REVIEW URRENT C OPINION

PET imaging for brain tumor diagnostics Bogdana Suchorska a, Joerg C. Tonn a, and Nathalie L. Jansen b

Purpose of review Brain tumors differ in histology, biology, prognosis and treatment options. Although structural magnetic resonance is still the gold standard for morphological tumor characterization, molecular imaging has gained an increasing importance in assessment of tumor activity and malignancy. Recent findings Amino acid PET is frequently used for surgery and biopsy planning as well as therapy monitoring in suspected primary brain tumors as well as metastatic lesions, whereas 18F-fluorodeoxyglucose (18F-FDG) remains the tracer of choice for evaluation of patients with primary central nervous system lymphoma. Application of somatostatin receptor ligands has improved tumor delineation in skull base meningioma and concurrently opened up new treatment possibilities in recurrent or surgically not assessable tumors. Recent development focuses on the implementation of hybrid PET/MRI as well as on the development of new tracers targeting tumor hypoxia, enzymes involved in neoplastic metabolic pathways and the combination of PET tracers with therapeutic agents. Summary Implementation of molecular imaging in the clinical routine continues to improve management in patients with brain tumors. However, more prospective large sample studies are needed to validate the additional informative value of PET. Keywords brain tumor, PET, therapy monitoring

INTRODUCTION In the past decades, molecular imaging methods have been increasingly implemented in clinical routine for neuro-oncological settings [1]. Although the most widely used radiotracer for metabolic imaging is still 18F-fluorodeoxyglucose (18F-FDG), many other radiopharmaceuticals have been developed, promising higher sensitivity as well as specificity for certain tumor entities [2]. Particularly, 18F-FDG has been replaced by radiolabeled amino acids or their analogues [3], which were shown to overcome the limitations of 18F-FDG. Due to physiologically low uptake in healthy brain tissue, amino acid PET enables a high tumor-to-background contrast, and due to absent or low uptake in inflammatory lesions, it allows for differentiation between tumor tissue and inflammation [4]. Tracer development is an evolving domain which promises to provide new insights and opportunities for brain tumor imaging. In the following, this review will discuss recent publications in the vast rapidly growing field of molecular imaging in brain tumors.

GLIOMA Gliomas constitute a heterogeneous group of histological subtypes: astrocytoma, oligodendroglioma,

mixed oligoastrocytoma and glioblastoma, which, depending on the WHO grade, substantially differ in natural disease course. Low-grade glioma (LGG) typically affects younger adults with a mean onset age of 35 years and has a variably long life expectancy depending on tumor location, size and initial treatment [5]. In contrast, survival probability in high-grade glioma (HGG) WHO III and IV is low. Median overall survival (OS) in glioblastoma, the most malignant of all primary brain tumors, is as short as 12–15 months and treatment options are limited [6]. Anaplastic astrocytoma, although of a slightly better prognosis with a median OS of 2–5 years, mostly leads to a rapid worsening of neurological functions and thus to a reduced life quality. In malignant glioma entities, outcome depends on histological features, performance a

Department of Neurosurgery and bDepartment of Nuclear Medicine, University Hospital, Ludwig Maximilians University, Munich, Germany Correspondence to Dr Bogdana Suchorska, Department of Neurosurgery, University Hospital, Ludwig Maximilians University, Marchioninistr. 15, 81377 Munich, Germany. Tel: +49 89 4400 72693; e-mail: bogdana. [email protected] Curr Opin Neurol 2014, 27:683–688 DOI:10.1097/WCO.0000000000000143

1350-7540 ß 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins

www.co-neurology.com

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Neoplasms

KEY POINTS  Dynamic analysis of amino acid tracer uptake provides further insights into metabolic properties and therapeutic effects in primary and metastatic brain tumors. 

18



68

F-FDG remains the tracer of choice in diagnosis and treatment monitoring of PCNSL. Ga-DODATATE PET becomes increasingly implemented for diagnostic and therapeutic purposes in meningiomas.

status, age at diagnosis and molecular subtypes. Although MRI still remains the keystone for assessment of morphological tumor features, such as location, mass effect and contrast enhancement, PET can provide valuable insights into biology and malignancy of the tumor tissue. Molecular imaging becomes increasingly implemented into surgical treatment planning in suspected LGG, in which identification of possible areas of increased metabolism is most essential to obtain the correct histological diagnosis. Interestingly, areas of increased PET uptake do not necessarily correlate with 5-aminolevulinic acid-induced fluorescence, thus integrating both preoperative PET and 5-aminolevulinic acid into surgery planning might provide additional insights and improve extent of resection [7]. In contrast, HGG patients benefit from an additional treatment monitoring by means of PET which can help identify early therapy failure and thus offers an opportunity for a more timely and individual therapy management.

