1
|
Puranik AD, Dev ID, Rangarajan V, Jain Y, Patra S, Purandare NC, Sahu A, Choudhary A, Bhattacharya K, Gupta T, Chatterjee A, Dasgupta A, Moiyadi A, Shetty P, Singh V, Sridhar E, Sahay A, Shah A, Menon N, Ghosh S, Choudhury S, Shah S, Agrawal A, Lakshminarayanan N, Kumar A, Gopalakrishna A. FET PET to differentiate between post-treatment changes and recurrence in high-grade gliomas: a single center multidisciplinary clinic controlled study. Neuroradiology 2025; 67:363-369. [PMID: 39527264 PMCID: PMC11893651 DOI: 10.1007/s00234-024-03495-9] [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: 03/14/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE The clinico-radiological dilemma in post-treatment high-grade gliomas, between disease recurrence (TR) and treatment-related changes (TRC), still persists. FET (Fluoro-ethyl-tyrosine) PET has been extensively used as problem-solving modality for cases where MR imaging is inconclusive. We incorporated a systematic imaging and clinical follow-up algorithm in a multi-disciplinary clinic (MDC) setting to analyse our cohort of FET PET in post-treatment gliomas. METHODS We retrospectively analyzed 171 patients of post-treatment grade III and IV glioma with equivocal findings on MRI. 185-222 MBq of 18 F-FET was injected and dedicated static imaging of brain was performed at 20 min. TBR (Tumor to background ratio) was used as semi-quantitative parameter. Cutoff of 2.5 was used for image interpretation. Imaging findings were confirmed with histopathological diagnosis, wherever available or in a multidisciplinary joint clinic based on serial imaging. RESULTS 121 of 171 patients showed recurrent disease on FET PET, on follow up, 109 were confirmed with recurrence; 7 patients showed TRC, whereas 5 were treated with bevacizumab, with no further clinico-radiological deterioration, thus confirming TRC. 50 patients showed TRC on FET PET, on follow up on follow up, 40 were confirmed as true-negative. 10 patients who showed TBR less than 2.5 had confirmed TR on subsequent MR imaging. The overall sensitivity and specificity was 91.6 and 76.9% respectively, with a diagnostic accuracy of 87.13%. CONCLUSION There is potential for FET PET to be used along with MRI in the post treatment algorithm of high-grade glial tumors.
Collapse
Affiliation(s)
- Ameya D Puranik
- Department of Nuclear Medicine and Molecular Imaging, Homi Bhabha National University, Tata Memorial Hospital, Mumbai, India.
| | - Indraja D Dev
- Department of Nuclear Medicine and Molecular Imaging, Homi Bhabha National University, Tata Memorial Hospital, Mumbai, India
| | - Venkatesh Rangarajan
- Department of Nuclear Medicine and Molecular Imaging, Homi Bhabha National University, Tata Memorial Hospital, Mumbai, India
| | - Yash Jain
- Department of Nuclear Medicine and Molecular Imaging, Homi Bhabha National University, Tata Memorial Hospital, Mumbai, India
| | - Sukriti Patra
- Department of Nuclear Medicine and Molecular Imaging, Homi Bhabha National University, Tata Memorial Hospital, Mumbai, India
| | - Nilendu C Purandare
- Department of Nuclear Medicine and Molecular Imaging, Homi Bhabha National University, Tata Memorial Hospital, Mumbai, India
| | - Arpita Sahu
- Department of Radiodiagnosis, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Mumbai, India
| | - Amitkumar Choudhary
- Department of Radiodiagnosis, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Mumbai, India
| | - Kajari Bhattacharya
- Department of Radiodiagnosis, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Mumbai, India
| | - Tejpal Gupta
- Department of Radiation Oncology, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Mumbai, India
| | - Abhishek Chatterjee
- Department of Radiation Oncology, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Mumbai, India
| | - Archya Dasgupta
- Department of Radiation Oncology, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Mumbai, India
| | - Aliasgar Moiyadi
- Department of Neurosurgery, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Mumbai, India
| | - Prakash Shetty
- Department of Neurosurgery, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Mumbai, India
| | - Vikas Singh
- Department of Neurosurgery, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Mumbai, India
| | - Epari Sridhar
- Department of Pathology, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Mumbai, India
| | - Ayushi Sahay
- Department of Pathology, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Mumbai, India
| | - Aekta Shah
- Department of Pathology, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Mumbai, India
| | - Nandini Menon
- Department of Medical Oncology, Tata Memorial Hospital and Advanced Center for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National University, Mumbai, India
| | - Suchismita Ghosh
- Department of Nuclear Medicine and Molecular Imaging, Homi Bhabha National University, Tata Memorial Hospital, Mumbai, India
| | - Sayak Choudhury
- Department of Nuclear Medicine and Molecular Imaging, Homi Bhabha National University, Tata Memorial Hospital, Mumbai, India
| | - Sneha Shah
- Department of Nuclear Medicine and Molecular Imaging, Homi Bhabha National University, Tata Memorial Hospital, Mumbai, India
| | - Archi Agrawal
- Department of Nuclear Medicine and Molecular Imaging, Homi Bhabha National University, Tata Memorial Hospital, Mumbai, India
| | - N Lakshminarayanan
- Medical Cyclotron Facility, Board of Radiation and Isotope Technology (BRIT), Bhabha Atomic Research Center, Mumbai, India
| | - Amit Kumar
- Medical Cyclotron Facility, Board of Radiation and Isotope Technology (BRIT), Bhabha Atomic Research Center, Mumbai, India
| | - Arjun Gopalakrishna
- Medical Cyclotron Facility, Board of Radiation and Isotope Technology (BRIT), Bhabha Atomic Research Center, Mumbai, India
| |
Collapse
|
2
|
Lee DY, Oh JS, Kim JW, Oh M, Oh SJ, Lee S, Kim YH, Kim JH, Nam SJ, Song SW, Kim JS. Pre-operative dual-time-point [ 18F]FET PET differentiates CDKN2A/B loss and PIK3CA mutation status in adult-type diffuse glioma: a single-center prospective study. Eur J Nucl Med Mol Imaging 2025; 52:669-682. [PMID: 39365462 DOI: 10.1007/s00259-024-06935-z] [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: 05/24/2024] [Accepted: 09/29/2024] [Indexed: 10/05/2024]
Abstract
PURPOSE While [18F]FET PET plays a complementary role in glioma imaging, it needs to be more comprehensively understood for improved characterization of glioma prior to surgery given the evolving landscape of molecular neuropathology. Thus, we investigated the utility of pre-operative dual-time-point [18F]FET PET in correlation with next-generation sequencing (NGS) data in patients with adult-type diffuse glioma (ADG). METHODS Adult patients who were suspected to have primary glioma were prospectively recruited between June 2021 and January 2024. They underwent pre-operative dual-time-point static PET/CT at 20 min (early) and 80 min (delay) after [18F]FET injection. Semi-quantitative parameters of the hottest lesion (SUVmax) of tumour and the hottest lesion-to-normal brain ratio (TBRmax) were assessed from each summed image. Furthermore, the percentage changes (△) of SUVmax and TBRmax between two images were calculated. Histopathology of glioma was determined according to the 2021 WHO classification and NGS data. RESULTS This study investigated a dozen genes in 76 patients, of whom 51 had isocitrate dehydrogenase (IDH)-wild-type glioblastoma, 13 had IDH-mutant astrocytoma, and 12 had IDH-mutant oligodendroglioma. Every tumour was [18F]FET-avid having TBRmax more than 1.6. Patients with CDKN2A/B loss had significantly higher values of SUVmax (5.7 ± 1.6 vs. 4.7 ± 1.3, p = 0.004; 5.0 ± 1.4 vs. 4.4 ± 1.2, p = 0.026) and TBRmax (6.5 ± 1.8 vs. 5.1 ± 1.7, p = 0.001; 5.3 ± 1.5 vs. 4.3 ± 1.3, p = 0.004) in both scans than patients without CDKN2A/B loss, even after adjustment for age, MRI enhancement, tumor grade and type of pathology. Furthermore, patients with PIK3CA mutation (16.2 ± 11.8 vs. 6.7 ± 11.6, p = 0.007) had significantly higher △SUVmax than patients without PIK3CA mutation, even after adjustment for age, MRI enhancement, tumor grade, and type of pathology. CONCLUSION Among the dozen genes investigated in this prospective study in patients with ADG, we found out that CDKN2A/B loss and PIK3CA mutation status could be differentiated by pre-operative dual-time-point [18F]FET PET/CT.
