Advertisement

A PET approach to brain tumors

Positron emission tomography, or PET, is a common method of imaging tumors by detecting their uptake of a radioactively labeled tracer. Radiolabeled glucose, in particular, is often used for this type of imaging, because tumor cells are often highly dependent on glycolysis and require large amounts of glucose to maintain their metabolism. Unfortunately, this method cannot be used to image brain tumors, because regular brain cells are also highly dependent on glucose. Now, Venneti et al. have used mouse models and human patients to show that radiolabeled glutamine, which is also taken up by tumor cells, can be used to image brain tumors and distinguish them from normal brain and even from tumors that are no longer growing.

Abstract

Glucose and glutamine are the two principal nutrients that cancer cells use to proliferate and survive. Many cancers show altered glucose metabolism, which constitutes the basis for in vivo positron emission tomography (PET) imaging with 18F-fluorodeoxyglucose (18F-FDG). However, 18F-FDG is ineffective in evaluating gliomas because of high background uptake in the brain. Glutamine metabolism is also altered in many cancers, and we demonstrate that PET imaging in vivo with the glutamine analog 4-18F-(2S,4R)-fluoroglutamine (18F-FGln) shows high uptake in gliomas but low background brain uptake, facilitating clear tumor delineation. Chemo/radiation therapy reduced 18F-FGln tumor avidity, corresponding with decreased tumor burden. 18F-FGln uptake was not observed in animals with a permeable blood-brain barrier or neuroinflammation. We translated these findings to human subjects, where 18F-FGln showed high tumor/background ratios with minimal uptake in the surrounding brain in human glioma patients with progressive disease. These data suggest that 18F-FGln is avidly taken up by gliomas, can be used to assess metabolic nutrient uptake in gliomas in vivo, and may serve as a valuable tool in the clinical management of gliomas.

Get full access to this article

View all available purchase options and get full access to this article.

Supplementary Material

Summary

Materials and Methods
Fig. S1. Glutamine is a major TCA cycle substrate and generates 2-HG.
Fig. S2. 19F-FGln is not metabolized in gliomas.
Fig. S3. 18F-FGln shows minimal, but specific, uptake in the normal brain.
Fig. S4. Mouse glioma models mimic human gliomas.
Fig. S5. Fasting and perfusion do not affect 18F-FGln uptake.
Fig. S6. 18F-FGln shows uptake in mouse glioma xenografts.
Fig. S7. Gln is metabolized to 2-HG without altering 18F-FGln uptake.
Fig. S8. Glioma animal models show 18F-FGln tumor uptake.
Fig. S9. Neuroinflammatory mouse models do not show 18F-FGln uptake.
Fig. S10. Chemoradiation decreases tumor burden and improves survival.
Fig. S11. Chemoradiation therapy results in increased gliosis.
Fig. S12. SLC1A5 partly mediates 18F-FGln uptake and is expressed in human gliomas.
Fig. S13. Patients imaged with 18F-FGln show glioma SLC1A5 expression.
Fig. S14. 18F-FGln shows uptake in a clinically progressive glioma.
Fig. S15. 18F-FGln does not show uptake in a clinically stable glioma.
Fig. S16. 18F-FGln shows clearance from blood and plasma.
Table S1. Patient characteristics.
Table S2. Comparison of 18F-FGln and 18F-FDG imaging in human subjects.
Table S3. Biodistribution of 18F-FGln in human subjects.
Table S4. 18F-FGln parent compound and radiometabolites in plasma at 1, 30, and 60 min after tracer injection.
Table S5. 18F-FGln dosimetry in human subjects.
Table S6. P values (provided as a separate Excel file).
Table S7. Data used to generate graphs (provided as a separate Excel file).
References (29, 30)

Resources

File (7-274ra17_sm.pdf)
File (7-274ra17_tables_s6_and_s7.zip)