barrier (BBB), delineation of tumor extent is reported to be more accurate with amino acid PET, and the areas with increased amino acid uptake in PET are often larger than areas with pathological contrast enhancement on MRI [20]. Therefore, amino acid PET can be effectively used for treatment planning, and a better outcome was reported for patients with radiotherapy planning on the basis of tumor extent definition by amino acid uptake [21]. Amino acid PET is not only useful in the initial setting, but also provides important information during the follow-up of glioma patients. It is a valuable tool for treatment monitoring as it helps to assess treatment response, to evaluate patients’ prognosis after therapy and to detect tumor recurrence with high accuracy [4,22,23,24 ,25,26]. The high sensitivity and specificity after different therapies is also because of the independence of the BBB, which is often the limiting factor in post-treatment evaluation of magnetic resonance (MR) images. Following locally aggressive therapies, such as combined radio–chemotherapy or stereotactic brachytherapy, an unspecific contrast enhancement can occur on MRI, which can mimic tumor progression and makes MRI-based treatment monitoring challenging. Contrariwise, the evolvement of antiangiogenic therapies has brought new challenges for conventional MRI by influencing BBB integrity toward mimicking treatment response. Inhibition of neovascularization and reduction of fragile blood vessels result in decreased pathologic contrast enhancement on MRI. Recent studies have reported amino acid PET to detect pseudoresponse in vascular endothelial growth factor antibody-treated patients and to be cost-effective [24 ,27–29,30 ]. In the direct comparison of 18F-DOPA and 18 F-FET, 18F-FET seems to have some advantages. Apart from the absent striatal tracer uptake (which can hamper tumor delineation in this location by 18 F-DOPA), 18F-FET is characterized by typical uptake behavior in LGG and HGG, which can be assessed by dynamic PET acquisition [31–34]. This property of distinguishing different time-activity curves (TACs) is not yet clarified in 18F-DOPA, as there were conflicting reports regarding this point [35 ,36,37]. In addition to tumor grading, TACs seem to provide prognostic information for patient outcome and malignant progression [38 ,39 ,40 ]. &

&

Different tracers: different applications The first promising amino acid that has been investigated in many studies for glioma imaging was 11 C-methionine which was shown to be suitable for glioma imaging [8–11]. However, the short half-life of 11C, requiring an on-site cyclotron, has led to the evolution of 18F-labeled amino acid tracers and brought 18F-3,4-dihydroxyphenylalanine (18F-DOPA) and O-(2–18F-Fluoroethyl)-l-Tyrosine (18FET) into clinical use [12–15]. Interestingly, 18 F-DOPA is widely spread in the USA, whereas 18FET is more common in Europe. Amino acid tracers were shown to be extremely valuable for different clinical settings [16 ,17]. At primary diagnosis, amino acid PET is helpful for detecting anaplastic foci, so-called ‘‘hot-spots’’ [18,19], which is especially useful for biopsy guidance in noncontrast-enhancing glioma. As the tracer uptake does not depend on the blood–brain

&

&&

&

&

&

&

684

www.co-neurology.com

New targets Emerging PET tracers for glioma imaging aim at different targets. The pyrimidine analogue 3’-deoxy3’-18F-fluorothymidine reflects thymidine kinase-1 activity, which is the principle enzyme in the Volume 27  Number 6  December 2014

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

PET imaging for brain tumor diagnostics Suchorska et al.

pathway of DNA synthesis and therefore depicts tumor proliferation rate. Although it is a promising tool for the differentiation between radionecrosis and vital tumor tissue [41,42], and was shown to be predictive of survival after bevacizumab therapy [43], it has limitations for clinical use because of permeability restrictions of the BBB [44]. Tumor hypoxia can be assessed by 18F-fluoromisonidazole. 18 F-Fluoromisonidazole has potential to predict response or resistance to radiotherapy and may be useful for treatment response assessment afterwards [45]. So far, it has mainly been used in a preclinical setting.

Combined PET/MRI: new prospects for tumor characterization Many recent studies focus on direct comparison between amino acid PET and sophisticated MR methods, such as MR spectroscopy, diffusionweighted imaging and perfusion MRI. So far, most studies revealed that the information gained by the different imaging methods is complementary; therefore, combination of all imaging methods might provide the best result for assessment of tumor characteristics [46–52]. The usefulness of combined PET/MR scanners is currently under investigation. Although the effect of image fusion does not play an essential role in the case of brain imaging, in which an accurate image fusion can be easily obtained by image co-registration based on fixed points (e.g., skull, sinus), it offers the chance to deliver simultaneous images, enabling the direct comparison of uptake kinetics and perfusion. However, the best effect might be provided by the introduction of a routinely combined interpretation of PET and MRI information by collaborative reading of a nuclear physician and a radiologist, as it has become the standard in clinical PET/computed tomography evaluation, which will certainly improve report quality in the future. Another important point concerning technical issues that needs to be addressed in future studies is the standardization of PET evaluation. Acquisition (dynamic vs. static, optimal time point after tracer injection and scan duration), reconstruction algorithms and method of analysis of common PET parameters [maximal and mean tumor-to-brain ratios (TBRs) biological tumor volume, region-ofinterest definition for TACs] should be harmonized to assure comparability between studies.

