1
|
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
|
2
|
Xue C, Li G, Zheng Q, Gu X, Shi Q, Su Y, Chu Q, Yuan X, Bao Z, Lu J, Li L. Tryptophan metabolism in health and disease. Cell Metab 2023; 35:1304-1326. [PMID: 37352864 DOI: 10.1016/j.cmet.2023.06.004] [Citation(s) in RCA: 110] [Impact Index Per Article: 110.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/10/2023] [Accepted: 06/05/2023] [Indexed: 06/25/2023]
Abstract
Tryptophan (Trp) metabolism primarily involves the kynurenine, 5-hydroxytryptamine, and indole pathways. A variety of bioactive compounds produced via Trp metabolism can regulate various physiological functions, including inflammation, metabolism, immune responses, and neurological function. Emerging evidence supports an intimate relationship between Trp metabolism disorder and diseases. The levels or ratios of Trp metabolites are significantly associated with many clinical features. Additionally, studies have shown that disease progression can be controlled by modulating Trp metabolism. Indoleamine-2,3-dioxygenase, Trp-2,3-dioxygenase, kynurenine-3-monooxygenase, and Trp hydroxylase are the rate-limiting enzymes that are critical for Trp metabolism. These key regulatory enzymes can be targeted for treating several diseases, including tumors. These findings provide novel insights into the treatment of diseases. In this review, we have summarized the recent research progress on the role of Trp metabolites in health and disease along with their clinical applications.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Ganglei Li
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qiuxian Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xinyu Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qingmiao Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuanshuai Su
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhengyi Bao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
3
|
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
|
4
|
Advanced Neuroimaging Approaches to Pediatric Brain Tumors. Cancers (Basel) 2022; 14:cancers14143401. [PMID: 35884462 PMCID: PMC9318188 DOI: 10.3390/cancers14143401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [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
|
5
|
van Genugten EAJ, Weijers JAM, Heskamp S, Kneilling M, van den Heuvel MM, Piet B, Bussink J, Hendriks LEL, Aarntzen EHJG. Imaging the Rewired Metabolism in Lung Cancer in Relation to Immune Therapy. Front Oncol 2022; 11:786089. [PMID: 35070990 PMCID: PMC8779734 DOI: 10.3389/fonc.2021.786089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/10/2021] [Indexed: 12/14/2022] Open
Abstract
Metabolic reprogramming is recognized as one of the hallmarks of cancer. Alterations in the micro-environmental metabolic characteristics are recognized as important tools for cancer cells to interact with the resident and infiltrating T-cells within this tumor microenvironment. Cancer-induced metabolic changes in the micro-environment also affect treatment outcomes. In particular, immune therapy efficacy might be blunted because of somatic mutation-driven metabolic determinants of lung cancer such as acidity and oxygenation status. Based on these observations, new onco-immunological treatment strategies increasingly include drugs that interfere with metabolic pathways that consequently affect the composition of the lung cancer tumor microenvironment (TME). Positron emission tomography (PET) imaging has developed a wide array of tracers targeting metabolic pathways, originally intended to improve cancer detection and staging. Paralleling the developments in understanding metabolic reprogramming in cancer cells, as well as its effects on stromal, immune, and endothelial cells, a wave of studies with additional imaging tracers has been published. These tracers are yet underexploited in the perspective of immune therapy. In this review, we provide an overview of currently available PET tracers for clinical studies and discuss their potential roles in the development of effective immune therapeutic strategies, with a focus on lung cancer. We report on ongoing efforts that include PET/CT to understand the outcomes of interactions between cancer cells and T-cells in the lung cancer microenvironment, and we identify areas of research which are yet unchartered. Thereby, we aim to provide a starting point for molecular imaging driven studies to understand and exploit metabolic features of lung cancer to optimize immune therapy.
Collapse
Affiliation(s)
- Evelien A J van Genugten
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Jetty A M Weijers
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Manfred Kneilling
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University, Tuebingen, Germany.,Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | | | - Berber Piet
- Department of Respiratory Diseases, Radboudumc, Nijmegen, Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Netherlands
| | - Lizza E L Hendriks
- Department of Pulmonary Diseases, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre (UMC), Maastricht, Netherlands
| | - Erik H J G Aarntzen
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| |
Collapse
|
6
|
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: 3.3] [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
|
7
|
John F, Michelhaugh SK, Barger GR, Mittal S, Juhász C. Depression and tryptophan metabolism in patients with primary brain tumors: Clinical and molecular imaging correlates. Brain Imaging Behav 2021; 15:974-985. [PMID: 32767048 DOI: 10.1007/s11682-020-00305-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Patients with brain tumors have an increased risk for depression, whose underlying pathomechanism may involve dysregulated tryptophan/kynurenine metabolism. In this study, we analyzed the relation of depressive symptoms to clinical and tumor characteristics as well as cerebral and systemic tryptophan metabolism in patients with primary brain tumors. Sixty patients with newly-diagnosed or recurrent primary brain tumor underwent testing with the Beck Depression Inventory-II (BDI-II), and 34 patients also had positron emission tomography (PET) imaging with alpha-[11C]methyl-L-tryptophan (AMT). BDI-II scores were correlated with clinical and tumor-related variables, cerebral regional AMT metabolism measured in the non-tumoral hemisphere, and plasma tryptophan metabolite levels. Sixteen patients (27%) had BDI-II scores indicating depression, including 6 with moderate/severe depression. High BDI-II scores were independent of clinical and tumor-related variables except lower Karnofsky Performance Status scores. In patients with recurrent malignant gliomas, depression was associated with shorter survival (hazard ratio: 3.7; p = 0.048). High BDI-II total and somatic subscale scores were associated with higher frontal cortical and thalamic AMT metabolic values measured on PET. In contrast, plasma tryptophan and kynurenine metabolite levels did not correlate with the BDI-II scores. In conclusion, our results confirm previous data that depression affects more than ¼ of patients with primary brain tumors, it is largely independent of tumor characteristics and is associated with shorter survival in patients with recurrent malignant gliomas. On PET imaging, higher tryptophan metabolism in the frontal cortex and thalamus was found in those with brain tumor-associated depression and supports the role of dysregulated tryptophan/kynurenine metabolism in this condition.
Collapse
Affiliation(s)
- Flóra John
- Department of Pediatrics, Wayne State University and PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, 3901 Beaubien St, MI, Detroit, 48201, USA
| | - Sharon K Michelhaugh
- Department of Neurosurgery, Wayne State University, 4201 St. Antoine St., Detroit, MI, 48201, USA
| | - Geoffrey R Barger
- Department of Neurology, Wayne State University, 4201 St. Antoine St, Detroit, MI, 48201, USA
- Karmanos Cancer Institute, 4100 John R. St, Detroit, MI, 48201, USA
| | - Sandeep Mittal
- Department of Neurosurgery, Wayne State University, 4201 St. Antoine St., Detroit, MI, 48201, USA
- Karmanos Cancer Institute, 4100 John R. St, Detroit, MI, 48201, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24014, USA
- Virginia Tech School of Neuroscience, Blacksburg, VA, 24061, USA
| | - Csaba Juhász
- Department of Pediatrics, Wayne State University and PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, 3901 Beaubien St, MI, Detroit, 48201, USA.
- Department of Neurosurgery, Wayne State University, 4201 St. Antoine St., Detroit, MI, 48201, USA.
- Department of Neurology, Wayne State University, 4201 St. Antoine St, Detroit, MI, 48201, USA.
- Karmanos Cancer Institute, 4100 John R. St, Detroit, MI, 48201, USA.
| |
Collapse
|
8
|
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: 3.3] [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
|
9
|
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: 0.3] [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
|
10
|
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: 0.8] [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
|
11
|
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: 5.8] [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
|
12
|
PET imaging of medulloblastoma with an 18F-labeled tryptophan analogue in a transgenic mouse model. Sci Rep 2020; 10:3800. [PMID: 32123231 PMCID: PMC7051973 DOI: 10.1038/s41598-020-60728-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 02/10/2020] [Indexed: 02/07/2023] Open
Abstract
In vivo positron emission tomography (PET) imaging is a key modality to evaluate disease status of brain tumors. In recent years, tremendous efforts have been made in developing PET imaging methods for pediatric brain tumors. Carbon-11 labelled tryptophan derivatives are feasible as PET imaging probes in brain tumor patients with activation of the kynurenine pathway, but the short half-life of carbon-11 limits its application. Using a transgenic mouse model for the sonic hedgehog (Shh) subgroup of medulloblastoma, here we evaluated the potential of the newly developed 1-(2-[18F]fluoroethyl)-L-tryptophan (1-L-[18F]FETrp) as a PET imaging probe for this common malignant pediatric brain tumor. 1-L-[18F]FETrp was synthesized on a PETCHEM automatic synthesizer with good chemical and radiochemical purities and enantiomeric excess values. Imaging was performed in tumor-bearing Smo/Smo medulloblastoma mice with constitutive actvation of the Smoothened (Smo) receptor using a PerkinElmer G4 PET-X-Ray scanner. Medulloblastoma showed significant and specific accumulation of 1-L-[18F]FETrp. 1-L-[18F]FETrp also showed significantly higher tumor uptake than its D-enantiomer, 1-D-[18F]FETrp. The uptake of 1-L-[18F]FETrp in the normal brain tissue was low, suggesting that 1-L-[18F]FETrp may prove a valuable PET imaging probe for the Shh subgroup of medulloblastoma and possibly other pediatric and adult brain tumors.