Collapse
Affiliation(s)
- Dong Yun Lee
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Jungsu S Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Jeong Won Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Minyoung Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Seung Jun Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Seungjoo Lee
- Department of Neurosurgery, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Young-Hoon Kim
- Department of Neurosurgery, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Jeong Hoon Kim
- Department of Neurosurgery, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Soo Jeong Nam
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea
| | - Sang Woo Song
- Department of Neurosurgery, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea.
| | - Jae Seung Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea.
| |
Collapse
|
3
|
Prokop G, Wiestler B, Hieber D, Withake F, Mayer K, Gempt J, Delbridge C, Schmidt-Graf F, Pfarr N, Märkl B, Schlegel J, Liesche-Starnecker F. Multiscale quantification of morphological heterogeneity with creation of a predictor of longer survival in glioblastoma. Int J Cancer 2023; 153:1658-1670. [PMID: 37501565 DOI: 10.1002/ijc.34665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023]
Abstract
Intratumor heterogeneity is a main cause of the dismal prognosis of glioblastoma (GBM). Yet, there remains a lack of a uniform assessment of the degree of heterogeneity. With a multiscale approach, we addressed the hypothesis that intratumor heterogeneity exists on different levels comprising traditional regional analyses, but also innovative methods including computer-assisted analysis of tumor morphology combined with epigenomic data. With this aim, 157 biopsies of 37 patients with therapy-naive IDH-wildtype GBM were analyzed regarding the intratumor variance of protein expression of glial marker GFAP, microglia marker Iba1 and proliferation marker Mib1. Hematoxylin and eosin stained slides were evaluated for tumor vascularization. For the estimation of pixel intensity and nuclear profiling, automated analysis was used. Additionally, DNA methylation profiling was conducted separately for the single biopsies. Scoring systems were established to integrate several parameters into one score for the four examined modalities of heterogeneity (regional, cellular, pixel-level and epigenomic). As a result, we could show that heterogeneity was detected in all four modalities. Furthermore, for the regional, cellular and epigenomic level, we confirmed the results of earlier studies stating that a higher degree of heterogeneity is associated with poorer overall survival. To integrate all modalities into one score, we designed a predictor of longer survival, which showed a highly significant separation regarding the OS. In conclusion, multiscale intratumor heterogeneity exists in glioblastoma and its degree has an impact on overall survival. In future studies, the implementation of a broadly feasible heterogeneity index should be considered.
Collapse
Affiliation(s)
- Georg Prokop
- Pathology, Medical Faculty, University of Augsburg, Augsburg, Germany
- Institute of Pathology, School of Medicine, Technical University Munich, Munich, Germany
| | - Benedikt Wiestler
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University Munich, Munich, Germany
| | - Daniel Hieber
- Pathology, Medical Faculty, University of Augsburg, Augsburg, Germany
- Institute DigiHealth, Neu-Ulm University of Applied Sciences, Neu-Ulm, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| | - Fynn Withake
- Department of Neuroradiology, Klinikum rechts der Isar, School of Medicine, Technical University Munich, Munich, Germany
| | - Karoline Mayer
- Institute of Pathology, School of Medicine, Technical University Munich, Munich, Germany
| | - Jens Gempt
- Department of Neurosurgery, Klinikum rechts der Isar, School of Medicine, Technical University Munich, Munich, Germany
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claire Delbridge
- Institute of Pathology, School of Medicine, Technical University Munich, Munich, Germany
| | - Friederike Schmidt-Graf
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University Munich, Munich, Germany
| | - Nicole Pfarr
- Institute of Pathology, School of Medicine, Technical University Munich, Munich, Germany
| | - Bruno Märkl
- Pathology, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Jürgen Schlegel
- Pathology, Medical Faculty, University of Augsburg, Augsburg, Germany
- Institute of Pathology, School of Medicine, Technical University Munich, Munich, Germany
| | - Friederike Liesche-Starnecker
- Pathology, Medical Faculty, University of Augsburg, Augsburg, Germany
- Institute of Pathology, School of Medicine, Technical University Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Augsburg, Germany
| |
Collapse
|
4
|
Becker H, Castaneda-Vega S, Patzwaldt K, Przystal JM, Walter B, Michelotti FC, Canjuga D, Tatagiba M, Pichler B, Beck SC, Holland EC, la Fougère C, Tabatabai G. Multiparametric Longitudinal Profiling of RCAS-tva-Induced PDGFB-Driven Experimental Glioma. Brain Sci 2022; 12:1426. [PMID: 36358353 PMCID: PMC9688186 DOI: 10.3390/brainsci12111426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 12/31/2023] Open
Abstract
Glioblastomas are incurable primary brain tumors harboring a heterogeneous landscape of genetic and metabolic alterations. Longitudinal imaging by MRI and [18F]FET-PET measurements enable us to visualize the features of evolving tumors in a dynamic manner. Yet, close-meshed longitudinal imaging time points for characterizing temporal and spatial metabolic alterations during tumor evolution in patients is not feasible because patients usually present with already established tumors. The replication-competent avian sarcoma-leukosis virus (RCAS)/tumor virus receptor-A (tva) system is a powerful preclinical glioma model offering a high grade of spatial and temporal control of somatic gene delivery in vivo. Consequently, here, we aimed at using MRI and [18F]FET-PET to identify typical neuroimaging characteristics of the platelet-derived growth factor B (PDGFB)-driven glioma model using the RCAS-tva system. Our study showed that this preclinical glioma model displays MRI and [18F]FET-PET features that highly resemble the corresponding established human disease, emphasizing the high translational relevance of this experimental model. Furthermore, our investigations unravel exponential growth dynamics and a model-specific tumor microenvironment, as assessed by histology and immunochemistry. Taken together, our study provides further insights into this preclinical model and advocates for the imaging-stratified design of preclinical therapeutic interventions.
