1
|
Islam S, Inglese M, Grech-Sollars M, Aravind P, Dubash S, Barwick TD, O'Neill K, Wang J, Saleem A, O'Callaghan J, Anchini G, Williams M, Waldman A, Aboagye EO. Feasibility of [ 18F]fluoropivalate hybrid PET/MRI for imaging lower and higher grade glioma: a prospective first-in-patient pilot study. Eur J Nucl Med Mol Imaging 2023; 50:3982-3995. [PMID: 37490079 PMCID: PMC10611885 DOI: 10.1007/s00259-023-06330-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/04/2023] [Indexed: 07/26/2023]
Abstract
PURPOSE MRI and PET are used in neuro-oncology for the detection and characterisation of lesions for malignancy to target surgical biopsy and to plan surgical resections or stereotactic radiosurgery. The critical role of short-chain fatty acids (SCFAs) in brain tumour biology has come to the forefront. The non-metabolised SCFA radiotracer, [18F]fluoropivalate (FPIA), shows low background signal in most tissues except eliminating organs and has appropriate human dosimetry. Tumour uptake of the radiotracer is, however, unknown. We investigated the uptake characteristics of FPIA in this pilot PET/MRI study. METHODS Ten adult glioma subjects were identified based on radiological features using standard-of-care MRI prior to any surgical intervention, with subsequent histopathological confirmation of glioma subtype and grade (lower-grade - LGG - and higher-grade - HGG - patients). FPIA was injected as an intravenous bolus injection (range 342-368 MBq), and dynamic PET and MRI data were acquired simultaneously over 66 min. RESULTS All patients tolerated the PET/MRI protocol. Three patients were reclassified following resection and histology. Tumour maximum standardised uptake value (SUVmax,60) increased in the order LGG (WHO grade 2) < HGG (WHO grade 3) < HGG (WHO grade 4). The net irreversible solute transfer, Ki, and influx rate constant, K1, were significantly higher in HGG (p < 0.05). Of the MRI variables studied, DCE-MRI-derived extravascular-and-extracellular volume fraction (ve) was high in tumours of WHO grade 4 compared with other grades (p < 0.05). SLC25A20 protein expression was higher in HGG compared with LGG. CONCLUSION Tumoural FPIA PET uptake is higher in HGG compared to LGG. This study supports further investigation of FPIA PET/MRI for brain tumour imaging in a larger patient population. CLINICAL TRIAL REGISTRATION Clinicaltrials.gov, NCT04097535.
Collapse
Affiliation(s)
- Shahriar Islam
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Marianna Inglese
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Matthew Grech-Sollars
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Preetha Aravind
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Suraiya Dubash
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Tara D Barwick
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Kevin O'Neill
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - James Wang
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Azeem Saleem
- Invicro Limited, Burlington Danes Building, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
- Hull York Medical School, University of Hull, Cottingham Road, Hull, HU6 7RX, UK
| | - James O'Callaghan
- Invicro Limited, Burlington Danes Building, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Giulio Anchini
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Matthew Williams
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Adam Waldman
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Eric O Aboagye
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
2
|
Cheng H, Tang Y, Li Z, Guo Z, Heath JR, Xue M, Wei W. Non-Mass Spectrometric Targeted Single-Cell Metabolomics. Trends Analyt Chem 2023; 168:117300. [PMID: 37840599 PMCID: PMC10569257 DOI: 10.1016/j.trac.2023.117300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Metabolic assays serve as pivotal tools in biomedical research, offering keen insights into cellular physiological and pathological states. While mass spectrometry (MS)-based metabolomics remains the gold standard for comprehensive, multiplexed analyses of cellular metabolites, innovative technologies are now emerging for the targeted, quantitative scrutiny of metabolites and metabolic pathways at the single-cell level. In this review, we elucidate an array of these advanced methodologies, spanning synthetic and surface chemistry techniques, imaging-based methods, and electrochemical approaches. We summarize the rationale, design principles, and practical applications for each method, and underscore the synergistic benefits of integrating single-cell metabolomics (scMet) with other single-cell omics technologies. Concluding, we identify prevailing challenges in the targeted scMet arena and offer a forward-looking commentary on future avenues and opportunities in this rapidly evolving field.
