1
|
Stauff E, Xu W, Kecskemethy HH, Langhans SA, Kandula VVR, Averill LW, Yue X. Tryptophan Kynurenine Pathway-Based Imaging Agents for Brain Disorders and Oncology-From Bench to Bedside. Biomolecules 2025; 15:47. [PMID: 39858441 PMCID: PMC11762981 DOI: 10.3390/biom15010047] [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: 11/15/2024] [Revised: 12/23/2024] [Accepted: 12/28/2024] [Indexed: 01/27/2025] Open
Abstract
Tryptophan (Trp)-based radiotracers have excellent potential for imaging many different types of brain pathology because of their involvement with both the serotonergic and kynurenine (KYN) pathways. However, radiotracers specific to the kynurenine metabolism pathway are limited. In addition, historically Trp-based radiopharmaceuticals were synthesized with the short-lived isotope carbon-11. A newer generation of Trp-based imaging agents using the longer half-lived and commercially available isotopes, such as fluorine-18 and iodine-124, are being developed. The newly developed amino acid-based tracers have been demonstrated to have favorable radiochemical and imaging characteristics in pre-clinical studies. However, many barriers still exist in the clinical translation of KYN pathway-specific radiotracers.
Collapse
Affiliation(s)
- Erik Stauff
- Department of Radiology, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA; (E.S.); (W.X.); (H.H.K.); (V.V.R.K.); (L.W.A.)
- Diagnostic & Research PET/MR Center, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA;
| | - Wenqi Xu
- Department of Radiology, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA; (E.S.); (W.X.); (H.H.K.); (V.V.R.K.); (L.W.A.)
- Diagnostic & Research PET/MR Center, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA;
| | - Heidi H. Kecskemethy
- Department of Radiology, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA; (E.S.); (W.X.); (H.H.K.); (V.V.R.K.); (L.W.A.)
- Diagnostic & Research PET/MR Center, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA;
| | - Sigrid A. Langhans
- Diagnostic & Research PET/MR Center, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA;
- Division of Neurology, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA
| | - Vinay V. R. Kandula
- Department of Radiology, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA; (E.S.); (W.X.); (H.H.K.); (V.V.R.K.); (L.W.A.)
- Diagnostic & Research PET/MR Center, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA;
| | - Lauren W. Averill
- Department of Radiology, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA; (E.S.); (W.X.); (H.H.K.); (V.V.R.K.); (L.W.A.)
- Diagnostic & Research PET/MR Center, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA;
| | - Xuyi Yue
- Department of Radiology, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA; (E.S.); (W.X.); (H.H.K.); (V.V.R.K.); (L.W.A.)
- Diagnostic & Research PET/MR Center, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA;
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
2
|
Horsley PJ, Bailey DL, Schembri G, Hsiao E, Drummond J, Back MF. The role of amino acid PET in radiotherapy target volume delineation for adult-type diffuse gliomas: A review of the literature. Crit Rev Oncol Hematol 2025; 205:104552. [PMID: 39521308 DOI: 10.1016/j.critrevonc.2024.104552] [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: 06/13/2024] [Revised: 10/31/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
PURPOSE To summarise existing literature examining amino acid positron emission tomography (AA-PET) for radiotherapy target volume delineation in patients with gliomas. METHODS Systematic search of MEDLINE and EMBASE databases. RESULTS Twenty studies met inclusion criteria. Studies comparing MRI- and AA-PET- derived target volumes consistently found these to be complementary. Across studies, AA-PET was a strong predictor of the site of subsequent relapse. In studies examining AA-PET-guided radiotherapy at standard doses, including one study using reduced margins, survival outcomes were similar to historical cohorts whose volumes were generated using MRI alone. Four prospective single-arm trials examining AA-PET-guided dose-escalated radiotherapy reported mixed results. The two trials that used both a higher biologically-effective dose and boost-volumes defined using both MRI and AA-PET reported promising outcomes. CONCLUSION AA-PET is a promising complementary tool to MRI for radiotherapy target volume delineation, with potential benefits requiring further validation including margin reduction and facilitation of dose-escalation.
Collapse
Affiliation(s)
- Patrick J Horsley
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, New South Wales, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, Australia.
| | - Dale L Bailey
- Department of Nuclear Medicine, Royal North Shore Hospital, St Leonards, New South Wales, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Geoffrey Schembri
- Department of Nuclear Medicine, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Edward Hsiao
- Department of Nuclear Medicine, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - James Drummond
- Department of Radiology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Michael F Back
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, New South Wales, Australia; The Brain Cancer Group, Sydney, New South Wales, Australia; Northern Clinical School, Sydney Medical School, University of Sydney, Sydney, Australia; Central Coast Cancer Centre, Gosford Hospital, Gosford, New South Wales, Australia
| |
Collapse
|
3
|
Kumar P, Kumar A, Nagaraj C, Sadashiva N, Saini J, Mangalore S, Rajan A, Sitani K, Beniwal M, Santosh V, Basavaraja H, Hazari PP, Mishra AK. Evaluating the Diagnostic Efficacy of 99mTc-Methionine Single-Photon Emission Computed Tomography-Computed Tomography: A Head-to-Head Comparison with 11C-Methionine Positron Emission Tomography-Magnetic Resonance Imaging in Glioma Patients. Cancer Biother Radiopharm 2024; 39:349-357. [PMID: 38324045 DOI: 10.1089/cbr.2023.0147] [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] [Indexed: 02/08/2024] Open
Abstract
Background: Amino acid positron emission tomography (PET) imaging plays a significant role in the diagnosis of gliomas and in differentiating tumor recurrence from necrosis. In this study, the authors evaluated the diagnostic efficacy of [99mTc]Tc-methionine single-photon emission computed tomography-computed tomography (SPECT-CT) in comparison with [11C]methionine PET-magnetic resonance imaging (MRI) in delineating tumors. Methods: Thirty-one (primary: 16 and postoperative: 15) patients of confirmed (either MRI or histopathological proven) glioma underwent both [99mTc]Tc-methionine SPECT-CT and [11C]methionine PET-MRI. A comparative analysis was performed between SPECT, PET, and MR images to calculate the concordance between the modalities and to evaluate the diagnostic efficacy of the [99mTc]Tc-methionine SPECT. Results: [99mTc]Tc-methionine SPECT showed comparable uptake in the tumor lesions in comparison to [11C]methionine PET. A significant and strong positive correlation was observed between the volume of tumor (Vt) in PET and Vt MR (p < 0.004). Likewise, a significant and strong positive correlation was found between Vt SPECT and Vt MR. [99mTc]-methionine has a sensitivity and specificity of 91% and 75%, respectively, compared with 82% and 100% for [11C]methionine in postoperative cases to differentiate the tumor recurrence from necrosis. The sensitivity and specificity of [99mTc]Tc-methionine was 92% and 100%, respectively, compared with 92% and 67% for [11C]methionine in primary tumors. Conclusion: [99mTc]Tc-methionine SPECT-CT is as equally good as [11C]methionine for diagnosing and differentiating it from necrosis especially in high-grade glioma.
Collapse
Affiliation(s)
- Pardeep Kumar
- Department of Neuroimaging and Interventional Radiology (NI&IR), National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Aishwarya Kumar
- Department of Neuroimaging and Interventional Radiology (NI&IR), National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Chandana Nagaraj
- Department of Neuroimaging and Interventional Radiology (NI&IR), National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Nishanth Sadashiva
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Jitender Saini
- Department of Neuroimaging and Interventional Radiology (NI&IR), National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Sandhya Mangalore
- Department of Neuroimaging and Interventional Radiology (NI&IR), National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Archith Rajan
- Department of Neuroimaging and Interventional Radiology (NI&IR), National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Keerti Sitani
- Department of Neuroimaging and Interventional Radiology (NI&IR), National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Manish Beniwal
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Vani Santosh
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| | - Harish Basavaraja
- Department of Nuclear Medicine, Kidwai Memorial Institute of Oncology, Bengaluru, India
| | | | - Anil Kumar Mishra
- Institute of Nuclear Medicine and Allied Sciences (INMAS), Delhi, India
| |
Collapse
|
4
|
Heidari M, Shokrani P. Imaging Role in Diagnosis, Prognosis, and Treatment Response Prediction Associated with High-grade Glioma. JOURNAL OF MEDICAL SIGNALS & SENSORS 2024; 14:7. [PMID: 38993200 PMCID: PMC11111132 DOI: 10.4103/jmss.jmss_30_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 07/31/2022] [Accepted: 03/14/2023] [Indexed: 07/13/2024]
Abstract
Background Glioma is one of the most drug and radiation-resistant tumors. Gliomas suffer from inter- and intratumor heterogeneity which makes the outcome of similar treatment protocols vary from patient to patient. This article is aimed to overview the potential imaging markers for individual diagnosis, prognosis, and treatment response prediction in malignant glioma. Furthermore, the correlation between imaging findings and biological and clinical information of glioma patients is reviewed. Materials and Methods The search strategy in this study is to select related studies from scientific websites such as PubMed, Scopus, Google Scholar, and Web of Science published until 2022. It comprised a combination of keywords such as Biomarkers, Diagnosis, Prognosis, Imaging techniques, and malignant glioma, according to Medical Subject Headings. Results Some imaging parameters that are effective in glioma management include: ADC, FA, Ktrans, regional cerebral blood volume (rCBV), cerebral blood flow (CBF), ve, Cho/NAA and lactate/lipid ratios, intratumoral uptake of 18F-FET (for diagnostic application), RD, ADC, ve, vp, Ktrans, CBFT1, rCBV, tumor blood flow, Cho/NAA, lactate/lipid, MI/Cho, uptakes of 18F-FET, 11C-MET, and 18F-FLT (for prognostic and predictive application). Cerebral blood volume and Ktrans are related to molecular markers such as vascular endothelial growth factor (VEGF). Preoperative ADCmin value of GBM tumors is associated with O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status. 2-hydroxyglutarate metabolite and dynamic 18F-FDOPA positron emission tomography uptake are related to isocitrate dehydrogenase (IDH) mutations. Conclusion Parameters including ADC, RD, FA, rCBV, Ktrans, vp, and uptake of 18F-FET are useful for diagnosis, prognosis, and treatment response prediction in glioma. A significant correlation between molecular markers such as VEGF, MGMT, and IDH mutations with some diffusion and perfusion imaging parameters has been identified.
Collapse
Affiliation(s)
- Maryam Heidari
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parvaneh Shokrani
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
5
|
Chen S, Jin C, Ohgaki R, Xu M, Okanishi H, Kanai Y. Structure-activity characteristics of phenylalanine analogs selectively transported by L-type amino acid transporter 1 (LAT1). Sci Rep 2024; 14:4651. [PMID: 38409393 PMCID: PMC10897196 DOI: 10.1038/s41598-024-55252-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 02/21/2024] [Indexed: 02/28/2024] Open
Abstract
L-type amino acid transporter 1 (LAT1) is a transmembrane protein responsible for transporting large neutral amino acids. While numerous LAT1-targeted compound delivery for the brain and tumors have been investigated, their LAT1 selectivity often remains ambiguous despite high LAT1 affinity. This study assessed the LAT1 selectivity of phenylalanine (Phe) analogs, focusing on their structure-activity characteristics. We discovered that 2-iodo-L-phenylalanine (2-I-Phe), with an iodine substituent at position 2 in the benzene ring, markedly improves LAT1 affinity and selectivity compared to parent amino acid Phe, albeit at the cost of reduced transport velocity. L-Phenylglycine (Phg), one carbon shorter than Phe, was found to be a substrate for LAT1 with a lower affinity, exhibiting a low level of selectivity for LAT1 equivalent to Phe. Notably, (R)-2-amino-1,2,3,4-tetrahydro-2-naphthoic acid (bicyclic-Phe), with an α-methylene moiety akin to the α-methyl group in α-methyl-L-phenylalanine (α-methyl-Phe), a known LAT1-selective compound, showed similar LAT1 transport maximal velocity to α-methyl-Phe, but with higher LAT1 affinity and selectivity. In vivo studies revealed tumor-specific accumulation of bicyclic-Phe, underscoring the importance of LAT1-selectivity in targeted delivery. These findings emphasize the potential of bicyclic-Phe as a promising LAT1-selective component, providing a basis for the development of LAT1-targeting compounds based on its structural framework.
Collapse
Affiliation(s)
- Sihui Chen
- Department of Bio-System Pharmacology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Chunhuan Jin
- Department of Bio-System Pharmacology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ryuichi Ohgaki
- Department of Bio-System Pharmacology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, 565-0871, Japan
| | - Minhui Xu
- Department of Bio-System Pharmacology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hiroki Okanishi
- Department of Bio-System Pharmacology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshikatsu Kanai
- Department of Bio-System Pharmacology, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, 565-0871, Japan.