BRAIN METASTASES Early detection of brain metastases provided by advanced screening programs as well as

improvements of treatment in general oncology has positively influenced the course of the disease and OS in patients with metastatic disease [53,54]. With the advent of new therapeutic agents, especially immunogenic and oncogene-derived targeted inhibitors of human epidermal growth factor receptor type 2 (HER2), epidermal growth factor receptor, BRAF, however, new challenges for disease monitoring have risen [55–58]. As the new therapeutic possibilities become more important, there is an increasing demand for new imaging methods which are independent of the applied therapy. The interpretation of findings based on follow-up MRI is difficult, as post-therapeutic changes associated with treatment response can be mistaken for tumor recurrence and vice versa [59]. Unlike new treatment response criteria for solid somatic tumors [Response Evaluation Criteria in Solid Tumors (RECIST)] or updated evaluation criteria for glioma [Response Assessment in Neuro-Oncology (RANO)], the evaluation basis for treated brain metastases still remains uncertain [60–62]. PET has proven to be helpful as far as discrimination of treatment response vs. recurrence after different irradiation modalities is concerned. Current studies include evaluation of viable tumor tissue after radiotherapy and chemotherapy and differentiation of progression from radiation injury [9,63]. As far as the applied tracer is concerned, FDG is not sensitive enough to differentiate vital brain metastasis from unspecific nontumor changes related to therapy [64]. Amino acid PET is more useful as it provides a more precise contrast between normal brain and hypermetabolic tumor tissue [65]. In a recent 18F-FET-PET study, TBRmax and TBRmean as well as TACs were compared for treatment evaluation in 31 treated brain metastases. A good discrimination of TBRmean as well as TAC patterns for differentiation of responders from nonresponders was found [66]. Altogether, dynamic evaluation of tracer uptake seems to provide new promising possibilities in estimation of cerebral tumor burden in analogy to dynamic analyses which have already been performed with 18F-FET and choline PET in glioma [38 ,67]. To satisfy the need for establishment of diagnostic criteria for monitoring disease course of brain metastases after treatment with upcoming current chemotherapeutics, a baseline evaluation of the uptake characteristics of the primary untreated metastases of different histological origin is necessary. An important point for future studies would also be a correlation with established as well as upcoming molecular markers. So far, only few studies have performed a correlation between biomarkers and PET tracer uptake [58]. Dijkers et al. [68]

1350-7540 ß 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins

&

www.co-neurology.com

685

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Neoplasms

used 89Zr to image HER2-positive metastases throughout the body, which revealed previously undetected brain metastases.

published until now; however, those were mostly retrospective or small-cohort, and recent publications are rare.

CENTRAL NERVOUS SYSTEM LYMPHOMA AND ASSOCIATED DISEASES

MENINGIOMA

Primary central nervous system lymphoma (PCNSL), although accounting for only 3–5% of all brain tumors, shows a rising incidence in the non-HIV population, as the life expectancy in the elderly increases [69,70]. Typical presentation on MRI is a large, well enhancing periventricular lesion and the basal ganglia; however, multiple differential diagnoses can present with a similar clinical and imaging picture. PCNSL tends to have a high cellular density and increased glycolytic metabolism and thus shows high uptake of 18F-FDG [71]. However, there is a high interference with 18F-FDG uptake in the basal ganglia, thalamus and grey matter. Furthermore, 18 F-FDG also shows a considerably high uptake in HGG, multiple sclerosis and other inflammatory processes [72,73]. Although a high sensitivity of FDG in differentiation between malignant glioma, metastases and PCNSL has been reported in small cohort studies, its additional use for PCNSL diagnostics remains unclear [74,75]. However, 18F-FDG has proven to be helpful for differentiation between HIV-related opportunistic infections such as toxoplasmosis from PCNSL-related lesions as toxoplasmosis shows a typically decreased 18F-FDG uptake [76]. Furthermore, 18F-FDG has been reported to provide sufficient and prognostic information after methotrexate treatment in PCNSL patients. Treatment response could even be detected earlier than by MRI alone [77,78]. As far as amino acid tracers are concerned, 11MET-PET was the second frequently used tracer, as lymphoma cells are dependent on the external supply of methionine. Thus, 11MET provides a better contrast than 18F-FDG and represents a good option for therapy monitoring after radiotherapy [79]. However, comparing sensitivity and specificity, similar results were obtained in a study comparing 11MET with 18F-FDG in PCNSL [71]. A new promising PET-based treatment approach might be the application of 90Yttrium-radiolabeled or 111Indium-radiolabeled mAbs aimed against cluster of differentiation-20 (CD-20) which can be found in membrane-associated phosphoproteins of B-cellderived PCNSL [80–82]. However, permeation of the BBB and thus the necessity of administration of BBB-disrupting agents are still limiting factors for anti-CD20 immunotherapy in PCNSL. Altogether, numerous studies investigating lymphoma with PET, mainly 18F-FDG, have been 686