Collapse
|
13
|
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: 0.5] [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
|
14
|
Riess C, Schneider B, Kehnscherper H, Gesche J, Irmscher N, Shokraie F, Classen CF, Wirthgen E, Domanska G, Zimpfer A, Strüder D, Junghanss C, Maletzki C. Activation of the Kynurenine Pathway in Human Malignancies Can Be Suppressed by the Cyclin-Dependent Kinase Inhibitor Dinaciclib. Front Immunol 2020; 11:55. [PMID: 32117235 PMCID: PMC7034242 DOI: 10.3389/fimmu.2020.00055] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 01/09/2020] [Indexed: 01/08/2023] Open
Abstract
Indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO2) are the key enzymes of tryptophan (TRP) metabolism in the kynurenine pathway (KP). Both enzymes function as indicators of immunosuppression and poor survival in cancer patients. Direct or indirect targeting of either of these substances seems thus reasonable to improve therapy options for patients. In this study, glioblastoma multiforme (GBM) as well as head and neck squamous cell carcinomas (HNSCC) were examined because of their different mechanisms of spontaneous and treatment-induced immune escape. Effects on gene expression and protein levels were examined. Accompanying assessment of TRP metabolites from treated GBM cell culture supernatants was conducted. Our results show a heterogeneous and inversely correlated expression profile of TRP-metabolizing genes among GBM and HNSCC cells, with low, but inducible IDO1 expression upon IFNγ treatment. TDO2 expression was higher in GBM cells, while genes encoding kynurenine aminotransferases were mainly confined to HNSCC cells. These data indicate that the KP is active in both entities, with however different enzymes involved in TRP catabolism. Upon treatment with Temozolomide, the standard of care for GBM patients, IDO1 was upregulated. Comparable, although less pronounced effects were seen in HNSCC upon Cetuximab and conventional drugs (i.e., 5-fluorouracil, Gemcitabine). Here, IDO1 and additional genes of the KP (KYAT1, KYAT2, and KMO) were induced. Vice versa, the novel yet experimental cyclin-dependent kinase inhibitor Dinaciclib suppressed KP in both entities. Our comprehensive data imply inhibition of the TRP catabolism by Dinaciclib, while conventional chemotherapeutics tend to activate this pathway. These data point to limitations of conventional therapy and highlight the potential of targeted therapies to interfere with the cells' metabolism more than anticipated.
Collapse
Affiliation(s)
- Christin Riess
- University Children's Hospital, Rostock University Medical Centre, Rostock, Germany.,Institute for Medical Microbiology, Virology, and Hygiene, Rostock University Medical Centre, Rostock, Germany.,Medical Clinic III - Hematology, Oncology, Palliative Care, Department of Internal Medicine, Rostock University Medical Center, Rostock, Germany
| | - Björn Schneider
- Institute of Pathology, Rostock University Medical Center, University of Rostock, Rostock, Germany
| | - Hanna Kehnscherper
- Medical Clinic III - Hematology, Oncology, Palliative Care, Department of Internal Medicine, Rostock University Medical Center, Rostock, Germany
| | - Julia Gesche
- Medical Clinic III - Hematology, Oncology, Palliative Care, Department of Internal Medicine, Rostock University Medical Center, Rostock, Germany
| | - Nina Irmscher
- Medical Clinic III - Hematology, Oncology, Palliative Care, Department of Internal Medicine, Rostock University Medical Center, Rostock, Germany
| | - Fatemeh Shokraie
- University Children's Hospital, Rostock University Medical Centre, Rostock, Germany
| | | | - Elisa Wirthgen
- University Children's Hospital, Rostock University Medical Centre, Rostock, Germany
| | - Grazyna Domanska
- Institute of Immunology and Transfusion Medicine, University of Greifswald, Greifswald, Germany
| | - Annette Zimpfer
- Institute of Pathology, Rostock University Medical Center, University of Rostock, Rostock, Germany
| | - Daniel Strüder
- Department of Otorhinolaryngology, Head and Neck Surgery "Otto Koerner", Rostock University Medical Center, Rostock, Germany
| | - Christian Junghanss
- Medical Clinic III - Hematology, Oncology, Palliative Care, Department of Internal Medicine, Rostock University Medical Center, Rostock, Germany
| | - Claudia Maletzki
- Medical Clinic III - Hematology, Oncology, Palliative Care, Department of Internal Medicine, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
15
|
Qiao Z, Mardon K, Stimson DHR, Migotto MA, Reutens DC, Bhalla R. Synthesis and evaluation of 6-[18F]fluoro-3-(pyridin-3-yl)-1H-indole as potential PET tracer for targeting tryptophane 2, 3-dioxygenase (TDO). Nucl Med Biol 2020; 84-85:1-10. [PMID: 31927462 DOI: 10.1016/j.nucmedbio.2019.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/17/2019] [Accepted: 12/17/2019] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The increase in expression of tryptophan 2, 3-dioxygenases (TDO) and indoleamine 2,3-dioxygenase (IDO) have been reported as potential tumor biomarkers. TDO and IDO are enzymes that catalyze the first and rate-limiting step of the kynurenine pathway. Positron emitting tomography (PET) tracers investigating the kynurenine pathway may allow for the detection of different disease pathologies in vivo including cancer. However, current PET tracers being developed for TDO and IDO have suffered from either multi-step low yielding syntheses or de-fluorination of the tracer in vivo. RESULTS TDO inhibitors based on 6-fluoroindole with C3 substituents are a class of small molecules that have been shown to bind to TDO effectively, restore tryptophan concentration and decrease the production of immunosuppressive metabolites. The compound 6-fluoro-3-(pyridine-3-yl)-1H-indole has been reported to have high in vitro affinity for TDO. Herein we report the fully automated radiosynthesis of 6-[18F]fluoro-3-(pyridine-3-yl)-1H-indole [18F]4 using a copper-mediated nucleophilic 18F-fluorination resulting in a non-corrected yield of 5 to 6% of the tracer with a radiochemical purity of >99% after 4 h. Small animal dynamic PET/CT imaging of [18F]4 intravenously injected into normal C57BL/6 mice revealed rapid accumulation in heart and brain, reaching maximum occupancy in heart (10.9% ID/g) and brain (8.1% ID/g) at 1.75 min and 2.25 min, respectively. Furthermore, these in vivo studies revealed no de-fluorination of the tracer, as evidence by the absence of [18F]fluoride accumulation in bone. CONCLUSION In vitro studies demonstrate that 4 has good affinity for hTDO and the radiolabeled analogue [18F]4 can be synthesized with suitable radiochemical yields. [18F]4 demonstrates good uptake in the brain and the radiolabeled compound shows no de-fluorination in vivo in C57BL/6 mice.
Collapse
Affiliation(s)
- Zheng Qiao
- Centre for Advanced Imaging, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Karine Mardon
- Centre for Advanced Imaging, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia; National Imaging Facility, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Damion H R Stimson
- Centre for Advanced Imaging, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Mary-Anne Migotto
- Centre for Advanced Imaging, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - David C Reutens
- Centre for Advanced Imaging, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Rajiv Bhalla
- Centre for Advanced Imaging, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
16
|
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: 0.5] [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
|
17
|
Tracers for non-invasive radionuclide imaging of immune checkpoint expression in cancer. EJNMMI Radiopharm Chem 2019; 4:29. [PMID: 31696402 PMCID: PMC6834817 DOI: 10.1186/s41181-019-0078-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/21/2019] [Indexed: 12/11/2022] Open
Abstract
Abstract Immunotherapy with checkpoint inhibitors demonstrates impressive improvements in the treatment of several types of cancer. Unfortunately, not all patients respond to therapy while severe immune-related adverse effects are prevalent. Currently, patient stratification is based on immunotherapy marker expression through immunohistochemical analysis on biopsied material. However, expression can be heterogeneous within and between tumor lesions, amplifying the sampling limitations of biopsies. Analysis of immunotherapy target expression by non-invasive quantitative molecular imaging with PET or SPECT may overcome this issue. In this review, an overview of tracers that have been developed for preclinical and clinical imaging of key immunotherapy targets, such as programmed cell death-1, programmed cell death ligand-1, IDO1 and cytotoxic T lymphocyte-associated antigen-4 is presented. We discuss important aspects to consider when developing such tracers and outline the future perspectives of molecular imaging of immunotherapy markers. Graphical abstract Current techniques in immune checkpoint imaging and its potential for future applications ![]()
Collapse
|
18
|
Mehta K, Atak A, Sahu A, Srivastava S, C MK. An early investigative serum Raman spectroscopy study of meningioma. Analyst 2019; 143:1916-1923. [PMID: 29620771 DOI: 10.1039/c8an00224j] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Meningiomas represent one of the most frequently reported non-glial, primary brain and central nervous system (CNS) tumors. Meningiomas often display a spectrum of anomalous locations and morphological attributes, deterring their timely diagnosis. Majority of them are sporadic in nature and thus the present-day screening strategies, including radiological investigations, often result in misdiagnosis due to their aberrant and equivocal radiological facets. Therefore, it is pertinent to explore less invasive and patient-friendly biofluids such as serum for their screening and diagnostics. The utility of serum Raman spectroscopy in diagnosis and therapeutic monitoring of cancers has been reported in the literature. In the present study, for the first time, to the best of our knowledge, we have explored Raman spectroscopy to classify the sera of meningioma and control subjects. For this exploration, 35 samples each of meningioma and control subjects were accrued and the spectra revealed variance in the levels of DNA, proteins, lipids, amino acids and β-carotene, i.e., a relatively higher protein, DNA and lipid content in meningioma. Subsequent Principal Component Analysis (PCA) and Principal Component-Linear Discriminant Analysis (PC-LDA) followed by Leave-One-Out Cross-Validation (LOOCV) and limited independent test data, in a patient-wise approach, yielded a classification efficiency of 92% and 80% for healthy and meningioma, respectively. Additionally, in the analogous analysis between healthy and different grades of meningioma, similar results were obtained. These results indicate the potential of Raman spectroscopy in differentiating meningioma. As present methods suffer from known limitations, with the prospective validation on a larger cohort, serum Raman spectroscopy could be an adjuvant/alternative approach in the clinical management of meningioma.