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
6
|
Muzik O, Shields AF, Barger GR, Jiang H, Chamiraju P, Juhász C. The First Human Application of an F-18-Labeled Tryptophan Analog for PET Imaging of Cancer. Mol Imaging Biol 2024; 26:29-35. [PMID: 38012510 DOI: 10.1007/s11307-023-01877-8] [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: 09/15/2023] [Revised: 11/01/2023] [Accepted: 11/10/2023] [Indexed: 11/29/2023]
Abstract
PURPOSE Preclinical studies showed the tryptophan analog PET radiotracer 1-(2-18F-fluoroethyl)-L-tryptophan (18F-FETrp) to accumulate in various tumors, including gliomas, and being metabolized via the immunosuppressive kynurenine pathway. In this first-in-human study, we tested the use 18F-FETrp-PET in patients with neuroendocrine and brain tumors. PROCEDURES We applied dynamic brain imaging in patients with gliomas (n = 2) and multi-pass 3D whole-body PET scans in patients with neuroendocrine tumors (n =4). Semiquantitative analysis of organ and tumor tracer uptake was performed using standardized uptake values (SUVs). In addition, organ dosimetry was performed based on extracted time-activity curves and the OLINDA software. RESULTS Neuroendocrine tumors showed an early peak (10-min post-injection) followed by washout. Both gliomas showed prolonged 18F-FETrp accumulation plateauing around 40 min and showing heterogeneous uptake including non-enhancing tumor regions. Biodistribution showed moderate liver uptake and fast clearance of radioactivity into the urinary bladder; the estimated effective doses were similar to other 18F-labeled radioligands. CONCLUSIONS The study provides proof-of-principle data for the safety and potential clinical value of 18F-FETrp-PET for molecular imaging of human gliomas.
Collapse
Affiliation(s)
- Otto Muzik
- Department of Pediatrics, Wayne State University, Detroit, MI, USA.
- Department of Neurology, Wayne State University, Detroit, MI, USA.
- PET Center, Karmanos Cancer Institute, Detroit, MI, USA.
| | - Anthony F Shields
- PET Center, Karmanos Cancer Institute, Detroit, MI, USA
- Department of Oncology, Wayne State University, Detroit, MI, USA
| | | | - Huailei Jiang
- PET Center, Karmanos Cancer Institute, Detroit, MI, USA
- Department of Oncology, Wayne State University, Detroit, MI, USA
| | | | - Csaba Juhász
- Department of Pediatrics, Wayne State University, Detroit, MI, USA
- Department of Neurology, Wayne State University, Detroit, MI, USA
- PET Center, Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
7
|
Kim EY, Vavere AL, Snyder SE, Chiang J, Li Y, Patni T, Qaddoumi I, Merchant TE, Robinson GW, Holtrop JL, Shulkin BL, Bag AK. [11C]-methionine positron emission tomography in the evaluation of pediatric low-grade gliomas. Neurooncol Adv 2024; 6:vdae056. [PMID: 38680989 PMCID: PMC11055465 DOI: 10.1093/noajnl/vdae056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024] Open
Abstract
Background [11C]-Methionine positron emission tomography (PET; [11C]-MET-PET) is principally used for the evaluation of brain tumors in adults. Although amino acid PET tracers are more commonly used in the evaluation of pediatric brain tumors, data on [11C]-MET-PET imaging of pediatric low-grade gliomas (pLGG) is scarce. This study aimed to investigate the roles of [11C]-MET-PET in the evaluation of pLGGs. Methods Eighteen patients with newly diagnosed pLGG and 26 previously treated pLGG patients underwent [11C]-MET-PET met the inclusion and exclusion criteria. Tumor-to-brain uptake ratio (TBR) and metabolic tumor volumes were assessed for diagnostic performances (newly diagnosed, 15; previously treated 26), change with therapy (newly diagnosed, 9; previously treated 7), and variability among different histology (n = 12) and molecular markers (n = 7) of pLGGs. Results The sensitivity of [11C]-MET-PET for diagnosing pLGG, newly diagnosed, and previously treated combined was 93% for both TBRmax and TBRpeak, 76% for TBRmean, and 95% for qualitative evaluation. TBRmax showed a statistically significant reduction after treatment, while other PET parameters showed a tendency to decrease. Median TBRmax, TBRpeak, and TBRmean values were slightly higher in the BRAFV600E mutated tumors compared to the BRAF fused tumors. Median TBRmax, and TBRpeak in diffuse astrocytomas were higher compared to pilocytic astrocytomas, but median TBRmean, was slightly higher in pilocytic astrocytomas. However, formal statistical analysis was not done due to the small sample size. Conclusions Our study shows that [11C]-MET-PET reliably characterizes new and previously treated pLGGs. Our study also shows that quantitative parameters tend to decrease with treatment, and differences may exist between various pLGG types.
Collapse
Affiliation(s)
- Emily Y Kim
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Amy L Vavere
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Scott E Snyder
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Jason Chiang
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Yimei Li
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Tushar Patni
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Ibrahim Qaddoumi
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Thomas E Merchant
- Department of Radiation Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Giles W Robinson
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Joseph L Holtrop
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Barry L Shulkin
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Asim K Bag
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
8
|
Phillips KA, Kamson DO, Schiff D. Disease Assessments in Patients with Glioblastoma. Curr Oncol Rep 2023; 25:1057-1069. [PMID: 37470973 DOI: 10.1007/s11912-023-01440-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2023] [Indexed: 07/21/2023]
Abstract
PURPOSE OF REVIEW The neuro-oncology team faces a unique challenge when assessing treatment response in patients diagnosed with glioblastoma. Magnetic resonance imaging (MRI) remains the standard imaging modality for measuring therapeutic response in both clinical practice and clinical trials. However, even for the neuroradiologist, MRI interpretations are not straightforward because of tumor heterogeneity, as evidenced by varying degrees of enhancement, infiltrating tumor patterns, cellular densities, and vasogenic edema. The situation is even more perplexing following therapy since treatment-related changes can mimic viable tumor. Additionally, antiangiogenic therapies can dramatically decrease contrast enhancement giving the false impression of decreasing tumor burden. Over the past few decades, several approaches have emerged to augment and improve visual interpretation of glioblastoma response to therapeutics. Herein, we summarize the state of the art for evaluating the response of glioblastoma to standard therapies and investigational agents as well as challenges and future directions for assessing treatment response in neuro-oncology. RECENT FINDINGS Monitoring glioblastoma responses to standard therapy and novel agents has been fraught with many challenges and limitations over the past decade. Excitingly, new promising methods are emerging to help address these challenges. Recently, the Response Assessment in Neuro-Oncology (RANO) working group proposed an updated response criteria (RANO 2.0) for the evaluation of all grades of glial tumors regardless of IDH status or therapies being evaluated. In addition, advanced neuroimaging techniques, such as histogram analysis, parametric response maps, morphometric segmentation, radio pharmacodynamics approaches, and the integrating of amino acid radiotracers in the tumor evaluation algorithm may help resolve equivocal lesion interpretations without operative intervention. Moreover, the introduction of other techniques, such as liquid biopsy and artificial intelligence could complement conventional visual assessment of glioblastoma response to therapies. Neuro-oncology has evolved over the past decade and has achieved significant milestones, including the establishment of new standards of care, emerging therapeutic options, and novel clinical, translational, and basic research. More recently, the integration of histopathology with molecular features for tumor classification has marked an important paradigm shift in brain tumor diagnosis. In a similar manner, treatment response monitoring in neuro-oncology has made considerable progress. While most techniques are still in their inception, there is an emerging body of evidence for clinical application. Further research will be critically important for the development of impactful breakthroughs in this area of the field.
Collapse
Affiliation(s)
- Kester A Phillips
- The Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment at Swedish Neuroscience Institute, 550 17Th Ave Suite 540, Seattle, WA, 98122, USA
| | - David O Kamson
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, 201 North Broadway, Skip Viragh Outpatient Cancer Building, 9Th Floor, Room 9177, Mailbox #3, Baltimore, MD, 21218, USA
| | - David Schiff
- Division of Neuro-Oncology, University of Virginia Health System, 1300 Jefferson Park Avenue, West Complex, Room 6225, Charlottesville, VA, 22903, USA.
| |
Collapse
|
9
|
Jiang H, Guo Y, Cai H, Viola N, Shields AF, Muzik O, Juhasz C. Automated radiosynthesis of 1-(2-[ 18 F]fluoroethyl)-L-tryptophan ([ 18 F]FETrp) for positron emission tomography (PET) imaging of cancer in humans. J Labelled Comp Radiopharm 2023; 66:180-188. [PMID: 37118900 PMCID: PMC10330273 DOI: 10.1002/jlcr.4027] [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: 03/14/2023] [Revised: 04/18/2023] [Accepted: 04/26/2023] [Indexed: 04/30/2023]
Abstract
The radiotracer 1-(2-[18 F]fluoroethyl)-L-tryptophan (L-[18 F]FETrp or [18 F]FETrp) is a substrate of indoleamine 2,3-dioxygenase, the initial and key enzyme of the kynurenine pathway associated with tumoral immune resistance. In preclinical positron emission tomography studies, [18 F]FETrp is highly accumulated in a wide range of primary and metastatic cancers, such as lung cancer, prostate cancer, and gliomas. However, the clinical translation of this radiotracer into the first-in-human trial has not been reported, partially due to its racemization during radiofluorination which renders the purification of the final product challenging. However, efficient purification is essential for human studies in order to assure radiochemical and enantiomeric purity. In this work, we report a fully automated radiosynthesis of [18 F]FETrp on a Synthra RNPlus research module, including a one-pot two steps radiosynthesis, dual independent chiral and reverse-phase semipreparative high-performance liquid chromatography purifications, and solid-phase extraction-assisted formulation. The presented approach has led to its Investigational New Drug application and approval that allows the testing of this tracer in humans.
Collapse
Affiliation(s)
- Huailei Jiang
- Cyclotron and Radiochemistry Core, Karmanos Cancer Institute, Detroit, MI, USA
- PET Center, Karmanos Cancer Institute, Detroit, MI, USA
- Department of Oncology, Wayne State University, Detroit, MI, USA
| | - Yan Guo
- Cyclotron and Radiochemistry Core, Karmanos Cancer Institute, Detroit, MI, USA
- PET Center, Karmanos Cancer Institute, Detroit, MI, USA
- Department of Oncology, Wayne State University, Detroit, MI, USA
| | - Hancheng Cai
- PET Radiochemistry Facility, Mayo Clinic, Jacksonville, FL, USA
| | - Nerissa Viola
- PET Center, Karmanos Cancer Institute, Detroit, MI, USA
- Department of Oncology, Wayne State University, Detroit, MI, USA
| | - Anthony F. Shields
- PET Center, Karmanos Cancer Institute, Detroit, MI, USA
- Department of Oncology, Wayne State University, Detroit, MI, USA
| | - Otto Muzik
- PET Center, Karmanos Cancer Institute, Detroit, MI, USA
- Departments of Pediatrics and Neurology, Wayne State University, Detroit, MI, USA
| | - Csaba Juhasz
- PET Center, Karmanos Cancer Institute, Detroit, MI, USA
- Departments of Pediatrics and Neurology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
10
|
Laudicella R, Mantarro C, Catalfamo B, Alongi P, Gaeta M, Minutoli F, Baldari S, Bisdas S. PET Imaging in Gliomas. RADIOLOGY‐NUCLEAR MEDICINE DIAGNOSTIC IMAGING 2023:194-218. [DOI: 10.1002/9781119603627.ch6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
11
|
Muthukumar S, Darden J, Crowley J, Witcher M, Kiser J. A Comparison of PET Tracers in Recurrent High-Grade Gliomas: A Systematic Review. Int J Mol Sci 2022; 24:ijms24010408. [PMID: 36613852 PMCID: PMC9820099 DOI: 10.3390/ijms24010408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022] Open
Abstract
Humans with high-grade gliomas have a poor prognosis, with a mean survival time of just 12-18 months for patients who undergo standard-of-care tumor resection and adjuvant therapy. Currently, surgery and chemoradiotherapy serve as standard treatments for this condition, yet these can be complicated by the tumor location, growth rate and recurrence. Currently, gadolinium-based, contrast-enhanced magnetic resonance imaging (CE-MRI) serves as the predominant imaging modality for recurrent high-grade gliomas, but it faces several drawbacks, including its inability to distinguish tumor recurrence from treatment-related changes and its failure to reveal the entirety of tumor burden (de novo or recurrent) due to limitations inherent to gadolinium contrast. As such, alternative imaging modalities that can address these limitations, including positron emission tomography (PET), are worth pursuing. To this end, the identification of PET-based markers for use in imaging of recurrent high-grade gliomas is paramount. This review will highlight several PET radiotracers that have been implemented in clinical practice and provide a comparison between them to assess the efficacy of these tracers.