www.co-neurology.com

Meningiomas are the most frequently encountered primary brain tumors and usually affect more frequently women, typically after their 3rd decade. In 90% of all cases, they are benign, whereas in up to 10% of patients, atypical and anaplastic tumors occur [83]. Surgical resection is the first treatment whenever possible. However, in selected cases, radiosurgery or fractionated stereotactic radiotherapy is an alternative treatment option [84]. Although morphological imaging by means of computed tomography and MRI is sufficient for surgical and radiation target definition in most cases, skull base or orbital affection as well as recurrent tumor within scar tissue requires a more accurate biological imaging. In the past years, several studies have been conducted applying different amino acid tracers, such as choline, tyrosine and methionine in meningioma [85–87]. However, none has proven superior to standard MRI methods in allowing an additional differentiation between the different WHO grades or to improve tumor-tobone contrast in skull affecting tumors. Recently, although due to the high level of somatostatin receptor subtype 2 expression in meningioma, PET imaging with somatostatin receptor ligands, such as 68Ga tetra-azacyclododecane tetra-acetic acid-octreotate (68Ga-DOTATATE) or 1,4,7,10-tetraazacyclododecane-N,N’,N,N"-tetraacetic-acid-D-Phe1-Tyr3-octreotide (68Ga-DOTATOC), has opened up new possibilities to solve the problem of target delineation in skull base tumors before surgery and prior to radiotherapy [88–90]. Furthermore, 68Ga-DOTATE/TOC not only enables accurate target volume delineation for therapy planning, it also enables treatment delivery when labelled with 90Yttrium or 177Lutetium [91,92] and might disclose a high potential for combined treatment planning and monitoring. Of note, treatment success is linked to WHO grade, as grade I meningioma responds better than higher tumor grades [92].

CONCLUSION Application of molecular imaging has already improved therapy planning and management of brain tumors. However, most studies, published so far, were still small sample-sized, of retrospective nature and lacking comparability because of different acquisition and data evaluation methods. Volume 27  Number 6  December 2014

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

PET imaging for brain tumor diagnostics Suchorska et al.

Further tracers for brain tumor imaging are currently under development. Combination of different tracers might provide more detailed information on tumor biology and course of the disease, thus overcoming currently existing limitations in the near future. Acknowledgements None. Conflicts of interest There are no conflicts of interest.

