Pilot Preclinical and Clinical Evaluation of (4S)-4-(3-[18F]Fluoropropyl)-L-Glutamate (18F-FSPG) for PET/CT Imaging of Intracranial Malignancies

Erik S Mittra, Norman Koglin, Camila Mosci, Meena Kumar, Aileen Hoehne, Khun Visith Keu, Andrei H Iagaru, Andre Mueller, Mathias Berndt, Santiago Bullich, Matthias Friebe, Heribert Schmitt-Willich, Volker Gekeler, Lüder M Fels, Claudia Bacher-Stier, Dae Hyuk Moon, Frederick T Chin, Andrew W Stephens, Ludger M Dinkelborg, Sanjiv S Gambhir, Erik S Mittra, Norman Koglin, Camila Mosci, Meena Kumar, Aileen Hoehne, Khun Visith Keu, Andrei H Iagaru, Andre Mueller, Mathias Berndt, Santiago Bullich, Matthias Friebe, Heribert Schmitt-Willich, Volker Gekeler, Lüder M Fels, Claudia Bacher-Stier, Dae Hyuk Moon, Frederick T Chin, Andrew W Stephens, Ludger M Dinkelborg, Sanjiv S Gambhir

Abstract

Purpose: (S)-4-(3-[18F]Fluoropropyl)-L-glutamic acid (18F-FSPG) is a novel radiopharmaceutical for Positron Emission Tomography (PET) imaging. It is a glutamate analogue that can be used to measure xC- transporter activity. This study was performed to assess the feasibility of 18F-FSPG for imaging orthotopic brain tumors in small animals and the translation of this approach in human subjects with intracranial malignancies.

Experimental design: For the small animal study, GS9L glioblastoma cells were implanted into brains of Fischer rats and studied with 18F-FSPG, the 18F-labeled glucose derivative 18F-FDG and with the 18F-labeled amino acid derivative 18F-FET. For the human study, five subjects with either primary or metastatic brain cancer were recruited (mean age 50.4 years). After injection of 300 MBq of 18F-FSPG, 3 whole-body PET/Computed Tomography (CT) scans were obtained and safety parameters were measured. The three subjects with brain metastases also had an 18F-FDG PET/CT scan. Quantitative and qualitative comparison of the scans was performed to assess kinetics, biodistribution, and relative efficacy of the tracers.

Results: In the small animals, the orthotopic brain tumors were visualized well with 18F-FSPG. The high tumor uptake of 18F-FSPG in the GS9L model and the absence of background signal led to good tumor visualization with high contrast (tumor/brain ratio: 32.7). 18F-FDG and 18F-FET showed T/B ratios of 1.7 and 2.8, respectively. In the human pilot study, 18F-FSPG was well tolerated and there was similar distribution in all patients. All malignant lesions were positive with 18F-FSPG except for one low-grade primary brain tumor. In the 18F-FSPG-PET-positive tumors a similar T/B ratio was observed as in the animal model.

Conclusions: 18F-FSPG is a novel PET radiopharmaceutical that demonstrates good uptake in both small animal and human studies of intracranial malignancies. Future studies on larger numbers of subjects and a wider array of brain tumors are planned.

Trial registration: ClinicalTrials.gov NCT01186601.

Conflict of interest statement

Competing Interests: The authors have the following interests: The study was sponsored by Bayer Healthcare. NK, AM, MB, HSW, VG, MF, LMD, AWS, LMF, and CBS were employed by Bayer Pharma AG at the time of study conduct. NK, AM, MB, SB, HSW, MF, LMD, and AWS are currently employed by Piramal Imaging GmbH. On the basis of prior work, patent applications were filed from Bayer to cover the compound investigated here. The compound rights were transferred to Piramal Imaging. The investigated compound is currently in clinical development and is not marketed. NK, AM, MB, HSW, AWS, MF, LMD are co-inventors of the compound and/or have ownership interests in Piramal Imaging. DHM, ESM and SSG received research grants from Bayer Healthcare and Piramal Imaging related to this compound. This does not alter the authors' adherence to all the PLOS ONE policies on sharing data and materials, as detailed online in the guide for authors.