REFERENCES AND NOTES

1
Ward P. S., Thompson C. B., Metabolic reprogramming: A cancer hallmark even Warburg did not anticipate. Cancer Cell 21, 297–308 (2012).
2
Vander Heiden M. G., Cantley L. C., Thompson C. B., Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009).
3
Weber W. A., Schwaiger M., Avril N., Quantitative assessment of tumor metabolism using FDG-PET imaging. Nucl. Med. Biol. 27, 683–687 (2000).
4
Kläsner B. D., Krause B. J., Beer A. J., Drzezga A., PET imaging of gliomas using novel tracers: A sleeping beauty waiting to be kissed. Expert Rev. Anticancer Ther. 10, 609–613 (2010).
5
Petrirena G. J., Goldman S., Delattre J. Y., Advances in PET imaging of brain tumors: A referring physician’s perspective. Curr. Opin. Oncol. 23, 617–623 (2011).
6
DeBerardinis R. J., Mancuso A., Daikhin E., Nissim I., Yudkoff M., Wehrli S., Thompson C. B., Beyond aerobic glycolysis: Transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis. Proc. Natl. Acad. Sci. U.S.A. 104, 19345–19350 (2007).
7
Wise D. R., Thompson C. B., Glutamine addiction: A new therapeutic target in cancer. Trends Biochem. Sci. 35, 427–433 (2010).
8
Lieberman B. P., Ploessl K., Wang L., Qu W., Zha Z., Wise D. R., Chodosh L. A., Belka G., Thompson C. B., Kung H. F., PET imaging of glutaminolysis in tumors by 18F-(2S,4R)4-fluoroglutamine. J. Nucl. Med. 52, 1947–1955 (2011).
9
Huse J. T., Holland E. C., Targeting brain cancer: Advances in the molecular pathology of malignant glioma and medulloblastoma. Nat. Rev. Cancer 10, 319–331 (2010).
10
Dang L., White D. W., Gross S., Bennett B. D., Bittinger M. A., Driggers E. M., Fantin V. R., Jang H. G., Jin S., Keenan M. C., Marks K. M., Prins R. M., Ward P. S., Yen K. E., Liau L. M., Rabinowitz J. D., Cantley L. C., Thompson C. B., Vander Heiden M. G., Su S. M., Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 465, 966 (2010).
11
Rohle D., Popovici-Muller J., Palaskas N., Turcan S., Grommes C., Campos C., Tsoi J., Clark O., Oldrini B., Komisopoulou E., Kunii K., Pedraza A., Schalm S., Silverman L., Miller A., Wang F., Yang H., Chen Y., Kernytsky A., Rosenblum M. K., Liu W., Biller S. A., Su S. M., Brennan C. W., Chan T. A., Graeber T. G., Yen K. E., Mellinghoff I. K., An inhibitor of mutant IDH1 delays growth and promotes differentiation of glioma cells. Science 340, 626–630 (2013).
12
Hambardzumyan D., Amankulor N. M., Helmy K. Y., Becher O. J., Holland E. C., Modeling adult gliomas using RCAS/t-va technology. Transl. Oncol. 2, 89–95 (2009).
13
Mills C. D., M1 and M2 macrophages: Oracles of health and disease. Crit. Rev. Immunol. 32, 463–488 (2012).
14
Wu C., Li F., Niu G., Chen X., PET imaging of inflammation biomarkers. Theranostics 3, 448–466 (2013).
15
Carman A. J., Mills J. H., Krenz A., Kim D. G., Bynoe M. S., Adenosine receptor signaling modulates permeability of the blood–brain barrier. J. Neurosci. 31, 13272–13280 (2011).
16
Gao X., Qian J., Zheng S., Changyi Y., Zhang J., Ju S., Zhu J., Li C., Overcoming the blood-brain barrier for delivering drugs into the brain by using adenosine receptor nanoagonist. ACS Nano 8, 3678–3689 (2014).
17
Leder K., Pitter K., Laplant Q., Hambardzumyan D., Ross B. D., Chan T. A., Holland E. C., Michor F., Mathematical modeling of PDGF-driven glioblastoma reveals optimized radiation dosing schedules. Cell 156, 603–616 (2014).
18