REFERENCES AND RECOMMENDED READING Papers of particular interest, published within the annual period of review, have been highlighted as: & of special interest && of outstanding interest 1. El-Deiry WS, Sigman CC, Kelloff GJ. Imaging and oncologic drug development. J Clin Oncol 2006; 24:3261–3273. 2. Koopmans KP, Glaudemans AW. Rationale for the use of radiolabelled peptides in diagnosis and therapy. Eur J Nucl Med Mol Imaging 2012; 39 (Suppl 1):S4–S10. 3. Gulyas B, Halldin C. New PET radiopharmaceuticals beyond FDG for brain tumor imaging. Q J Nucl Med Mol Imaging 2012; 56:173–190. 4. Karunanithi S, Sharma P, Kumar A, et al. 18F-FDOPA PET/CT for detection of recurrence in patients with glioma: prospective comparison with 18F-FDG PET/CT. Eur J Nucl Med Mol Imaging 2013; 40:1025–1035. 5. Pignatti F, van den Bent M, Curran D, et al. Prognostic factors for survival in adult patients with cerebral low-grade glioma. J Clin Oncol 2002; 20:2076– 2084. 6. Weller M, van den Bent M, Hopkins K, et al. EANO guideline for the diagnosis and treatment of anaplastic gliomas and glioblastoma. Lancet Oncol 2014; 15:e395–e403. 7. Arita H, Kinoshita M, Kagawa N, et al. 11C-methionine uptake and intraoperative 5-aminolevulinic acid-induced fluorescence as separate index markers of cell density in glioma: a stereotactic image-histological analysis. Cancer 2012; 118:1619–1627. 8. Mosskin M, Ericson K, Hindmarsh T, et al. Positron emission tomography compared with magnetic resonance imaging and computed tomography in supratentorial gliomas using multiple stereotactic biopsies as reference. Acta Radiol 1989; 30:225–232. 9. Terakawa Y, Tsuyuguchi N, Iwai Y, et al. Diagnostic accuracy of 11Cmethionine PET for differentiation of recurrent brain tumors from radiation necrosis after radiotherapy. J Nucl Med 2008; 49:694–699. 10. Ullrich RT, Kracht L, Brunn A, et al. Methyl-L-11C-methionine PET as a diagnostic marker for malignant progression in patients with glioma. J Nucl Med 2009; 50:1962–1968. 11. Yamane T, Sakamoto S, Senda M. Clinical impact of 11C-methionine PET on expected management of patients with brain neoplasm. Eur J Nucl Med Mol Imaging 2010; 37:685–690. 12. Weber WA, Wester HJ, Grosu AL, et al. O-(2-[18F]fluoroethyl)-L-tyrosine and L-[methyl-11C]methionine uptake in brain tumours: initial results of a comparative study. Eur J Nucl Med 2000; 27:542–549. 13. Wester HJ, Herz M, Weber W, et al. Synthesis and radiopharmacology of O-(2-[18F]fluoroethyl)-L-tyrosine for tumor imaging. J Nucl Med 1999; 40:205–212. 14. Bergmann R, Pietzsch J, Fuechtner F, et al. 3-O-methyl-6-18F-fluoro-L-dopa, a new tumor imaging agent: investigation of transport mechanism in vitro. J Nucl Med 2004; 45:2116–2122. 15. Heiss WD, Wienhard K, Wagner R, et al. F-Dopa as an amino acid tracer to detect brain tumors. J Nucl Med 1996; 37:1180–1182. 16. Hutterer M, Nowosielski M, Putzer D, et al. [18F]-fluoro-ethyl-L-tyrosine PET: a & valuable diagnostic tool in neuro-oncology, but not all that glitters is glioma. Neuro Oncol 2013; 15:341–351. Gives a good overview of FET-uptake characteristics in a broad range of brain tumors and nontumoral lesions. 17. Smits A, Baumert BG. The clinical value of PET with amino acid tracers for gliomas WHO grade II. Int J Mol Imaging 2011; 2011:372509. 18. Kunz M, Thon N, Eigenbrod S, et al. Hot spots in dynamic 18FET-PET delineate malignant tumor parts within suspected WHO grade II gliomas. Neuro Oncol 2011; 13:307–316.