Figures

Fig 1. CONSORT Participant Flow Diagram.
Fig 1. CONSORT Participant Flow Diagram.
Fig 2
Fig 2
PET imaging of rat GS9L brain tumors with (A) 18F-FSPG (B) 18F-FDG and (C) 18F-FET. Selected sagittal (upper images) and coronal (lower images) slices are shown (dose: 15 MBq each, imaging time: 0–30 min p.i.).
Fig 3. Time-activity curves of 18F-FSPG, 18F-FDG…
Fig 3. Time-activity curves of 18F-FSPG, 18F-FDG and 18F-FET tumor-uptake ratio in orthotopic GS9L rat brain tumor lesions.
A volume of interest (VOI) analysis over time was performed from the tumor lesions and the ratio calculated using the VOI from the healthy brain for each tracer. 18F-FSPG showed increasing VOI ratio activity over time. In contrast, 18F-FDG and 18F-FET VOI ratios remained rather constant and at a lower level.
Fig 4. Comparison of axial post-contrast T1…
Fig 4. Comparison of axial post-contrast T1 MRI (left column) and 18F-FSPG PET (right column) in the two subjects with primary brain tumor.
Subject A has recurrent high-grade glioblastoma, while subject B has an enlarging, partially resected low-grade oligodendroglioma. In each case, the arrows point to the primary lesions. For subject A, there is intense accumulation of 18F-FSPG in the enhancing lesion (SUV 5.2), while in subject B, there is no accumulation of the radiotracer (SUV 0.7) in the non-enhancing lesion. Comparatively, the background brain SUV is 0.1 for both subjects. There is incidental note of prominent physiologic uptake of 18F-FSPG in the scalp.
Fig 5. 18F-FSPG PET images, in comparison…
Fig 5. 18F-FSPG PET images, in comparison with MRI and 18F-FDG PET images of the three patients with lung cancer metastases to the brain.
For each subject, the whole-body Maximum Intensity Projection (MIP) image of the 18F-FSPG PET scan is shown on the left. In the right column, the axial images through the level of the brain metastasis include the post-contrast T1 MRI (top), 18F-FSPG PET (middle), and 18F-FDG PET (bottom). Physiologic distribution to normal organs are highlighted on the MIP image for subject A, including the liver (l), pancreas (p), kidneys (k), and bladder (b). For the first two subjects (A, B), the small lesions (below 1.5 cm) are clearly visible on MRI and with 18F-FSPG (SUV-A: 11.0, SUV-B: 4.7). With 18F-FDG, however, there is no discernible activity in these lesions. The larger lesion for subject C, who had previously been treated in this region, is again clearly discernable with MRI although the etiology of the enhancement was unclear whether representing residual/recurrent disease versus post-therapy changes. Both the 18F-FDG (SUV 10.1) and 18F-FSPG PET (SUV 21.8) are positive for this subject, but the accumulation of the latter is stronger, and further enhanced by the lack of uptake in the surrounding normal brain tissue (FDG SUV 4.8, FSPG SUV 0.1).