Collapse
Affiliation(s)
- Hannes Becker
- Department of Neurology & Interdisciplinary Neuro-Oncology, Hertie Institute for Clinical Brain Research, Center for Neuro-Oncology, Comprehensive Cancer Center, University Hospital Tübingen, Eberhard Karls University Tubingen, 72072 Tubingen, Germany
- Department of Neurosurgery, University Hospital Tubingen, Eberhard Karls University Tubingen, 72072 Tubingen, Germany
| | - Salvador Castaneda-Vega
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, 72072 Tubingen, Germany
- Department of Nuclear Medicine and Clinical Molecular Imaging, Eberhard Karls University Tuebingen, 72072 Tubingen, Germany
| | - Kristin Patzwaldt
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, 72072 Tubingen, Germany
| | - Justyna M. Przystal
- Department of Neurology & Interdisciplinary Neuro-Oncology, Hertie Institute for Clinical Brain Research, Center for Neuro-Oncology, Comprehensive Cancer Center, University Hospital Tübingen, Eberhard Karls University Tubingen, 72072 Tubingen, Germany
- German Translational Cancer Consortium (DKTK), DKFZ Partner Site, 72072 Tubingen, Germany
| | - Bianca Walter
- Department of Neurology & Interdisciplinary Neuro-Oncology, Hertie Institute for Clinical Brain Research, Center for Neuro-Oncology, Comprehensive Cancer Center, University Hospital Tübingen, Eberhard Karls University Tubingen, 72072 Tubingen, Germany
| | - Filippo C. Michelotti
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, 72072 Tubingen, Germany
| | - Denis Canjuga
- Department of Neurology & Interdisciplinary Neuro-Oncology, Hertie Institute for Clinical Brain Research, Center for Neuro-Oncology, Comprehensive Cancer Center, University Hospital Tübingen, Eberhard Karls University Tubingen, 72072 Tubingen, Germany
| | - Marcos Tatagiba
- Department of Neurology & Interdisciplinary Neuro-Oncology, Hertie Institute for Clinical Brain Research, Center for Neuro-Oncology, Comprehensive Cancer Center, University Hospital Tübingen, Eberhard Karls University Tubingen, 72072 Tubingen, Germany
- Department of Neurosurgery, University Hospital Tubingen, Eberhard Karls University Tubingen, 72072 Tubingen, Germany
| | - Bernd Pichler
- Department of Nuclear Medicine and Clinical Molecular Imaging, Eberhard Karls University Tuebingen, 72072 Tubingen, Germany
- German Translational Cancer Consortium (DKTK), DKFZ Partner Site, 72072 Tubingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University, 72072 Tubingen, Germany
| | - Susanne C. Beck
- Department of Neurology & Interdisciplinary Neuro-Oncology, Hertie Institute for Clinical Brain Research, Center for Neuro-Oncology, Comprehensive Cancer Center, University Hospital Tübingen, Eberhard Karls University Tubingen, 72072 Tubingen, Germany
| | - Eric C. Holland
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, DC 98109, USA
| | - Christian la Fougère
- Department of Nuclear Medicine and Clinical Molecular Imaging, Eberhard Karls University Tuebingen, 72072 Tubingen, Germany
- German Translational Cancer Consortium (DKTK), DKFZ Partner Site, 72072 Tubingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University, 72072 Tubingen, Germany
| | - Ghazaleh Tabatabai
- Department of Neurology & Interdisciplinary Neuro-Oncology, Hertie Institute for Clinical Brain Research, Center for Neuro-Oncology, Comprehensive Cancer Center, University Hospital Tübingen, Eberhard Karls University Tubingen, 72072 Tubingen, Germany
- German Translational Cancer Consortium (DKTK), DKFZ Partner Site, 72072 Tubingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University, 72072 Tubingen, Germany
| |
Collapse
|
5
|
Diagnostic yield of simultaneous dynamic contrast-enhanced magnetic resonance perfusion measurements and [ 18F]FET PET in patients with suspected recurrent anaplastic astrocytoma and glioblastoma. Eur J Nucl Med Mol Imaging 2022; 49:4677-4691. [PMID: 35907033 PMCID: PMC9605929 DOI: 10.1007/s00259-022-05917-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 07/16/2022] [Indexed: 11/04/2022]
Abstract
Purpose Both amino acid positron emission tomography (PET) and magnetic resonance imaging (MRI) blood volume (BV) measurements are used in suspected recurrent high-grade gliomas. We compared the separate and combined diagnostic yield of simultaneously acquired dynamic contrast-enhanced (DCE) perfusion MRI and O-(2-[18F]-fluoroethyl)-L-tyrosine ([18F]FET) PET in patients with anaplastic astrocytoma and glioblastoma following standard therapy. Methods A total of 76 lesions in 60 hybrid [18F]FET PET/MRI scans with DCE MRI from patients with suspected recurrence of anaplastic astrocytoma and glioblastoma were included retrospectively. BV was measured from DCE MRI employing a 2-compartment exchange model (2CXM). Diagnostic performances of maximal tumour-to-background [18F]FET uptake (TBRmax), maximal BV (BVmax) and normalised BVmax (nBVmax) were determined by ROC analysis using 6-month histopathological (n = 28) or clinical/radiographical follow-up (n = 48) as reference. Sensitivity and specificity at optimal cut-offs were determined separately for enhancing and non-enhancing lesions. Results In progressive lesions, all BV and [18F]FET metrics were higher than in non-progressive lesions. ROC analyses showed higher overall ROC AUCs for TBRmax than both BVmax and nBVmax in both lesion-wise (all lesions, p = 0.04) and in patient-wise analysis (p < 0.01). Combining TBRmax with BV metrics did not increase ROC AUC. Lesion-wise positive fraction/sensitivity/specificity at optimal cut-offs were 55%/91%/84% for TBRmax, 45%/77%/84% for BVmax and 59%/84%/72% for nBVmax. Combining TBRmax and best-performing BV cut-offs yielded lesion-wise sensitivity/specificity of 75/97%. The fraction of progressive lesions was 11% in concordant negative lesions, 33% in lesions only BV positive, 64% in lesions only [18F]FET positive and 97% in concordant positive lesions. Conclusion The overall diagnostic accuracy of DCE BV imaging is good, but lower than that of [18F]FET PET. Adding DCE BV imaging did not improve the overall diagnostic accuracy of [18F]FET PET, but may improve specificity and allow better lesion-wise risk stratification than [18F]FET PET alone. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-022-05917-3.