19. Galldiks N, Kracht LW, Dunkl V, et al. Imaging of non or very subtle contrastenhancing malignant gliomas with [11C]-methionine positron emission tomography. Mol Imaging 2011; 10:453–459. 20. Pafundi DH, Laack NN, Youland RS, et al. Biopsy validation of 18F-DOPA PET and biodistribution in gliomas for neurosurgical planning and radiotherapy target delineation: results of a prospective pilot study. Neuro Oncol 2013; 15:1058–1067. 21. Grosu AL, Weber WA, Franz M, et al. Reirradiation of recurrent high-grade gliomas using amino acid PET (SPECT)/CT/MRI image fusion to determine gross tumor volume for stereotactic fractionated radiotherapy. Int J Radiat Oncol Biol Phys 2005; 63:511–519. 22. Galldiks N, Langen KJ, Holy R, et al. Assessment of treatment response in patients with glioblastoma using O-(2-18F-fluoroethyl)-L-tyrosine PET in comparison to MRI. J Nucl Med 2012; 53:1048–1057. 23. Karunanithi S, Sharma P, Kumar A, et al. Can 18F-FDOPA PET/CT predict survival in patients with suspected recurrent glioma? A prospective study. Eur J Radiol 2014; 83:219–225. 24. Schwarzenberg J, Czernin J, Cloughesy TF, et al. Treatment response & evaluation using 18F-FDOPA PET in patients with recurrent malignant glioma on bevacizumab therapy. Clin Cancer Res 2014; 20:3550–3559. Prospective study for early treatment evaluation after bevacizumab therapy. 25. Niyazi M, Jansen N, Ganswindt U, et al. Re-irradiation in recurrent malignant glioma: prognostic value of [18F]FET-PET. J Neurooncol 2012; 110:389– 395. 26. Jansen NL, Suchorska B, Schwarz SB, et al. [18F]fluoroethyltyrosine-positron emission tomography-based therapy monitoring after stereotactic iodine-125 brachytherapy in patients with recurrent high-grade glioma. Mol Imaging 2013; 12:137–147. 27. Galldiks N, Filss CP, Goldbrunner R, Langen KJ. Discrepant MR and [18F]fluoroethyl-L-tyrosine PET imaging findings in a patient with bevacizumab failure. Case Rep Oncol 2012; 5:490–494. 28. Hutterer M, Nowosielski M, Putzer D, et al. O-(2-18F-fluoroethyl)-L-tyrosine PET predicts failure of antiangiogenic treatment in patients with recurrent high-grade glioma. J Nucl Med 2011; 52:856–864. 29. Galldiks N, Rapp M, Stoffels G, et al. Response assessment of bevacizumab in patients with recurrent malignant glioma using [18F]Fluoroethyl-L-tyrosine PET in comparison to MRI. Eur J Nucl Med Mol Imaging 2013; 40:22–33. 30. Heinzel A, Muller D, Langen KJ, et al. The use of O-(2-18F-fluoroethyl)-L& tyrosine PET for treatment management of bevacizumab and irinotecan in patients with recurrent high-grade glioma: a cost-effectiveness analysis. J Nucl Med 2013; 54:1217–1222. Interesting insight into economic aspects of FET-PET imaging. 31. Jansen NL, Graute V, Armbruster L, et al. MRI-suspected low-grade glioma: is there a need to perform dynamic FET PET? Eur J Nucl Med Mol Imaging 2012; 39:1021–1029. 32. Calcagni ML, Galli G, Giordano A, et al. Dynamic O-(2-[18F]fluoroethyl)-Ltyrosine (F-18 FET) PET for glioma grading: assessment of individual probability of malignancy. Clin Nucl Med 2011; 36:841–847. 33. Popperl G, Kreth FW, Herms J, et al. Analysis of 18F-FET PET for grading of recurrent gliomas: is evaluation of uptake kinetics superior to standard methods? J Nucl Med 2006; 47:393–403. 34. Popperl G, Kreth FW, Mehrkens JH, et al. FET PET for the evaluation of untreated gliomas: correlation of FET uptake and uptake kinetics with tumour grading. Eur J Nucl Med Mol Imaging 2007; 34:1933–1942. 35. Kratochwil C, Combs SE, Leotta K, et al. Intra-individual comparison of 18 && F-FET and 18F-DOPA in PET imaging of recurrent brain tumors. Neuro Oncol 2014; 16:434–440. Evaluation of differences between the two most interesting and most often used amino acid tracers for imaging protocols and the interpretation of findings. 36. Schiepers C, Chen W, Cloughesy T, et al. 18F-FDOPA kinetics in brain tumors. J Nucl Med 2007; 48:1651–1661. 37. Fueger BJ, Czernin J, Cloughesy T, et al. Correlation of 6-18F-fluoro-L-dopa PET uptake with proliferation and tumor grade in newly diagnosed and recurrent gliomas. J Nucl Med 2010; 51:1532–1538. 38. Jansen NL, Suchorska B, Wenter V, et al. Dynamic 18F-FET PET in newly & diagnosed astrocytic low-grade glioma identifies high-risk patients. J Nucl Med 2014; 55:198–203. Presents interesting new findings concerning the role of FET kinetics. 39. Galldiks N, Stoffels G, Ruge MI, et al. Role of O-(2-18F-fluoroethyl)-L-tyrosine & PET as a diagnostic tool for detection of malignant progression in patients with low-grade glioma. J Nucl Med 2013; 54:2046–2054. Presents interesting new findings concerning the role of FET kinetics. 40. Pyka T, Gempt J, Ringel F, et al. Prediction of glioma recurrence using & dynamic 18F-fluoroethyltyrosine PET. AJNR Am J Neuroradiol 2014. [Epub ahead of print] Presents interesting new findings concerning the role of FET kinetics, although small patient collective. 41. Hatakeyama T, Kawai N, Nishiyama Y, et al. 11C-methionine (MET) and 18F-fluorothymidine (FLT) PET in patients with newly diagnosed glioma. Eur J Nucl Med Mol Imaging 2008; 35:2009–2017. 42. Jacobs AH, Thomas A, Kracht LW, et al. 18F-fluoro-L-thymidine and 11C-methylmethionine as markers of increased transport and proliferation in brain tumors. J Nucl Med 2005; 46:1948–1958.