Collapse
Affiliation(s)
- Hanjun Cheng
- Institute for Systems Biology, Seattle, WA, 98109, United States
| | - Yin Tang
- Institute for Systems Biology, Seattle, WA, 98109, United States
| | - Zhonghan Li
- Department of Chemistry, University of California, Riverside, CA, 92521, United States
| | - Zhili Guo
- Department of Chemistry, University of California, Riverside, CA, 92521, United States
| | - James R. Heath
- Institute for Systems Biology, Seattle, WA, 98109, United States
| | - Min Xue
- Department of Chemistry, University of California, Riverside, CA, 92521, United States
| | - Wei Wei
- Institute for Systems Biology, Seattle, WA, 98109, United States
| |
Collapse
|
3
|
Epigenetic potentiation of somatostatin-2 by guadecitabine in neuroendocrine neoplasias as a novel method to allow delivery of peptide receptor radiotherapy. Eur J Cancer 2022; 176:110-120. [PMID: 36208569 DOI: 10.1016/j.ejca.2022.09.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/08/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Somatostatin receptor-2 (SSTR2) is expressed on cell surface of neuroendocrine neoplasias; its presence is exploited for the delivery of peptide receptor radionuclide therapy (PRRT). Patients with no or low expression of SSTR2 are not candidates for PRRT. SSTR2 promotor undergoes epigenetic modification, known to regulate gene expression. We investigated whether the demethylation agent, guadecitabine, could enhance the expression of SSTR2 in NET models, using radioligand uptake/PET imaging as a biomarker of epigenetic modification. METHODS The effects of guadecitabine on the transcriptional, translational, and functional regulation of SSTR2 both in vitro and in vivo using low (QGP-1) and high (BON-1) methylated neuroendocrine neoplasia models was characterised. Promotor region methylation profiling of clinical samples (n = 61) was undertaken. Safety of combination guadecitabine and PRRT was assessed in vivo. RESULTS Pyrosequencing of cell lines illustrated differential methylation indices - BON: 1 94%, QGP: 1 21%. Following guadecitabine treatment, a dose-dependent increase in SSTR2 in BON-1 at a transcriptional, translational, and functional levels using the SSTR2-directed radioligand, 18F-FET-βAG-TOCA ([18F]-FETO) (150% increase [18F]-FETO uptake, p < 0.05) was observed. In vivo, guadecitabine treatment resulted in a 70% increase in [18F]-FETO uptake in BON-1 tumour models compared models with low baseline percentage methylation (p < 0.05). No additive toxicity was observed with the combination treatment of PRRT and guadecitabine in vivo. Methylation index in clinical samples was 10.5% compared to 5.2% in controls (p = 0.03) and correlated with SSTR2 expression (Wilcoxon rank sign -3.75,p < 0.01). CONCLUSION Guadecitabine increases SSTR2 expression both in vitro and in vivo. The combination of demethylation agents with PRRT warrants further investigation.
Collapse
|
4
|
Guo Z, Cheng H, Li Z, Shao S, Sarkar P, Wang S, Chaudhuri R, Perkins NG, Ji F, Wei W, Xue M. Single-Cell Profiling of Fatty Acid Uptake Using Surface-Immobilized Dendrimers. J Am Chem Soc 2021; 143:11191-11198. [PMID: 34266234 DOI: 10.1021/jacs.1c05103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We present a chemical approach to profile fatty acid uptake in single cells. We use azide-modified analogues to probe the fatty acid influx and surface-immobilized dendrimers with dibenzocyclooctyne (DBCO) groups for detection. A competition between the fatty acid probes and BHQ2-azide quencher molecules generates fluorescence signals in a concentration-dependent manner. By integrating this method onto a microfluidics-based multiplex protein analysis platform, we resolved the relationships between fatty acid influx, oncogenic signaling activities, and cell proliferation in single glioblastoma cells. We found that p70S6K and 4EBP1 differentially correlated with fatty acid uptake. We validated that cotargeting p70S6K and fatty acid metabolism synergistically inhibited cell proliferation. Our work provided the first example of studying fatty acid metabolism in the context of protein signaling at single-cell resolution and generated new insights into cancer biology.
Collapse
Affiliation(s)
- Zhili Guo
- Department of Chemistry, University of California, Riverside, Riverside, California 92521, United States
| | - Hanjun Cheng
- Institute for Systems Biology, Seattle, Washington 98109, United States
| | - Zhonghan Li
- Department of Chemistry, University of California, Riverside, Riverside, California 92521, United States
| | - Shiqun Shao
- Department of Chemistry, University of California, Riverside, Riverside, California 92521, United States.,Institute for Systems Biology, Seattle, Washington 98109, United States
| | - Priyanka Sarkar
- Department of Chemistry, University of California, Riverside, Riverside, California 92521, United States
| | - Siwen Wang
- Department of Chemistry, University of California, Riverside, Riverside, California 92521, United States
| | - Rohit Chaudhuri
- Department of Chemistry, University of California, Riverside, Riverside, California 92521, United States
| | - Nicole G Perkins
- Department of Chemistry, University of California, Riverside, Riverside, California 92521, United States
| | - Fei Ji
- Department of Chemistry, University of California, Riverside, Riverside, California 92521, United States
| | - Wei Wei
- Institute for Systems Biology, Seattle, Washington 98109, United States
| | - Min Xue
- Department of Chemistry, University of California, Riverside, Riverside, California 92521, United States
| |
Collapse
|
5
|
Vassileva V, Braga M, Barnes C, Przystal J, Ashek A, Allott L, Brickute D, Abrahams J, Suwan K, Carcaboso AM, Hajitou A, Aboagye EO. Effective Detection and Monitoring of Glioma Using [ 18F]FPIA PET Imaging. Biomedicines 2021; 9:811. [PMID: 34356874 PMCID: PMC8301305 DOI: 10.3390/biomedicines9070811] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/25/2021] [Accepted: 07/09/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Reprogrammed cellular metabolism is a cancer hallmark. In addition to increased glycolysis, the oxidation of acetate in the citric acid cycle is another common metabolic phenotype. We have recently developed a novel fluorine-18-labelled trimethylacetate-based radiotracer, [18F]fluoro-pivalic acid ([18F]FPIA), for imaging the transcellular flux of short-chain fatty acids, and investigated whether this radiotracer can be used for the detection of glioma growth. METHODS We evaluated the potential of [18F]FPIA PET to monitor tumor growth in orthotopic patient-derived (HSJD-GBM-001) and cell line-derived (U87, LN229) glioma xenografts, and also included [18F]FDG PET for comparison. We assessed proliferation (Ki-67) and the expression of lipid metabolism and transport proteins (CPT1, SLC22A2, SLC22A5, SLC25A20) by immunohistochemistry, along with etomoxir treatment to provide insights into [18F]FPIA uptake. RESULTS Longitudinal PET imaging showed gradual increase in [18F]FPIA uptake in orthotopic glioma models with disease progression (p < 0.0001), and high tumor-to-brain contrast compared to [18F]FDG (p < 0.0001). [18F]FPIA uptake correlated positively with Ki-67 (p < 0.01), SLC22A5 (p < 0.001) and SLC25A20 (p = 0.001), and negatively with CPT1 (p < 0.01) and SLC22A2 (p < 0.01). Etomoxir reduced [18F]FPIA uptake, which correlated with decreased Ki-67 (p < 0.05). CONCLUSIONS Our findings support the use of [18F]FPIA PET for the detection and longitudinal monitoring of glioma, showing a positive correlation with tumor proliferation, and suggest transcellular flux-mediated radiotracer uptake.