Collapse
Affiliation(s)
- Kanika Mehta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India.
| | | | | | | | | |
Collapse
|
19
|
Lukas RV, Juhász C, Wainwright DA, James CD, Kennedy E, Stupp R, Lesniak MS. Imaging tryptophan uptake with positron emission tomography in glioblastoma patients treated with indoximod. J Neurooncol 2019; 141:111-120. [PMID: 30415456 PMCID: PMC6414051 DOI: 10.1007/s11060-018-03013-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 09/13/2018] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Glioblastoma (GBM) is the most frequent and aggressive primary tumor of the central nervous system, accounting for over 50% of all primary malignant gliomas arising in the adult brain. Even after surgical resection, adjuvant radiotherapy (RT) and temozolomide (TMZ) chemotherapy, as well as tumor-treating fields, the median survival is only 15-20 months. We have identified a pathogenic mechanism that contributes to the tumor-induced immunosuppression in the form of increased indoleamine 2,3 dioxygenase 1 (IDO1) expression; an enzyme that metabolizes the essential amino acid, tryptophan (Trp), into kynurenine (Kyn). However, real-time measurements of IDO1 activity has yet to become mainstream in clinical protocols for assessing IDO1 activity in GBM patients. METHODS Pre-treatment and on-treatment α-[11C]-methyl-L-Trp (AMT) positron emission tomography (PET) with co-registered MRI was performed on patients with recurrent GBM treated with the IDO1 pathway inhibitor indoximod (D1-MT) and TMZ. RESULTS Regional intratumoral variability of AMT within enhancing and non-enhancing tumor was noted at baseline. On treatment imaging revealed decreased regional uptake suggesting IDO1 pathway modulation with treatment. CONCLUSIONS Here, we have validated the ability to use PET of the Trp probe, AMT, for use in visualizing and quantifying intratumoral Trp uptake in GBM patients treated with an IDO1 pathway inhibitor. These data serve as rationale to utilize AMT-PET imaging in the future evaluation of GBM patients treated with IDO1 enzyme inhibitors.
Collapse
Affiliation(s)
- Rimas V Lukas
- Department of Neurology, Northwestern University, 710 N. Lake Shore Drive, Abbott Hall 1114, Chicago, IL, 60611, USA.
- Lurie Cancer Center, Northwestern University, Chicago, USA.
- Lou & Jean Malnati Brain Tumor Institute, Northwestern University, Chicago, USA.
| | - Csaba Juhász
- Neurology, and Neurosurgery, Department of Pediatrics, Wayne State University, Detroit, USA
- Karmanos Cancer Institute, Wayne State University, Detroit, USA
| | - Derek A Wainwright
- Department of Neurosurgery, Northwestern University, Chicago, USA
- Lurie Cancer Center, Northwestern University, Chicago, USA
- Lou & Jean Malnati Brain Tumor Institute, Northwestern University, Chicago, USA
| | - Charles David James
- Department of Neurosurgery, Northwestern University, Chicago, USA
- Lurie Cancer Center, Northwestern University, Chicago, USA
- Lou & Jean Malnati Brain Tumor Institute, Northwestern University, Chicago, USA
| | | | - Roger Stupp
- Department of Neurology, Northwestern University, 710 N. Lake Shore Drive, Abbott Hall 1114, Chicago, IL, 60611, USA
- Department of Neurosurgery, Northwestern University, Chicago, USA
- Lurie Cancer Center, Northwestern University, Chicago, USA
- Lou & Jean Malnati Brain Tumor Institute, Northwestern University, Chicago, USA
| | - Maciej S Lesniak
- Department of Neurosurgery, Northwestern University, Chicago, USA
- Lurie Cancer Center, Northwestern University, Chicago, USA
- Lou & Jean Malnati Brain Tumor Institute, Northwestern University, Chicago, USA
| |
Collapse
|
20
|
Zorofchian S, Iqbal F, Rao M, Aung PP, Esquenazi Y, Ballester LY. Circulating tumour DNA, microRNA and metabolites in cerebrospinal fluid as biomarkers for central nervous system malignancies. J Clin Pathol 2018; 72:271-280. [DOI: 10.1136/jclinpath-2018-205414] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/11/2018] [Accepted: 10/21/2018] [Indexed: 01/06/2023]
Abstract
Central nervous system (CNS) malignancies can be difficult to diagnose and many do not respond satisfactorily to existing therapies. Monitoring patients with CNS malignancies for treatment response and tumour recurrence can be challenging because of the difficulty and risks of brain biopsies, and the low specificity and sensitivity of the less invasive methodologies that are currently available. Uncertainty about tumour diagnosis or whether a tumour has responded to treatment or has recurred can cause delays in therapeutic decisions that can impact patient outcome. Therefore, there is an urgent need to develop and validate reliable and minimally invasive biomarkers for CNS tumours that can be used alone or in combination with current clinical practices. Blood-based biomarkers can be informative in the diagnosis and monitoring of various types of cancer. However, blood-based biomarkers have proven suboptimal for analysis of CNS tumours. In contrast, circulating biomarkers in cerebrospinal fluid (CSF), including circulating tumour DNA, microRNAs and metabolites, hold promise for accurate and minimally invasive assessment of CNS tumours. This review summarises the current understanding of these three types of CSF biomarkers and their potential use in neuro-oncologic clinical practice.
Collapse
|
21
|
Stables R, Clemens G, Butler HJ, Ashton KM, Brodbelt A, Dawson TP, Fullwood LM, Jenkinson MD, Baker MJ. Feature driven classification of Raman spectra for real-time spectral brain tumour diagnosis using sound. Analyst 2018; 142:98-109. [PMID: 27757448 DOI: 10.1039/c6an01583b] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Spectroscopic diagnostics have been shown to be an effective tool for the analysis and discrimination of disease states from human tissue. Furthermore, Raman spectroscopic probes are of particular interest as they allow for in vivo spectroscopic diagnostics, for tasks such as the identification of tumour margins during surgery. In this study, we investigate a feature-driven approach to the classification of metastatic brain cancer, glioblastoma (GB) and non-cancer from tissue samples, and we provide a real-time feedback method for endoscopic diagnostics using sound. To do this, we first evaluate the sensitivity and specificity of three classifiers (SVM, KNN and LDA), when trained with both sub-band spectral features and principal components taken directly from Raman spectra. We demonstrate that the feature extraction approach provides an increase in classification accuracy of 26.25% for SVM and 25% for KNN. We then discuss the molecular assignment of the most salient sub-bands in the dataset. The most salient sub-band features are mapped to parameters of a frequency modulation (FM) synthesizer in order to generate audio clips from each tissue sample. Based on the properties of the sub-band features, the synthesizer was able to maintain similar sound timbres within the disease classes and provide different timbres between disease classes. This was reinforced via listening tests, in which participants were able to discriminate between classes with mean classification accuracy of 71.1%. Providing intuitive feedback via sound frees the surgeons' visual attention to remain on the patient, allowing for greater control over diagnostic and surgical tools during surgery, and thus promoting clinical translation of spectroscopic diagnostics.
Collapse
Affiliation(s)
- Ryan Stables
- Digital Media Technology Laboratory, Millennium Point, City Centre Campus Birmingham City University, West Midlands, B47XG, UK
| | - Graeme Clemens
- WestCHEM, Department of Pure and Applied Chemistry, University of Strathclyde, Technology and Innovation Centre, 99 George Street, Glasgow, G11RD, UK. Twitter:@ChemistryBaker and Centre for Materials Science, Division of Chemistry, University of Central Lancashire, Preston, PR12HE, UK
| | - Holly J Butler
- WestCHEM, Department of Pure and Applied Chemistry, University of Strathclyde, Technology and Innovation Centre, 99 George Street, Glasgow, G11RD, UK. Twitter:@ChemistryBaker
| | - Katherine M Ashton
- Neuropathology, Lancashire Teaching Hospitals NHS Trust, Royal Preston Hospital, Sharoe Green Lane North, Preston, PR29HT, UK
| | - Andrew Brodbelt
- Neuropathology, Lancashire Teaching Hospitals NHS Trust, Royal Preston Hospital, Sharoe Green Lane North, Preston, PR29HT, UK
| | - Timothy P Dawson
- Neuropathology, Lancashire Teaching Hospitals NHS Trust, Royal Preston Hospital, Sharoe Green Lane North, Preston, PR29HT, UK
| | - Leanne M Fullwood
- Centre for Materials Science, Division of Chemistry, University of Central Lancashire, Preston, PR12HE, UK
| | - Michael D Jenkinson
- The Walton Centre for Neurology and Neurosurgery, The Walton Centre NHS Trust, Lower Lane, Liverpool, L97LJ, UK
| | - Matthew J Baker
- WestCHEM, Department of Pure and Applied Chemistry, University of Strathclyde, Technology and Innovation Centre, 99 George Street, Glasgow, G11RD, UK. Twitter:@ChemistryBaker
| |
Collapse
|
22
|
Anna I, Bartosz P, Lech P, Halina A. Novel strategies of Raman imaging for brain tumor research. Oncotarget 2017; 8:85290-85310. [PMID: 29156720 PMCID: PMC5689610 DOI: 10.18632/oncotarget.19668] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 04/29/2017] [Indexed: 01/07/2023] Open
Abstract
Raman diagnostics and imaging have been shown to be an effective tool for the analysis and discrimination of human brain tumors from normal structures. Raman spectroscopic methods have potential to be applied in clinical practice as they allow for identification of tumor margins during surgery. In this study, we investigate medulloblastoma (grade IV WHO) (n= 5), low-grade astrocytoma (grades I-II WHO) (n =4), ependymoma (n=3) and metastatic brain tumors (n= 1) and the tissue from the negative margins used as normal controls. We compare a high grade medulloblastoma, low grade astrocytoma and non-tumor samples from human central nervous system (CNS) tissue. Based on the properties of the Raman vibrational features and Raman images we provide a real–time feedback method that is label-free to monitor tumor metabolism that reveals reprogramming of biosynthesis of lipids, proteins, DNA and RNA. Our results indicate marked metabolic differences between low and high grade brain tumors. We discuss molecular mechanisms causing these metabolic changes, particularly lipid alterations in malignant medulloblastoma and low grade gliomas that may shed light on the mechanisms driving tumor recurrence thereby revealing new approaches for the treatment of malignant glioma. We have found that the high-grade tumors of central nervous system (medulloblastoma) exhibit enhanced level of β-sheet conformation and down-regulated level of α-helix conformation when comparing against normal tissue. We have found that almost all tumors studied in the paper have increased Raman signals of nucleic acids. This increase can be interpreted as increased DNA/RNA turnover in brain tumors. We have shown that the ratio of Raman intensities I2930/I2845 at 2930 and 2845 cm-1 is a good source of information on the ratio of lipid and protein contents. We have found that the ratio reflects the different lipid and protein contents of cancerous brain tissue compared to the non-tumor tissue. We found that levels of the saturated fatty acids were significantly reduced in the high grade medulloblastoma samples compared with non-tumor brain samples and low grade astrocytoma. Differences were also noted in the n-6/n-3 polyunsaturated fatty acids (PUFA) content between medulloblastoma and non-tumor brain samples. The content of the oleic acid (OA) was significantly smaller in almost all brain high grade brain tumors than that observed in the control samples. It indicates that the fatty acid composition of human brain tumors differs from that found in non-tumor brain tissue. The iodine number NI for the normal brain tissue is 60. For comparison OA has 87, docosahexaenoic acid (DHA) 464, α-linolenic acid (ALA) 274. The high grade tumors have the iodine numbers between that for palmitic acid, stearic acid, arachidic acid (NI=0) and oleic acid (NI=87). Most low grade tumors have NI similar to that of OA. The iodine number for arachidonic acid (AA) (NI=334) is much higher than those observed for all studied samples.