Collapse
|
6
|
Scarpelli ML, Healey DR, Mehta S, Quarles CC. Imaging Glioblastoma With 18F-Fluciclovine Amino Acid Positron Emission Tomography. Front Oncol 2022; 12:829050. [PMID: 35174096 PMCID: PMC8841434 DOI: 10.3389/fonc.2022.829050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 01/12/2022] [Indexed: 11/13/2022] Open
Abstract
IntroductionConventional methods of imaging brain tumors fail to assess metabolically active tumor regions, which limits their capabilities for tumor detection, localization, and response assessment. Positron emission tomography (PET) with 18F-fluciclovine (fluciclovine) provides regional assessment of amino acid uptake in tumors that could overcome some of the limitations of conventional imaging. However, the biological basis of enhanced fluciclovine uptake is insufficiently characterized in brain tumors, which confounds clinical interpretation and application. This study sought to address this gap by correlating multiple biologic quantities with fluciclovine PET uptake across a range of human glioblastoma xenograft models.MethodsThirty-one rats underwent orthotopic implantations with one of five different human glioblastoma cell lines. After tumors were established, fluciclovine PET and magnetic resonance imaging (MRI) scans were performed. The fluciclovine tumor-to-normal-brain (TN) uptake ratio was used to quantify fluciclovine uptake. MRI scans were used to assess tumor volume and gadolinium enhancement status. Histologic assessments quantified tumor cell proliferation, tumor cell density, and tumor cell amino acid transporters (LAT1 and ASCT2). Multivariate linear regression models related fluciclovine uptake with the other measured quantities.ResultsWithin the multivariate regression, the fluciclovine TN uptake ratio (measured 15 to 35 minutes after fluciclovine injection) was most strongly associated with tumor ASCT2 levels (β=0.64; P=0.001). The fluciclovine TN uptake ratio was also significantly associated with tumor volume (β=0.45; P=0.001) and tumor enhancement status (β=0.40; P=0.01). Tumor cell proliferation, tumor cell density, and LAT1 levels were not significantly associated with fluciclovine uptake in any of the multivariate models. In general, both enhancing and non-enhancing tumors could be visualized on fluciclovine PET images, with the median TN uptake ratio across the five tumor lines being 2.4 (range 1.1 to 8.9).ConclusionsIncreased fluciclovine PET uptake was associated with increased levels of the amino acid transporter ASCT2, suggesting fluciclovine PET may be useful for assessing brain tumor amino acid metabolism. Fluciclovine PET uptake was elevated in both enhancing and non-enhancing tumors but the degree of uptake was greater in larger tumors and tumors with enhancement, indicating these variables could confound fluciclovine metabolic measurements if not accounted for.
Collapse
Affiliation(s)
| | - Debbie R. Healey
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Shwetal Mehta
- Ivy Brain Tumor Center, Barrow Neurological Institute, Phoenix, AZ, United States
| | - C. Chad Quarles
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, Phoenix, AZ, United States
- *Correspondence: C. Chad Quarles,
| |
Collapse
|
7
|
Laudicella R, Bauckneht M, Cuppari L, Donegani MI, Arnone A, Baldari S, Burger IA, Quartuccio N. Emerging applications of imaging in glioma: focus on PET/MRI and radiomics. Clin Transl Imaging 2021; 9:609-623. [DOI: 10.1007/s40336-021-00464-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/17/2021] [Indexed: 02/07/2023]
|
8
|
Verger A, Imbert L, Zaragori T. Dynamic amino-acid PET in neuro-oncology: a prognostic tool becomes essential. Eur J Nucl Med Mol Imaging 2021; 48:4129-4132. [PMID: 34518904 DOI: 10.1007/s00259-021-05530-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Antoine Verger
- Department of Nuclear Medicine & Nancyclotep Imaging Platform, CHRU-Nancy, Université de Lorraine, F-54000, Nancy, France.
- INSERM, IADI, UMR 1254 Université de Lorraine, F-54000, Nancy, France.
- Médecine Nucléaire, Hôpital de Brabois, CHRU-Nancy, Allée du Morvan, 54500, Vandoeuvre-les-Nancy, France.