References

    1. Howlader N NA, Krapcho M, Neyman N, Aminou R, Waldron W, Altekruse SF, Kosary CL, Ruhl J, Tatalovich Z, Cho H, Mariotto A, Eisner MP, Lewis DR, Chen HS, Feuer EJ, Cronin KA, Edwards BK (eds). SEER Cancer Statistics Review, 1975–2008. National Cancer Institute Bethesda, MD [Internet]. 2008. Available: .
    1. Marshall D, Mitchell DA, Graner MW, Bigner DD. Immunotherapy of brain tumors. Handb Clin Neurol. 2012;104:309–30. Epub 2012/01/11. 10.1016/b978-0-444-52138-5.00020-7 .
    1. Parney IF, Berger MS. Principles of brain tumor surgery. Handb Clin Neurol. 2012;104:187–213. Epub 2012/01/11. 10.1016/b978-0-444-52138-5.00015-3 .
    1. Penas-Prado M, Armstrong TS, Gilbert MR. Glioblastoma. Handb Clin Neurol. 2012;105:485–506. Epub 2012/01/11. 10.1016/b978-0-444-53502-3.00004-5 .
    1. Ferguson SD. Malignant gliomas: diagnosis and treatment. Dis Mon. 2011;57(10):558–69. Epub 2011/11/01. 10.1016/j.disamonth.2011.08.020 .
    1. Cha S. Neuroimaging in neuro-oncology. Neurotherapeutics. 2009;6(3):465–77. Epub 2009/06/30. 10.1016/j.nurt.2009.05.002 .
    1. Alexiou GA, Tsiouris S, Kyritsis AP, Voulgaris S, Argyropoulou MI, Fotopoulos AD. Glioma recurrence versus radiation necrosis: accuracy of current imaging modalities. J Neurooncol. 2009;95(1):1–11. Epub 2009/04/22. 10.1007/s11060-009-9897-1 .
    1. Heiss WD, Raab P, Lanfermann H. Multimodality assessment of brain tumors and tumor recurrence. J Nucl Med. 2011;52(10):1585–600. Epub 2011/08/16. 10.2967/jnumed.110.084210 .
    1. Fahlbusch R, Samii A. A review of cranial imaging techniques: potential and limitations. Clin Neurosurg. 2007;54:100–4. Epub 2008/05/29. .
    1. la Fougere C, Suchorska B, Bartenstein P, Kreth FW, Tonn JC. Molecular imaging of gliomas with PET: opportunities and limitations. Neuro Oncol. 2011;13(8):806–19. Epub 2011/07/16. 10.1093/neuonc/nor054
    1. Heiss WD. Clinical Impact of Amino Acid PET in Gliomas. J Nucl Med. 2014. Epub 2014/07/10. 10.2967/jnumed.114.142661 .
    1. Venneti S, Dunphy MP, Zhang H, Pitter KL, Zanzonico P, Campos C, et al. Glutamine-based PET imaging facilitates enhanced metabolic evaluation of gliomas in vivo. Sci Transl Med. 2015;7(274):274ra17 Epub 2015/02/13. 10.1126/scitranslmed.aaa1009 .
    1. Gao H, Jiang X. Progress on the diagnosis and evaluation of brain tumors. Cancer Imaging. 2013;13(4):466–81. Epub 2013/12/18. 10.1102/1470-7330.2013.0039 ; PubMed Central PMCID: PMCPmc3864167.
    1. Watkins S, Robel S, Kimbrough IF, Robert SM, Ellis-Davies G, Sontheimer H. Disruption of astrocyte-vascular coupling and the blood-brain barrier by invading glioma cells. Nat Commun. 2014;5:4196 Epub 2014/06/20. 10.1038/ncomms5196 ; PubMed Central PMCID: PMCPmc4127490.
    1. Robert SM, Sontheimer H. Glutamate transporters in the biology of malignant gliomas. Cell Mol Life Sci. 2014;71(10):1839–54. Epub 2013/11/28. 10.1007/s00018-013-1521-z ; PubMed Central PMCID: PMCPmc3999209.
    1. Lewerenz J, Klein M, Methner A. Cooperative action of glutamate transporters and cystine/glutamate antiporter system Xc- protects from oxidative glutamate toxicity. J Neurochem. 2006;98(3):916–25. Epub 2006/06/15. 10.1111/j.1471-4159.2006.03921.x .
    1. Robert SM, Buckingham SC, Campbell SL, Robel S, Holt KT, Ogunrinu-Babarinde T, et al. SLC7A11 expression is associated with seizures and predicts poor survival in patients with malignant glioma. Sci Transl Med. 2015;7(289):289ra86 10.1126/scitranslmed.aaa8103
    1. Koglin N, Mueller A, Berndt M, Schmitt-Willich H, Toschi L, Stephens AW, et al. Specific PET imaging of xC- transporter activity using a (18)F-labeled glutamate derivative reveals a dominant pathway in tumor metabolism. Clin Cancer Res. 2011;17(18):6000–11. Epub 2011/07/14. 10.1158/1078-0432.CCR-11-0687 .
