[11C]Methionine and [11C]PBR28 as PET Imaging Tracers to Differentiate Metastatic Tumor Recurrence or Radiation Necrosis

Thuy T Tran, Jean-Dominique Gallezot, Lucia B Jilaveanu, Christopher Zito, Gabriela Turcu, Keunpoong Lim, Nabeel Nabulsi, Henry Huang, Anita Huttner, Harriet M Kluger, Veronica L Chiang, Richard Carson, Thuy T Tran, Jean-Dominique Gallezot, Lucia B Jilaveanu, Christopher Zito, Gabriela Turcu, Keunpoong Lim, Nabeel Nabulsi, Henry Huang, Anita Huttner, Harriet M Kluger, Veronica L Chiang, Richard Carson

Abstract

Purpose: As stereotactic radiosurgery (SRS) and immunotherapy are increasingly used to treat brain metastases, incidence of radiation necrosis (RN) is consequently rising. Differentiating tumor regrowth (TR) from RN is vital in management but difficult to assess using MRI. We hypothesized that tumor methionine levels would be elevated given increased metabolism and high amino acid uptake, whereas RN would increase inflammation marked by upregulated translocator protein (PBR-TSPO), which can be quantified with specific PET tracers.

Procedures: We performed a feasibility study to prospectively evaluate [11C]methionine and [11C]PBR28 using PET in 5 patients with 7 previously SRS-treated brain metastases demonstrating regrowth to differentiate TR from RN.

Results: Sequential imaging with dual tracers was well-tolerated. [11C]methionine was accurate for detecting pathologically confirmed TR in 7/7 lesions, whereas [11C]PBR28 was only accurate in 3/7 lesions. Tumor PBR-TSPO expression was elevated in both melanoma and lung cancer cells, contributing to lack of specificity of [11C]PBR28-PET.

Conclusion: Sequential use of PET tracers is safe and effective. [11C]Methionine was a reliable TR marker, but [11C]PBR28 was not a reliable marker of RN. Studies are needed to determine the causes of post-radiation inflammation and identify specific markers of RN to improve diagnostic imaging.

Trial registration: ClinicalTrials.gov NCT02433171.

Keywords: PET; [11C]PBR28; [11C]methionine; brain metastasis; radiation necrosis.

Conflict of interest statement

Declaration of Conflicting Interests: The author(s) declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article. Dr. Harriet Kluger reports research grants from Merck, Bristol-Myers Squibb, and Apexigen during the conduct of the study, and personal fees from Merck, Regeneron, Alexion, Prometheus, Corvus, Nektar, Biodesix, Roche-Genetech, Pfizer, Iovance, Immunocore, Array Biopharma, and Celldex, outside of the submitted work. Dr. Chiang reports personal fees from Brainlab, Monteris Medical, and MRI Interventions outside of the submitted work. The other authors declare no conflicts of interest.

Figures

Figure 1.
Figure 1.
MRI and PET imaging for [11C]MET and [11C]PBR28. A, Representative images of suspicious lesions for TR or RN as seen on post-gadolinium MRI sequences. Corresponding lesions as seen on PET for [11C]MET and [11C]PBR28. B, Patient 2 had histologically confirmed TR, but had uptake of both [11C]MET and [11C]PBR28 radiotracers. Patient 3 had histologically confirmed RN but had absent uptake of both [11C]MET and [11C]PBR28 radiotracers. Patient 5 had histologically confirmed RN and demonstrated uptake of only [11C]PBR28. Tumor is outlined in a dashed line (top photo, taken at 20X). Characteristic features of RN include vessel hyalinization (arrows), increased immune infiltrate (arrowhead) (bottom photo, taken at 10X), and paucicelluar coagulative necrosis (*) (middle photo, taken at 4X, and bottom photo).
Figure 2.
Figure 2.
Quantitation of PBR-TSPO in NSCLC and melanoma. (A-B) PBR, also known as TSPO, protein is present at high levels in human monocytes, but expression is decreased in human NSCLC (P = 0.0038 by t test, mean 0.38 ± 0.045 standard error of the mean [SEM]) and human melanoma (P = 0.0008, 0.40 ± 0.089) cell lines. (C) Human NSCLC (n = 28) and (D) melanoma (n = 13) tissue microarrays were stained with either cytokeratin or S100, respectively, to create a tumor mask. PBR-TSPO immunofluorescence staining intensity was quantitated in tumor cells by AQUA. Representative sections demonstrating tissue with higher and lower PBR-TSPO expression is shown for NSCLC and melanoma cases.