Collapse
Affiliation(s)
- Vessela Vassileva
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (M.B.); (C.B.); (L.A.); (D.B.); (J.A.)
| | - Marta Braga
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (M.B.); (C.B.); (L.A.); (D.B.); (J.A.)
| | - Chris Barnes
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (M.B.); (C.B.); (L.A.); (D.B.); (J.A.)
| | - Justyna Przystal
- Department of Medicine, Division of Brain Sciences, Imperial College London, Hammersmith Campus, Burlington Danes, London W12 0NN, UK; (J.P.); (K.S.); (A.H.)
| | - Ali Ashek
- Department of Medicine, Faculty of Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK;
| | - Louis Allott
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (M.B.); (C.B.); (L.A.); (D.B.); (J.A.)
| | - Diana Brickute
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (M.B.); (C.B.); (L.A.); (D.B.); (J.A.)
| | - Joel Abrahams
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (M.B.); (C.B.); (L.A.); (D.B.); (J.A.)
| | - Keittisak Suwan
- Department of Medicine, Division of Brain Sciences, Imperial College London, Hammersmith Campus, Burlington Danes, London W12 0NN, UK; (J.P.); (K.S.); (A.H.)
| | | | - Amin Hajitou
- Department of Medicine, Division of Brain Sciences, Imperial College London, Hammersmith Campus, Burlington Danes, London W12 0NN, UK; (J.P.); (K.S.); (A.H.)
| | - Eric O. Aboagye
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; (M.B.); (C.B.); (L.A.); (D.B.); (J.A.)
| |
Collapse
|
6
|
Dubash SR, Keat N, Kozlowski K, Barnes C, Allott L, Brickute D, Hill S, Huiban M, Barwick TD, Kenny L, Aboagye EO. Clinical translation of 18F-fluoropivalate - a PET tracer for imaging short-chain fatty acid metabolism: safety, biodistribution, and dosimetry in fed and fasted healthy volunteers. Eur J Nucl Med Mol Imaging 2020; 47:2549-2561. [PMID: 32123971 PMCID: PMC7515955 DOI: 10.1007/s00259-020-04724-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 02/14/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Fatty acids derived de novo or taken up from the extracellular space are an essential source of nutrient for cell growth and proliferation. Radiopharmaceuticals including 11C-acetate, and 18F-FAC (2-18F-fluoroacetate), have previously been used to study short-chain fatty acid (SCFA) metabolism. We developed 18F-fluoropivalate (18F-FPIA; 3-18F-fluoro-2,2-dimethylpropionic acid) bearing a gem-dimethyl substituent to assert metabolic stability for studying SCFA metabolism. We report the safety, biodistribution, and internal radiation dosimetry profile of 18F-FPIA in 24 healthy volunteers and the effect of dietary conditions. MATERIALS AND METHODS Healthy volunteer male and female subjects were enrolled (n = 24), and grouped into 12 fed and 12 fasted. Non-esterified fatty acids (NEFA) and carnitine blood measurements were assessed. Subjects received 159.48 MBq (range, 47.31-164.66 MBq) of 18F-FPIA. Radiochemical purity was > 99%. Safety data were obtained during and 24 h after radiotracer administration. Subjects underwent detailed multiple whole-body PET/CT scanning with sampling of venous bloods for radioactivity and radioactive metabolite quantification. Regions of interest were defined to derive individual and mean organ residence times; effective dose was calculated using OLINDA 1.1. RESULTS All subjects tolerated 18F-FPIA with no adverse events. Over 90% of radiotracer was present in plasma at 60 min post-injection. The organs receiving highest absorbed dose (in mGy/MBq) were the liver (0.070 ± 0.023), kidneys (0.043 ± 0.013), gallbladder wall (0.026 ± 0.003), and urinary bladder (0.021 ± 0.004); otherwise there was low tissue uptake. The calculated effective dose using mean organ residence times over all 24 subjects was 0.0154 mSv/MBq (SD ± 0.0010). No differences in biodistribution or dosimetry were seen in fed and fasted subjects, though systemic NEFA and carnitine levels reflected fasted and fed states. CONCLUSION The favourable safety, imaging, and dosimetric profile makes 18F-FPIA a promising candidate radiotracer for tracing SCFA metabolism.