Collapse
Affiliation(s)
- Imiela Anna
- Lodz University of Technology, Institute of Applied Radiation Chemistry, Laboratory of Laser Molecular Spectroscopy, 93-590 Lodz, Poland
| | - Polis Bartosz
- Polish Mother's Memorial Hospital Research Institute, Department of Neurosurgery and Neurotraumatology, 3-338 Lodz, Poland
| | - Polis Lech
- Polish Mother's Memorial Hospital Research Institute, Department of Neurosurgery and Neurotraumatology, 3-338 Lodz, Poland
| | - Abramczyk Halina
- Lodz University of Technology, Institute of Applied Radiation Chemistry, Laboratory of Laser Molecular Spectroscopy, 93-590 Lodz, Poland
| |
Collapse
|
23
|
Bosnyák E, Michelhaugh SK, Klinger NV, Kamson DO, Barger GR, Mittal S, Juhász C. Prognostic Molecular and Imaging Biomarkers in Primary Glioblastoma. Clin Nucl Med 2017; 42:341-347. [PMID: 28195901 DOI: 10.1097/rlu.0000000000001577] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE Several molecular glioma markers (including isocitrate dehydrogenase 1 [IDH1] mutation, amplification of the epidermal growth factor receptor [EGFR], and methylation of the O6-methylguanine-DNA methyltransferase [MGMT] promoter) have been associated with glioblastoma survival. In this study, we examined the association between tumoral amino acid uptake, molecular markers, and overall survival in patients with IDH1 wild-type (primary) glioblastoma. PATIENTS AND METHODS Twenty-one patients with newly diagnosed IDH1 wild-type glioblastomas underwent presurgical MRI and PET scanning with alpha[C-11]-L-methyl-tryptophan (AMT). MRI characteristics (T2- and T1-contrast volume), tumoral tryptophan uptake, PET-based metabolic tumor volume, and kinetic variables were correlated with prognostic molecular markers (EGFR and MGMT) and overall survival. RESULTS EGFR amplification was associated with lower T1-contrast volume (P = 0.04) as well as lower T1-contrast/T2 volume (P = 0.04) and T1-contrast/PET volume ratios (P = 0.02). Tumors with MGMT promoter methylation showed lower metabolic volume (P = 0.045) and lower tumor/cortex AMT unidirectional uptake ratios than those with unmethylated MGMT promoter (P = 0.009). While neither EGFR amplification nor MGMT promoter methylation was significantly associated with survival, high AMT tumor/cortex uptake ratios on PET were strongly prognostic for longer survival (hazards ratio, 30; P = 0.002). Estimated mean overall survival was 26 months in patients with high versus 8 months in those with low tumoral AMT uptake ratios. CONCLUSIONS The results demonstrate specific MRI and amino acid PET imaging characteristics associated with EGFR amplification and MGMT promoter methylation in patients with primary glioblastoma. High tryptophan uptake on PET may identify a subgroup with prolonged survival.
Collapse
Affiliation(s)
- Edit Bosnyák
- From the Department of *Pediatrics, Wayne State University, Detroit; †PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit; Departments of ‡Neurosurgery, and §Neurology, Wayne State University, Detroit; ∥Karmanos Cancer Institute, Detroit; and ¶Deparment of Oncology, Wayne State University, Detroit, Michigan
| | | | | | | | | | | | | |
Collapse
|
24
|
Michelhaugh SK, Muzik O, Guastella AR, Klinger NV, Polin LA, Cai H, Xin Y, Mangner TJ, Zhang S, Juhász C, Mittal S. Assessment of Tryptophan Uptake and Kinetics Using 1-(2-18F-Fluoroethyl)-l-Tryptophan and α-11C-Methyl-l-Tryptophan PET Imaging in Mice Implanted with Patient-Derived Brain Tumor Xenografts. J Nucl Med 2016; 58:208-213. [PMID: 27765857 DOI: 10.2967/jnumed.116.179994] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/14/2016] [Indexed: 11/16/2022] Open
Abstract
Abnormal tryptophan metabolism via the kynurenine pathway is involved in the pathophysiology of a variety of human diseases including cancers. α-11C-methyl-l-tryptophan (11C-AMT) PET imaging demonstrated increased tryptophan uptake and trapping in epileptic foci and brain tumors, but the short half-life of 11C limits its widespread clinical application. Recent in vitro studies suggested that the novel radiotracer 1-(2-18F-fluoroethyl)-l-tryptophan (18F-FETrp) may be useful to assess tryptophan metabolism via the kynurenine pathway. In this study, we tested in vivo organ and tumor uptake and kinetics of 18F-FETrp in patient-derived xenograft mouse models and compared them with 11C-AMT uptake. METHODS Xenograft mouse models of glioblastoma and metastatic brain tumors (from lung and breast cancer) were developed by subcutaneous implantation of patient tumor fragments. Dynamic PET scans with 18F-FETrp and 11C-AMT were obtained for mice bearing human brain tumors 1-7 d apart. The biodistribution and tumoral SUVs for both tracers were compared. RESULTS 18F-FETrp showed prominent uptake in the pancreas and no bone uptake, whereas 11C-AMT showed higher uptake in the kidneys. Both tracers showed uptake in the xenograft tumors, with a plateau of approximately 30 min after injection; however, 18F-FETrp showed higher tumoral SUV than 11C-AMT in all 3 tumor types tested. The radiation dosimetry for 18F-FETrp determined from the mouse data compared favorably with the clinical 18F-FDG PET tracer. CONCLUSION 18F-FETrp tumoral uptake, biodistribution, and radiation dosimetry data provide strong preclinical evidence that this new radiotracer warrants further studies that may lead to a broadly applicable molecular imaging tool to examine abnormal tryptophan metabolism in human tumors.
Collapse
Affiliation(s)
| | - Otto Muzik
- Department of Pediatrics, Wayne State University, Detroit, Michigan.,Department of Radiology, Wayne State University, Detroit, Michigan.,PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, Michigan
| | - Anthony R Guastella
- Department of Neurosurgery, Wayne State University, Detroit, Michigan.,Department of Oncology, Wayne State University, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan
| | - Neil V Klinger
- Department of Neurosurgery, Wayne State University, Detroit, Michigan
| | - Lisa A Polin
- Department of Oncology, Wayne State University, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan
| | - Hancheng Cai
- Department of Radiology and Advanced Imaging Research Center, University of Texas Southwestern, Dallas, Texasand
| | - Yangchun Xin
- Department of Radiology and Advanced Imaging Research Center, University of Texas Southwestern, Dallas, Texasand
| | - Thomas J Mangner
- Department of Radiology, Wayne State University, Detroit, Michigan.,PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, Michigan
| | - Shaohui Zhang
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, Michigan
| | - Csaba Juhász
- Department of Pediatrics, Wayne State University, Detroit, Michigan.,PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, Michigan.,Karmanos Cancer Institute, Detroit, Michigan.,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.,Karmanos Cancer Institute, Detroit, Michigan
| |
Collapse
|
25
|
Juhász C, Bosnyák E. PET and SPECT studies in children with hemispheric low-grade gliomas. Childs Nerv Syst 2016; 32:1823-32. [PMID: 27659825 PMCID: PMC5120676 DOI: 10.1007/s00381-016-3125-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 05/20/2016] [Indexed: 10/21/2022]
Abstract
Molecular imaging is playing an increasing role in the pretreatment evaluation of low-grade gliomas. While glucose positron emission tomography (PET) can be helpful to differentiate low-grade from high-grade tumors, PET imaging with amino acid radiotracers has several advantages, such as better differentiation between tumors and non-tumorous lesions, optimized biopsy targeting, and improved detection of tumor recurrence. This review provides a brief overview of single-photon emission computed tomography (SPECT) studies followed by a more detailed review of the clinical applications of glucose and amino acid PET imaging in low-grade hemispheric gliomas. We discuss key differences in the performance of the most commonly utilized PET radiotracers and highlight the advantage of PET/MRI fusion to obtain optimal information about tumor extent, heterogeneity, and metabolism. Recent data also suggest that simultaneous acquisition of PET/MR images and the combination of advanced MRI techniques with quantitative PET can further improve the pretreatment and post-treatment evaluation of pediatric brain tumors.