1350-7540 ß 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins

www.co-neurology.com

687

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Neoplasms 43. Schwarzenberg J, Czernin J, Cloughesy TF, et al. 3’-deoxy-3’-18F-fluorothymidine PET and MRI for early survival predictions in patients with recurrent malignant glioma treated with bevacizumab. J Nucl Med 2012; 53:29–36. 44. Chen W, Cloughesy T, Kamdar N, et al. Imaging proliferation in brain tumors with 18F-FLT PET: comparison with 18F-FDG. J Nucl Med 2005; 46:945– 952. 45. Spence AM, Muzi M, Swanson KR, et al. Regional hypoxia in glioblastoma multiforme quantified with [18F]fluoromisonidazole positron emission tomography before radiotherapy: correlation with time to progression and survival. Clin Cancer Res 2008; 14:2623–2630. 46. Filss CP, Galldiks N, Stoffels G, et al. Comparison of 18F-FET PET and perfusion-weighted MR imaging: a PET/MR imaging hybrid study in patients with brain tumors. J Nucl Med 2014; 55:540–545. 47. Rahm V, Boxheimer L, Bruehlmeier M, et al. Focal changes in diffusivity on apparent diffusion coefficient MR imaging and amino acid uptake on PET do not colocalize in nonenhancing low-grade gliomas. J Nucl Med 2014; 55:546–550. 48. Berntsson SG, Falk A, Savitcheva I, et al. Perfusion and diffusion MRI combined with 11C-methionine PET in the preoperative evaluation of suspected adult low-grade gliomas. J Neurooncol 2013; 114:241–249. 49. Gempt J, Soehngen E, Forster S, et al. Multimodal imaging in cerebral gliomas and its neuropathological correlation. Eur J Radiol 2014; 83:829– 834. 50. Yoon JH, Kim JH, Kang WJ, et al. Grading of cerebral glioma with multiparametric MR imaging and 18F-FDG-PET: concordance and accuracy. Eur Radiol 2014; 24:380–389. 51. Rose S, Fay M, Thomas P, et al. Correlation of MRI-derived apparent diffusion coefficients in newly diagnosed gliomas with [18F]-fluoro-L-dopa PET: what are we really measuring with minimum ADC? AJNR Am J Neuroradiol 2013; 34:758–764. 52. Nowosielski M, DiFranco MD, Putzer D, et al. An intra-individual comparison of MRI, [18F]-FET and [18F]-FLT PET in patients with high-grade gliomas. PLoS One 2014; 9:e95830. 53. Sperduto PW, Kased N, Roberge D, et al. Summary report on the graded prognostic assessment: an accurate and facile diagnosis-specific tool to estimate survival for patients with brain metastases. J Clin Oncol 2012; 30:419–425. 54. Chen AM, Jahan TM, Jablons DM, et al. Risk of cerebral metastases and neurological death after pathological complete response to neoadjuvant therapy for locally advanced nonsmall-cell lung cancer: clinical implications for the subsequent management of the brain. Cancer 2007; 109:1668– 1675. 55. Cufer T, Knez L. Update on systemic therapy of advanced nonsmall-cell lung cancer. Expert Rev Anticancer Ther 2014; 14:1189–203. 56. Jackson SE, Chester JD. Personalised cancer medicine. Int J Cancer 2014. [Epub ahead of print] 57. Andrews MC, Woods K, Cebon J, Behren A. Evolving role of tumor antigens for future melanoma therapies. Future Oncol 2014; 10:1457–1468. 58. van Kruchten M, de Vries EG, Brown M, et al. PET imaging of oestrogen receptors in patients with breast cancer. Lancet Oncol 2013; 14:e465– e475. 59. Kickingereder P, Dorn F, Blau T, et al. Differentiation of local tumor recurrence from radiation-induced changes after stereotactic radiosurgery for treatment of brain metastasis: case report and review of the literature. Radiat Oncol 2013; 8:52. 60. Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst 2000; 92:205–216. 61. Lin NU, Lee EQ, Aoyama H, et al. Challenges relating to solid tumour brain metastases in clinical trials, part 1: patient population, response, and progression. A report from the RANO group. Lancet Oncol 2013; 14:e396– e406. 62. Wen PY, Macdonald DR, Reardon DA, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol 2010; 28:1963–1972. 63. Tsuyuguchi N, Sunada I, Iwai Y, et al. Methionine positron emission tomography of recurrent metastatic brain tumor and radiation necrosis after stereotactic radiosurgery: is a differential diagnosis possible? J Neurosurg 2003; 98:1056–1064. 64. Kruger S, Mottaghy FM, Buck AK, et al. Brain metastasis in lung cancer. Comparison of cerebral MRI and 18F-FDG-PET/CT for diagnosis in the initial staging. Nuklearmedizin 2011; 50:101–106. 65. Grosu AL, Astner ST, Riedel E, et al. An interindividual comparison of O-(2[18F]fluoroethyl)-L-tyrosine (FET)- and L-[methyl-11C]methionine (MET)-PET in patients with brain gliomas and metastases. Int J Radiat Oncol Biol Phys 2011; 81:1049–1058.