Collapse
Affiliation(s)
| | - Jordan Darden
- Carilion Clinic Neurosurgery, Roanoke, VA 24016, USA
| | | | - Mark Witcher
- Carilion Clinic Neurosurgery, Roanoke, VA 24016, USA
| | - Jackson Kiser
- Carilion Clinic Radiology, Roanoke, VA 24016, USA
- Correspondence:
| |
Collapse
|
12
|
A Systematic Review of Amino Acid PET Imaging in Adult-Type High-Grade Glioma Surgery: A Neurosurgeon's Perspective. Cancers (Basel) 2022; 15:cancers15010090. [PMID: 36612085 PMCID: PMC9817716 DOI: 10.3390/cancers15010090] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/04/2022] [Accepted: 12/13/2022] [Indexed: 12/29/2022] Open
Abstract
Amino acid PET imaging has been used for a few years in the clinical and surgical management of gliomas with satisfactory results in diagnosis and grading for surgical and radiotherapy planning and to differentiate recurrences. Biological tumor volume (BTV) provides more meaningful information than standard MR imaging alone and often exceeds the boundary of the contrast-enhanced nodule seen in MRI. Since a gross total resection reflects the resection of the contrast-enhanced nodule and the majority of recurrences are at a tumor's margins, an integration of PET imaging during resection could increase PFS and OS. A systematic review of the literature searching for "PET" [All fields] AND "glioma" [All fields] AND "resection" [All fields] was performed in order to investigate the diffusion of integration of PET imaging in surgical practice. Integration in a neuronavigation system and intraoperative use of PET imaging in the primary diagnosis of adult high-grade gliomas were among the criteria for article selection. Only one study has satisfied the inclusion criteria, and a few more (13) have declared to use multimodal imaging techniques with the integration of PET imaging to intentionally perform a biopsy of the PET uptake area. Despite few pieces of evidence, targeting a biologically active area in addition to other tools, which can help intraoperatively the neurosurgeon to increase the amount of resected tumor, has the potential to provide incremental and complementary information in the management of brain gliomas. Since supramaximal resection based on the extent of MRI FLAIR hyperintensity resulted in an advantage in terms of PFS and OS, PET-based biological tumor volume, avoiding new neurological deficits, deserves further investigation.
Collapse
|
13
|
18F-Radiolabeled Translocator Protein (TSPO) PET Tracers: Recent Development of TSPO Radioligands and Their Application to PET Study. Pharmaceutics 2022; 14:pharmaceutics14112545. [PMID: 36432736 PMCID: PMC9697781 DOI: 10.3390/pharmaceutics14112545] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
Translocator protein 18 kDa (TSPO) is a transmembrane protein in the mitochondrial membrane, which has been identified as a peripheral benzodiazepine receptor. TSPO is generally present at high concentrations in steroid-producing cells and plays an important role in steroid synthesis, apoptosis, and cell proliferation. In the central nervous system, TSPO expression is relatively modest under normal physiological circumstances. However, some pathological disorders can lead to changes in TSPO expression. Overexpression of TSPO is associated with several diseases, such as neurodegenerative diseases, neuroinflammation, brain injury, and cancers. TSPO has therefore become an effective biomarker of related diseases. Positron emission tomography (PET), a non-invasive molecular imaging technique used for the clinical diagnosis of numerous diseases, can detect diseases related to TSPO expression. Several radiolabeled TSPO ligands have been developed for PET. In this review, we describe recent advances in the development of TSPO ligands, and 18F-radiolabeled TSPO in particular, as PET tracers. This review covers pharmacokinetic studies, preclinical and clinical trials of 18F-labeled TSPO PET ligands, and the synthesis of TSPO ligands.
Collapse
|
14
|
Quigg M, Kundu B. Dynamic FDG-PET demonstration of functional brain abnormalities. Ann Clin Transl Neurol 2022; 9:1487-1497. [PMID: 36069052 PMCID: PMC9463948 DOI: 10.1002/acn3.51546] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 11/27/2022] Open
Abstract
Positron emission tomography with fluorine‐18 fluorodeoxyglucose (18F‐FDG‐PET) has been used over 3 decades to map patterns of brain glucose metabolism to evaluate normal brain function or demonstrate abnormalities of metabolism in brain disorders. Traditional PET maps patterns of absolute tracer uptake but has demonstrated shortcomings in disorders such as brain neoplasm or focal epilepsy in the ability to resolve normally from pathological tissue. In this review, we describe an alternative process of metabolic mapping, dynamic PET. This new technology quantifies the dynamics of tracer uptake and decays with the goal of improving the functional mapping of the desired metabolic activity in the target organ. We discuss technical implementation and findings of initial pilot studies in brain tumor treatment and epilepsy surgery.
Collapse
Affiliation(s)
- Mark Quigg
- Department of Neurology, University of Virginia, Charlottesville, Virginia, 22908, USA
| | - Bijoy Kundu
- Departments of Radiology & Medical Imaging and Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
15
|
Advanced Neuroimaging Approaches to Pediatric Brain Tumors. Cancers (Basel) 2022; 14:cancers14143401. [PMID: 35884462 PMCID: PMC9318188 DOI: 10.3390/cancers14143401] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 07/08/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary After leukemias, brain tumors are the most common cancers in children, and early, accurate diagnosis is critical to improve patient outcomes. Beyond the conventional imaging methods of computed tomography (CT) and magnetic resonance imaging (MRI), advanced neuroimaging techniques capable of both structural and functional imaging are moving to the forefront to improve the early detection and differential diagnosis of tumors of the central nervous system. Here, we review recent developments in neuroimaging techniques for pediatric brain tumors. Abstract Central nervous system tumors are the most common pediatric solid tumors; they are also the most lethal. Unlike adults, childhood brain tumors are mostly primary in origin and differ in type, location and molecular signature. Tumor characteristics (incidence, location, and type) vary with age. Children present with a variety of symptoms, making early accurate diagnosis challenging. Neuroimaging is key in the initial diagnosis and monitoring of pediatric brain tumors. Conventional anatomic imaging approaches (computed tomography (CT) and magnetic resonance imaging (MRI)) are useful for tumor detection but have limited utility differentiating tumor types and grades. Advanced MRI techniques (diffusion-weighed imaging, diffusion tensor imaging, functional MRI, arterial spin labeling perfusion imaging, MR spectroscopy, and MR elastography) provide additional and improved structural and functional information. Combined with positron emission tomography (PET) and single-photon emission CT (SPECT), advanced techniques provide functional information on tumor metabolism and physiology through the use of radiotracer probes. Radiomics and radiogenomics offer promising insight into the prediction of tumor subtype, post-treatment response to treatment, and prognostication. In this paper, a brief review of pediatric brain cancers, by type, is provided with a comprehensive description of advanced imaging techniques including clinical applications that are currently utilized for the assessment and evaluation of pediatric brain tumors.
Collapse
|
16
|
Fibroblast Activation Protein Inhibitor Theranostics. PET Clin 2022; 17:453-464. [PMID: 35717101 DOI: 10.1016/j.cpet.2022.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The evolution of the fibroblast activation protein inhibitor molecules over the past decade has brought into the forefront a novel theranostic agent that has the potential of matching the workhorse of PET/computed tomography, [fluorine-18] fluoro-2-deoxy-d-glucose (18F-FDG). It is hoped that in the next decade it can act as a complementary tracer to 18F-FDG, in providing phenotypic and biomarker information and also in directing fibroblast activation protein-targeted therapies.
Collapse
|
17
|
Galldiks N, Langen KJ, Albert NL, Law I, Kim MM, Villanueva-Meyer JE, Soffietti R, Wen PY, Weller M, Tonn JC. Investigational PET tracers in neuro-oncology-What's on the horizon? A report of the PET/RANO group. Neuro Oncol 2022; 24:1815-1826. [PMID: 35674736 DOI: 10.1093/neuonc/noac131] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Many studies in patients with brain tumors evaluating innovative PET tracers have been published in recent years, and the initial results are promising. Here, the Response Assessment in Neuro-Oncology (RANO) PET working group provides an overview of the literature on novel investigational PET tracers for brain tumor patients. Furthermore, newer indications of more established PET tracers for the evaluation of glucose metabolism, amino acid transport, hypoxia, cell proliferation, and others are also discussed. Based on the preliminary findings, these novel investigational PET tracers should be further evaluated considering their promising potential. In particular, novel PET probes for imaging of translocator protein and somatostatin receptor overexpression as well as for immune system reactions appear to be of additional clinical value for tumor delineation and therapy monitoring. Progress in developing these radiotracers may contribute to improving brain tumor diagnostics and advancing clinical translational research.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937 Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany.,Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Düsseldorf, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany.,Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Düsseldorf, Germany.,Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig Maximilians-University of Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Michelle M Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Javier E Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Riccardo Soffietti
- Department of Neuro-Oncology, University and City of Health and Science Hospital, Turin, Italy
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts, USA
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center University Hospital and University of Zurich, Zurich, Switzerland
| | - Joerg C Tonn
- Department of Neurosurgery, University Hospital of Munich (LMU), Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
18
|
Diao W, Su D, Cao Y, Jia Z. The diagnostic accuracy of O-(2-18F-fluoroethyl)-L-tyrosine parameters for the differentiation of brain tumour progression from treatment-related changes. Nucl Med Commun 2022; 43:350-358. [PMID: 35102078 DOI: 10.1097/mnm.0000000000001524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND 18F-fluoro-ethyl-tyrosine (18F-FET) is recommended to distinguish brain tumours post-therapeutic true progression (including recurrent and metastatic brain tumours) and treatment-related change (TRC). However, many parameters of 18F-FET can be used for this differential diagnosis. Our purpose was to investigate the diagnostic accuracy of various 18F-FET parameters to differentiate true progression from TRC. METHODS We performed a literature search using the following databases: the PubMed, Embase and Web of Science databases up to 29 November 2020. We included studies that reported the diagnostic test results of 18F-FET to distinguish true progression from TRC. The Quality Assessment of Diagnostic Accuracy Studies-2 tool was used to evaluate the quality of the included studies. The diagnostic accuracy of various parameters was pooled using a random-effects model. RESULTS We included 17 eligible studies (nine parameters). For static parameters of 18F-FET, the maximum and mean tumour-to-brain ratios (TBRmax and TBRmean) showed similar pooled sensitivities of 82% [95% confidence interval (CI), 80-85%) and 82% (95% CI, 78-85%), respectively. Among the three kinetic parameters (slope, time to peak and kinetic pattern), the kinetic pattern presented the optimal diagnostic value with a pooled sensitivity of 81% (95% CI, 75-86%). When combining the static and kinetic parameters, the diagnostic performance of 18F-FET was significantly improved, with a pooled sensitivity of 90% (95% CI, 84-94%) in the combination of TBR and kinetic patterns. CONCLUSIONS 18F-FET static parameters alone showed a comparably high sensitivity in the differentiation between brain tumour true progression and TRC. Combining static and kinetic parameters provided improved diagnostic performance.
Collapse
Affiliation(s)
- Wei Diao
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu, Peoples Republic of China
| | | | | | | |
Collapse
|
19
|
Prather KY, O’Neal CM, Westrup AM, Tullos HJ, Hughes KL, Conner AK, Glenn CA, Battiste JD. A systematic review of amino acid PET in assessing treatment response to temozolomide in glioma. Neurooncol Adv 2022; 4:vdac008. [PMID: 35300149 PMCID: PMC8923003 DOI: 10.1093/noajnl/vdac008] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The response assessment in neuro-oncology (RANO) criteria have been the gold standard for monitoring treatment response in glioblastoma (GBM) and differentiating tumor progression from pseudoprogression. While the RANO criteria have played a key role in detecting early tumor progression, their ability to identify pseudoprogression is limited by post-treatment damage to the blood-brain barrier (BBB), which often leads to contrast enhancement on MRI and correlates poorly to tumor status. Amino acid positron emission tomography (AA PET) is a rapidly growing imaging modality in neuro-oncology. While contrast-enhanced MRI relies on leaky vascularity or a compromised BBB for delivery of contrast agents, amino acid tracers can cross the BBB, making AA PET particularly well-suited for monitoring treatment response and diagnosing pseudoprogression. The authors performed a systematic review of PubMed, MEDLINE, and Embase through December 2021 with the search terms “temozolomide” OR “Temodar,” “glioma” OR “glioblastoma,” “PET,” and “amino acid.” There were 19 studies meeting inclusion criteria. Thirteen studies utilized [18F]FET, five utilized [11C]MET, and one utilized both. All studies used static AA PET parameters to evaluate TMZ treatment in glioma patients, with nine using dynamic tracer parameters in addition. Throughout these studies, AA PET demonstrated utility in TMZ treatment monitoring and predicting patient survival.
Collapse
Affiliation(s)
- Kiana Y Prather
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Christen M O’Neal
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Alison M Westrup
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Hurtis J Tullos
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kendall L Hughes
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Andrew K Conner
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Chad A Glenn
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - James D Battiste
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
20
|
Enhancing the accumulation level of 3-[ 18F]fluoro-L-α-methyltyrosine in tumors by preloading probenecid. Nucl Med Biol 2021; 104-105:47-52. [PMID: 34896813 DOI: 10.1016/j.nucmedbio.2021.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 11/23/2022]
Abstract
INTRODUCTION 3-[18F]fluoro-α-methyl-L-tyrosine ([18F]FAMT) is a promising amino acid tracer targeting L-type amino acid transporter 1 (LAT1). One concern regarding the diagnosis using [18F]FAMT is the possibility of false-negative findings because of its relatively low accumulation level even in malignant tumors. Moreover, preloading probenecid, an organic anion transporter inhibitor, markedly increased the tumor accumulation level of radioiodine-labeled α-methyltyrosine. In this study, we evaluated the usefulness of preloading probenecid in improving the tumor-imaging capability of [18F]FAMT. METHODS Three biodistribution studies of [18F]FAMT were conducted in normal mice to elucidate the usefulness of probenecid preloading. Later, a biodistribution study and positron emission tomography (PET) imaging of [18F]FAMT were conducted with or without probenecid injection in tumor-bearing mice. RESULTS Probenecid preloading significantly delayed blood clearance and consequently enhanced the accumulation of [18F]FAMT in the pancreas, a LAT1-positive organ. The effects of probenecid preloading were independent of the administration route. Tumor accumulation level in the biodistribution study and the maximum standardized uptake value in tumors on PET imaging of the probenecid preloading group were significantly higher than those of the control (without probenecid injection) group in tumor-bearing mice. CONCLUSIONS Preloading probenecid significantly delayed blood clearance and consequently enhanced the accumulation of [18F]FAMT in tumors. These results indicate that preloading probenecid could improve the diagnostic accuracy of [18F]FAMT.