Tran D. K., Jensen R. L., Treatment-related brain tumor imaging changes: So-called “pseudoprogression” vs. tumor progression: Review and future research opportunities. Surg. Neurol. Int. 4, S129–S135 (2013).
19
Ploessl K., Wang L. M., Lieberman B. P., Qu W. C., Kung H. F., Comparative evaluation of 18F-labeled glutamic acid and glutamine as tumor metabolic imaging agents. J. Nucl. Med. 53, 1616–1624 (2012).
20
Dhermain F. G., Hau P., Lanfermann H., Jacobs A. H., van den Bent M. J., Advanced MRI and PET imaging for assessment of treatment response in patients with gliomas. Lancet Neurol. 9, 906–920 (2010).
21
Shiroishi M. S., Booker M. T., Agarwal M., Jain N., Naghi I., Lerner A., Law M., Posttreatment evaluation of central nervous system gliomas. Magn. Reson. Imaging Clin. N. Am. 21, 241–268 (2013).
22
Brandão L. A., Shiroishi M. S., Law M., Brain tumors: A multimodality approach with diffusion-weighted imaging, diffusion tensor imaging, magnetic resonance spectroscopy, dynamic susceptibility contrast and dynamic contrast-enhanced magnetic resonance imaging. Magn. Reson. Imaging Clin. N. Am. 21, 199–239 (2013).
23
la Fougère C., Suchorska B., Bartenstein P., Kreth F. W., Tonn J. C., Molecular imaging of gliomas with PET: Opportunities and limitations. Neuro Oncol. 13, 806–819 (2011).
24
Crippa F., Alessi A., Serafini G. L., PET with radiolabeled aminoacid. Q. J. Nucl. Med. Mol. Imaging 56, 151–162 (2012).
25
Koglin N., Mueller A., Berndt M., Schmitt-Willich H., Toschi L., Stephens A. W., Gekeler V., Friebe M., Dinkelborg L. M., Specific PET imaging of xC transporter activity using a 18F-labeled glutamate derivative reveals a dominant pathway in tumor metabolism. Clin. Cancer Res. 17, 6000–6011 (2011).
26
Baek S., Mueller A., Lim Y. S., Lee H. C., Lee Y. J., Gong G., Kim J. S., Ryu J. S., Oh S. J., Lee S. J., Bacher-Stier C., Fels L., Koglin N., Schatz C. A., Dinkelborg L. M., Moon D. H., (4S)-4-(3-18F-fluoropropyl)-l-glutamate for imaging of xC transporter activity in hepatocellular carcinoma using PET: Preclinical and exploratory clinical studies. J. Nucl. Med. 54, 117–123 (2013).
27
Wise D. R., Ward P. S., Shay J. E., Cross J. R., Gruber J. J., Sachdeva U. M., Platt J. M., Dematteo R. G., Simon M. C., Thompson C. B., Hypoxia promotes isocitrate dehydrogenase-dependent carboxylation of α-ketoglutarate to citrate to support cell growth and viability. Proc. Natl. Acad. Sci. U.S.A. 108, 19611–19616 (2011).
28
Qu W. C., Zha Z. H., Ploessl K., Lieberman B. P., Zhu L., Wise D. R., Thompson C. B., Kung H. F., Synthesis of optically pure 4-fluoro-glutamines as potential metabolic imaging agents for tumors. J. Am. Chem. Soc. 133, 1122–1133 (2011).
29
Venneti S., Lopresti B. J., Wang G., Slagel S. L., Mason N. S., Mathis C. A., Fischer M. L., Larsen N. J., Mortimer A. D., Hastings T. G., Smith A. D., Zigmond M. J., Suhara T., Higuchi M., Wiley C. A., A comparison of the high-affinity peripheral benzodiazepine receptor ligands DAA1106 and (R)-PK11195 in rat models of neuroinflammation: Implications for PET imaging of microglial activation. J. Neurochem. 102, 2118–2131 (2007).
30
Phillips E., Penate-Medina O., Zanzonico P. B., Carvajal R. D., Mohan P., Ye Y., Humm J., Gonen M., Kalaigian H., Schoder H., Strauss H. W., Larson S. M., Wiesner U., Bradbury M. S., Clinical translation of an ultrasmall inorganic optical-PET imaging nanoparticle probe. Sci. Transl. Med. 6, 260ra149 (2014).