Collapse
Affiliation(s)
- Csaba Juhász
- Departments of Pediatrics, Wayne State University, Detroit, MI, USA. .,Departments of Neurology, Wayne State University, Detroit, MI, USA. .,PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Wayne State University School of Medicine, 3901 Beaubien Street, Detroit, MI, 48201, USA. .,Karmanos Cancer Institute, Detroit, MI, USA.
| | - Edit Bosnyák
- Department of Pediatrics, Wayne State University, Detroit, MI, USA,PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit Medical Center, Detroit, MI, USA
| |
Collapse
|
26
|
Abstract
Tryptophan-2, 3-dioxygenase (TDO) is a heme-containing protein catalyzing the first reaction in the kynurenine pathway, which incorporates oxygen into the indole moiety of tryptophan and catalyzes it into kynurenine (KYN). The activation of TDO results in the depletion of tryptophan and the accumulation of kynurenine and its metabolites. These metabolites can affect the function of neurons and inhibit the proliferation of T cells. Increasing evidence demonstrates that TDO is a potential therapeutic target in the treatment of brain diseases as well as in the antitumor and transplant fields. Despite its growing popularity, there are few reviews only focusing on TDO. Hence, we herein review TDO by providing a comprehensive overview of TDO, including its biological functions as well as the evolution, structure and catalytic process of TDO. Additionally, this review will focus on the role of TDO in the pathology of three groups of brain diseases: Schizophrenia, Alzheimer's disease (AD) and Glioma. Finally, we will also provide an opinion regarding the future developmental directions of TDO in brain diseases, especially whether TDO has a potential role in other brain diseases as well as the development and applications of TDO inhibitors as treatments.
Collapse
Affiliation(s)
- Cheng-Peng Yu
- The Second Clinic Medical College, School of Medicine, Nanchang University, Nanchang, China
| | - Ze-Zheng Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, China
| | - Da-Ya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, China.
| |
Collapse
|
27
|
Guastella AR, Michelhaugh SK, Klinger NV, Kupsky WJ, Polin LA, Muzik O, Juhász C, Mittal S. Tryptophan PET Imaging of the Kynurenine Pathway in Patient-Derived Xenograft Models of Glioblastoma. Mol Imaging 2016; 15:15/0/1536012116644881. [PMID: 27151136 PMCID: PMC4887573 DOI: 10.1177/1536012116644881] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 03/18/2016] [Indexed: 11/17/2022] Open
Abstract
Increasing evidence demonstrates the immunosuppressive kynurenine pathway's (KP) role in the pathophysiology of human gliomas. To study the KP in vivo, we used the noninvasive molecular imaging tracer α-[(11)C]-methyl-l-tryptophan (AMT). The AMT-positron emission tomography (PET) has shown high uptake in high-grade gliomas and predicted survival in patients with recurrent glioblastoma (GBM). We generated patient-derived xenograft (PDX) models from dissociated cells, or tumor fragments, from 5 patients with GBM. Mice bearing subcutaneous tumors were imaged with AMT-PET, and tumors were analyzed to detect the KP enzymes indoleamine 2,3-dioxygenase (IDO) 1, IDO2, tryptophan 2,3-dioxygenase, kynureninase, and kynurenine 3-monooxygenase. Overall, PET imaging showed robust tumoral AMT uptake in PDX mice with prolonged tracer accumulation over 60 minutes, consistent with AMT trapping seen in humans. Immunostained tumor tissues demonstrated positive detection of multiple KP enzymes. Furthermore, intracranial implantation of GBM cells was performed with imaging at both 9 and 14 days postimplant, with a marked increase in AMT uptake at 14 days and a corresponding high level of tissue immunostaining for KP enzymes. These results indicate that our PDX mouse models recapitulate human GBM, including aberrant tryptophan metabolism, and offer an in vivo system for development of targeted therapeutics for patients with GBM.
Collapse
Affiliation(s)
- Anthony R Guastella
- Department of Neurosurgery, Wayne State University, Detroit, MI, USA Department of Oncology, Wayne State University, Detroit, MI, USA
| | | | - Neil V Klinger
- Department of Neurosurgery, Wayne State University, Detroit, MI, USA
| | - William J Kupsky
- Department of Pathology, Wayne State University, Detroit, MI, USA Karmanos Cancer Institute, Detroit, MI, USA
| | - Lisa A Polin
- Department of Pathology, Wayne State University, Detroit, MI, USA Karmanos Cancer Institute, Detroit, MI, USA
| | - Otto Muzik
- Department of Pediatrics, Wayne State University, Detroit, MI, USA Department of Radiology, Wayne State University, Detroit, MI, USA PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, USA
| | - Csaba Juhász
- Karmanos Cancer Institute, Detroit, MI, USA Department of Pediatrics, Wayne State University, Detroit, MI, USA PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, USA Department of Neurology, Wayne State University, Detroit, MI, USA
| | - Sandeep Mittal
- Department of Neurosurgery, Wayne State University, Detroit, MI, USA Department of Oncology, Wayne State University, Detroit, MI, USA Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
28
|
Challapalli A, Aboagye EO. Positron Emission Tomography Imaging of Tumor Cell Metabolism and Application to Therapy Response Monitoring. Front Oncol 2016; 6:44. [PMID: 26973812 PMCID: PMC4770188 DOI: 10.3389/fonc.2016.00044] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/12/2016] [Indexed: 12/12/2022] Open
Abstract
Cancer cells do reprogram their energy metabolism to enable several functions, such as generation of biomass including membrane biosynthesis, and overcoming bioenergetic and redox stress. In this article, we review both established and evolving radioprobes developed in association with positron emission tomography (PET) to detect tumor cell metabolism and effect of treatment. Measurement of enhanced tumor cell glycolysis using 2-deoxy-2-[(18)F]fluoro-D-glucose is well established in the clinic. Analogs of choline, including [(11)C]choline and various fluorinated derivatives are being tested in several cancer types clinically with PET. In addition to these, there is an evolving array of metabolic tracers for measuring intracellular transport of glutamine and other amino acids or for measuring glycogenesis, as well as probes used as surrogates for fatty acid synthesis or precursors for fatty acid oxidation. In addition to providing us with opportunities for examining the complex regulation of reprogramed energy metabolism in living subjects, the PET methods open up opportunities for monitoring pharmacological activity of new therapies that directly or indirectly inhibit tumor cell metabolism.
Collapse
Affiliation(s)
| | - Eric O. Aboagye
- Department of Surgery and Cancer, Imperial College London, London, UK
| |
Collapse
|
29
|
Design and automated production of 11C-alpha-methyl-l-tryptophan (11C-AMT). Nucl Med Biol 2016; 43:303-8. [PMID: 27150033 DOI: 10.1016/j.nucmedbio.2016.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/08/2016] [Accepted: 02/10/2016] [Indexed: 11/21/2022]
Abstract
(11)C-alpha-methyl-l-tryptophan ([(11)C]AMT), a tryptophan metabolism PET tracer, has successfully been employed for brain serotonin pathway and indoleamine 2,3-dioxygenase (IDO) pathway related tumor imaging. We here report a reliable, automated procedure for routine synthesis of [(11)C]AMT based on an Eckert and Ziegler Modular-Lab system. The semi-preparative HPLC was incorporated into the system to improve chemical purity and specific activity. The 6-step radiosynthesis followed by HPLC-purification provided [(11)C]AMT in 5.3±1.2% (n=6, non-decay-corrected) overall radiochemical yield with radiochemical purity >99% and specific activity of 35-116GBq/μmol. Usually, 2.95±0.65GBq (n=6, EOS) patient ready dose was produced from about 55.5GBq [(11)C]CO2 in 50min.
Collapse
|
30
|
Bosnyák E, Kamson DO, Behen ME, Barger GR, Mittal S, Juhász C. Imaging cerebral tryptophan metabolism in brain tumor-associated depression. EJNMMI Res 2015; 5:56. [PMID: 26475140 PMCID: PMC4608955 DOI: 10.1186/s13550-015-0136-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 10/09/2015] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Depression in patients with brain tumors is associated with impaired quality of life and shorter survival. Altered metabolism of tryptophan to serotonin and kynurenine metabolites may play a role in tumor-associated depression. Our recent studies with alpha[(11)C]methyl-L-tryptophan (AMT)-PET in brain tumor patients indicated abnormal tryptophan metabolism not only in the tumor mass but also in normal-appearing contralateral brain. In the present study, we explored if tryptophan metabolism in such brain regions is associated with depression. METHODS Twenty-one patients (mean age: 57 years) with a brain tumor (10 meningiomas, 8 gliomas, and 3 brain metastases) underwent AMT-PET scanning. MRI and AMT-PET images were co-registered, and AMT kinetic parameters, including volume of distribution (VD', an estimate of net tryptophan transport) and K (unidirectional uptake, related to tryptophan metabolism), were measured in the tumor mass and in unaffected cortical and subcortical regions contralateral to the tumor. Depression scores (based on the Beck Depression Inventory-II [BDI-II]) were correlated with tumor size, grade, type, and AMT-PET variables. RESULTS The mean BDI-II score was 12 ± 10 (range: 2-33); clinical levels of depression were identified in seven patients (33 %). High BDI-II scores were most strongly associated with high thalamic AMT K values both in the whole group (Spearman's rho = 0.63, p = 0.004) and in the subgroup of 18 primary brain tumors (r = 0.68, p = 0.004). Frontal and striatal VD' values were higher in the depressed subgroup than in non-depressed patients (p < 0.05); the group difference was even more robust when moderately/severely depressed patients were compared to patients with no/mild depression (frontal: p = 0.005; striatal: p < 0.001). Tumor size, grade, and tumor type were not related to depression scores. CONCLUSIONS Abnormalities of tryptophan transport and metabolism in the thalamus, striatum, and frontal cortex, measured by PET, are associated with depression in patients with brain tumor. These changes may indicate an imbalance between the serotonin and kynurenine pathways and serve as a molecular imaging marker of brain tumor-associated depression. TRIAL REGISTRATION ClinicalTrials.gov NCT02367469.