Collapse
|
21
|
Moussaron A, Jouan-Hureaux V, Collet C, Pierson J, Thomas N, Choulier L, Veran N, Doyen M, Arnoux P, Maskali F, Dumas D, Acherar S, Barberi-Heyob M, Frochot C. Preliminary Study of New Gallium-68 Radiolabeled Peptide Targeting NRP-1 to Detect Brain Metastases by Positron Emission Tomography. Molecules 2021; 26:7273. [PMID: 34885871 PMCID: PMC8659110 DOI: 10.3390/molecules26237273] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/11/2021] [Accepted: 11/23/2021] [Indexed: 12/11/2022] Open
Abstract
Due to their very poor prognosis and a fatal outcome, secondary brain tumors are one of the biggest challenges in oncology today. From the point of view of the early diagnosis of these brain micro- and macro-tumors, the sensitivity and specificity of the diagnostic tools constitute an obstacle. Molecular imaging, such as Positron Emission Tomography (PET), is a promising technique but remains limited in the search for cerebral localizations, given the commercially available radiotracers. Indeed, the [18F]FDG PET remains constrained by the physiological fixation of the cerebral cortex, which hinders the visualization of cerebral metastases. Tumor angiogenesis is recognized as a crucial phenomenon in the progression of malignant tumors and is correlated with overexpression of the neuropilin-1 (NRP-1) receptor. Here, we describe the synthesis and the photophysical properties of the new gallium-68 radiolabeled peptide to target NRP-1. The KDKPPR peptide was coupled with gallium-68 anchored into a bifunctional NODAGA chelating agent, as well as Cy5 for fluorescence detection. The Cy5 absorbance spectra did not change, whereas the molar extinction coefficient (ε) decreased drastically. An enhancement of the fluorescence quantum yield (φF) could be observed due to the better water solubility of Cy5. [68Ga]Ga-NODAGA-K(Cy5)DKPPR was radiosynthesized efficiently, presented hydrophilic properties (log D = -1.86), and had high in vitro stability (>120 min). The molecular affinity and the cytotoxicity of this new chelated radiotracer were evaluated in vitro on endothelial cells (HUVEC) and MDA-MB-231 cancer cells (hormone-independent and triple-negative line) and in vivo on a brain model of metastasis in a nude rat using the MDA-MB-231 cell line. No in vitro toxicity has been observed. The in vivo preliminary experiments showed promising results, with a high contrast between the healthy brain and metastatic foci for [68Ga]Ga-NODAGA-K(Cy5)DKPPR.
Collapse
Affiliation(s)
- Albert Moussaron
- Université de Lorraine, CNRS, LRGP, F-54000 Nancy, France; (A.M.); (P.A.)
| | - Valérie Jouan-Hureaux
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (V.J.-H.); (J.P.); (N.T.); (M.B.-H.)
| | - Charlotte Collet
- Nancyclotep Molecular Imaging Platform, F-54500 Vandœuvre-lès-Nancy, France; (C.C.); (N.V.); (M.D.); (F.M.)
- Université de Lorraine, INSERM, U1254, IADI, F-54500 Vandœuvre-lès-Nancy, France
| | - Julien Pierson
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (V.J.-H.); (J.P.); (N.T.); (M.B.-H.)
| | - Noémie Thomas
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (V.J.-H.); (J.P.); (N.T.); (M.B.-H.)
| | | | - Nicolas Veran
- Nancyclotep Molecular Imaging Platform, F-54500 Vandœuvre-lès-Nancy, France; (C.C.); (N.V.); (M.D.); (F.M.)
| | - Matthieu Doyen
- Nancyclotep Molecular Imaging Platform, F-54500 Vandœuvre-lès-Nancy, France; (C.C.); (N.V.); (M.D.); (F.M.)
- Université de Lorraine, INSERM, U1254, IADI, F-54500 Vandœuvre-lès-Nancy, France
| | - Philippe Arnoux
- Université de Lorraine, CNRS, LRGP, F-54000 Nancy, France; (A.M.); (P.A.)
| | - Fatiha Maskali
- Nancyclotep Molecular Imaging Platform, F-54500 Vandœuvre-lès-Nancy, France; (C.C.); (N.V.); (M.D.); (F.M.)
| | | | - Samir Acherar
- Université de Lorraine, CNRS, LCPM, F-54000 Nancy, France;
| | - Muriel Barberi-Heyob
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; (V.J.-H.); (J.P.); (N.T.); (M.B.-H.)
| | - Céline Frochot
- Université de Lorraine, CNRS, LRGP, F-54000 Nancy, France; (A.M.); (P.A.)
| |
Collapse
|
22
|
Krämer F, Gröner B, Hoffmann C, Craig A, Brugger M, Drzezga A, Timmer M, Neumaier F, Zlatopolskiy BD, Endepols H, Neumaier B. Evaluation of 3-l- and 3-d-[ 18F]Fluorophenylalanines as PET Tracers for Tumor Imaging. Cancers (Basel) 2021; 13:cancers13236030. [PMID: 34885141 PMCID: PMC8656747 DOI: 10.3390/cancers13236030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The early detection and treatment of malignant brain tumors can significantly improve the survival time and life quality of affected patients. Whereas positron emission tomography (PET) with O-(2-[18F]fluoroethyl)tyrosine ([18F]FET) offers improved diagnostic accuracy compared to other imaging methods, there is still a need for PET tracers with better tumor-specificity. A higher protein incorporation rate, as well as a higher affinity for the amino acid transporter LAT1, could provide probes with superior image quality compared to [18F]FET. The aim of the present study was a preclinical evaluation of the two enantiomeric phenylalanine (Phe) analogues, 3-l- and 3-d-[18F]fluorophenylalanine ([18F]FPhes), as possible alternatives to [18F]FET. Based on promising in vitro evaluation results, the radiolabeled amino acids were studied in vivo in two subcutaneous and one orthotopic rodent tumor xenograft models using µPET. The results show that 3-l- and 3-d-[18F]FPhe enable high-quality visualization of tumors with certain advantages over [18F]FET, making them promising candidates for further preclinical and clinical evaluations. Abstract Purpose: The preclinical evaluation of 3-l- and 3-d-[18F]FPhe in comparison to [18F]FET, an established tracer for tumor imaging. Methods: In vitro studies were conducted with MCF-7, PC-3, and U87 MG human tumor cell lines. In vivo µPET studies were conducted in healthy rats with/without the inhibition of peripheral aromatic l-amino acid decarboxylase by benserazide pretreatment (n = 3 each), in mice bearing subcutaneous MCF-7 or PC-3 tumor xenografts (n = 10), and in rats bearing orthotopic U87 MG tumor xenografts (n = 14). Tracer accumulation was quantified by SUVmax, SUVmean and tumor-to-brain ratios (TBrR). Results: The uptake of 3-l-[18F]FPhe in MCF-7 and PC-3 cells was significantly higher relative to [18F]FET. The uptake of all three tracers was significantly reduced by the suppression of amino acid transport systems L or ASC. 3-l-[18F]FPhe but not 3-d-[18F]FPhe exhibited protein incorporation. In benserazide-treated healthy rats, brain uptake after 42–120 min was significantly higher for 3-d-[18F]FPhe vs. 3-l-[18F]FPhe. [18F]FET showed significantly higher uptake into subcutaneous MCF-7 tumors (52–60 min p.i.), while early uptake into orthotopic U87 MG tumors was significantly higher for 3-l-[18F]FPhe (SUVmax: 3-l-[18F]FPhe, 107.6 ± 11.3; 3-d-[18F]FPhe, 86.0 ± 4.3; [18F]FET, 90.2 ± 7.7). Increased tumoral expression of LAT1 and ASCT2 was confirmed immunohistologically. Conclusion: Both novel tracers enable accurate tumor delineation with an imaging quality comparable to [18F]FET.
Collapse
Affiliation(s)
- Felicia Krämer
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.K.); (B.G.); (C.H.); (A.C.); (F.N.); (B.D.Z.); (H.E.)
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany;
| | - Benedikt Gröner
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.K.); (B.G.); (C.H.); (A.C.); (F.N.); (B.D.Z.); (H.E.)
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany;
| | - Chris Hoffmann
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.K.); (B.G.); (C.H.); (A.C.); (F.N.); (B.D.Z.); (H.E.)
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany;
| | - Austin Craig
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.K.); (B.G.); (C.H.); (A.C.); (F.N.); (B.D.Z.); (H.E.)
| | - Melanie Brugger
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany;
| | - Alexander Drzezga
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany;
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn-Cologne, Germany
- Molecular Organization of the Brain (INM-2), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany
| | - Marco Timmer
- Center for Neurosurgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany;
| | - Felix Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.K.); (B.G.); (C.H.); (A.C.); (F.N.); (B.D.Z.); (H.E.)
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany;
| | - Boris D. Zlatopolskiy
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.K.); (B.G.); (C.H.); (A.C.); (F.N.); (B.D.Z.); (H.E.)
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany;
- Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
| | - Heike Endepols
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.K.); (B.G.); (C.H.); (A.C.); (F.N.); (B.D.Z.); (H.E.)
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany;
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany;
| | - Bernd Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (F.K.); (B.G.); (C.H.); (A.C.); (F.N.); (B.D.Z.); (H.E.)
- Nuclear Chemistry (INM-5), Institute of Neuroscience and Medicine, Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428 Jülich, Germany;
- Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
- Correspondence:
| |
Collapse
|
23
|
Lim TX, Ahamed M, Reutens DC. The aryl hydrocarbon receptor: A diagnostic and therapeutic target in glioma. Drug Discov Today 2021; 27:422-435. [PMID: 34624509 DOI: 10.1016/j.drudis.2021.09.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 07/29/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022]
Abstract
Glioblastoma multiforme (GBM) is a deadly disease; 5-year survival rates have shown little improvement over the past 30 years. In vivo positron emission tomography (PET) imaging is an important method of identifying potential diagnostic and therapeutic molecular targets non-invasively. The aryl hydrocarbon receptor (AhR) is a transcription factor that regulates multiple genes involved in immune response modulation and tumorigenesis. The AhR is an attractive potential drug target and studies have shown that its activation by small molecules can modulate innate and adaptive immunity beneficially and prevent AhR-mediated tumour promotion in several cancer types. In this review, we provide an overview of the role of the AhR in glioma tumorigenesis and highlight its potential as an emerging biomarker for glioma therapies targeting the tumour immune response and PET diagnostics.
Collapse
Affiliation(s)
- Ting Xiang Lim
- ARC Centre for Innovation in Biomedical Imaging Technology, Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - Muneer Ahamed
- ARC Centre for Innovation in Biomedical Imaging Technology, Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia
| | - David C Reutens
- ARC Centre for Innovation in Biomedical Imaging Technology, Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
24
|
Lan X, Fan K, Cai W. First-in-human study of an 18F-labeled boramino acid: a new class of PET tracers. Eur J Nucl Med Mol Imaging 2021; 48:3037-3040. [PMID: 33547555 DOI: 10.1007/s00259-021-05227-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. .,Hubei Key Laboratory of Molecular Imaging, Wuhan, China.
| | - Kevin Fan
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, 1111 Highland Avenue, Madison, WI, USA
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, 1111 Highland Avenue, Madison, WI, USA.
| |
Collapse
|
25
|
Fluorine-18-Labeled PET Radiotracers for Imaging Tryptophan Uptake and Metabolism: a Systematic Review. Mol Imaging Biol 2021; 22:805-819. [PMID: 31512038 DOI: 10.1007/s11307-019-01430-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Due to its metabolism via the serotonin and kynurenine pathways, tryptophan plays a key role in multiple disease processes including cancer. Imaging tryptophan uptake and metabolism in vivo can be achieved with tryptophan derivative positron emission tomography (PET) radiotracers. While human studies with such tracers have been confined to C-11-labeled compounds, preclinical development of F-18-labeled tryptophan-based radiotracers has surged in recent years. We performed a systematic review of studies reporting on such F-18-labeled tryptophan tracers to summarize and compare their biological characteristics and their potential for tumor imaging, with a particular focus on key enzymes of the kynurenine pathway (indoleamine 2,3-dioxygenase [IDO] and tryptophan 2,3-dioxygenase [TDO]), which play an important role in tumoral immune resistance. From a PubMed search, English language articles including data on the preparation and radiochemical and/or biological characteristics of F-18-labeled tryptophan derivative radiotracers were reviewed. A total of 19 original papers included data on 15 unique radiotracers, the majority of which were synthesized with an adequate radiochemical yield. Automated synthesis was reported for 1-(2-[18F]fluoroethyl)-L-tryptophan, the most extensively evaluated tracer thus far. Biodistribution studies showed high uptake in the pancreas, while the L-type amino acid transporter was the dominant transport mechanism for most of the reviewed tracers. Tracers tested for tumor uptake showed accumulation in tumor cell lines in vitro and in xenografts in vivo, often with favorable tumor-to-background uptake ratios in comparison with clinically used F-18-labeled radiotracers. Five tracers showed promise for imaging IDO activity, including 1-(2-[18F]fluoroethyl)-L-tryptophan and a F-18-labeled analog of alpha-[11C]methyl-L-tryptophan tested clinically in previous studies. Two radiotracers were metabolized by TDO but showed defluorination in vivo. In summary, most F-18-labeled tryptophan derivative PET tracers share common transport mechanisms and biodistribution characteristics. Several reported tracers could be candidates for further testing and validation toward PET imaging applications in a variety of human diseases.