(0)eLetters

eLetters is a forum for ongoing peer review. eLetters are not edited, proofread, or indexed, but they are screened. eLetters should provide substantive and scholarly commentary on the article. Embedded figures cannot be submitted, and we discourage the use of figures within eLetters in general. If a figure is essential, please include a link to the figure within the text of the eLetter. Please read our Terms of Service before submitting an eLetter.

Log In to Submit a Response

No eLetters have been published for this article yet.

Information & Authors

Information

Published In

Science Translational Medicine
Volume 7 | Issue 274
February 2015

Submission history

Received: 16 October 2014
Accepted: 9 January 2015

Permissions

Request permissions for this article.

Acknowledgments

We thank E. M. Burnazi and S. Cai of the MSKCC Radiochemistry and Molecular Imaging Probe Core; V. Longo of the MSKCC Small Animal Imaging Core; D. Yarilin of the MSKCC Cytology Core Facility; J. Chou and K. Panageas from the Epidemiology and Biostatistics program; P. Desai, Y. Koike, and A. Ku for technical assistance; A. Pedraza for maintaining and distributing glioma cell lines; and the MSKCC Organic Synthesis Core. We also thank A. Lashley and B. Gansukh for facilitating human studies and C. Le, M. Lupu, and D. Winkleman for animal MRI studies. We thank T. Lindsten, D. Pozega, and members of the Thompson Laboratory for critical reading of the manuscript. Funding: Supported by a Stand Up to Cancer Dream Team Translational Research Grant, grant number SU2C-AACR-DT0509 (C.B.T.). Stand Up to Cancer is a program of the Entertainment Industry Foundation administered by the American Association for Cancer Research. This work was supported by grants from the MSKCC Brain Tumor Center (S.V. and H.Z.), National Cancer Institute (NCI) K08 CA181475 (S.V.), National Institute of General Medical Sciences Medical Scientist Training Program (NIGMS MSTP) GM07739 (K.L.P.), National Institute of Neurological Disorders and Stroke (NINDS) F31 NS076028 (K.L.P.), NCI P50 CA086438 (J.S.L.), NIH R01 NS080944 (I.K.M.), and NCI R01-CA164490 (H.F.K). The MSKCC Cores were supported by the NIH Cancer Center Support Grant P30 CA08748 (C.B.T.). Author contributions: S.V., J.S.L, and C.B.T. conceived the experiments and wrote the paper. S.V. and H.Z. performed the animal experiments and analyzed the data. M.P.D. conducted the human subject study with guidance from A.M.O. and W.A.W. M.P.D., B.B., and P.Z. analyzed human data. K.L.P., C.C., S.D.C., D.R., I.K.M., G.L.R., and E.C.H. contributed to the animal experiments. M.P.D., H.Z., K.L.P., W.A.W., I.K.M., and H.F.K. provided critical insights. J.R.C. helped with the metabolism experiments, and C.W.B. helped in establishing cell lines. S.L., K.P., and H.F.K. enabled synthesis of the radioligand. Competing interests: C.B.T. is cofounder of Agios Pharmaceuticals and has financial interest in Agios. He is also a director of Charles River Laboratories and Merck Pharmaceuticals and owns stocks in these companies. H.F.K. is the CEO of Five Eleven Pharma, which has no conflict of interest with the subject matter in this manuscript. Data and materials availability: Patents have been filed concerning F-glutamine PET tracer preparation and imaging. These patents do not restrict the research or noncommercial use of the probe or technique.

Authors

Affiliations

Sriram Venneti*, [email protected]
Department of Pathology, University of Michigan, Ann Arbor, MI 41809, USA.
Mark P. Dunphy*
Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY 10065, USA.
Hanwen Zhang*
Molecular Pharmacology and Chemistry Program, MSKCC, New York, NY 10065, USA.
Kenneth L. Pitter
Cancer Biology and Genetics Program, MSKCC, New York, NY 10065, USA.
Patrick Zanzonico
Medical Physics, MSKCC, New York, NY 10065, USA.
Carl Campos
Human Oncology and Pathogenesis Program, MSKCC, New York, NY 10065, USA.
Sean D. Carlin
Radiochemistry and Imaging Sciences Service, Department of Radiology, MSKCC, New York, NY 10065, USA.
Gaspare La Rocca
Cancer Biology and Genetics Program, MSKCC, New York, NY 10065, USA.
Serge Lyashchenko
Radiochemistry and Molecular Imaging Probe Core, MSKCC, New York, NY 10065, USA.
Karl Ploessl
Departments of Radiology and Pharmacology, University of Pennsylvania, Philadelphia, PA 19104, USA.
Daniel Rohle
Cancer Biology and Genetics Program, MSKCC, New York, NY 10065, USA.
Human Oncology and Pathogenesis Program, MSKCC, New York, NY 10065, USA.
Antonio M. Omuro
Department of Neurology, MSKCC, New York, NY 10065, USA.
Justin R. Cross
Donald B. and Catherine C. Marron Cancer Metabolism Center, MSKCC, New York, NY 10065, USA.
Cameron W. Brennan
Medical Physics, MSKCC, New York, NY 10065, USA.
Department of Neurosurgery, MSKCC, New York, NY 10065, USA.
Wolfgang A. Weber
Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY 10065, USA.
Molecular Pharmacology and Chemistry Program, MSKCC, New York, NY 10065, USA.
Eric C. Holland
Director, Solid Tumor Translational Research, Division of Human Biology, Fred Hutchinson Cancer Research Center, and Alvord Brain Tumor Center, University of Washington, Seattle, WA 98109, USA.
Ingo K. Mellinghoff
Human Oncology and Pathogenesis Program, MSKCC, New York, NY 10065, USA.
Department of Neurology, MSKCC, New York, NY 10065, USA.
Hank F. Kung
Departments of Radiology and Pharmacology, University of Pennsylvania, Philadelphia, PA 19104, USA.
Jason S. Lewis [email protected]
Molecular Pharmacology and Chemistry Program, MSKCC, New York, NY 10065, USA.
Radiochemistry and Imaging Sciences Service, Department of Radiology, MSKCC, New York, NY 10065, USA.
Radiochemistry and Molecular Imaging Probe Core, MSKCC, New York, NY 10065, USA.
Craig B. Thompson [email protected]
Cancer Biology and Genetics Program, MSKCC, New York, NY 10065, USA.
Human Oncology and Pathogenesis Program, MSKCC, New York, NY 10065, USA.