Collapse
Affiliation(s)
- Edit Bosnyák
- Department of Pediatrics, Wayne State University, 3901 Beaubien Street, Detroit, MI, 48201, USA.
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, 3901 Beaubien Street, Detroit, MI, 48201, USA.
| | - David O Kamson
- Department of Pediatrics, Wayne State University, 3901 Beaubien Street, Detroit, MI, 48201, USA.
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, 3901 Beaubien Street, Detroit, MI, 48201, USA.
| | - Michael E Behen
- Department of Pediatrics, Wayne State University, 3901 Beaubien Street, Detroit, MI, 48201, USA.
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, 3901 Beaubien Street, Detroit, MI, 48201, USA.
| | - Geoffrey R Barger
- Department of Neurology, Wayne State University, 4201 St. Antoine, Detroit, MI, 48201, USA.
- Karmanos Cancer Institute, Detroit, MI, USA.
| | - Sandeep Mittal
- Department of Neurosurgery, Wayne State University, 4160 John R., Suite 930, Detroit, MI, 48201, USA.
- Department of Oncology, Wayne State University, Detroit, MI, USA.
- Karmanos Cancer Institute, Detroit, MI, USA.
| | - Csaba Juhász
- Department of Pediatrics, Wayne State University, 3901 Beaubien Street, Detroit, MI, 48201, USA.
- Department of Neurology, Wayne State University, 4201 St. Antoine, Detroit, MI, 48201, USA.
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, 3901 Beaubien Street, Detroit, MI, 48201, USA.
- Karmanos Cancer Institute, Detroit, MI, USA.
| |
Collapse
|
31
|
Tryptophan PET predicts spatial and temporal patterns of post-treatment glioblastoma progression detected by contrast-enhanced MRI. J Neurooncol 2015; 126:317-25. [PMID: 26514361 DOI: 10.1007/s11060-015-1970-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 10/25/2015] [Indexed: 10/22/2022]
Abstract
Amino acid PET is increasingly utilized for the detection of recurrent gliomas. Increased amino acid uptake is often observed outside the contrast-enhancing brain tumor mass. In this study, we evaluated if non-enhancing PET+ regions could predict spatial and temporal patterns of subsequent MRI progression in previously treated glioblastomas. Twelve patients with a contrast-enhancing area suspicious for glioblastoma recurrence on MRI underwent PET scanning with the amino acid radiotracer alpha-[(11)C]-methyl-L-tryptophan (AMT). Brain regions showing increased AMT uptake in and outside the contrast-enhancing volume were objectively delineated to include high uptake consistent with glioma (as defined by previous studies). Volume and tracer uptake of such non-enhancing PET+ regions were compared to spatial patterns and timing of subsequent progression of the contrast-enhancing lesion, as defined by serial surveillance MRI. Non-enhancing PET+ volumes varied widely across patients and extended up to 24 mm from the edge of MRI contrast enhancement. In ten patients with clear progression of the contrast-enhancing lesion, the non-enhancing PET+ volumes predicted the location of new enhancement, which extended beyond the PET+ brain tissue in six. In two patients, with no PET+ area beyond the initial contrast enhancement, MRI remained stable. There was a negative correlation between AMT uptake in non-enhancing brain and time to subsequent progression (r = -0.77, p = 0.003). Amino acid PET imaging could complement MRI not only for detecting glioma recurrence but also predicting the location and timing of subsequent tumor progression. This could support decisions for surgical intervention or other targeted therapies for recurrent gliomas.
Collapse
|
32
|
Jeong JW, Juhász C, Mittal S, Bosnyák E, Kamson DO, Barger GR, Robinette NL, Kupsky WJ, Chugani DC. Multi-modal imaging of tumor cellularity and Tryptophan metabolism in human Gliomas. Cancer Imaging 2015; 15:10. [PMID: 26245742 PMCID: PMC4527188 DOI: 10.1186/s40644-015-0045-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/23/2015] [Indexed: 01/12/2023] Open
Abstract
Background To assess gliomas using image-based estimation of cellularity, we utilized isotropic diffusion spectrum imaging (IDSI) on clinically feasible diffusion tensor imaging (DTI) and compared it with amino acid uptake measured by α[11C]methyl-L-tryptophan positron emission tomography (AMT-PET). Methods In 10 patients with a newly-diagnosed glioma, metabolically active tumor regions were defined in both FLAIR hyperintense areas and based on increased uptake on AMT-PET. A recently developed independent component analysis with a ball and stick model was extended to perform IDSI in clinical DTI data. In tumor regions, IDSI was used to define tumor cellularity which was compared between low and high grade glioma and correlated with the glioma proliferative index. Results The IDSI-derived cellularity values were elevated in both FLAIR and AMT-PET-derived regions of high-grade gliomas. ROC curve analysis found that the IDSI-derived cellularity can provide good differentiation of low-grade from high-grade gliomas (accuracy/sensitivity/specificity of 0.80/0.80/0.80). . Both apparent diffusion coefficient (ADC) and IDSI-derived cellularity showed a significant correlation with the glioma proliferative index (based on Ki-67 labeling; R = 0.95, p < 0.001), which was particularly strong when the tumor regions were confined to areas with high tryptophan uptake excluding areas with peritumoral edema. Conclusion IDSI-MRI combined with AMT-PET may provide a multi-modal imaging tool to enhance pretreatment assessment of human gliomas by evaluating tumor cellularity and differentiate low-grade form high-grade gliomas. Electronic supplementary material The online version of this article (doi:10.1186/s40644-015-0045-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jeong-Won Jeong
- Departments of Pediatrics and Neurology, Wayne State University School of Medicine, 3901 Beaubien St., Detroit, MI, 48201, USA. .,PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, USA.
| | - Csaba Juhász
- Departments of Pediatrics and Neurology, Wayne State University School of Medicine, 3901 Beaubien St., Detroit, MI, 48201, USA. .,PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, USA. .,Karmanos Cancer Institute, Detroit, MI, USA.
| | - Sandeep Mittal
- Karmanos Cancer Institute, Detroit, MI, USA. .,Departments of Neurosurgery and Oncology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Edit Bosnyák
- Departments of Pediatrics and Neurology, Wayne State University School of Medicine, 3901 Beaubien St., Detroit, MI, 48201, USA. .,PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, USA.
| | - David O Kamson
- Departments of Pediatrics and Neurology, Wayne State University School of Medicine, 3901 Beaubien St., Detroit, MI, 48201, USA. .,PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, USA.
| | - Geoffrey R Barger
- Karmanos Cancer Institute, Detroit, MI, USA. .,Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Natasha L Robinette
- Karmanos Cancer Institute, Detroit, MI, USA. .,Department of Radiology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - William J Kupsky
- Karmanos Cancer Institute, Detroit, MI, USA. .,Department of Pathology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Diane C Chugani
- Departments of Pediatrics and Neurology, Wayne State University School of Medicine, 3901 Beaubien St., Detroit, MI, 48201, USA. .,PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, USA.
| |
Collapse
|
33
|
Wei WZ, Jones RF, Juhasz C, Gibson H, Veenstra J. Evolution of animal models in cancer vaccine development. Vaccine 2015; 33:7401-7407. [PMID: 26241945 DOI: 10.1016/j.vaccine.2015.07.075] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 07/02/2015] [Indexed: 12/29/2022]
Abstract
Advances in cancer vaccine development are facilitated by animal models reflecting key features of human cancer and its interface with host immunity. Several series of transplantable preneoplastic and neoplastic mouse mammary lesions have been used to delineate mechanisms of anti-tumor immunity. Mimicking immune tolerance to tumor-associated antigens (TAA) such as HER2/neu, transgenic mice developing spontaneous mammary tumors are strong model systems for pre-clinical vaccine testing. In these models, HER2 DNA vaccines are easily administered, well-tolerated, and induce both humoral and cellular immunity. Although engineered mouse strains have advanced cancer immunotherapy, basic shortcomings remain. For example, multiple mouse strains have to be tested to recapitulate genetic regulation of immune tolerance in humans. Outbred domestic felines more closely parallel humans in the natural development of HER2 positive breast cancer and their varying genetic background. Electrovaccination with heterologous HER2 DNA induces robust adaptive immune responses in cats. Importantly, homologous feline HER2 DNA with a single amino acid substitution elicits unique antibodies to feline mammary tumor cells, unlocking a new vaccine principle. As an alternative approach to targeted vaccination, non-surgical tumor ablation such as cryoablation induces anti-tumor immunity via in situ immunization, particularly when combined with toll-like receptor (TLR) agonist. As strategies for vaccination advance, non-invasive monitoring of host response becomes imperative. As an example, magnetic resonance imaging (MRI) and positron emission tomography (PET) scanning following administration of tryptophan metabolism tracer [11C]-alpha-methyl-tryptophan (AMT) provides non-invasive imaging of both tumor growth and metabolic activities. Because AMT is a substrate of indoleamine-pyrrole 2,3-dioxygenase (IDO), an enzyme that produces the immune regulatory molecule kynurenine, AMT imaging can provide novel insight of host response. In conclusion, new feline models improve the predictive power of cancer immunotherapy and real-time PET imaging enables mechanistic monitoring of host immunity. Strategic utilization of these new tools will expedite cancer vaccine development.