Collapse
|
26
|
An YS, Kim SH, Roh TH, Park SH, Kim TG, Kim JH. Correlation Between 18F-FDG Uptake and Immune Cell Infiltration in Metastatic Brain Lesions. Front Oncol 2021; 11:618705. [PMID: 34249674 PMCID: PMC8266210 DOI: 10.3389/fonc.2021.618705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 06/09/2021] [Indexed: 11/13/2022] Open
Abstract
Background The purpose of this study was to investigate the correlation between 18F-fluorodeoxyglucose (FDG) uptake and infiltrating immune cells in metastatic brain lesions. Methods This retrospective study included 34 patients with metastatic brain lesions who underwent brain 18F-FDG positron emission tomography (PET)/computed tomography (CT) followed by surgery. 18F-FDG uptake ratio was calculated by dividing the standardized uptake value (SUV) of the metastatic brain lesion by the contralateral normal white matter uptake value. We investigated the clinicopathological characteristics of the patients and analyzed the correlation between 18F-FDG uptake and infiltration of various immune cells. In addition, we evaluated immune-expression levels of glucose transporter 1 (GLUT1), hexokinase 2 (HK2), and Ki-67 in metastatic brain lesions. Results The degree of 18F-FDG uptake of metastatic brain lesions was not significantly correlated with clinical parameters. There was no significant relationship between the 18F-FDG uptake and degree of immune cell infiltration in brain metastasis. Furthermore, other markers, such as GLUT1, HK2, and Ki-67, were not correlated with degree of 18F-FDG uptake. In metastatic brain lesions that originated from breast cancer, a higher degree of 18F-FDG uptake was observed in those with high expression of CD68. Conclusions In metastatic brain lesions, the degree of 18F-FDG uptake was not significantly associated with infiltration of immune cells. The 18F-FDG uptake of metastatic brain lesions from breast cancer, however, might be associated with macrophage activity.
Collapse
Affiliation(s)
- Young-Sil An
- Department of Nuclear Medicine and Molecular Imaging, Ajou University School of Medicine, Suwon, South Korea
| | - Se-Hyuk Kim
- Department of Neurosurgery, Ajou University School of Medicine, Suwon, South Korea
| | - Tae Hoon Roh
- Department of Neurosurgery, Ajou University School of Medicine, Suwon, South Korea
| | - So Hyun Park
- Department of Pathology, Ajou University School of Medicine, Suwon, South Korea
| | - Tae-Gyu Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, South Korea
| | - Jang-Hee Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, South Korea
| |
Collapse
|
27
|
Ma H, Zhao J, Liu S, Xie D, Zhang Z, Nie D, Wen F, Yang Z, Tang G. 18F-Trifluoromethylated D-Cysteine as a Promising New PET Tracer for Glioma Imaging: Comparative Analysis With MRI and Histopathology in Orthotopic C6 Models. Front Oncol 2021; 11:645162. [PMID: 33996562 PMCID: PMC8117348 DOI: 10.3389/fonc.2021.645162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/15/2021] [Indexed: 11/13/2022] Open
Abstract
Comparing MRI and histopathology, this study aims to comprehensively explore the potential application of 18F-trifluoromethylated D-cysteine (S-[18F]CF3-D-CYS) in evaluating glioma by using orthotopic C6 glioma models. Sprague-Dawley (SD) rats (n = 9) were implanted with C6 glioma cells. Tumor growth was monitored every week by multiparameter MRI [including dynamic contrast-enhanced MRI (DCE-MRI)], [18F]FDG, S-[18F]CF3-D-CYS, and [18F]FDOPA PET imaging. Repeated scans of the same rat with the two or three [18F]-labeled radiotracers were investigated. Initial regions of interest were manually delineated on T2WI and set on the same level of PET images, and tumor-to-normal brain uptake ratios (TNRs) were calculated to semiquantitatively assess the tracer accumulation in the tumor. The tumor volume in PET and histopathology was calculated. HE and Ki67 immunohistochemical staining were further performed. The correlations between the uptake of S-[18F]CF3-D-CYS and Ki67 were analyzed. Dynamic S-[18F]CF3-D-CYS PET imaging showed tumor uptake rapidly reached a peak, maintained plateau during 10-30 min after injection, then decreased slowly. Compared with [18F]FDG and [18F]FDOPA PET imaging, S-[18F]CF3-D-CYS PET demonstrated the highest TNRs (P < 0.05). There were no significant differences in the tumor volume measured on S-[18F]CF3-D-CYS PET or HE specimen. Furthermore, our results showed that the uptake of S-[18F]CF3-D-CYS was significantly positively correlated with tumor Ki67, and the poor accumulated S-[18F]CF3-D-CYS was consistent with tumor hemorrhage. There was no significant correlation between the S-[18F]CF3-D-CYS uptakes and the Ktrans values derived from DCE-MRI. In comparison with MRI and histopathology, S-[18F]CF3-D-CYS PET performs well in the diagnosis and evaluation of glioma. S-[18F]CF3-D-CYS PET may serve as a valuable tool in the clinical management of gliomas.
Collapse
Affiliation(s)
- Hui Ma
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Nuclear Medicine, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing Zhao
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shaoyu Liu
- Department of Nuclear Medicine, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Nuclear Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dingxiang Xie
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhanwen Zhang
- Department of Nuclear Medicine, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Nuclear Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dahong Nie
- Department of Nuclear Medicine, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fuhua Wen
- Department of Nuclear Medicine, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhiyun Yang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ganghua Tang
- Department of Nuclear Medicine, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Nanfang PET Center, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
28
|
Tripathi SK, Kean R, Bongiorno E, Hooper DC, Jin YY, Wickstrom E, McCue PA, Thakur ML. Targeting VPAC1 Receptors for Imaging Glioblastoma. Mol Imaging Biol 2021; 22:293-302. [PMID: 31292914 DOI: 10.1007/s11307-019-01388-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE Scintigraphic imaging of malignant glioblastoma (MG) continues to be challenging. We hypothesized that VPAC1 cell surface receptors can be targeted for positron emission tomography (PET) imaging of orthotopically implanted MG in a mouse model, using a VPAC1-specific peptide [64Cu]TP3805. PROCEDURES The expression of VPAC1 in mouse GL261 and human U87 glioma cell lines was determined by western blot. The ability of [64Cu]TP3805 to bind to GL261 and U87 cells was studied by cell-binding. Receptor-blocking studies were performed to validate receptor specificity. GL261 tumors were implanted orthotopically in syngeneic T-bet knockout C57BL/6 mouse brain (N = 15) and allowed to grow for 2-3 weeks. Mice were injected i.v., first with ~ 150 μCi of 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) then 24 h later with ~ 200 μCi of [64Cu]TP3805. In another set of tumor-bearing mice, (N = 5), ionic [64Cu]Cl2 was injected as a control. Mice were imaged at a 2-h post-injection using an Inveon micro-PET/CT, sacrificed and % ID/g of [64Cu]TP3805 and [64Cu]Cl2 were calculated in a tumor, normal brain, and other tissues. For histologic tissue examination, 3-μm thick sections of the tumors and normal brain were prepared, digital autoradiography (DAR) was performed, and then the sections were H&E stained for histologic examination. RESULTS Western blots showed a strong signal for VPAC1 on both cell lines. [64Cu]TP3805 cell-binding was 87 ± 1.5 %. Receptor-blocking reduced cell-binding to 24.3 ± 1.5 % (P < 0.01). PET imaging revealed remarkable accumulation of [64Cu]TP3805 in GL261 MG with a negligible background in the normal brain, as compared to [18F]FDG. Micro-PET/CT image analyses and tissue distribution showed that the brain tumor uptake for [64Cu]TP3805 was 8.2 ± 1.7 % ID/g and for [64Cu]Cl2 2.1 ± 0.5 % ID/g as compared to 1.0 ± 0.3 % ID/g and 1.4 ± 0.3 % ID/g for normal mouse brains, respectively. The high tumor/normal brain ratio for [64Cu]TP3805 (8.1 ± 1.1) allowed tumors to be visualized unequivocally. Histology and [64Cu]TP3805 DAR differentiated malignant tumors from healthy brain and confirmed PET findings. CONCLUSION Targeting VPAC1 receptors using [64Cu]TP3805 for PET imaging of MG is a promising novel approach and calls for further investigation.
Collapse
Affiliation(s)
- Sushil K Tripathi
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rhonda Kean
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Emily Bongiorno
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Douglas C Hooper
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Eric Wickstrom
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Peter A McCue
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mathew L Thakur
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Urology, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Radiology, Laboratories of Radiopharmaceutical Research and Molecular Imaging, 1020 Locust Street, 359-JAH, Philadelphia, PA, 19107, USA.
| |
Collapse
|
29
|
Goud NS, Bhattacharya A, Joshi RK, Nagaraj C, Bharath RD, Kumar P. Carbon-11: Radiochemistry and Target-Based PET Molecular Imaging Applications in Oncology, Cardiology, and Neurology. J Med Chem 2021; 64:1223-1259. [PMID: 33499603 DOI: 10.1021/acs.jmedchem.0c01053] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The positron emission tomography (PET) molecular imaging technique has gained its universal value as a remarkable tool for medical diagnosis and biomedical research. Carbon-11 is one of the promising radiotracers that can report target-specific information related to its pharmacology and physiology to understand the disease status. Currently, many of the available carbon-11 (t1/2 = 20.4 min) PET radiotracers are heterocyclic derivatives that have been synthesized using carbon-11 inserted different functional groups obtained from primary and secondary carbon-11 precursors. A spectrum of carbon-11 PET radiotracers has been developed against many of the upregulated and emerging targets for the diagnosis, prognosis, prediction, and therapy in the fields of oncology, cardiology, and neurology. This review focuses on the carbon-11 radiochemistry and various target-specific PET molecular imaging agents used in tumor, heart, brain, and neuroinflammatory disease imaging along with its associated pathology.
Collapse
Affiliation(s)
- Nerella Sridhar Goud
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Ahana Bhattacharya
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Raman Kumar Joshi
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Chandana Nagaraj
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Rose Dawn Bharath
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Pardeep Kumar
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| |
Collapse
|
30
|
Evaluation of Glycolytic Response to Multiple Classes of Anti-glioblastoma Drugs by Noninvasive Measurement of Pyruvate Kinase M2 Using [ 18F]DASA-23. Mol Imaging Biol 2021; 22:124-133. [PMID: 30989436 DOI: 10.1007/s11307-019-01353-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Pyruvate kinase M2 (PKM2) catalyzes the final step in glycolysis, the key process of tumor metabolism. PKM2 is found in high levels in glioblastoma (GBM) cells with marginal expression within healthy brain tissue, rendering it a key biomarker of GBM metabolic re-programming. Our group has reported the development of a novel radiotracer, 1-((2-fluoro- 6-[18F]fluorophenyl)sulfonyl)-4-((4-methoxyphenyl)sulfonyl)piperazine ([18F]DASA- 23), to non-invasively detect PKM2 levels with positron emission tomography (PET). PROCEDURE U87 human GBM cells were treated with the IC50 concentration of various agents used in the treatment of GBM, including alkylating agents (temozolomide, carmustine, lomustine, procarbazine), inhibitor of topoisomerase I (irinotecan), vascular endothelial and epidermal growth factor receptor inhibitors (cediranib and erlotinib, respectively) anti-metabolite (5-fluorouracil), microtubule inhibitor (vincristine), and metabolic agents (dichloroacetate and IDH1 inhibitor ivosidenib). Following drug exposure for three or 6 days (n = 6 replicates per condition), the radiotracer uptake of [18F]DASA-23 and 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) was assessed. Changes in PKM2 protein levels were determined via Western blot and correlated to radiotracer uptake. RESULTS Significant interactions were found between the treatment agent (n = 12 conditions total comprised 11 drugs and vehicle) and the duration of treatment (3- or 6-day exposure to each drug) on the cellular uptake of [18F]DASA-23 (p = 0.0001). The greatest change in the cellular uptake of [18F]DASA-23 was found after exposure to alkylating agents (p < 0. 0001) followed by irinotecan (p = 0. 0012), erlotinib (p = 0. 02), and 5-fluorouracil (p = 0. 005). Correlation of PKM2 protein levels and [18F]DASA-23 cellular uptake revealed a moderate correlation (r = 0.44, p = 0.15). CONCLUSIONS These proof of principle studies emphasize the superiority of [18F]DASA-23 to [18F]FDG in detecting the glycolytic response of GBM to multiple classes of anti-neoplastic drugs in cell culture. A clinical trial evaluating the diagnostic utility of [18F]DASA-23 PET in GBM patients (NCT03539731) is ongoing.