Collapse
Affiliation(s)
- Suraiya R Dubash
- Department of Surgery and Cancer, Imperial College London, GN1 Commonwealth Building, Hammersmith Hospital, DuCane Rd, London, W12 0NN, UK
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| | | | - Kasia Kozlowski
- Department of Surgery and Cancer, Imperial College London, GN1 Commonwealth Building, Hammersmith Hospital, DuCane Rd, London, W12 0NN, UK
| | - Chris Barnes
- Department of Surgery and Cancer, Imperial College London, GN1 Commonwealth Building, Hammersmith Hospital, DuCane Rd, London, W12 0NN, UK
| | - Louis Allott
- Department of Surgery and Cancer, Imperial College London, GN1 Commonwealth Building, Hammersmith Hospital, DuCane Rd, London, W12 0NN, UK
| | - Diana Brickute
- Department of Surgery and Cancer, Imperial College London, GN1 Commonwealth Building, Hammersmith Hospital, DuCane Rd, London, W12 0NN, UK
| | | | | | - Tara D Barwick
- Department of Surgery and Cancer, Imperial College London, GN1 Commonwealth Building, Hammersmith Hospital, DuCane Rd, London, W12 0NN, UK
- Department of Radiology/Nuclear Medicine, Imperial College Healthcare NHS Trust, London, UK
| | - Laura Kenny
- Department of Surgery and Cancer, Imperial College London, GN1 Commonwealth Building, Hammersmith Hospital, DuCane Rd, London, W12 0NN, UK
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - Eric O Aboagye
- Department of Surgery and Cancer, Imperial College London, GN1 Commonwealth Building, Hammersmith Hospital, DuCane Rd, London, W12 0NN, UK.
| |
Collapse
|
7
|
Allott L, Aboagye EO. Chemistry Considerations for the Clinical Translation of Oncology PET Radiopharmaceuticals. Mol Pharm 2020; 17:2245-2259. [DOI: 10.1021/acs.molpharmaceut.0c00328] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Louis Allott
- Comprehensive Cancer Imaging Centre, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, United Kingdom
| | - Eric O. Aboagye
- Comprehensive Cancer Imaging Centre, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, United Kingdom
| |
Collapse
|
8
|
Lord SR, Collins JM, Cheng WC, Haider S, Wigfield S, Gaude E, Fielding BA, Pinnick KE, Harjes U, Segaran A, Jha P, Hoefler G, Pollak MN, Thompson AM, Roy PG, English R, Adams RF, Frezza C, Buffa FM, Karpe F, Harris AL. Transcriptomic analysis of human primary breast cancer identifies fatty acid oxidation as a target for metformin. Br J Cancer 2020; 122:258-265. [PMID: 31819193 PMCID: PMC6986920 DOI: 10.1038/s41416-019-0665-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 11/12/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Epidemiological studies suggest that metformin may reduce the incidence of cancer in patients with diabetes and multiple late phase clinical trials assessing the potential of repurposing this drug are underway. Transcriptomic profiling of tumour samples is an excellent tool to understand drug bioactivity, identify candidate biomarkers and assess for mechanisms of resistance to therapy. METHODS Thirty-six patients with untreated primary breast cancer were recruited to a window study and transcriptomic profiling of tumour samples carried out before and after metformin treatment. RESULTS Multiple genes that regulate fatty acid oxidation were upregulated at the transcriptomic level and there was a differential change in expression between two previously identified cohorts of patients with distinct metabolic responses. Increase in expression of a mitochondrial fatty oxidation gene composite signature correlated with change in a proliferation gene signature. In vitro assays showed that, in contrast to previous studies in models of normal cells, metformin reduces fatty acid oxidation with a subsequent accumulation of intracellular triglyceride, independent of AMPK activation. CONCLUSIONS We propose that metformin at clinical doses targets fatty acid oxidation in cancer cells with implications for patient selection and drug combinations. CLINICAL TRIAL REGISTRATION NCT01266486.
Collapse
Affiliation(s)
- Simon R Lord
- Department of Oncology, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK.
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK.
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| | - Jennifer M Collins
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Wei-Chen Cheng
- Department of Oncology, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Syed Haider
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Simon Wigfield
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Edoardo Gaude
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Barbara A Fielding
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7WG, UK
| | - Katherine E Pinnick
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Ulrike Harjes
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Ashvina Segaran
- Department of Oncology, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Pooja Jha
- Institut für Pathologie, Medizinische Universität Graz, Auenbruggerplatz 25, 8036, Graz, Austria
| | - Gerald Hoefler
- Institut für Pathologie, Medizinische Universität Graz, Auenbruggerplatz 25, 8036, Graz, Austria
| | - Michael N Pollak
- Department of Oncology, McGill University, Montreal, QC, H3T 1E2, Canada
| | - Alastair M Thompson
- Division of Surgical Oncology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Pankaj G Roy
- Breast Surgery Unit, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Ruth English
- Oxford Breast Imaging Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Rosie F Adams
- Oxford Breast Imaging Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Christian Frezza
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Francesca M Buffa
- Department of Oncology, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Fredrik Karpe
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Adrian L Harris
- Department of Oncology, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
9
|
Window of opportunity clinical trial designs to study cancer metabolism. Br J Cancer 2019; 122:45-51. [PMID: 31819180 PMCID: PMC6964681 DOI: 10.1038/s41416-019-0621-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 10/16/2019] [Accepted: 10/18/2019] [Indexed: 02/06/2023] Open
Abstract
Window of opportunity trials exploit the ‘window’ of time after cancer diagnosis, typically prior to initiation of cancer therapy. In recent years this study design has become a more regular feature of drug development, as this ‘window’ provides an opportunity to carry out a thorough pharmacodynamic assessment of a therapy of interest in tumours that are unperturbed by prior treatment. Many of the first window trials interrogated the bioactivity of drugs being repurposed for cancer treatment, in particular the anti-mitochondrial agent, metformin. In this review, we describe examples of window study designs that have been used to assess drugs that target cancer metabolism with a focus on metformin. In addition, we discuss how window studies may aid the development of molecular metabolic cancer imaging.