| | - Laëtitia Imbert
- Department of Nuclear Medicine & Nancyclotep Imaging Platform, CHRU-Nancy, Université de Lorraine, F-54000, Nancy, France
- INSERM, IADI, UMR 1254 Université de Lorraine, F-54000, Nancy, France
| | - Timothée Zaragori
- Department of Nuclear Medicine & Nancyclotep Imaging Platform, CHRU-Nancy, Université de Lorraine, F-54000, Nancy, France
- INSERM, IADI, UMR 1254 Université de Lorraine, F-54000, Nancy, France
| |
Collapse
|
9
|
Liesche-Starnecker F, Prokop G, Yakushev I, Preibisch C, Delbridge C, Meyer HS, Aftahy K, Barz M, Meyer B, Zimmer C, Schlegel J, Wiestler B, Gempt J. Visualizing cellularity and angiogenesis in newly-diagnosed glioblastoma with diffusion and perfusion MRI and FET-PET imaging. EJNMMI Res 2021; 11:72. [PMID: 34398358 PMCID: PMC8368421 DOI: 10.1186/s13550-021-00817-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 07/28/2021] [Indexed: 12/31/2022] Open
Abstract
Purpose Combining imaging modalities has become an essential tool for assessment of tumor biology in glioblastoma (GBM) patients. Aim of this study is to understand how tumor cellularity and neovascularization are reflected in O-(2-[18F]fluoroethyl)-L-tyrosine positron emission tomography ([18F] FET PET) and magnetic resonance imaging (MRI) parameters, including cerebral blood volume (CBV), fractional anisotropy (FA) and mean diffusivity (MD). Methods In this prospective cohort, 162 targeted biopsies of 43 patients with therapy-naïve, isocitrate dehydrogenase (IDH) wildtype GBM were obtained after defining areas of interest based on imaging parameters [18F] FET PET, CBV, FA and MD. Histopathological analysis of cellularity and neovascularization was conducted and results correlated to imaging data. Results ANOVA analysis showed a significant increase of CBV in areas with high neovascularization. For diffusion metrics, and in particular FA, a trend for inverse association with neovascularization was found. [18F] FET PET showed a significant positive correlation to cellularity, while CBV also showed a trend towards correlation with cellularity, not reaching significant levels. In contrast, MD and FA were negatively associated with cellularity. Conclusion Our study confirms that amino acid PET and MR imaging parameters are indicative of histological tumor properties in glioblastoma and highlights the ability of multimodal imaging to assess tumor biology non-invasively.
Collapse
Affiliation(s)
- Friederike Liesche-Starnecker
- Department of Neuropathology, Institute of Pathology, School of Medicine, Technical University Munich, Munich, Germany
| | - Georg Prokop
- Department of Neuropathology, Institute of Pathology, School of Medicine, Technical University Munich, Munich, Germany
| | - Igor Yakushev
- Department of Nuclear Medicine, Klinikum rechts der isar, School of Medicine, Technical University Munich, Munich, Germany
| | - Christine Preibisch
- Department of Neuroradiology, Klinikum rechts der isar, School of Medicine, Technical University Munich, Munich, Germany
| | - Claire Delbridge
- Department of Neuropathology, Institute of Pathology, School of Medicine, Technical University Munich, Munich, Germany
| | - Hanno S Meyer
- Department of Neurosurgery, Klinikum rechts der isar, School of Medicine, Technical University Munich, Ismaningerstr. 22, 81675, Munich, Germany
| | - Kaywan Aftahy
- Department of Neurosurgery, Klinikum rechts der isar, School of Medicine, Technical University Munich, Ismaningerstr. 22, 81675, Munich, Germany
| | - Melanie Barz
- Department of Neurosurgery, Klinikum rechts der isar, School of Medicine, Technical University Munich, Ismaningerstr. 22, 81675, Munich, Germany
| | - Bernhard Meyer
- Department of Neurosurgery, Klinikum rechts der isar, School of Medicine, Technical University Munich, Ismaningerstr. 22, 81675, Munich, Germany
| | - Claus Zimmer
- Department of Neuroradiology, Klinikum rechts der isar, School of Medicine, Technical University Munich, Munich, Germany
| | - Jürgen Schlegel
- Department of Neuropathology, Institute of Pathology, School of Medicine, Technical University Munich, Munich, Germany
| | - Benedikt Wiestler
- Department of Neuroradiology, Klinikum rechts der isar, School of Medicine, Technical University Munich, Munich, Germany.,TranslaTUM (Zentralinstitut für translationale Krebsforschung der Technischen Universität München), Einsteinstraße 25, Munich, Germany
| | - Jens Gempt
- Department of Neurosurgery, Klinikum rechts der isar, School of Medicine, Technical University Munich, Ismaningerstr. 22, 81675, Munich, Germany.
| |
Collapse
|
10
|
Puranik AD, Rangarajan V, Dev ID, Jain Y, Purandare NC, Sahu A, Choudhary A, Gupta T, Chatterjee A, Moiyadi A, Shetty P, Sridhar E, Sahay A, Patil VM, Shah S, Agrawal A. Brain FET PET tumor-to-white mater ratio to differentiate recurrence from post-treatment changes in high-grade gliomas. J Neuroimaging 2021; 31:1211-1218. [PMID: 34388273 DOI: 10.1111/jon.12914] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/17/2021] [Accepted: 07/17/2021] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND AND PURPOSE Highergrade glial neoplasms undergo standard treatment with surgery, radiotherapy, and alkylating agents. There is often a clinical/neuroimaging dilemma in the post-treatment setting to differentiate disease recurrence from treatment-related changes. FET (fluoro-ethyl-tyrosine) PET has emerged as a molecular imaging modality for cases where MR imaging is inconclusive. This study aims to develop a cutoff on FET PET for differentiating true recurrence from post-treatment changes. METHODS We retrospectively analyzed72 patientswith post-treatment grade 3 or 4 brain gliomas. Five to six mCi of 18 F-FET was injected and static imaging of the brain was performed at 20 min. A tumor-to-white matter (T/Wm) ratio was used as semiquantitative parameter. A T/Wm cutoff of 2.5 was used for image interpretation. Imaging findings were confirmed by either histopathologic diagnosis in a multidisciplinary joint clinic or based on follow-up of clinical and neuroimaging findings. RESULTS Forty-one of 72 patients (57%) showed recurrent disease on FET PET. Thirty-five of them were confirmed to have tumor recurrence; six patients showed post-treatment changes. Thirty-one of 72 patients (43%) showed post-treatment changes on FET PET; 27 were confirmed as post-treatment change and four patients had tumor recurrence on subsequent MR imaging. An optimum T/Wm cutoff of 2.65 was derived based on receiver operating characteristic analysis with a sensitivity of 80% and specificity of 87.5%. CONCLUSION Static FET PET can be used as problem-solving imaging modality with a T/Wm cutoff of 2.65 to differentiate late recurrence from post-treatment changes in grade 3 or 4 brain gliomas with equivocal MR features.