    1. Smolarz K, Krause BJ, Graner FP, Wagner FM, Hultsch C, Bacher-Stier C, et al. (S)-4-(3-18F-fluoropropyl)-L-glutamic acid: an 18F-labeled tumor-specific probe for PET/CT imaging—dosimetry. J Nucl Med. 2013;54(6):861–6. Epub 2013/04/10. 10.2967/jnumed.112.112581 .
    1. Baek S, Choi CM, Ahn SH, Lee JW, Gong G, Ryu JS, et al. Exploratory clinical trial of (4S)-4-(3-[18F]fluoropropyl)-L-glutamate for imaging xC- transporter using positron emission tomography in patients with non-small cell lung or breast cancer. Clin Cancer Res. 2012;18(19):5427–37. Epub 2012/08/16. 10.1158/1078-0432.ccr-12-0214 .
    1. Baek S, Mueller A, Lim YS, Lee HC, Lee YJ, Gong G, et al. (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. 2013;54(1):117–23. Epub 2012/12/13. 10.2967/jnumed.112.108704 .
    1. Webster JM, Morton CA, Johnson BF, Yang H, Rishel MJ, Lee BD, et al. Functional imaging of oxidative stress with a novel PET imaging agent, 18F-5-fluoro-L-aminosuberic acid. J Nucl Med. 2014;55(4):657–64. Epub 2014/03/01. 10.2967/jnumed.113.126664 ; PubMed Central PMCID: PMCPmc4009729.
    1. Iagaru A, Kundu R, Jadvar H, Nagle D. Evaluation by 18F-FDG-PET of patients with anal squamous cell carcinoma. Hell J Nucl Med. 2009;12(1):26–9. Epub 2009/03/31. .
    1. Iagaru AH, Mittra ES, McDougall IR, Quon A, Gambhir SS. 18F-FDG PET/CT evaluation of patients with ovarian carcinoma. Nucl Med Commun. 2008;29(12):1046–51. Epub 2008/11/07. 10.1097/MNM.0b013e32831089cb
    1. Heiss P, Mayer S, Herz M, Wester HJ, Schwaiger M, Senekowitsch-Schmidtke R. Investigation of transport mechanism and uptake kinetics of O-(2-[18F]fluoroethyl)-L-tyrosine in vitro and in vivo. J Nucl Med. 1999;40(8):1367–73. Epub 1999/08/18. .
    1. Nedergaard MK, Kristoffersen K, Michaelsen SR, Madsen J, Poulsen HS, Stockhausen MT, et al. The use of longitudinal 18F-FET MicroPET imaging to evaluate response to irinotecan in orthotopic human glioblastoma multiforme xenografts. PLoS One. 2014;9(2):e100009 Epub 2014/06/12. 10.1371/journal.pone.0100009 ; PubMed Central PMCID: PMCPmc4053391.
    1. Takeuchi S, Wada K, Toyooka T, Shinomiya N, Shimazaki H, Nakanishi K, et al. Increased xCT expression correlates with tumor invasion and outcome in patients with glioblastomas. Neurosurgery. 2013;72(1):33–41; discussion Epub 2012/10/26. 10.1227/NEU.0b013e318276b2de .
    1. Prabhu A, Sarcar B, Kahali S, Yuan Z, Johnson JJ, Adam KP, et al. Cysteine catabolism: a novel metabolic pathway contributing to glioblastoma growth. Cancer Res. 2014;74(3):787–96. Epub 2013/12/20. 10.1158/0008-5472.can-13-1423 .
    1. Ishimoto T, Nagano O, Yae T, Tamada M, Motohara T, Oshima H, et al. CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc(-) and thereby promotes tumor growth. Cancer Cell. 2011;19(3):387–400. Epub 2011/03/15. 10.1016/j.ccr.2011.01.038 .
    1. Bridges RJ, Natale NR, Patel SA. System xc cystine/glutamate antiporter: an update on molecular pharmacology and roles within the CNS. Br J Pharmacol. 2012;165(1):20–34. Epub 2011/05/14. 10.1111/j.1476-5381.2011.01480.x
    1. Lyons SA, Chung WJ, Weaver AK, Ogunrinu T, Sontheimer H. Autocrine glutamate signaling promotes glioma cell invasion. Cancer Res. 2007;67(19):9463–71. Epub 2007/10/03. 10.1158/0008-5472.can-07-2034 ; PubMed Central PMCID: PMCPmc2045073.
    1. Lapa C, Linsenmann T, Monoranu CM, Samnick S, Buck AK, Bluemel C, et al. Comparison of the amino acid tracers 18F-FET and 18F-DOPA in high-grade glioma patients. J Nucl Med. 2014;55(10):1611–6. Epub 2014/08/16. 10.2967/jnumed.114.140608 .

Source: PubMed

3
Sottoscrivi