Collapse
|
10
|
Brickute D, Braga M, Kaliszczak MA, Barnes C, Lau D, Carroll L, Stevens E, Trousil S, Alam IS, Nguyen QD, Aboagye EO. Development and Evaluation of an 18F-Radiolabeled Monocyclam Derivative for Imaging CXCR4 Expression. Mol Pharm 2019; 16:2106-2117. [PMID: 30883140 PMCID: PMC6522096 DOI: 10.1021/acs.molpharmaceut.9b00069] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/14/2019] [Accepted: 03/18/2019] [Indexed: 01/02/2023]
Abstract
In humans, C-X-C chemokine receptor type 4 (CXCR4) is a protein that is encoded by the CXCR4 gene and binds the ligand CXCL12 (also known as SDF-1). The CXCR4-CXCL12 interaction in cancer elicits biological activities that result in tumor progression and has accordingly been the subject of significant investigation for detection and treatment of the disease. Peptidic antagonists have been labeled with a variety of radioisotopes for the detection of CXCR4, but the methodology utilizing small molecules has predominantly used radiometals. We report here the development of a 18F-radiolabeled cyclam-based small molecule radioprobe, [18F]MCFB, for imaging CXCR4 expression. The IC50 value of [19F]MCFB for CXCR4 was similar to that of AMD3465 (111.3 and 89.8 nM, respectively). In vitro binding assays show that the tracer depicted a differential CXCR4 expression, which was blocked in the presence of AMD3465, demonstrating the specificity of [18F]MCFB. Positron emission tomography (PET) imaging studies showed a distinct uptake of the radioprobe in lymphoma and breast cancer xenografts. High liver and kidney uptakes were seen with [18F]MCFB, leading us to further examine the basis of its pharmacokinetics in relation to the tracer's cationic nature and thus the role of organic cation transporters (OCTs). Substrate competition following the intravenous injection of metformin led to a marked decrease in the urinary excretion of [18F]MCFB, with moderate changes observed in other organs, including the liver. Our results suggest involvement of OCTs in the renal elimination of the tracer. In conclusion, the 18F-radiolabeled monocyclam, [18F]MCFB, has potential to detect tumor CXCR4 in nonhepatic tissues.
Collapse
Affiliation(s)
| | | | - Maciej A. Kaliszczak
- Cancer Imaging Centre, Department
of
Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, U.K.
| | - Chris Barnes
- Cancer Imaging Centre, Department
of
Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, U.K.
| | - Doreen Lau
- Cancer Imaging Centre, Department
of
Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, U.K.
| | - Laurence Carroll
- Cancer Imaging Centre, Department
of
Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, U.K.
| | - Elizabeth Stevens
- Cancer Imaging Centre, Department
of
Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, U.K.
| | - Sebastian Trousil
- Cancer Imaging Centre, Department
of
Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, U.K.
| | - Israt S. Alam
- Cancer Imaging Centre, Department
of
Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, U.K.
| | - Quang-Dé Nguyen
- Cancer Imaging Centre, Department
of
Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, U.K.
| | - Eric O. Aboagye
- Cancer Imaging Centre, Department
of
Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, U.K.