Collapse
Affiliation(s)
- Ameya D Puranik
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Venkatesh Rangarajan
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Indraja D Dev
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Yash Jain
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Nilendu C Purandare
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Arpita Sahu
- Department of Radiodiagnosis, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Amitkumar Choudhary
- Department of Radiodiagnosis, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Tejpal Gupta
- Department of Radiation Oncology, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Navi Mumbai, India
| | - Abhishek Chatterjee
- Department of Radiation Oncology, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Navi Mumbai, India
| | - Aliasgar Moiyadi
- Department of Neuro-surgery, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Navi Mumbai, India
| | - Prakash Shetty
- Department of Neuro-surgery, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Navi Mumbai, India
| | - Epari Sridhar
- Department of Pathology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Ayushi Sahay
- Department of Pathology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Vijay M Patil
- Department of Medical Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Sneha Shah
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Archi Agrawal
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
11
|
Zhou Y, Tan W, Zou J, Cao J, Huang Q, Jiang B, Yoshida S, Li Y. Metabolomics Analyses of Mouse Retinas in Oxygen-Induced Retinopathy. Invest Ophthalmol Vis Sci 2021; 62:9. [PMID: 34374743 PMCID: PMC8363770 DOI: 10.1167/iovs.62.10.9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purpose Retinal neovascularization is a severe pathological process leading to irreversible blindness. This study aims to identify the altered metabolites and their related pathways that are involved in retinal neovascularization. Methods To reveal the global metabolomic profile change in the retinal neovascularization process, an untargeted metabolomics analysis of oxygen-induced retinopathy (OIR) mice retinas was carried out first, followed by the validation of amino acids and their derivatives through a targeted metabolomics analysis. The involved pathways were predicted by bioinformatic analysis. Results By untargeted metabolomics, a total of 58 and 49 metabolites altered significantly in OIR retinas under cationic and anionic modes, respectively. By bioinformatics analysis, “ABC transporters,” “central carbon metabolism in cancer.” and “alanine, aspartate, and glutamate metabolism” were the most enriched Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways associated with the changed metabolites. By targeted metabolomics, no significant change was found in the assessed amino acids and their derivatives at postnatal day (P) 12, whereas significantly altered amino acids and their derivatives were recognized at P13, P17, and P42 in OIR retinas. Conclusions The metabolomic profile was significantly altered in the neovascularized retinas. In particular, numerous amino acids and their derivatives were significantly changed in OIR retinas. These altered metabolites, together with their associated pathways, might be involved in the pathogenesis of retinal neovascular diseases.
Collapse
Affiliation(s)
- Yedi Zhou
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Wei Tan
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Jingling Zou
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Jian Cao
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Qian Huang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Bing Jiang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Shigeo Yoshida
- Department of Ophthalmology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Yun Li
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| |
Collapse
|
12
|
Lerche CW, Radomski T, Lohmann P, Caldeira L, Brambilla CR, Tellmann L, Scheins J, Kops ER, Galldiks N, Langen KJ, Herzog H, Jon Shah N. A Linearized Fit Model for Robust Shape Parameterization of FET-PET TACs. IEEE TRANSACTIONS ON MEDICAL IMAGING 2021; 40:1852-1862. [PMID: 33735076 DOI: 10.1109/tmi.2021.3067169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The kinetic analysis of [Formula: see text]-FET time-activity curves (TAC) can provide valuable diagnostic information in glioma patients. The analysis is most often limited to the average TAC over a large tissue volume and is normally assessed by visual inspection or by evaluating the time-to-peak and linear slope during the late uptake phase. Here, we derived and validated a linearized model for TACs of [Formula: see text]-FET in dynamic PET scans. Emphasis was put on the robustness of the numerical parameters and how reliably automatic voxel-wise analysis of TAC kinetics was possible. The diagnostic performance of the extracted shape parameters for the discrimination between isocitrate dehydrogenase (IDH) wildtype (wt) and IDH-mutant (mut) glioma was assessed by receiver-operating characteristic in a group of 33 adult glioma patients. A high agreement between the adjusted model and measured TACs could be obtained and relative, estimated parameter uncertainties were small. The best differentiation between IDH-wt and IDH-mut gliomas was achieved with the linearized model fitted to the averaged TAC values from dynamic FET PET data in the time interval 4-50 min p.i.. When limiting the acquisition time to 20-40 min p.i., classification accuracy was only slightly lower (-3%) and was comparable to classification based on linear fits in this time interval. Voxel-wise fitting was possible within a computation time ≈ 1 min per image slice. Parameter uncertainties smaller than 80% for all fits with the linearized model were achieved. The agreement of best-fit parameters when comparing voxel-wise fits and fits of averaged TACs was very high (p < 0.001).
Collapse
|
13
|
Fully automated analysis combining [ 18F]-FET-PET and multiparametric MRI including DSC perfusion and APTw imaging: a promising tool for objective evaluation of glioma progression. Eur J Nucl Med Mol Imaging 2021; 48:4445-4455. [PMID: 34173008 PMCID: PMC8566389 DOI: 10.1007/s00259-021-05427-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/24/2021] [Indexed: 12/15/2022]
Abstract
Purpose To evaluate diagnostic accuracy of fully automated analysis of multimodal imaging data using [18F]-FET-PET and MRI (including amide proton transfer-weighted (APTw) imaging and dynamic-susceptibility-contrast (DSC) perfusion) in differentiation of tumor progression from treatment-related changes in patients with glioma. Material and methods At suspected tumor progression, MRI and [18F]-FET-PET data as part of a retrospective analysis of an observational cohort of 66 patients/74 scans (51 glioblastoma and 23 lower-grade-glioma, 8 patients included at two different time points) were automatically segmented into necrosis, FLAIR-hyperintense, and contrast-enhancing areas using an ensemble of deep learning algorithms. In parallel, previous MR exam was processed in a similar way to subtract preexisting tumor areas and focus on progressive tumor only. Within these progressive areas, intensity statistics were automatically extracted from [18F]-FET-PET, APTw, and DSC-derived cerebral-blood-volume (CBV) maps and used to train a Random Forest classifier with threefold cross-validation. To evaluate contribution of the imaging modalities to the classifier’s performance, impurity-based importance measures were collected. Classifier performance was compared with radiology reports and interdisciplinary tumor board assessments. Results In 57/74 cases (77%), tumor progression was confirmed histopathologically (39 cases) or via follow-up imaging (18 cases), while remaining 17 cases were diagnosed as treatment-related changes. The classification accuracy of the Random Forest classifier was 0.86, 95% CI 0.77–0.93 (sensitivity 0.91, 95% CI 0.81–0.97; specificity 0.71, 95% CI 0.44–0.9), significantly above the no-information rate of 0.77 (p = 0.03), and higher compared to an accuracy of 0.82 for MRI (95% CI 0.72–0.9), 0.81 for [18F]-FET-PET (95% CI 0.7–0.89), and 0.81 for expert consensus (95% CI 0.7–0.89), although these differences were not statistically significant (p > 0.1 for all comparisons, McNemar test). [18F]-FET-PET hot-spot volume was single-most important variable, with relevant contribution from all imaging modalities. Conclusion Automated, joint image analysis of [18F]-FET-PET and advanced MR imaging techniques APTw and DSC perfusion is a promising tool for objective response assessment in gliomas. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-021-05427-8.