Notes

*
These authors contributed equally to this work.
Corresponding authors. E-mail: [email protected] (S.V.); [email protected] (C.B.T.); [email protected] (J.S.L.)

Metrics & Citations

Metrics

Article Usage

Altmetrics

Citations

Cite as

Export citation

Select the format you want to export the citation of this publication.

Cited by

  1. Integrin α6 targeted cancer imaging and therapy, Visualized Cancer Medicine, 4, (4), (2023).https://doi.org/10.1051/vcm/2022007
    Crossref
  2. Characteristic Evaluation of a 11 C-Labeled Leucine Analog, l -α-[5- 11 C]methylleucine, as a Tracer for Brain Tumor Imaging by Positron Emission Tomography , Molecular Pharmaceutics, 20, 3, (1842-1849), (2023).https://doi.org/10.1021/acs.molpharmaceut.2c01069
    Crossref
  3. What is cancer metabolism?, Cell, 186, 8, (1670-1688), (2023).https://doi.org/10.1016/j.cell.2023.01.038
    Crossref
  4. Imaging Techniques in Pharmacological Precision Medicine, , (2023).https://doi.org/10.1007/164_2023_641
    Crossref
  5. Rewired Metabolism of Amino Acids and Its Roles in Glioma Pathology, Metabolites, 12, 10, (918), (2022).https://doi.org/10.3390/metabo12100918
    Crossref
  6. A Comparison of PET Tracers in Recurrent High-Grade Gliomas: A Systematic Review, International Journal of Molecular Sciences, 24, 1, (408), (2022).https://doi.org/10.3390/ijms24010408
    Crossref
  7. Application of Metabolic Reprogramming to Cancer Imaging and Diagnosis, International Journal of Molecular Sciences, 23, 24, (15831), (2022).https://doi.org/10.3390/ijms232415831
    Crossref
  8. Monitoring of Current Cancer Therapy by Positron Emission Tomography and Possible Role of Radiomics Assessment, International Journal of Molecular Sciences, 23, 16, (9394), (2022).https://doi.org/10.3390/ijms23169394
    Crossref
  9. 7T HR FID-MRSI Compared to Amino Acid PET: Glutamine and Glycine as Promising Biomarkers in Brain Tumors, Cancers, 14, 9, (2163), (2022).https://doi.org/10.3390/cancers14092163
    Crossref
  10. Advancing Cancer Treatment by Targeting Glutamine Metabolism—A Roadmap, Cancers, 14, 3, (553), (2022).https://doi.org/10.3390/cancers14030553
    Crossref
  11. See more
Loading...

View Options

Check Access

Log in to view the full text

AAAS ID LOGIN

AAAS login provides access to Science for AAAS Members, and access to other journals in the Science family to users who have purchased individual subscriptions.

Log in via OpenAthens.
Log in via Shibboleth.

More options

Register for free to read this article

As a service to the community, this article is available for free. Login or register for free to read this article.

View options

PDF format

Download this article as a PDF file

Download PDF

Full Text

FULL TEXT

Media

Figures

Multimedia

Tables

Share

Share

Share article link

Share on social media