| |
Collapse
|
11
|
Senni N, Savall M, Bossard P. [β-catenin mutated hepatocarcinoma metabolic rewiring at the heart of their transformation]. Med Sci (Paris) 2019; 34:1029-1031. [PMID: 30623757 DOI: 10.1051/medsci/2018285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Nadia Senni
- Inserm, U1016, Institut Cochin, 24, rue du faubourg Saint-Jacques, 75014 Paris, France - Cnrs, UMR8104, Paris, France - Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Mathilde Savall
- Inserm, U1016, Institut Cochin, 24, rue du faubourg Saint-Jacques, 75014 Paris, France - Cnrs, UMR8104, Paris, France - Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Pascale Bossard
- Inserm, U1016, Institut Cochin, 24, rue du faubourg Saint-Jacques, 75014 Paris, France - Cnrs, UMR8104, Paris, France - Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
12
|
Heinzmann K, Nguyen QD, Honess D, Smith DM, Stribbling S, Brickute D, Barnes C, Griffiths J, Aboagye E. Depicting Changes in Tumor Biology in Response to Cetuximab Monotherapy or Combination Therapy by Apoptosis and Proliferation Imaging Using 18F-ICMT-11 and 18F-FLT PET. J Nucl Med 2018; 59:1558-1565. [PMID: 29794225 PMCID: PMC6167530 DOI: 10.2967/jnumed.118.209304] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 04/09/2018] [Indexed: 12/26/2022] Open
Abstract
Imaging biomarkers must demonstrate their value in monitoring treatment. Two PET tracers, the caspase-3/7-specific isatin-5-sulfonamide 18F-ICMT-11 (18F-(S)-1-((1-(2-fluoroethyl)-1H-[1,2,3]-triazol-4-yl)methyl)-5-(2(2,4-difluoro-phenoxymethyl)-pyrrolidine-1-sulfonyl)isatin) and 18F-FLT (3'-deoxy-3'-18F-fluorothymidine), were used to detect early treatment-induced changes in tumor biology and determine whether any of these changes indicate a response to cetuximab, administered as monotherapy or combination therapy with gemcitabine. Methods: In mice bearing cetuximab-sensitive H1975 tumors (non-small lung cancer), the effects of single or repeated doses of the antiepidermal growth factor receptor antibody cetuximab (10 mg/kg on day 1 only or on days 1 and 2) or a single dose of gemcitabine (125 mg/kg on day 2) were investigated by 18F-ICMT-11 or 18F-FLT on day 3. Imaging was also performed after 2 doses of cetuximab (days 1 and 2) in mice bearing cetuximab-insensitive HCT116 tumors (colorectal cancer). For imaging-histology comparison, tumors were evaluated for proliferation (Ki-67 and thymidine kinase 1 [TK1]), cell death (cleaved caspase-3 and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling [TUNEL]), and target engagement (epidermal growth factor receptor expression) by immunohistochemistry, immunofluorescence, and immunoblotting, respectively. Tumor and plasma were analyzed for thymidine and gemcitabine metabolites by liquid chromatography-mass spectrometry. Results: Retention of both tracers was sensitive to cetuximab in H1975 tumors. 18F-ICMT-11 uptake and ex vivo cleaved caspase-3 staining notably increased in tumors treated with repeated doses of cetuximab (75%) and combination treatment (46%). Although a single dose of cetuximab was insufficient to induce apoptosis, it did affect proliferation. Significant reductions in tumor 18F-FLT uptake (44%-50%; P < 0.001) induced by cetuximab monotherapy and combination therapy were paralleled by a clear decrease in proliferation (Ki-67 decrease, 72%-95%; P < 0.0001), followed by a marked tumor growth delay. TK1 expression and tumor thymidine concentrations were profoundly reduced. Neither imaging tracer depicted the gemcitabine-induced tumor changes. However, cleaved caspase-3 and Ki-67 staining did not significantly differ after gemcitabine treatment whereas TK1 expression and thymidine concentrations increased. No cetuximab-induced modulation of the imaging tracers or other response markers was detected in the insensitive model of HCT116. Conclusion:18F-ICMT-11 and 18F-FLT are valuable tools to assess cetuximab sensitivity depicting distinct and time-variant aspects of treatment response.
Collapse
Affiliation(s)
- Kathrin Heinzmann
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom; and
| | - Quang-Dé Nguyen
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom; and
| | - Davina Honess
- Cancer Research U.K. Cambridge Institute, Cambridge, United Kingdom
| | | | - Stephen Stribbling
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom; and
| | - Diana Brickute
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom; and
| | - Chris Barnes
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom; and
| | - John Griffiths
- Cancer Research U.K. Cambridge Institute, Cambridge, United Kingdom
| | - Eric Aboagye
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom; and
| |
Collapse
|
13
|
Kranz M, Bergmann R, Kniess T, Belter B, Neuber C, Cai Z, Deng G, Fischer S, Zhou J, Huang Y, Brust P, Deuther-Conrad W, Pietzsch J. Bridging from Brain to Tumor Imaging: (S)-(-)- and (R)-(+)-[ 18F]Fluspidine for Investigation of Sigma-1 Receptors in Tumor-Bearing Mice. Molecules 2018; 23:E702. [PMID: 29558382 PMCID: PMC6017399 DOI: 10.3390/molecules23030702] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 03/12/2018] [Accepted: 03/18/2018] [Indexed: 12/12/2022] Open
Abstract
Sigma-1 receptors (Sig1R) are highly expressed in various human cancer cells and hence imaging of this target with positron emission tomography (PET) can contribute to a better understanding of tumor pathophysiology and support the development of antineoplastic drugs. Two Sig1R-specific radiolabeled enantiomers (S)-(-)- and (R)-(+)-[18F]fluspidine were investigated in several tumor cell lines including melanoma, squamous cell/epidermoid carcinoma, prostate carcinoma, and glioblastoma. Dynamic PET scans were performed in mice to investigate the suitability of both radiotracers for tumor imaging. The Sig1R expression in the respective tumors was confirmed by Western blot. Rather low radiotracer uptake was found in heterotopically (subcutaneously) implanted tumors. Therefore, a brain tumor model (U87-MG) with orthotopic implantation was chosen to investigate the suitability of the two Sig1R radiotracers for brain tumor imaging. High tumor uptake as well as a favorable tumor-to-background ratio was found. These results suggest that Sig1R PET imaging of brain tumors with [18F]fluspidine could be possible. Further studies with this tumor model will be performed to confirm specific binding and the integrity of the blood-brain barrier (BBB).
Collapse
Affiliation(s)
- Mathias Kranz
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 04318 Leipzig, Germany.