Collapse
|
14
|
Steidl E, Langen KJ, Hmeidan SA, Polomac N, Filss CP, Galldiks N, Lohmann P, Keil F, Filipski K, Mottaghy FM, Shah NJ, Steinbach JP, Hattingen E, Maurer GD. Sequential implementation of DSC-MR perfusion and dynamic [ 18F]FET PET allows efficient differentiation of glioma progression from treatment-related changes. Eur J Nucl Med Mol Imaging 2021; 48:1956-1965. [PMID: 33241456 PMCID: PMC8113145 DOI: 10.1007/s00259-020-05114-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/08/2020] [Indexed: 11/21/2022]
Abstract
PURPOSE Perfusion-weighted MRI (PWI) and O-(2-[18F]fluoroethyl-)-l-tyrosine ([18F]FET) PET are both applied to discriminate tumor progression (TP) from treatment-related changes (TRC) in patients with suspected recurrent glioma. While the combination of both methods has been reported to improve the diagnostic accuracy, the performance of a sequential implementation has not been further investigated. Therefore, we retrospectively analyzed the diagnostic value of consecutive PWI and [18F]FET PET. METHODS We evaluated 104 patients with WHO grade II-IV glioma and suspected TP on conventional MRI using PWI and dynamic [18F]FET PET. Leakage corrected maximum relative cerebral blood volumes (rCBVmax) were obtained from dynamic susceptibility contrast PWI. Furthermore, we calculated static (i.e., maximum tumor to brain ratios; TBRmax) and dynamic [18F]FET PET parameters (i.e., Slope). Definitive diagnoses were based on histopathology (n = 42) or clinico-radiological follow-up (n = 62). The diagnostic performance of PWI and [18F]FET PET parameters to differentiate TP from TRC was evaluated by analyzing receiver operating characteristic and area under the curve (AUC). RESULTS Across all patients, the differentiation of TP from TRC using rCBVmax or [18F]FET PET parameters was moderate (AUC = 0.69-0.75; p < 0.01). A rCBVmax cutoff > 2.85 had a positive predictive value for TP of 100%, enabling a correct TP diagnosis in 44 patients. In the remaining 60 patients, combined static and dynamic [18F]FET PET parameters (TBRmax, Slope) correctly discriminated TP and TRC in a significant 78% of patients, increasing the overall accuracy to 87%. A subgroup analysis of isocitrate dehydrogenase (IDH) mutant tumors indicated a superior performance of PWI to [18F]FET PET (AUC = 0.8/< 0.62, p < 0.01/≥ 0.3). CONCLUSION While marked hyperperfusion on PWI indicated TP, [18F]FET PET proved beneficial to discriminate TP from TRC when PWI remained inconclusive. Thus, our results highlight the clinical value of sequential use of PWI and [18F]FET PET, allowing an economical use of diagnostic methods. The impact of an IDH mutation needs further investigation.
Collapse
Affiliation(s)
- Eike Steidl
- Institute of Neuroradiology, University Hospital, Goethe University Frankfurt am Main, Schleusenweg 2-16, Frankfurt am Main, 60528, Germany.
- University Cancer Center Frankfurt (UCT), University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Karl-Josef Langen
- Inst. of Neuroscience and Medicine, Medical Imaging Physics (INM-4), Research Center Juelich, Juelich, Germany
- Dept. of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Duesseldorf, Aachen, Germany
| | - Sarah Abu Hmeidan
- Institute of Neuroradiology, University Hospital, Goethe University Frankfurt am Main, Schleusenweg 2-16, Frankfurt am Main, 60528, Germany
- University Cancer Center Frankfurt (UCT), University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Nenad Polomac
- Institute of Neuroradiology, University Hospital, Goethe University Frankfurt am Main, Schleusenweg 2-16, Frankfurt am Main, 60528, Germany
- University Cancer Center Frankfurt (UCT), University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Christian P Filss
- Inst. of Neuroscience and Medicine, Medical Imaging Physics (INM-4), Research Center Juelich, Juelich, Germany
- Dept. of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Norbert Galldiks
- Inst. of Neuroscience and Medicine, Cognitive Neuroscience (INM-3), Research Center Juelich, Juelich, Germany
- Dept. of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Duesseldorf, Cologne, Germany
| | - Philipp Lohmann
- Inst. of Neuroscience and Medicine, Cognitive Neuroscience (INM-3), Research Center Juelich, Juelich, Germany
- Department of Stereotaxy and Functional Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Fee Keil
- Institute of Neuroradiology, University Hospital, Goethe University Frankfurt am Main, Schleusenweg 2-16, Frankfurt am Main, 60528, Germany
- University Cancer Center Frankfurt (UCT), University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Katharina Filipski
- University Cancer Center Frankfurt (UCT), University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Felix M Mottaghy
- Dept. of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Duesseldorf, Aachen, Germany
- Dept. of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Nadim Jon Shah
- Inst. of Neuroscience and Medicine, Medical Imaging Physics (INM-4), Research Center Juelich, Juelich, Germany
- Inst. of Neuroscience and Medicine, Molecular Neuroscience and Neuroimaging (INM-11), JARA, Research Center Juelich, Juelich, Germany
- Dept. of Neurology, University Hospital RWTH Aachen, Aachen, Germany
- JARA - BRAIN - Translational Medicine, Aachen, Germany
| | - Joachim P Steinbach
- University Cancer Center Frankfurt (UCT), University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Dr. Senckenberg Institute of Neurooncology, University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Elke Hattingen
- Institute of Neuroradiology, University Hospital, Goethe University Frankfurt am Main, Schleusenweg 2-16, Frankfurt am Main, 60528, Germany
- University Cancer Center Frankfurt (UCT), University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gabriele D Maurer
- University Cancer Center Frankfurt (UCT), University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Dr. Senckenberg Institute of Neurooncology, University Hospital, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
15
|
Li Z, Kong Z, Chen J, Li J, Li N, Yang Z, Wang Y, Liu Z. 18F-Boramino acid PET/CT in healthy volunteers and glioma patients. Eur J Nucl Med Mol Imaging 2021; 48:3113-3121. [PMID: 33590273 DOI: 10.1007/s00259-021-05212-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/18/2021] [Indexed: 01/08/2023]
Abstract