688

www.co-neurology.com

66. Galldiks N, Stoffels G, Filss CP, et al. Role of O-(2-(18)F-fluoroethyl)-Ltyrosine PET for differentiation of local recurrent brain metastasis from radiation necrosis. J Nucl Med 2012; 53:1367–1374. 67. Mertens K, Bolcaen J, Ham H, et al. The optimal timing for imaging brain tumours and other brain lesions with 18F-labelled fluoromethylcholine: a dynamic positron emission tomography study. Nucl Med Commun 2012; 33:954–959. 68. Dijkers EC, Oude Munnink TH, Kosterink JG, et al. Biodistribution of 89Zrtrastuzumab and PET imaging of HER2-positive lesions in patients with metastatic breast cancer. Clin Pharmacol Ther 2010; 87:586–592. 69. Olson JE, Janney CA, Rao RD, et al. The continuing increase in the incidence of primary central nervous system non-Hodgkin lymphoma: a surveillance, epidemiology, and end results analysis. Cancer 2002; 95:1504–1510. 70. Sierra del Rio M, Rousseau A, Soussain C, et al. Primary CNS lymphoma in immunocompetent patients. Oncologist 2009; 14:526–539. 71. Kawase Y, Yamamoto Y, Kameyama R, et al. Comparison of 11C-methionine PET and 18F-FDG PET in patients with primary central nervous system lymphoma. Mol Imaging Biol 2011; 13:1284–1289. 72. Tsuyuguchi N, Sunada I, Ohata K, et al. Evaluation of treatment effects in brain abscess with positron emission tomography: comparison of fluorine-18-fluorodeoxyglucose and carbon-11-methionine. Ann Nucl Med 2003; 17:47–51. 73. Schiepers C, Van Hecke P, Vandenberghe R, et al. Positron emission tomography, magnetic resonance imaging and proton NMR spectroscopy of white matter in multiple sclerosis. Mult Scler 1997; 3:8–17. 74. Kosaka N, Tsuchida T, Uematsu H, et al. 18F-FDG PET of common enhancing malignant brain tumors. AJR Am J Roentgenol 2008; 190:W365–W369. 75. Kawai N, Zhen HN, Miyake K, et al. Prognostic value of pretreatment 18F-FDG PET in patients with primary central nervous system lymphoma: SUV-based assessment. J Neurooncol 2010; 100:225–232. 76. Villringer K, Jager H, Dichgans M, et al. Differential diagnosis of CNS lesions in AIDS patients by FDG-PET. J Comput Assist Tomogr 1995; 19:532–536. 77. Kawai N, Nishiyama Y, Miyake K, et al. Evaluation of tumor FDG transport and metabolism in primary central nervous system lymphoma using [18F]fluorodeoxyglucose (FDG) positron emission tomography (PET) kinetic analysis. Ann Nucl Med 2005; 19:685–690. 78. Palmedo H, Urbach H, Bender H, et al. FDG-PET in immunocompetent patients with primary central nervous system lymphoma: correlation with MRI and clinical follow-up. Eur J Nucl Med Mol Imaging 2006; 33:164–168. 79. Ogawa T, Kanno I, Hatazawa J, et al. Methionine PET for follow-up of radiation therapy of primary lymphoma of the brain. Radiographics 1994; 14:101–110. 80. Muldoon LL, Lewin SJ, Dosa E, et al. Imaging and therapy with rituximab antiCD20 immunotherapy in an animal model of central nervous system lymphoma. Clin Cancer Res 2011; 17:2207–2215. 81. Doolittle ND, Jahnke K, Belanger R, et al. Potential of chemo-immunotherapy and radioimmunotherapy in relapsed primary central nervous system (CNS) lymphoma. Leuk Lymphoma 2007; 48:1712–1720. 82. Maza S, Kiewe P, Munz DL, et al. First report on a prospective trial with yttrium90-labeled ibritumomab tiuxetan (Zevalin) in primary CNS lymphoma. Neuro Oncol 2009; 11:423–429. 83. Mawrin C, Perry A. Pathological classification and molecular genetics of meningiomas. J Neurooncol 2010; 99:379–391. 84. Combs SE, Ganswindt U, Foote RL, et al. State-of-the-art treatment alternatives for base of skull meningiomas: complementing and controversial indications for neurosurgery, stereotactic and robotic based radiosurgery or modern fractionated radiation techniques. Radiat Oncol 2012; 7:226. 85. Giovacchini G, Fallanca F, Landoni C, et al. C-11 choline versus F-18 fluorodeoxyglucose for imaging meningiomas: an initial experience. Clin Nucl Med 2009; 34:7–10. 86. Rutten I, Cabay JE, Withofs N, et al. PET/CT of skull base meningiomas using 2-18F-fluoro-L-tyrosine: initial report. J Nucl Med 2007; 48:720–725. 87. Grosu AL, Weber WA, Astner ST, et al. 11C-methionine PET improves the target volume delineation of meningiomas treated with stereotactic fractionated radiotherapy. Int J Radiat Oncol Biol Phys 2006; 66:339–344. 88. Graf R, Nyuyki F, Steffen IG, et al. Contribution of 68Ga-DOTATOC PET/CT to target volume delineation of skull base meningiomas treated with stereotactic radiation therapy. Int J Radiat Oncol Biol Phys 2013; 85:68–73. 89. Ikeda H, Tsuyuguchi N, Kunihiro N, et al. Analysis of progression and recurrence of meningioma using 11C-methionine PET. Ann Nucl Med 2013; 27:772–780. 90. Afshar-Oromieh A, Wolf MB, Kratochwil C, et al. Comparison of 68GaDOTATOC-PET/CT and PET/MRI hybrid systems in patients with cranial meningioma: initial results. Neuro Oncol 2014. [Epub ahead of print] 91. Hanscheid H, Sweeney RA, Flentje M, et al. PET SUV correlates with radionuclide uptake in peptide receptor therapy in meningioma. Eur J Nucl Med Mol Imaging 2012; 39:1284–1288. 92. Bartolomei M, Bodei L, De Cicco C, et al. Peptide receptor radionuclide therapy with 90Y-DOTATOC in recurrent meningioma. Eur J Nucl Med Mol Imaging 2009; 36:1407–1416.

Volume 27  Number 6  December 2014

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

PET imaging for brain tumor diagnostics.

Brain tumors differ in histology, biology, prognosis and treatment options. Although structural magnetic resonance is still the gold standard for morp...
227KB Sizes 0 Downloads 7 Views