- Department of Diagnostic Radiology, PET Center, Yale University School of Medicine, New Haven, CT 06519, USA.
| | - Ralf Bergmann
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
| | - Torsten Kniess
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
| | - Birgit Belter
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
| | - Christin Neuber
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
| | - Zhengxin Cai
- Department of Diagnostic Radiology, PET Center, Yale University School of Medicine, New Haven, CT 06519, USA.
| | - Gang Deng
- Department of Neurosurgery and Biomedical Engineering, Yale University School of Medicine, New Haven, CT 06519, USA.
| | - Steffen Fischer
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 04318 Leipzig, Germany.
| | - Jiangbing Zhou
- Department of Neurosurgery and Biomedical Engineering, Yale University School of Medicine, New Haven, CT 06519, USA.
| | - Yiyun Huang
- Department of Diagnostic Radiology, PET Center, Yale University School of Medicine, New Haven, CT 06519, USA.
| | - Peter Brust
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 04318 Leipzig, Germany.
| | - Winnie Deuther-Conrad
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 04318 Leipzig, Germany.
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
- Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, 01062 Dresden, Germany.
| |
Collapse
|
14
|
Zhang Z, Liu S, Tang X, Nie D, Tang G, Sun A, Xiong Y, Ma H, Wen F, Hu P. Radiosynthesis and preliminary biological evaluation of the 2-[18F]fluoropropionic acid enantiomers for tumor PET imaging. J Radioanal Nucl Chem 2018. [DOI: 10.1007/s10967-018-5753-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
15
|
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: 13.8] [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
|
16
|
Challapalli A, Aboagye EO. Positron Emission Tomography Imaging of Tumor Cell Metabolism and Application to Therapy Response Monitoring. Front Oncol 2016; 6:44. [PMID: 26973812 PMCID: PMC4770188 DOI: 10.3389/fonc.2016.00044] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/12/2016] [Indexed: 12/12/2022] Open
Abstract
Cancer cells do reprogram their energy metabolism to enable several functions, such as generation of biomass including membrane biosynthesis, and overcoming bioenergetic and redox stress. In this article, we review both established and evolving radioprobes developed in association with positron emission tomography (PET) to detect tumor cell metabolism and effect of treatment. Measurement of enhanced tumor cell glycolysis using 2-deoxy-2-[(18)F]fluoro-D-glucose is well established in the clinic. Analogs of choline, including [(11)C]choline and various fluorinated derivatives are being tested in several cancer types clinically with PET. In addition to these, there is an evolving array of metabolic tracers for measuring intracellular transport of glutamine and other amino acids or for measuring glycogenesis, as well as probes used as surrogates for fatty acid synthesis or precursors for fatty acid oxidation. In addition to providing us with opportunities for examining the complex regulation of reprogramed energy metabolism in living subjects, the PET methods open up opportunities for monitoring pharmacological activity of new therapies that directly or indirectly inhibit tumor cell metabolism.
Collapse
Affiliation(s)
| | - Eric O. Aboagye
- Department of Surgery and Cancer, Imperial College London, London, UK
| |
Collapse
|
17
|
Sengupta D, Pratx G. Imaging metabolic heterogeneity in cancer. Mol Cancer 2016; 15:4. [PMID: 26739333 PMCID: PMC4704434 DOI: 10.1186/s12943-015-0481-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 12/10/2015] [Indexed: 01/01/2023] Open
Abstract
As our knowledge of cancer metabolism has increased, it has become apparent that cancer metabolic processes are extremely heterogeneous. The reasons behind this heterogeneity include genetic diversity, the existence of multiple and redundant metabolic pathways, altered microenvironmental conditions, and so on. As a result, methods in the clinic and beyond have been developed in order to image and study tumor metabolism in the in vivo and in vitro regimes. Both regimes provide unique advantages and challenges, and may be used to provide a picture of tumor metabolic heterogeneity that is spatially and temporally comprehensive. Taken together, these methods may hold the key to appropriate cancer diagnoses and treatments in the future.
Collapse
Affiliation(s)
- Debanti Sengupta
- Stanford University School of Medicine, A226 Building A, 1050 Arastradero Road, Palo Alto, CA, 94304, USA
| | - Guillem Pratx
- Stanford University School of Medicine, A226 Building A, 1050 Arastradero Road, Palo Alto, CA, 94304, USA.