Collapse
Affiliation(s)
- Wei-Zen Wei
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States.
| | - Richard F Jones
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States
| | - Csaba Juhasz
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States
| | - Heather Gibson
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States
| | - Jesse Veenstra
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States
| |
Collapse
|
34
|
Bosnyák E, Kamson DO, Guastella AR, Varadarajan K, Robinette NL, Kupsky WJ, Muzik O, Michelhaugh SK, Mittal S, Juhász C. Molecular imaging correlates of tryptophan metabolism via the kynurenine pathway in human meningiomas. Neuro Oncol 2015; 17:1284-92. [PMID: 26092774 DOI: 10.1093/neuonc/nov098] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/06/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Increased tryptophan metabolism via the kynurenine pathway (KP) is a key mechanism of tumoral immune suppression in gliomas. However, details of tryptophan metabolism in meningiomas have not been elucidated. In this study, we evaluated in vivo tryptophan metabolism in meningiomas and compared it with gliomas using α-[(11)C]-methyl-L-tryptophan (AMT)-PET. We also explored expression patterns of KP enzymes in resected meningiomas. METHODS Forty-seven patients with MRI-detected meningioma (n = 16) and glioma (n = 31) underwent presurgical AMT-PET scanning. Tumoral AMT uptake and tracer kinetic parameters (including K and k3' evaluating unidirectional uptake and trapping, respectively) were measured, correlated with meningioma grade, and compared between meningiomas and gliomas. Patterns of KP enzyme expression were assessed by immunohistochemistry in all meningiomas. RESULTS Meningioma grade showed a positive correlation with AMT k3' tumor/cortex ratio (r = 0.75, P = .003), and this PET parameter distinguished grade I from grade II/III meningiomas with 92% accuracy. Kinetic AMT parameters could differentiate meningiomas from both low-grade gliomas (97% accuracy by k3' ratios) and high-grade gliomas (83% accuracy by K ratios). Among 3 initial KP enzymes (indoleamine 2,3-dioxygenase 1/2, and tryptophan 2,3-dioxygenase 2 [TDO2]), TDO2 showed the strongest immunostaining, particularly in grade I meningiomas. TDO2 also showed a strong negative correlation with AMT k3' ratios (P = .001). CONCLUSIONS PET imaging of tryptophan metabolism can provide quantitative imaging markers for differentiating grade I from grade II/III meningiomas. TDO2 may be an important driver of in vivo tryptophan metabolism in these tumors. These results can have implications for pharmacological targeting of the KP in meningiomas.
Collapse
Affiliation(s)
- Edit Bosnyák
- Department of Pediatrics, Wayne State University, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Department of Neurology, Wayne State University, Detroit, Michigan (C.J.); Department of Neurosurgery, Wayne State University, Detroit, Michigan (A.R.G., K.V., S.K.M., S.M.); Department of Oncology, Wayne State University, Detroit, Michigan (A.R.G., S.M.); Department of Radiology, Wayne State University, , Detroit, Michigan (N.L.R., O.M.); Department of Pathology, Wayne State University, Detroit, Michigan (W.J.K.); PET Center, Children's Hospital of Michigan, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Karmanos Cancer Institute, Detroit, Michigan (N.L.R., W.J.K., S.M., C.J.)
| | - David O Kamson
- Department of Pediatrics, Wayne State University, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Department of Neurology, Wayne State University, Detroit, Michigan (C.J.); Department of Neurosurgery, Wayne State University, Detroit, Michigan (A.R.G., K.V., S.K.M., S.M.); Department of Oncology, Wayne State University, Detroit, Michigan (A.R.G., S.M.); Department of Radiology, Wayne State University, , Detroit, Michigan (N.L.R., O.M.); Department of Pathology, Wayne State University, Detroit, Michigan (W.J.K.); PET Center, Children's Hospital of Michigan, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Karmanos Cancer Institute, Detroit, Michigan (N.L.R., W.J.K., S.M., C.J.)
| | - Anthony R Guastella
- Department of Pediatrics, Wayne State University, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Department of Neurology, Wayne State University, Detroit, Michigan (C.J.); Department of Neurosurgery, Wayne State University, Detroit, Michigan (A.R.G., K.V., S.K.M., S.M.); Department of Oncology, Wayne State University, Detroit, Michigan (A.R.G., S.M.); Department of Radiology, Wayne State University, , Detroit, Michigan (N.L.R., O.M.); Department of Pathology, Wayne State University, Detroit, Michigan (W.J.K.); PET Center, Children's Hospital of Michigan, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Karmanos Cancer Institute, Detroit, Michigan (N.L.R., W.J.K., S.M., C.J.)
| | - Kaushik Varadarajan
- Department of Pediatrics, Wayne State University, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Department of Neurology, Wayne State University, Detroit, Michigan (C.J.); Department of Neurosurgery, Wayne State University, Detroit, Michigan (A.R.G., K.V., S.K.M., S.M.); Department of Oncology, Wayne State University, Detroit, Michigan (A.R.G., S.M.); Department of Radiology, Wayne State University, , Detroit, Michigan (N.L.R., O.M.); Department of Pathology, Wayne State University, Detroit, Michigan (W.J.K.); PET Center, Children's Hospital of Michigan, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Karmanos Cancer Institute, Detroit, Michigan (N.L.R., W.J.K., S.M., C.J.)
| | - Natasha L Robinette
- Department of Pediatrics, Wayne State University, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Department of Neurology, Wayne State University, Detroit, Michigan (C.J.); Department of Neurosurgery, Wayne State University, Detroit, Michigan (A.R.G., K.V., S.K.M., S.M.); Department of Oncology, Wayne State University, Detroit, Michigan (A.R.G., S.M.); Department of Radiology, Wayne State University, , Detroit, Michigan (N.L.R., O.M.); Department of Pathology, Wayne State University, Detroit, Michigan (W.J.K.); PET Center, Children's Hospital of Michigan, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Karmanos Cancer Institute, Detroit, Michigan (N.L.R., W.J.K., S.M., C.J.)
| | - William J Kupsky
- Department of Pediatrics, Wayne State University, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Department of Neurology, Wayne State University, Detroit, Michigan (C.J.); Department of Neurosurgery, Wayne State University, Detroit, Michigan (A.R.G., K.V., S.K.M., S.M.); Department of Oncology, Wayne State University, Detroit, Michigan (A.R.G., S.M.); Department of Radiology, Wayne State University, , Detroit, Michigan (N.L.R., O.M.); Department of Pathology, Wayne State University, Detroit, Michigan (W.J.K.); PET Center, Children's Hospital of Michigan, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Karmanos Cancer Institute, Detroit, Michigan (N.L.R., W.J.K., S.M., C.J.)
| | - Otto Muzik
- Department of Pediatrics, Wayne State University, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Department of Neurology, Wayne State University, Detroit, Michigan (C.J.); Department of Neurosurgery, Wayne State University, Detroit, Michigan (A.R.G., K.V., S.K.M., S.M.); Department of Oncology, Wayne State University, Detroit, Michigan (A.R.G., S.M.); Department of Radiology, Wayne State University, , Detroit, Michigan (N.L.R., O.M.); Department of Pathology, Wayne State University, Detroit, Michigan (W.J.K.); PET Center, Children's Hospital of Michigan, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Karmanos Cancer Institute, Detroit, Michigan (N.L.R., W.J.K., S.M., C.J.)
| | - Sharon K Michelhaugh
- Department of Pediatrics, Wayne State University, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Department of Neurology, Wayne State University, Detroit, Michigan (C.J.); Department of Neurosurgery, Wayne State University, Detroit, Michigan (A.R.G., K.V., S.K.M., S.M.); Department of Oncology, Wayne State University, Detroit, Michigan (A.R.G., S.M.); Department of Radiology, Wayne State University, , Detroit, Michigan (N.L.R., O.M.); Department of Pathology, Wayne State University, Detroit, Michigan (W.J.K.); PET Center, Children's Hospital of Michigan, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Karmanos Cancer Institute, Detroit, Michigan (N.L.R., W.J.K., S.M., C.J.)
| | - Sandeep Mittal
- Department of Pediatrics, Wayne State University, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Department of Neurology, Wayne State University, Detroit, Michigan (C.J.); Department of Neurosurgery, Wayne State University, Detroit, Michigan (A.R.G., K.V., S.K.M., S.M.); Department of Oncology, Wayne State University, Detroit, Michigan (A.R.G., S.M.); Department of Radiology, Wayne State University, , Detroit, Michigan (N.L.R., O.M.); Department of Pathology, Wayne State University, Detroit, Michigan (W.J.K.); PET Center, Children's Hospital of Michigan, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Karmanos Cancer Institute, Detroit, Michigan (N.L.R., W.J.K., S.M., C.J.)
| | - Csaba Juhász
- Department of Pediatrics, Wayne State University, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Department of Neurology, Wayne State University, Detroit, Michigan (C.J.); Department of Neurosurgery, Wayne State University, Detroit, Michigan (A.R.G., K.V., S.K.M., S.M.); Department of Oncology, Wayne State University, Detroit, Michigan (A.R.G., S.M.); Department of Radiology, Wayne State University, , Detroit, Michigan (N.L.R., O.M.); Department of Pathology, Wayne State University, Detroit, Michigan (W.J.K.); PET Center, Children's Hospital of Michigan, Detroit, Michigan (E.B., D.O.K., O.M., C.J.); Karmanos Cancer Institute, Detroit, Michigan (N.L.R., W.J.K., S.M., C.J.)