PURPOSE In this work, the safety, biodistribution, and radiation dosimetry of large neutral amino acid transporter type-1 (LAT-1) targeting PET tracer 18F-trifluorobborate-derived tyrosine (denoted as 18F-FBY) has been investigated. It is designed as a first-in-human study in healthy volunteers and to assay LAT-1 expression level in glioma patients. METHODS Six healthy volunteers (3 M, 3 F) underwent whole-body PET acquisitions at multiple time points after bolus injection of 18F-FBY. Regions of interest (ROIs) were mapped manually on major organs, and then the time-activity curves (TACs) were obtained. Dosimetry was calculated with the OLINDA/EXM software. Thirteen patients who were suspected of glioma were scanned with PET/CT at 30 min after 18F-FBY injection. Within 7 days after PET/CT, the tumor was removed surgically, and LAT-1 immunohistochemical staining for LAT-1 was performed on tumor samples and correlated with 18F-FBY PET imaging. RESULTS 18F-FBY was well tolerated by all healthy volunteers, and no adverse symptoms were observed or reported. 18F-FBY is rapidly cleared from the blood circulation and excreted mainly through the kidneys and urinary tract. The effective dose (ED) was 0.0039 ± 0.0006 mSv/MBq. In 14 surgical confirmed gliomas (one of the patiens had two gliomas), 18F-FBY uptake increased consistently with tumor grade, with maximum standard uptake values (SUVmax) of 0.28 ± 0.14 and 2.84 ± 0.46 and tumor-to-normal contralateral activity (T/N) ratio of 2.30 ± 1.26 and 24.56 ± 6.32 in low- and high-grade tumors, respectively. In addition to the significant difference in the uptakes between low- and high-grade gliomas (P < 0.001), the immunohistochemical staining confirmed the positive correlations between the SUVmax, LAT-1 expression (r2 = 0.80, P < 0.001), and Ki-67 labeling index (r2 = 0.79, P < 0.001). CONCLUSION 18F-FBY is a PET tracer with favorable dosimetry profile and pharmacokinetics. It has the potential to assay LAT-1 expression in glioma patients and may provide imaging guidance for further boron neutron capture therapy of gliomas. TRIAL REGISTRATION clinicaltrials.gov (NCT03980431).
Collapse
Affiliation(s)
- Zhu Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of nuclear medicine, Peking University Cancer Hospital & Institute, Beijing, 100871, China
| | - Ziren Kong
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junyi Chen
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Jiyuan Li
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Nan Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of nuclear medicine, Peking University Cancer Hospital & Institute, Beijing, 100871, China
| | - Zhi Yang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of nuclear medicine, Peking University Cancer Hospital & Institute, Beijing, 100871, China.
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Zhibo Liu
- Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China. .,Peking University-Tsinghua University Center for Life Sciences, Beijing, 100871, China.
| |
Collapse
|
16
|
Schön S, Cabello J, Liesche-Starnecker F, Molina-Romero M, Eichinger P, Metz M, Karimov I, Preibisch C, Keupp J, Hock A, Meyer B, Weber W, Zimmer C, Pyka T, Yakushev I, Gempt J, Wiestler B. Imaging glioma biology: spatial comparison of amino acid PET, amide proton transfer, and perfusion-weighted MRI in newly diagnosed gliomas. Eur J Nucl Med Mol Imaging 2020; 47:1468-1475. [PMID: 31953672 PMCID: PMC7188730 DOI: 10.1007/s00259-019-04677-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Imaging glioma biology holds great promise to unravel the complex nature of these tumors. Besides well-established imaging techniques such O-(2-[18F]fluoroethyl)-L-tyrosine (FET)-PET and dynamic susceptibility contrast (DSC) perfusion imaging, amide proton transfer-weighted (APTw) imaging has emerged as a promising novel MR technique. In this study, we aimed to better understand the relation between these imaging biomarkers and how well they capture cellularity and vascularity in newly diagnosed gliomas. METHODS Preoperative MRI and FET-PET data of 46 patients (31 glioblastoma and 15 lower-grade glioma) were segmented into contrast-enhancing and FLAIR-hyperintense areas. Using established cutoffs, we calculated hot-spot volumes (HSV) and their spatial overlap. We further investigated APTw and CBV values in FET-HSV. In a subset of 10 glioblastoma patients, we compared cellularity and vascularization in 34 stereotactically targeted biopsies with imaging. RESULTS In glioblastomas, the largest HSV was found for APTw, followed by PET and CBV (p < 0.05). In lower-grade gliomas, APTw-HSV was clearly lower than in glioblastomas. The spatial overlap of HSV was highest between APTw and FET in both tumor entities and regions. APTw correlated significantly with cellularity, similar to FET, while the association with vascularity was more pronounced in CBV and FET. CONCLUSIONS We found a relevant spatial overlap in glioblastomas between hotspots of APTw and FET both in contrast-enhancing and FLAIR-hyperintense tumor. As suggested by earlier studies, APTw was lower in lower-grade gliomas compared with glioblastomas. APTw meaningfully contributes to biological imaging of gliomas.
Collapse
Affiliation(s)
- S Schön
- Department of Neuroradiology, Klinikum Rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - J Cabello
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - F Liesche-Starnecker
- Department of Neuropathology, Institute of Pathology, Technical University of Munich, Munich, Germany
| | - M Molina-Romero
- Image-based Biomedical Modeling, Technical University of Munich, Munich, Germany
| | - P Eichinger
- Department of Neuroradiology, Klinikum Rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - M Metz
- Department of Neuroradiology, Klinikum Rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - I Karimov
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - C Preibisch
- Department of Neuroradiology, Klinikum Rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - J Keupp
- Philips Research, Hamburg, Germany
| | - A Hock
- Philips Health Systems, Zurich, Switzerland
| | - B Meyer
- Department of Neurosurgery, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - W Weber
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - C Zimmer
- Department of Neuroradiology, Klinikum Rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - T Pyka
- Department of Neuroradiology, Klinikum Rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - I Yakushev
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - J Gempt
- Department of Neurosurgery, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - B Wiestler
- Department of Neuroradiology, Klinikum Rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany.
| |
Collapse
|