Collapse
|
31
|
Comparison of Amino Acid PET to Advanced and Emerging MRI Techniques for Neurooncology Imaging: A Systematic Review of the Recent Studies. Mol Imaging 2021; 2021:8874078. [PMID: 34194287 PMCID: PMC8205602 DOI: 10.1155/2021/8874078] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/23/2020] [Accepted: 11/17/2020] [Indexed: 12/16/2022] Open
Abstract
Introduction Standard neuroimaging protocols for brain tumors have well-known limitations. The clinical use of additional modalities including amino acid PET (aaPET) and advanced MRI (aMRI) techniques (including DWI, PWI, and MRS) is emerging in response to the need for more accurate detection of brain tumors. In this systematic review of the past 2 years of the literature, we discuss the most recent studies that directly compare or combine aaPET and aMRI for brain tumor imaging. Methods A PubMed search was conducted for human studies incorporating both aaPET and aMRI and published between July 2018 and August 2020. Results A total of 22 studies were found in the study period. Recent studies of aaPET with DWI showed a superiority of MET, FET, FDOPA, and AMT PET for detecting tumor, predicting recurrence, diagnosing progression, and predicting survival. Combining modalities further improved performance. Comparisons of aaPET with PWI showed mixed results about spatial correlation. However, both modalities were able to detect high-grade tumors, identify tumor recurrence, differentiate recurrence from treatment effects, and predict survival. aaPET performed better on these measures than PWI, but when combined, they had the strongest results. Studies of aaPET with MRS demonstrated that both modalities have diagnostic potential but MET PET and FDOPA PET performed better than MRS. MRS suffered from some data quality issues that limited analysis in two studies, and, in one study that combined modalities, overall performance actually decreased. Four recent studies compared aaPET with emerging MRI approaches (such as CEST imaging, MR fingerprinting, and SISTINA), but the initial results remain inconclusive. Conclusions aaPET outperformed the aMRI imaging techniques in most recent studies. DWI and PWI added meaningful complementary data, and the combination of aaPET with aMRI yielded the best results in most studies.
Collapse
|
32
|
Kamson D, Tsien C. Novel Magnetic Resonance Imaging and Positron Emission Tomography in the RT Planning and Assessment of Response of Malignant Gliomas. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00078-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
33
|
Aasen SN, Espedal H, Keunen O, Adamsen TCH, Bjerkvig R, Thorsen F. Current landscape and future perspectives in preclinical MR and PET imaging of brain metastasis. Neurooncol Adv 2021; 3:vdab151. [PMID: 34988446 PMCID: PMC8704384 DOI: 10.1093/noajnl/vdab151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Brain metastasis (BM) is a major cause of cancer patient morbidity. Clinical magnetic resonance imaging (MRI) and positron emission tomography (PET) represent important resources to assess tumor progression and treatment responses. In preclinical research, anatomical MRI and to some extent functional MRI have frequently been used to assess tumor progression. In contrast, PET has only to a limited extent been used in animal BM research. A considerable culprit is that results from most preclinical studies have shown little impact on the implementation of new treatment strategies in the clinic. This emphasizes the need for the development of robust, high-quality preclinical imaging strategies with potential for clinical translation. This review focuses on advanced preclinical MRI and PET imaging methods for BM, describing their applications in the context of what has been done in the clinic. The strengths and shortcomings of each technology are presented, and recommendations for future directions in the development of the individual imaging modalities are suggested. Finally, we highlight recent developments in quantitative MRI and PET, the use of radiomics and multimodal imaging, and the need for a standardization of imaging technologies and protocols between preclinical centers.
Collapse
Affiliation(s)
- Synnøve Nymark Aasen
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Department of Health and Functioning, Western Norway University of Applied Sciences, Bergen, Norway
| | - Heidi Espedal
- The Molecular Imaging Center, Department of Biomedicine, University of Bergen, Bergen, Norway
- Mohn Medical Imaging and Visualization Centre, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Olivier Keunen
- Translational Radiomics, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Tom Christian Holm Adamsen
- Centre for Nuclear Medicine, Department of Radiology, Haukeland University Hospital, Bergen, Norway
- 180 °N – Bergen Tracer Development Centre, Department of Radiology, Haukeland University Hospital, Bergen, Norway
- Department of Chemistry, University of Bergen, Bergen, Norway
| | - Rolf Bjerkvig
- Department of Biomedicine, University of Bergen, Bergen, Norway
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Frits Thorsen
- Department of Biomedicine, University of Bergen, Bergen, Norway
- The Molecular Imaging Center, Department of Biomedicine, University of Bergen, Bergen, Norway
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, Shandong, Jinan, P.R. China
| |
Collapse
|
34
|
Snyder SE, Butch ER, Shulkin BL. Radiopharmaceuticals in Pediatric Nuclear Medicine. HANDBOOK OF RADIOPHARMACEUTICALS 2020:653-701. [DOI: 10.1002/9781119500575.ch21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
35
|
Juhász C, Mittal S. Molecular Imaging of Brain Tumor-Associated Epilepsy. Diagnostics (Basel) 2020; 10:diagnostics10121049. [PMID: 33291423 PMCID: PMC7762008 DOI: 10.3390/diagnostics10121049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 11/16/2022] Open
Abstract
Epilepsy is a common clinical manifestation and a source of significant morbidity in patients with brain tumors. Neuroimaging has a pivotal role in neuro-oncology practice, including tumor detection, differentiation, grading, treatment guidance, and posttreatment monitoring. In this review, we highlight studies demonstrating that imaging can also provide information about brain tumor-associated epileptogenicity and assist delineation of the peritumoral epileptic cortex to optimize postsurgical seizure outcome. Most studies focused on gliomas and glioneuronal tumors where positron emission tomography (PET) and advanced magnetic resonance imaging (MRI) techniques can detect metabolic and biochemical changes associated with altered amino acid transport and metabolism, neuroinflammation, and neurotransmitter abnormalities in and around epileptogenic tumors. PET imaging of amino acid uptake and metabolism as well as activated microglia can detect interictal or peri-ictal cortical increased uptake (as compared to non-epileptic cortex) associated with tumor-associated epilepsy. Metabolic tumor volumes may predict seizure outcome based on objective treatment response during glioma chemotherapy. Advanced MRI, especially glutamate imaging, can detect neurotransmitter changes around epileptogenic brain tumors. Recently, developed PET radiotracers targeting specific glutamate receptor types may also identify therapeutic targets for pharmacologic seizure control. Further studies with advanced multimodal imaging approaches may facilitate development of precision treatment strategies to control brain tumor-associated epilepsy.
Collapse
Affiliation(s)
- Csaba Juhász
- Departments of Pediatrics, Neurology, Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
- PET Center and Translational Imaging Laboratory, Barbara Ann Karmanos Cancer Institute, Detroit, MI 48201, USA
- Correspondence:
| | - Sandeep Mittal
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA;
- Carilion Clinic Neurosurgery, Roanoke, VA 24014, USA
- Fralin Biomedical Research Institute, Roanoke, VA 24016, USA
| |
Collapse
|
36
|
Huntoon K, Toland AMS, Dahiya S. Meningioma: A Review of Clinicopathological and Molecular Aspects. Front Oncol 2020; 10:579599. [PMID: 33194703 PMCID: PMC7645220 DOI: 10.3389/fonc.2020.579599] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/22/2020] [Indexed: 12/19/2022] Open
Abstract
Meningiomas are the most the common primary brain tumors in adults, representing approximately a third of all intracranial neoplasms. They classically are found to be more common in females, with the exception of higher grades that have a predilection for males, and patients of older age. Meningiomas can also be seen as a spectrum of inherited syndromes such as neurofibromatosis 2 as well as ionizing radiation. In general, the 5-year survival for a WHO grade I meningioma exceeds 80%; however, survival is greatly reduced in anaplastic meningiomas. The standard of care for meningiomas in a surgically-accessible location is gross total resection. Radiation therapy is generally saved for atypical, anaplastic, recurrent, and surgically inaccessible benign meningiomas with a total dose of ~60 Gy. However, the method of radiation, regimen and timing is still evolving and is an area of active research with ongoing clinical trials. While there are currently no good adjuvant chemotherapeutic agents available, recent advances in the genomic and epigenomic landscape of meningiomas are being explored for potential targeted therapy.
Collapse
Affiliation(s)
- Kristin Huntoon
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | | | - Sonika Dahiya
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
37
|
Bonomi R, John F, Patel S, Barger G, Robinette N, Amit-Yousif AJ, Dominello M, Juhasz C. Multimodal neuroimaging of gliomatosis cerebri: a case series of four patients. Acta Radiol Open 2020; 9:2058460120942789. [PMID: 32913666 PMCID: PMC7444143 DOI: 10.1177/2058460120942789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 06/23/2020] [Indexed: 01/18/2023] Open
Abstract
In the latest World Health Organization classification of brain tumors, gliomatosis cerebri has been redefined to varying subsets of diffuse gliomas; however, the term is still used to describe gliomas with infiltrative growth into three or more cerebral lobes. These tumors are frequently misdiagnosed and difficult to treat due to their atypical presentation using structural imaging modalities including computed tomography and T1/T2-weighted magnetic resonance imaging (MRI). In this retrospective case series, we compared clinical MRI to amino acid positron emission tomography (PET) to assess the potential value of PET in the assessment of the extent of tumor involvement and in monitoring disease progression. We report the clinical course and serial multimodal imaging findings of four patients. Each patient presented at varying points in disease progression with widespread glioma brain involvement and was evaluated at least once by amino acid PET using alpha-[11C]methyl-L-tryptophan ([11C]-AMT). Increased uptake of [11C]-AMT was detected in a subset of non-enhancing brain lesions and detected tumor invasion before MRI signs of tumor in some regions. Increased uptake of [11C]-AMT was also detected in tumorous regions not detected by perfusion MRI or MR spectroscopy. Metabolic response to treatment was also observed in two patients. Overall, these data are consistent with and expand upon previous reports using other amino acid PET tracers in gliomatosis and show the potential added value of this imaging modality to clinical MRI in the detection and monitoring of these diffusely infiltrative tumors.
Collapse
Affiliation(s)
- Robin Bonomi
- Departments of Pediatrics, Neurology, Radiology, Oncology, Wayne State University, Detroit, MI, USA
| | - Flora John
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, USA
| | | | | | | | | | | | - Csaba Juhasz
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, USA
| |
Collapse
|
38
|
Zlatopolskiy BD, Endepols H, Krasikova RN, Fedorova OS, Ermert J, Neumaier B. 11C- and 18F-labelled tryptophans as PET-tracers for imaging of altered tryptophan metabolism in age-associated disorders. RUSSIAN CHEMICAL REVIEWS 2020. [DOI: 10.1070/rcr4954] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The ageing of the world’s population is the result of increased life expectancy observed in almost all countries throughout the world. Consequently, a rising tide of ageing-associated disorders, like cancer and neurodegenerative diseases, represents one of the main global challenges of the 21st century. The ability of mankind to overcome these challenges is directly dependent on the capability to develop novel methods for therapy and diagnosis of age-associated diseases. One hallmark of age-related pathologies is an altered tryptophan metabolism. Numerous pathological processes including neurodegenerative and neurological diseases like epilepsy, Parkinson’s and Alzheimer’s diseases, cancer and diabetes exhibit marked changes in tryptophan metabolism. Visualization of key processes of tryptophan metabolic pathways, especially using positron emission tomography (PET) and related hybrid methods like PET/CT and PET/MRI, can be exploited to early detect the aforementioned disorders with considerable accuracy, allowing appropriate and timely treatment of patients. Here we review the published 11C- and 18F-labelled tryptophans with respect to the production and also preclinical and clinical evaluation as PET-tracers for visualization of different branches of tryptophan metabolism.
The bibliography includes 159 references.