| |
Collapse
|
18
|
Tahara T, Zhang Z, Ohno M, Hirao Y, Hosaka N, Doi H, Suzuki M, Onoe H. A novel (11)C-labeled thymidine analog, [(11)C]AZT, for tumor imaging by positron emission tomography. EJNMMI Res 2015; 5:124. [PMID: 26337804 PMCID: PMC4597405 DOI: 10.1186/s13550-015-0124-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 08/18/2015] [Indexed: 11/18/2022] Open
Abstract
Background Nucleoside analogs labeled with positrons, such as 11C and 18F, are considered valuable in visualizing the proliferative activity of tumor cells in vivo using positron emission tomography (PET). We recently developed the 11C-labeled thymidine analogs [11C]zidovudine ([11C]AZT) and [11C]stavudine ([11C]d4T) via the Pd(0)-Cu(I) co-mediated rapid C–C coupling reaction. In this study, to examine whether [11C]AZT and [11C]d4T might be useful for visualization of tumors in vivo, we performed PET imaging, tissue distribution studies, and metabolite analysis in tumor-bearing mice. Methods Mice bearing tumors (rat glioma C6 and human cervical adenocarcinoma HeLa cells) were injected with 50 MBq of [11C]AZT or [11C]d4T, and PET was performed immediately thereafter. After PET imaging, the radioactivity in several tissues, including tumor tissues, was measured using a γ-counter. In addition, radioactive metabolites in plasma, bile, intestinal contents, and tumor were analyzed using thin layer chromatography (TLC). Cellular uptake of [11C]AZT in C6 was measured in the presence or absence of non-labeled thymidine (0.1 mM). Results In PET studies, C6 and HeLa tumors in mice were clearly visualized using [11C]AZT. Time-activity curves using [11C]AZT showed that the accumulation of radioactivity in tumors plateaued at 10 min after injection and persisted for 60 min, while most of the radioactivity in other tissues was rapidly excreted into the urine. In various tissues of the body, tumor tissue showed the highest radioactivity at 80 min after injection (five to six times higher uptake than that of blood). Compared with tumor tissue, uptake was lower in other proliferative tissues such as the spleen, intestine, and bone marrow, resulting in a high tumor-to-bone marrow ratio. Cellular uptake of [11C]AZT in C6 cells was completely blocked by the application of thymidine, strongly indicating the specific involvement of nucleoside transporters. In contrast, the time-activity curve of [11C]d4T in the tumor showed transient and rapid excretion with almost no obvious tumor tissue accumulation. Conclusions Tumors can be detected by PET using [11C]AZT; therefore, [11C]AZT could be useful as a novel PET tracer for tumor imaging in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s13550-015-0124-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tsuyoshi Tahara
- Division of Bio-Function Dynamics Imaging, RIKEN Center for Life Science Technologies (CLST), 6-7-3 Minatojima, Minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan,
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Lucarelli G, Rutigliano M, Galleggiante V, Giglio A, Palazzo S, Ferro M, Simone C, Bettocchi C, Battaglia M, Ditonno P. Metabolomic profiling for the identification of novel diagnostic markers in prostate cancer. Expert Rev Mol Diagn 2015; 15:1211-24. [PMID: 26174441 DOI: 10.1586/14737159.2015.1069711] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Metabolomic profiling offers a powerful methodology for understanding the perturbations of biochemical systems occurring during a disease process. During neoplastic transformation, prostate cells undergo metabolic reprogramming to satisfy the demands of growth and proliferation. An early event in prostate cell transformation is the loss of capacity to accumulate zinc. This change is associated with a higher energy efficiency and increased lipid biosynthesis for cellular proliferation, membrane formation and cell signaling. Moreover, recent studies have shown that sarcosine, an N-methyl derivative of glycine, was significantly increased during disease progression from normal to localized to metastatic prostate cancer. Mapping the metabolomic profiles to their respective biochemical pathways showed an upregulation of androgen-induced protein synthesis, an increased amino acid metabolism and a perturbation of nitrogen breakdown pathways, along with high total choline-containing compounds and phosphocholine levels. In this review, the role of emerging biomarkers is summarized, based on the current understanding of the prostate cancer metabolome.
Collapse
Affiliation(s)
- Giuseppe Lucarelli
- a 1 Department of Emergency and Organ Transplantation - Urology, Andrology and Kidney Transplantation Unit, University of Bari, Bari, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Alam IS, Arshad MA, Nguyen QD, Aboagye EO. Radiopharmaceuticals as probes to characterize tumour tissue. Eur J Nucl Med Mol Imaging 2015; 42:537-61. [PMID: 25647074 DOI: 10.1007/s00259-014-2984-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 12/18/2014] [Indexed: 01/06/2023]
Abstract
Tumour cells exhibit several properties that allow them to grow and divide. A number of these properties are detectable by nuclear imaging methods. We discuss crucial tumour properties that can be described by current radioprobe technologies, further discuss areas of emerging radioprobe development, and finally articulate need areas that our field should aspire to develop. The review focuses largely on positron emission tomography and draws upon the seminal 'Hallmarks of Cancer' review article by Hanahan and Weinberg in 2011 placing into context the present and future roles of radiotracer imaging in characterizing tumours.
Collapse
Affiliation(s)
- Israt S Alam
- Comprehensive Cancer Imaging Centre, Imperial College London, London, W12 0NN, UK
| | | | | | | |
Collapse
|
21
|
Abstract
Positron emission tomography (PET) is an extraordinarily sensitive clinical imaging modality for interrogating tumor metabolism. Radiolabeled PET substrates can be traced at subphysiological concentrations, allowing noninvasive imaging of metabolism and intratumoral heterogeneity in systems ranging from advanced cancer models to patients in the clinic. There are a wide range of novel and more established PET radiotracers, which can be used to investigate various aspects of the tumor, including carbohydrate, amino acid, and fatty acid metabolism. In this review, we briefly discuss the more established metabolic tracers and describe recent work on the development of new tracers. Some of the unanswered questions in tumor metabolism are considered alongside new technical developments, such as combined PET/magnetic resonance imaging scanners, which could provide new imaging solutions to some of the outstanding diagnostic challenges facing modern cancer medicine.
Collapse
Affiliation(s)
- David Y. Lewis
- Cancer Research UK - Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Dmitry Soloviev
- Cancer Research UK - Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Kevin M. Brindle
- Cancer Research UK - Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| |
Collapse
|