49
|
Abstract
This article reviews current knowledge of the metabolism of drugs that contain fluorine. The strategic value of fluorine substitution in drug design is discussed in terms of chemical structure and basic concepts in drug metabolism and drug toxicity.
Collapse
Affiliation(s)
- B K Park
- Department of Pharmacology and Therapeutics, New Medical Building, University of Liverpool, Liverpool, United Kingdom.
| | | | | |
Collapse
|
50
|
Affiliation(s)
- C H Takimoto
- Department of Medicine, Division of Medical Oncology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|