Collapse
|
39
|
Galldiks N, Langen KJ, Albert NL, Chamberlain M, Soffietti R, Kim MM, Law I, Le Rhun E, Chang S, Schwarting J, Combs SE, Preusser M, Forsyth P, Pope W, Weller M, Tonn JC. PET imaging in patients with brain metastasis-report of the RANO/PET group. Neuro Oncol 2020; 21:585-595. [PMID: 30615138 DOI: 10.1093/neuonc/noz003] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 10/11/2018] [Accepted: 01/03/2019] [Indexed: 12/23/2022] Open
Abstract
Brain metastases (BM) from extracranial cancer are associated with significant morbidity and mortality. Effective local treatment options are stereotactic radiotherapy, including radiosurgery or fractionated external beam radiotherapy, and surgical resection. The use of systemic treatment for intracranial disease control also is improving. BM diagnosis, treatment planning, and follow-up is most often based on contrast-enhanced magnetic resonance imaging (MRI). However, anatomic imaging modalities including standard MRI have limitations in accurately characterizing posttherapeutic reactive changes and treatment response. Molecular imaging techniques such as positron emission tomography (PET) characterize specific metabolic and cellular features of metastases, potentially providing clinically relevant information supplementing anatomic MRI. Here, the Response Assessment in Neuro-Oncology working group provides recommendations for the use of PET imaging in the clinical management of patients with BM based on evidence from studies validated by histology and/or clinical outcome.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, University Hospital Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine 3, 4, Research Center Juelich, Juelich, Germany.,Center of Integrated Oncology, Universities of Cologne and Bonn, Cologne, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine 3, 4, Research Center Juelich, Juelich, Germany.,Department of Nuclear Medicine, University Hospital Aachen, Aachen, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig Maximilians-University of Munich, Munich, Germany
| | - Marc Chamberlain
- Departments of Neurology and Neurological Surgery, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington, USA
| | - Riccardo Soffietti
- Department of Neuro-Oncology, University and City of Health and Science Hospital, Turin, Italy
| | - Michelle M Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Denmark
| | - Emilie Le Rhun
- Department of Neurosurgery, University Hospital Lille, Lille, France
| | - Susan Chang
- Department of Neurosurgery, University of California, San Francisco, California, USA
| | - Julian Schwarting
- Department of Neurosurgery, Ludwig Maximilians-University of Munich, Munich, Germany.,German Cancer Consortium, Partner Site Munich, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Technical University Munich, Munich, Germany
| | - Matthias Preusser
- Department of Medicine I and Comprehensive Cancer Centre CNS Tumours Unit, Medical University of Vienna, Vienna, Austria
| | - Peter Forsyth
- Moffitt Cancer Center, University of South Florida, Tampa, Florida, USA
| | - Whitney Pope
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California , USA
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Jörg C Tonn
- Department of Neurosurgery, Ludwig Maximilians-University of Munich, Munich, Germany.,German Cancer Consortium, Partner Site Munich, Germany
| |
Collapse
|
40
|
John F, Robinette NL, Amit-Yousif AJ, Bosnyák E, Barger GR, Shah KD, Mittal S, Juhász C. Multimodal Imaging of Nonenhancing Glioblastoma Regions. Mol Imaging 2020; 18:1536012119885222. [PMID: 31736437 PMCID: PMC6862774 DOI: 10.1177/1536012119885222] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Clinical glioblastoma treatment mostly focuses on the contrast-enhancing tumor mass. Amino acid positron emission tomography (PET) can detect additional, nonenhancing glioblastoma-infiltrated brain regions that are difficult to distinguish on conventional magnetic resonance imaging (MRI). We combined MRI with perfusion imaging and amino acid PET to evaluate such nonenhancing glioblastoma regions. METHODS Structural MRI, relative cerebral blood volume (rCBV) maps from perfusion MRI, and α-[11C]-methyl-l-tryptophan (AMT)-PET images were analyzed in 20 patients with glioblastoma. The AMT uptake and rCBV (expressed as tumor to normal [T/N] ratios) were compared in nonenhancing tumor portions showing increased signal on T2/fluid-attenuated inversion recovery (T2/FLAIR) images. RESULTS Thirteen (65%) tumors showed robust heterogeneity in nonenhancing T2/FLAIR hyperintense areas on AMT-PET, whereas the nonenhancing regions in the remaining 7 cases had homogeneous AMT uptake (low in 6, high in 1). AMT and rCBV T/N ratios showed only a moderate correlation in the nonenhancing regions (r = 0.41, P = .017), but regions with very low rCBV (<0.79 T/N ratio) had invariably low AMT uptake. CONCLUSIONS The findings demonstrate the metabolic and perfusion heterogeneity of nonenhancing T2/FLAIR hyperintense glioblastoma regions. Amino acid PET imaging of such regions can detect glioma-infiltrated brain for treatment targeting; however, very low rCBV values outside the contrast-enhancing tumor mass make increased AMT uptake in nonenhancing glioblastoma regions unlikely.
Collapse
Affiliation(s)
- Flóra John
- Department of Pediatrics, Wayne State University and PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, USA
| | - Natasha L Robinette
- Department of Radiology, Wayne State University, Detroit, MI, USA.,Karmanos Cancer Institute, Detroit, MI, USA
| | - Alit J Amit-Yousif
- Department of Radiology, Wayne State University, Detroit, MI, USA.,Karmanos Cancer Institute, Detroit, MI, USA
| | - Edit Bosnyák
- Department of Pediatrics, Wayne State University and PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, USA
| | - Geoffrey R Barger
- Department of Neurology, Wayne State University, Detroit, MI, USA.,Karmanos Cancer Institute, Detroit, MI, USA
| | - Keval D Shah
- Department of Neurology, Wayne State University, Detroit, MI, USA
| | - Sandeep Mittal
- Department of Neurosurgery, Wayne State University, Detroit, MI, USA.,Karmanos Cancer Institute, Detroit, MI, USA.,Virginia Tech Carilion School of Medicine, Roanoke, VA, USA.,Virginia Tech School of Neuroscience, Blacksburg, VA, USA
| | - Csaba Juhász
- Department of Pediatrics, Wayne State University and PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, USA.,Department of Neurology, Wayne State University, Detroit, MI, USA.,Department of Neurosurgery, Wayne State University, Detroit, MI, USA.,Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
41
|
John F, Bosnyák E, Robinette NL, Amit-Yousif AJ, Barger GR, Shah KD, Michelhaugh SK, Klinger NV, Mittal S, Juhász C. Multimodal imaging-defined subregions in newly diagnosed glioblastoma: impact on overall survival. Neuro Oncol 2020; 21:264-273. [PMID: 30346623 DOI: 10.1093/neuonc/noy169] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Although glioblastomas are heterogeneous brain-infiltrating tumors, their treatment is mostly focused on the contrast-enhancing tumor mass. In this study, we combined conventional MRI, diffusion-weighted imaging (DWI), and amino acid PET to explore imaging-defined glioblastoma subregions and evaluate their potential prognostic value. METHODS Contrast-enhanced T1, T2/fluid attenuated inversion recovery (FLAIR) MR images, apparent diffusion coefficient (ADC) maps from DWI, and alpha-[11C]-methyl-L-tryptophan (AMT)-PET images were analyzed in 30 patients with newly diagnosed glioblastoma. Five tumor subregions were identified based on a combination of MRI contrast enhancement, T2/FLAIR signal abnormalities, and AMT uptake on PET. ADC and AMT uptake tumor/contralateral normal cortex (T/N) ratios in these tumor subregions were correlated, and their prognostic value was determined. RESULTS A total of 115 MRI/PET-defined subregions were analyzed. Most tumors showed not only a high-AMT uptake (T/N ratio > 1.65, N = 27) but also a low-uptake subregion (N = 21) within the contrast-enhancing tumor mass. High AMT uptake extending beyond contrast enhancement was also common (N = 25) and was associated with low ADC (r = -0.40, P = 0.05). Higher AMT uptake in the contrast-enhancing tumor subregions was strongly prognostic for overall survival (hazard ratio: 7.83; 95% CI: 1.98-31.02, P = 0.003), independent of clinical and molecular genetic prognostic variables. Nonresected high-AMT uptake subregions predicted the sites of tumor progression on posttreatment PET performed in 10 patients. CONCLUSIONS Glioblastomas show heterogeneous amino acid uptake with high-uptake regions often extending into non-enhancing brain with high cellularity; nonresection of these predict the site of posttreatment progression. High tryptophan uptake values in MRI contrast-enhancing tumor subregions are a strong, independent imaging marker for longer overall survival.
Collapse
Affiliation(s)
- Flóra John
- Department of Pediatrics, Wayne State University, Detroit, Michigan.,PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit Medical Center, Detroit, Michigan
| | - Edit Bosnyák
- Department of Pediatrics, Wayne State University, Detroit, Michigan.,PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit Medical Center, Detroit, Michigan
| | - Natasha L Robinette
- Department of Oncology, Wayne State University, Detroit, Michigan.,Department of Radiology, Wayne State University, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan
| | - Alit J Amit-Yousif
- Department of Oncology, Wayne State University, Detroit, Michigan.,Department of Radiology, Wayne State University, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan
| | - Geoffrey R Barger
- Department of Neurology, Wayne State University, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan
| | - Keval D Shah
- Department of Neurology, Wayne State University, Detroit, Michigan
| | | | | | - Sandeep Mittal
- Department of Neurosurgery, Wayne State University, Detroit, Michigan.,Department of Oncology, Wayne State University, Detroit, Michigan.,Department of Biomedical Engineering, Wayne State University, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan
| | - Csaba Juhász
- Department of Pediatrics, Wayne State University, Detroit, Michigan.,Department of Neurology, Wayne State University, Detroit, Michigan.,Department of Neurosurgery, Wayne State University, Detroit, Michigan.,PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit Medical Center, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan
| |
Collapse
|
42
|
Werner JM, Lohmann P, Fink GR, Langen KJ, Galldiks N. Current Landscape and Emerging Fields of PET Imaging in Patients with Brain Tumors. Molecules 2020; 25:E1471. [PMID: 32213992 PMCID: PMC7146177 DOI: 10.3390/molecules25061471] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/17/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Abstract
The number of positron-emission tomography (PET) tracers used to evaluate patients with brain tumors has increased substantially over the last years. For the management of patients with brain tumors, the most important indications are the delineation of tumor extent (e.g., for planning of resection or radiotherapy), the assessment of treatment response to systemic treatment options such as alkylating chemotherapy, and the differentiation of treatment-related changes (e.g., pseudoprogression or radiation necrosis) from tumor progression. Furthermore, newer PET imaging approaches aim to address the need for noninvasive assessment of tumoral immune cell infiltration and response to immunotherapies (e.g., T-cell imaging). This review summarizes the clinical value of the landscape of tracers that have been used in recent years for the above-mentioned indications and also provides an overview of promising newer tracers for this group of patients.
Collapse
Affiliation(s)
- Jan-Michael Werner
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937 Cologne, Germany; (J.-M.W.); (G.R.F.)
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Leo-Brandt-St., 52425 Juelich, Germany; (P.L.); (K.-J.L.)
| | - Gereon R. Fink
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937 Cologne, Germany; (J.-M.W.); (G.R.F.)
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Leo-Brandt-St., 52425 Juelich, Germany; (P.L.); (K.-J.L.)
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Leo-Brandt-St., 52425 Juelich, Germany; (P.L.); (K.-J.L.)
- Department of Nuclear Medicine, University Hospital Aachen, 52074 Aachen, Germany
| | - Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937 Cologne, Germany; (J.-M.W.); (G.R.F.)
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Leo-Brandt-St., 52425 Juelich, Germany; (P.L.); (K.-J.L.)
| |
Collapse
|
43
|
Automated production of 1-(2-[ 18F]fluoroethyl)-l-tryptophan for imaging of tryptophan metabolism. Appl Radiat Isot 2020; 156:109022. [PMID: 32056678 DOI: 10.1016/j.apradiso.2019.109022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/18/2019] [Accepted: 12/05/2019] [Indexed: 11/23/2022]
Abstract
Automated production of an fluorine-18 labeled tryptophan analogue, 1-(2-[18F]fluoroethyl)-l-tryptophan (1-L-[18F]FETrp) in a current Good Manufacturing Practice facility was achieved. 1-L-[18F]FETrp was produced by a one-pot, two-step strategy with an overall synthesis time of approximately 100 min, a radiochemical yield of 20 ± 5% (decay corrected), radiochemical purity and enantiomeric excess over 90%, and a molar activity of 103 ± 15 GBq/μmol at the end of synthesis (EOS). The dose mass of 1-L-FETrp in four consecutive batches was less than 5 μg. The radiopharmaceutical product met all quality control criteria for clinical use.
Collapse
|
44
|
Kamson DO. Hypometabolic gliomas on FET-PET-is there an inverted U-curve for survival? Neuro Oncol 2020; 21:1221-1222. [PMID: 31278863 DOI: 10.1093/neuonc/noz122] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- David Olayinka Kamson
- Department of Neurology and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
45
|
Human biodistribution and radiation dosimetry of [ 18F]DASA-23, a PET probe targeting pyruvate kinase M2. Eur J Nucl Med Mol Imaging 2020; 47:2123-2130. [PMID: 31938892 DOI: 10.1007/s00259-020-04687-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/05/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE To assess the safety, biodistribution, and radiation dosimetry of the novel positron emission tomography (PET) radiopharmaceutical 1-((2-fluoro-6-[[18F]]fluorophenyl)sulfonyl)-4-((4-methoxyphenyl)sulfonyl)piperazine ([18F]DASA-23) in healthy volunteers. METHODS We recruited 5 healthy volunteers who provided a written informed consent. Volunteers were injected with 295.0 ± 8.2 MBq of [18F]DASA-23 intravenously. Immediately following injection, a dynamic scan of the brain was acquired for 15 min. This was followed by serial whole-body PET/MRI scans acquired up to 3 h post-injection. Blood samples were collected at regular intervals, and vital signs monitored pre- and post-radiotracer administration. Regions of interest were drawn around multiple organs, time-activity curves were calculated, and organ uptake and dosimetry were estimated with OLINDA/EXM (version 1.1) software. RESULTS All subjects tolerated the PET/MRI examination, without adverse reactions to [18F]DASA-23. [18F]DASA-23 passively crossed the blood-brain barrier, followed by rapid clearance from the brain. High accumulation of [18F]DASA-23 was noted in organs such as the gallbladder, liver, small intestine, and urinary bladder, suggesting hepatobiliary and urinary clearance. The effective dose of [18F]DASA-23 was 23.5 ± 5.8 μSv/MBq. CONCLUSION We successfully completed a pilot first-in-human study of [18F]DASA-23. Our results indicate that [18F]DASA-23 can be used safely in humans to evaluate pyruvate kinase M2 levels. Ongoing studies are evaluating the ability of [18F]DASA-23 to visualize intracranial malignancies, NCT03539731. TRIAL REGISTRATION ClinicalTrials.gov , NCT03539731 (registered 28 May 2018).