References

    1. Miyatake S, Nonoguchi N, Furuse M, et al. Pathophysiology, diagnosis, and treatment of radiation necrosis in the brain. Neurol Med Chir (Tokyo). 2015;55(1):50–59. doi:10.2176/nmc.ra.2014-0188
    1. Vellayappan B, Tan CL, Yong C, et al. Diagnosis and management of radiation necrosis in patients with brain metastases. Front Oncol. 2018;8:395 doi:10.3389/fonc.2018.00395
    1. Gotz I, Grosu AL. [(18)F]FET-PET imaging for treatment and response monitoring of radiation therapy in malignant glioma patients—a review. Front Oncol. 2013;3:104 doi:10.3389/fonc.2013.00104
    1. Sogani SK, Jena A, Taneja S, et al. Potential for differentiation of glioma recurrence from radionecrosis using integrated (18)F-fluoroethyl-L-tyrosine (FET) positron emission tomography/magnetic resonance imaging: a prospective evaluation. Neurol India. 2017;65(2):293–301. doi:10.4103/neuroindia.NI_101_16
    1. Piroth MD, Liebenstund S, Galldiks N, et al. Monitoring of radiochemotherapy in patients with glioblastoma using O-(2-(1)(8)Fluoroethyl)-L-tyrosine positron emission tomography: is dynamic imaging helpful? Mol Imaging. 2013;12(6):388–395.
    1. Lohmann P, Kocher M, Ceccon G, et al. Combined FET PET/MRI radiomics differentiates radiation injury from recurrent brain metastasis. Neuroimage Clin. 2018;20:537–542. doi:10.1016/j.nicl.2018.08.024
    1. Zikou A, Sioka C, Alexiou GA, et al. Radiation necrosis, pseudoprogression, pseudoresponse, and tumor recurrence: imaging challenges for the evaluation of treated gliomas. Contrast Media Mol Imaging. 2018;2018:6828396 /01/11. doi:10.1155/2018/6828396
    1. Glaudemans AW, Enting RH, Heesters MA, et al. Value of 11C-methionine PET in imaging brain tumours and metastases. Eur J Nucl Med Mol Imaging. 2013;40(4):615–635. doi:10.1007/s00259-012-2295-5
    1. Yomo S, Oguchi K. Prospective study of (11)C-methionine PET for distinguishing between recurrent brain metastases and radiation necrosis: limitations of diagnostic accuracy and long-term results of salvage treatment. BMC Cancer. 2017;17(1):713 doi:10.1186/s12885-017-3702-x
    1. Garcia JR, Cozar M, Baquero M, et al. The value of (11)C-methionine PET in the early differentiation between tumour recurrence and radionecrosis in patients treated for a high-grade glioma and indeterminate MRI. Rev Esp Med Nucl Imagen Mol. 2017;36(2):85–90. doi:10.1016/j.remn.2016.06.002
    1. Deng SM, Zhang B, Wu YW, et al. Detection of glioma recurrence by (1)(1)C-methionine positron emission tomography and dynamic susceptibility contrast-enhanced magnetic resonance imaging: a meta-analysis. Nucl Med Commun. 2013;34(8):758–766. doi:10.1097/MNM.0b013e328361f598
    1. Nihashi T, Dahabreh IJ, Terasawa T. Diagnostic accuracy of PET for recurrent glioma diagnosis: a meta-analysis. AJNR Am J Neuroradiol. 2013;34(5):944–950 , S941-911. doi:10.3174/ajnr.A3324
    1. Law I, Albert NL, Arbizu J, et al. Joint EANM/EANO/RANO practice guidelines/SNMMI procedure standards for imaging of gliomas using PET with radiolabelled amino acids and [(18)F]FDG: version 1.0. Eur J Nucl Med Mol Imaging. 2019;46(3):540–557. doi:10.1007/s00259-018-4207-9
    1. Werry EL, Bright FM, Piguet O, et al. Recent developments in TSPO PET imaging as a biomarker of neuroinflammation in neurodegenerative disorders. Int J Mol Sci. 2019;20(13):3161 doi:10.3390/ijms20133161
    1. Ruksha T, Aksenenko M, Papadopoulos V. Role of translocator protein in melanoma growth and progression. Arch Dermatol Res. 2012;304(10):839–845. doi:10.1007/s00403-012-1294-5
    1. Han Z, Slack RS, Li W, Papadopoulos V. Expression of peripheral benzodiazepine receptor (PBR) in human tumors: relationship to breast, colorectal, and prostate tumor progression. J Recept Signal Transduct Res. 2003;23(2-3):225–238. doi:10.1081/RRS-120025210
    1. Lumniczky K, Szatmari T, Safrany G. Ionizing radiation-induced immune and inflammatory reactions in the brain. Front Immunol. 2017;8:517 doi:10.3389/fimmu.2017.00517
    1. Cuccurullo V, Di Stasio GD, Cascini GL, et al. The molecular effects of ionizing radiations on brain cells: radiation necrosis vs. tumor recurrence. Diagnostics (Basel). 2019;9(4):127 doi:10.3390/diagnostics9040127