Collapse
|
46
|
Jeong JW, Lee MH, John F, Robinette NL, Amit-Yousif AJ, Barger GR, Mittal S, Juhász C. Feasibility of Multimodal MRI-Based Deep Learning Prediction of High Amino Acid Uptake Regions and Survival in Patients With Glioblastoma. Front Neurol 2020; 10:1305. [PMID: 31920928 PMCID: PMC6928045 DOI: 10.3389/fneur.2019.01305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022] Open
Abstract
Purpose: Amino acid PET has shown high accuracy for the diagnosis and prognostication of malignant gliomas, however, this imaging modality is not widely available in clinical practice. This study explores a novel end-to-end deep learning framework ("U-Net") for its feasibility to detect high amino acid uptake glioblastoma regions (i.e., metabolic tumor volume) using clinical multimodal MRI sequences. Methods: T2, fluid-attenuated inversion recovery (FLAIR), apparent diffusion coefficient map, contrast-enhanced T1, and alpha-[11C]-methyl-L-tryptophan (AMT)-PET images were analyzed in 21 patients with newly-diagnosed glioblastoma. U-Net system with data augmentation was implemented to deeply learn non-linear voxel-wise relationships between intensities of multimodal MRI as the input and metabolic tumor volume from AMT-PET as the output. The accuracy of the MRI- and PET-based volume measures to predict progression-free survival was tested. Results: In the augmented dataset using all four MRI modalities to investigate the upper limit of U-Net accuracy in the full study cohort, U-Net achieved high accuracy (sensitivity/specificity/positive predictive value [PPV]/negative predictive value [NPV]: 0.85/1.00/0.81/1.00, respectively) to predict PET-defined tumor volumes. Exclusion of FLAIR from the MRI input set had a strong negative effect on sensitivity (0.60). In repeated hold out validation in randomly selected subjects, specificity and NPV remained high (1.00), but mean sensitivity (0.62), and PPV (0.68) were moderate. AMT-PET-learned MRI tumor volume from this U-net model within the contrast-enhancing volume predicted 6-month progression-free survival with 0.86/0.63 sensitivity/specificity. Conclusions: These data indicate the feasibility of PET-based deep learning for enhanced pretreatment glioblastoma delineation and prognostication by clinical multimodal MRI.
Collapse
Affiliation(s)
- Jeong-Won Jeong
- Department of Pediatrics, Wayne State University School of Medicine and PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, United States.,Department of Neurology, Wayne State University, Detroit, MI, United States.,Translational Neuroscience Program, Wayne State University, Detroit, MI, United States
| | - Min-Hee Lee
- Department of Pediatrics, Wayne State University School of Medicine and PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, United States
| | - Flóra John
- Department of Pediatrics, Wayne State University School of Medicine and PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, United States
| | - Natasha L Robinette
- Department of Oncology, Wayne State University, Detroit, MI, United States.,Karmanos Cancer Institute, Detroit, MI, United States
| | - Alit J Amit-Yousif
- Department of Oncology, Wayne State University, Detroit, MI, United States.,Karmanos Cancer Institute, Detroit, MI, United States
| | - Geoffrey R Barger
- Department of Neurology, Wayne State University, Detroit, MI, United States.,Karmanos Cancer Institute, Detroit, MI, United States
| | - Sandeep Mittal
- Department of Oncology, Wayne State University, Detroit, MI, United States.,Karmanos Cancer Institute, Detroit, MI, United States.,Department of Neurosurgery, Wayne State University, Detroit, MI, United States.,Virginia Tech Carilion School of Medicine and Carilion Clinic, Roanoke, VA, United States
| | - Csaba Juhász
- Department of Pediatrics, Wayne State University School of Medicine and PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, United States.,Department of Neurology, Wayne State University, Detroit, MI, United States.,Translational Neuroscience Program, Wayne State University, Detroit, MI, United States.,Karmanos Cancer Institute, Detroit, MI, United States.,Department of Neurosurgery, Wayne State University, Detroit, MI, United States
| |
Collapse
|
47
|
Seidensaal K, Harrabi SB, Debus J. Molecular Imaging for Particle Therapy: Current Approach and Future Directions. Recent Results Cancer Res 2020; 216:865-879. [PMID: 32594410 DOI: 10.1007/978-3-030-42618-7_28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
During the last decades, radiation oncology has been subject to a number of technological innovations. Particle therapy has evolved in parallel to the modern high-precision photon radiotherapy techniques and offers a superior dose distribution with decreased integral dose to healthy tissues. With advancing precision of treatment, the necessity for accurate and confident target volume delineation is rising. When morphological imaging reaches its limitations, molecular imaging can provide valuable information.
Collapse
Affiliation(s)
- Katharina Seidensaal
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Semi Ben Harrabi
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
48
|
Albano D, Tomasini D, Bonù M, Giubbini R, Bertagna F. 18F-Fluciclovine ( 18F-FACBC) PET/CT or PET/MRI in gliomas/glioblastomas. Ann Nucl Med 2019; 34:81-86. [PMID: 31773466 DOI: 10.1007/s12149-019-01426-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/05/2019] [Indexed: 02/06/2023]
Abstract
18F-fluciclovine (18F-FACBC) is a radiotracer already studied for prostate cancer, and its potential role in brain tumors (such as glioma) is not yet well investigated despite promising results. The aim of this review is to evaluate the possible diagnostic role of 18F-FACBC PET/CT or PET/MRI in patients with gliomas and glioblastomas. A comprehensive literature search of the PubMed/MEDLINE, Scopus, Embase, and Cochrane library databases was conducted to find the relevant published articles about the diagnostic performance of FACBC PET/CT or PET/MRI in patients affected by glioma and/or glioblastoma. Seven papers were included in the systematic review. From the analyses of the selected studies, the following main findings were obtained: glioma and glioblastoma are FACBC-avid tumors with a detection rate of about 100%; FACBC PET has high-diagnostic accuracy in defining tumor extent, volumes, and satellite lesions better than MR; compared to methionine, FACBC has similar accuracy but better tumor-to-background contrast; FACBC uptake may help to discriminate between low-grade and high-grade glioma. Radiolabelled fluciclovine (18F-FACBC) imaging seems to be useful in analyzing glioma/glioblastoma. Further studies enrolling a wider population are needed to clarify the real clinical and diagnostic role of 18F-FACBC in this setting and its possible position in the diagnostic flowchart.
Collapse
Affiliation(s)
- Domenico Albano
- Nuclear Medicine, University of Brescia and Spedali Civili Brescia, P.le Spedali Civili 1, 25123, Brescia, Italy.
| | - Davide Tomasini
- Department of Radiation Oncology, University of Brescia and ASST Spedali Civili Di Brescia, Brescia, Italy
| | - Marco Bonù
- Department of Radiation Oncology, University of Brescia and ASST Spedali Civili Di Brescia, Brescia, Italy
| | - Raffaele Giubbini
- Nuclear Medicine, University of Brescia and Spedali Civili Brescia, P.le Spedali Civili 1, 25123, Brescia, Italy
| | - Francesco Bertagna
- Nuclear Medicine, University of Brescia and Spedali Civili Brescia, P.le Spedali Civili 1, 25123, Brescia, Italy
| |
Collapse
|
49
|
Sogani SK, Jena A, Taneja S, Gambhir A, Mishra AK, D'Souza MM, Verma SM, Hazari PP, Negi P, Jadhav GKR. Potential for differentiation of glioma recurrence from radionecrosis using integrated 18F-fluoroethyl-L-tyrosine (FET) positron emission tomography/magnetic resonance imaging: A prospective evaluation. Neurol India 2019; 65:293-301. [PMID: 28290392 DOI: 10.4103/neuroindia.ni_101_16] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
PURPOSE To assess the utility of 18F-fluoroethyl-L-tyrosine (FET) positron emission tomography/magnetic resonance imaging (PET/MRI) in distinguishing recurrence from radionecrosis. MATERIALS AND METHODS Thirty-two patients (25 males, 7 females) of glioma who had already undergone surgery/chemoradiotherapy and had enhancing brain lesions suspicious of recurrence were evaluated using integrated 18F-FET PET/MRI, and followed up with histopathology or clinical follow-up and/or MRI/PET/MRI imaging. Manually drawn regions of interest over areas of maximal enhancement or FET uptake were used to calculate tumor to background ratios [TBRmax, TBRmean], choline: creatine ratio [Cho: Cr ratio], normalized relative cerebral blood volume [N rCBVmean] and apparent diffusion coefficient [ADCmean]. Correlations were evaluated using Pearson's coefficient. Accuracy of each parameter was calculated using independent t-test and receiver operator curve (ROC) analysis while utility of all four parameters together using multivariate analysis of variance (MANOVA) for differentiating recurrence vs. radionecrosis was evaluated. Positive histopathology and imaging/clinical follow up served as the gold standard. RESULTS Twenty-four of the 32 patients were diagnosed with recurrent disease and 8 with radiation necrosis. Significant correlations were observed between TBRmaxand N rCBVmean (ρ =0.503; P = 0.003), TBRmean, and N rCBVmean (ρ =0.414; P = 0.018), TBRmaxand ADCmean (ρ = -0.52; P = 0.002), and TBRmeanand ADCmean(ρ = -0.518; P = 0.002). TBRmax, TBRmean, ADCmean, Cho: Cr ratios, and N rCBVmeanwere significant in differentiating recurrence from radiation necrosis with an accuracy of 94.1%, 88.2%, 80.4%, 96.4%, and 89.9%, respectively. MANOVA indicated that combination of all parameters demonstrated better evaluation of recurrence vs. necrosis than any single parameter. The diagnostic accuracy, sensitivity, and specificity using all MRI parameters were 93.75%, 96%, and 85.7%, and using all FET PET/MRI parameters was 96.87%, 100%, and 85.7%, respectively. CONCLUSIONS Synergetic effect of multiple MR parameters evaluated together in addition to FET PET uptake highlights the fact that integrated 18F-FET PET/MRI might have the potential to impact management of patients with glioma by timely and conclusive recognition of true recurrence from radiation necrosis.
Collapse
Affiliation(s)
- Shanti K Sogani
- Department of Neurosurgery, Institute of Neuro Sciences, Indraprastha Apollo Hospitals, Sarita Vihar, New Delhi, India
| | - Amarnath Jena
- Department of Molecular Imaging and Nuclear Medicine, Indraprastha Apollo Hospitals, Sarita Vihar, New Delhi, India
| | - Sangeeta Taneja
- Department of Molecular Imaging and Nuclear Medicine, Indraprastha Apollo Hospitals, Sarita Vihar, New Delhi, India
| | - Aashish Gambhir
- Department of Molecular Imaging and Nuclear Medicine, Indraprastha Apollo Hospitals, Sarita Vihar, New Delhi, India
| | - Anil K Mishra
- Molecular Imaging and Research Centre, Institute of Nuclear Medicine and Allied Sciences, Brig. SK Mazumdar Road, Timarpur, Delhi, India
| | - Maria M D'Souza
- Molecular Imaging and Research Centre, Institute of Nuclear Medicine and Allied Sciences, Brig. SK Mazumdar Road, Timarpur, Delhi, India
| | - Sapna M Verma
- Institute of Radiation Oncology, Indraprastha Apollo Hospitals, Sarita Vihar, New Delhi, India
| | - Puja P Hazari
- Molecular Imaging and Research Centre, Institute of Nuclear Medicine and Allied Sciences, Brig. SK Mazumdar Road, Timarpur, Delhi, India
| | - Pradeep Negi
- Department of Molecular Imaging and Nuclear Medicine, Indraprastha Apollo Hospitals, Sarita Vihar, New Delhi, India
| | - Ganesh K R Jadhav
- Institute of Radiation Oncology, Indraprastha Apollo Hospitals, Sarita Vihar, New Delhi, India
| |
Collapse
|
50
|
Hanaoka H, Ohshima Y, Yamaguchi A, Suzuki H, Ishioka NS, Higuchi T, Arano Y, Tsushima Y. Novel 18F-Labeled α-Methyl-Phenylalanine Derivative with High Tumor Accumulation and Ideal Pharmacokinetics for Tumor-Specific Imaging. Mol Pharm 2019; 16:3609-3616. [DOI: 10.1021/acs.molpharmaceut.9b00446] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Hirofumi Hanaoka
- Department of Bioimaging Information Analysis, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Yasuhiro Ohshima
- Project “Medical Radioisotope Application”, Department of Radiation-Applied Biology Research, Takasaki Advanced Radiation Research Institute, Quantum Beam Advanced Research Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), Takasaki 370-1292, Japan
| | - Aiko Yamaguchi
- Department of Bioimaging Information Analysis, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Hiroyuki Suzuki
- Department of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Science, Chiba University, Chiba 260-8675, Japan
| | - Noriko S. Ishioka
- Project “Medical Radioisotope Application”, Department of Radiation-Applied Biology Research, Takasaki Advanced Radiation Research Institute, Quantum Beam Advanced Research Directorate, National Institutes for Quantum and Radiological Science and Technology (QST), Takasaki 370-1292, Japan
| | - Tetsuya Higuchi
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Yasushi Arano
- Department of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Science, Chiba University, Chiba 260-8675, Japan
| | - Yoshito Tsushima
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| |
Collapse
|