    1. Chen MK, Guilarte TR. Translocator protein 18 kDa (TSPO): molecular sensor of brain injury and repair. Pharmacol Ther. 2008;118(1):1–17. doi:10.1016/j.pharmthera.2007.12.004
    1. Alomari A, Rauch PJ, Orsaria M, et al. Radiologic and histologic consequences of radiosurgery for brain tumors. J Neurooncol. 2014;117(1):33–42. doi:10.1007/s11060-014-1359-8
    1. Kreisl WC, Fujita M, Fujimura Y, et al. Comparison of [(11)C]-(R)-PK 11195 and [(11)C]PBR28, two radioligands for translocator protein (18 kDa) in human and monkey: implications for positron emission tomographic imaging of this inflammation biomarker. NeuroImage. 2010;49(4):2924–2932. doi:10.1016/j.neuroimage.2009.11.056
    1. Deloar HM, Fujiwara T, Nakamura T, et al. Estimation of internal absorbed dose of L-[methyl-11C]methionine using whole-body positron emission tomography. Eur J Nucl Med. 1998;25(6):629–633.
    1. Brown AK, Fujita M, Fujimura Y, et al. Radiation dosimetry and biodistribution in monkey and man of 11C-PBR28: a PET radioligand to image inflammation. J Nucl Med. 2007;48(12):2072–2079. doi:10.2967/jnumed.107.044842
    1. Jin X, Mulnix T, Gallezot JD, Carson RE. Evaluation of motion correction methods in human brain PET imaging—a simulation study based on human motion data. Med Phys. 2013;40(10):10250 3. doi:10.1118/1.4819820
    1. Carson RE, Barker W, Liow J-S, et al. Design of a motion-compensation OSEM list-mode algorithm for resolution-recovery reconstruction of the HRRT. IEEE Nucl Sci Symp Conf Rec. 2003; M16–6.
    1. Patlak CS, Blasberg RG. Graphical evaluation of blood-to-brain transfer constants from multiple-time uptake data. Generalizations. J Cereb Blood Flow Metab. 1985;5(4):584–590. doi:10.1038/jcbfm.1985.87
    1. Patlak CS, Blasberg RG, Fenstermacher JD. Graphical evaluation of blood-to-brain transfer constants from multiple-time uptake data. J Cereb Blood Flow Metab. 1983;3(1):1–7. doi: 10.1038/jcbfm.1983.1
    1. Ichise M, Toyama H, Innis RB, Carson RE. Strategies to improve neuroreceptor parameter estimation by linear regression analysis. J Cereb Blood Flow Metab. 2002;22(10):1271–1281. doi:10.1097/00004647-200210000-00015
    1. Ichise M, Liow J-S, Lu J-Q, et al. Linearized reference tissue parametric imaging methods: application to [11C]DASB positron emission tomography studies of the serotonin transporter in human brain. J Cereb Blood Flow Metab. 2003;23(9):1096–1112. doi:10.1097/
    1. Camp RL, Chung GG, Rimm DL. Automated subcellular localization and quantification of protein expression in tissue microarrays. Nat Med. 2002;8(11):1323–1327. doi:10.1038/nm791
    1. Owen DR, Yeo AJ, Gunn RN, et al. An 18-kDa translocator protein (TSPO) polymorphism explains differences in binding affinity of the PET radioligand PBR28. J Cereb Blood Flow Metab. 2012;32(1):1–5. doi:10.1038/jcbfm.2011.147
    1. Bhoola NH, Mbita Z, Hull R, Dlamini Z. Translocator protein (TSPO) as a potential biomarker in human cancers. Int J Mol Sci. 2018;19(8):2176 doi:10.3390/ijms19082176
    1. Donabedian PL, Kossatz S, Engelbach JA, et al. Discriminating radiation injury from recurrent tumor with [(18)F]PARPi and amino acid PET in mouse models. EJNMMI Res. 2018;8(1):59 doi:10.1186/s13550-018-0399-z
    1. Furumoto S, Shinbo R, Iwata R, et al. In vitro and in vivo characterization of 2-deoxy-2-18F-fluoro-D-mannose as a tumor-imaging agent for PET. J Nucl Med. 2013;54(8):1354–1361. doi:10.2967/jnumed.112.113571
    1. Tjuvajev JG, Macapinlac HA, Daghighian F, et al. Imaging of brain tumor proliferative activity with iodine-131-iododeoxyuridine. J Nucl Med. 1994;35(9):1407–1417.
    1. Liu Y, Carpenter AB, Pirozzi CJ, et al. Non-invasive sensitive brain tumor detection using dual-modality bioimaging nanoprobe. Nanotechnology. 2019;30(27):275101 doi:10.1088/1361-6528/ab0e9c
    1. Weber W, Bartenstein P, Gross MW, et al. Fluorine-18-FDG PET and iodine-123-IMT SPECT in the evaluation of brain tumors. J Nucl Med. 1997;38(5):802–808.

Source: PubMed

3
Iratkozz fel