| |
Collapse
|
35
|
Juhász C, Dwivedi S, Kamson DO, Michelhaugh SK, Mittal S. Comparison of amino acid positron emission tomographic radiotracers for molecular imaging of primary and metastatic brain tumors. Mol Imaging 2015; 13. [PMID: 24825818 DOI: 10.2310/7290.2014.00015] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Positron emission tomography (PET) is an imaging technology that can detect and characterize tumors based on their molecular and biochemical properties, such as altered glucose, nucleoside, or amino acid metabolism. PET plays a significant role in the diagnosis, prognostication, and treatment of various cancers, including brain tumors. In this article, we compare uptake mechanisms and the clinical performance of the amino acid PET radiotracers (l-[methyl-11C]methionine [MET], 18F-fluoroethyl-tyrosine [FET], 18F-fluoro-l-dihydroxy-phenylalanine [FDOPA], and 11C-alpha-methyl-l-tryptophan [AMT]) most commonly used for brain tumor imaging. First, we discuss and compare the mechanisms of tumoral transport and accumulation, the basis of differential performance of these radioligands in clinical studies. Then we summarize studies that provided direct comparisons among these amino acid tracers and to clinically used 2-deoxy-2[18F]fluoro-d-glucose [FDG] PET imaging. We also discuss how tracer kinetic analysis can enhance the clinical information obtained from amino acid PET images. We discuss both similarities and differences in potential clinical value for each radioligand. This comparative review can guide which radiotracer to favor in future clinical trials aimed at defining the role of these molecular imaging modalities in the clinical management of brain tumor patients.
Collapse
|
36
|
Adams S, Teo C, McDonald KL, Zinger A, Bustamante S, Lim CK, Sundaram G, Braidy N, Brew BJ, Guillemin GJ. Involvement of the kynurenine pathway in human glioma pathophysiology. PLoS One 2014; 9:e112945. [PMID: 25415278 PMCID: PMC4240539 DOI: 10.1371/journal.pone.0112945] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 10/17/2014] [Indexed: 12/23/2022] Open
Abstract
The kynurenine pathway (KP) is the principal route of L-tryptophan (TRP) catabolism leading to the production of kynurenine (KYN), the neuroprotectants, kynurenic acid (KYNA) and picolinic acid (PIC), the excitotoxin, quinolinic acid (QUIN) and the essential pyridine nucleotide, nicotinamide adenine dinucleotide (NAD(+)). The enzymes indoleamine 2,3-dioxygenase-1 (IDO-1), indoleamine 2,3-dioxygenase-2 (IDO-2) and tryptophan 2,3-dioxygenase (TDO-2) initiate the first step of the KP. IDO-1 and TDO-2 induction in tumors are crucial mechanisms implicated to play pivotal roles in suppressing anti-tumor immunity. Here, we report the first comprehensive characterisation of the KP in 1) cultured human glioma cells and 2) plasma from patients with glioblastoma (GBM). Our data revealed that interferon-gamma (IFN-γ) stimulation significantly potentiated the expression of the KP enzymes, IDO-1 IDO-2, kynureninase (KYNU), kynurenine hydroxylase (KMO) and significantly down-regulated 2-amino-3-carboxymuconate semialdehyde decarboxylase (ACMSD) and kynurenine aminotransferase-I (KAT-I) expression in cultured human glioma cells. This significantly increased KP activity but significantly lowered the KYNA/KYN neuroprotective ratio in human cultured glioma cells. KP activation (KYN/TRP) was significantly higher, whereas the concentrations of the neuroreactive KP metabolites TRP, KYNA, QUIN and PIC and the KYNA/KYN ratio were significantly lower in GBM patient plasma (n = 18) compared to controls. These results provide further evidence for the involvement of the KP in glioma pathophysiology and highlight a potential role of KP products as novel and highly attractive therapeutic targets to evaluate for the treatment of brain tumors, aimed at restoring anti-tumor immunity and reducing the capacity for malignant cells to produce NAD(+), which is necessary for energy production and DNA repair.
Collapse
MESH Headings
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- Astrocytes/drug effects
- Astrocytes/metabolism
- Biosynthetic Pathways
- Brain Neoplasms/genetics
- Brain Neoplasms/metabolism
- Brain Neoplasms/physiopathology
- CD11b Antigen/metabolism
- Carboxy-Lyases/genetics
- Carboxy-Lyases/metabolism
- Cells, Cultured
- Chromatography, High Pressure Liquid
- Disaccharides
- Gene Expression/drug effects
- Glial Fibrillary Acidic Protein/metabolism
- Glioma/genetics
- Glioma/metabolism
- Glioma/physiopathology
- Glucuronates
- Humans
- Immunohistochemistry
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Interferon-gamma/pharmacology
- Kynurenic Acid/blood
- Kynurenic Acid/metabolism
- Kynurenine/biosynthesis
- Kynurenine/blood
- Picolinic Acids/blood
- Picolinic Acids/metabolism
- Quinolinic Acid/blood
- Quinolinic Acid/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Tryptophan/blood
- Tryptophan/metabolism
- Tryptophan Oxygenase/genetics
- Tryptophan Oxygenase/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Seray Adams
- MND and Neurodegenerative Diseases Research Centre, Australian School of Advanced Medicine, Macquarie University, Sydney, NSW, Australia
| | - Charles Teo
- Centre for Minimally Invasive Neurosurgery, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Kerrie L. McDonald
- Cure For Life Neuro-Oncology Group, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
| | - Anna Zinger
- School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| | - Sonia Bustamante
- Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, NSW, Australia
| | - Chai K. Lim
- MND and Neurodegenerative Diseases Research Centre, Australian School of Advanced Medicine, Macquarie University, Sydney, NSW, Australia
| | - Gayathri Sundaram
- St Vincent's Centre for Applied Medical Research, Sydney, NSW, Australia
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Bruce J. Brew
- St Vincent's Centre for Applied Medical Research, Sydney, NSW, Australia
- Department of Neurology, St Vincent's Hospital, Sydney, NSW, Australia
| | - Gilles J. Guillemin
- MND and Neurodegenerative Diseases Research Centre, Australian School of Advanced Medicine, Macquarie University, Sydney, NSW, Australia
- St Vincent's Centre for Applied Medical Research, Sydney, NSW, Australia
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
37
|
Kamson DO, Lee TJ, Varadarajan K, Robinette NL, Muzik O, Chakraborty PK, Snyder M, Barger GR, Mittal S, Juhász C. Clinical significance of tryptophan metabolism in the nontumoral hemisphere in patients with malignant glioma. J Nucl Med 2014; 55:1605-10. [PMID: 25189339 DOI: 10.2967/jnumed.114.141002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED α-(11)C-methyl-L-tryptophan (AMT) PET allows evaluation of brain serotonin synthesis and can also track upregulation of the immunosuppressive kynurenine pathway in tumor tissue. Increased AMT uptake is a hallmark of World Health Organization grade III-IV gliomas. Our recent study also suggested decreased frontal cortical AMT uptake in glioma patients contralateral to the tumor. The clinical significance of extratumoral tryptophan metabolism has not been established. In the present study, we investigated clinical correlates of tryptophan metabolic abnormalities in the nontumoral hemisphere of glioma patients. METHODS Standardized AMT uptake values (SUVs) and the uptake rate constant of AMT (K [mL/g/min], a measure proportional to serotonin synthesis in nontumoral gray matter) were quantified in the frontal and temporal cortex and thalamus in the nontumoral hemisphere in 77 AMT PET scans of 66 patients (41 men, 25 women; mean age ± SD, 55 ± 15 y) with grade III-IV gliomas. These AMT values were determined before treatment in 35 and after treatment in 42 patients and were correlated with clinical variables and survival. RESULTS AMT uptake in the thalamus showed a moderate age-related increase before treatment (SUV, r = 0.39, P = 0.02) but decrease after treatment (K, r = -0.33, P = 0.057). Women had higher thalamic SUVs before treatment (P = 0.037) and higher thalamic (P = 0.013) and frontal cortical K values (P = 0.023) after treatment. In the posttreatment glioma group, high thalamic SUVs and high thalamocortical SUV ratios were associated with short survival in Cox regression analysis. The thalamocortical ratio remained strongly prognostic (P < 0.01) when clinical predictors, including age, glioma grade, and time since radiotherapy, were entered in the regression model. Long interval between radiotherapy and posttreatment AMT PET as well as high radiation dose affecting the thalamus were associated with lower contralateral thalamic or cortical AMT uptake values. CONCLUSION These observations provide evidence for altered tryptophan uptake in contralateral cortical and thalamic brain regions in glioma patients after initial therapy, suggesting treatment effects on the serotonergic system. Low thalamic tryptophan uptake appears to be a strong, independent predictor of long survival in patients with previous glioma treatment.
Collapse
Affiliation(s)
- David O Kamson
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, Michigan Department of Pediatrics, Wayne State University, Detroit, Michigan
| | - Tiffany J Lee
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, Michigan
| | - Kaushik Varadarajan
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, Michigan
| | - Natasha L Robinette
- Department of Radiology, Wayne State University, Detroit, Michigan Karmanos Cancer Institute, Detroit, Michigan
| | - Otto Muzik
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, Michigan Department of Pediatrics, Wayne State University, Detroit, Michigan Department of Radiology, Wayne State University, Detroit, Michigan Department of Neurology, Wayne State University, Detroit, Michigan
| | - Pulak K Chakraborty
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, Michigan Department of Radiology, Wayne State University, Detroit, Michigan
| | | | - Geoffrey R Barger
- Karmanos Cancer Institute, Detroit, Michigan Department of Neurology, Wayne State University, Detroit, Michigan
| | - Sandeep Mittal
- Karmanos Cancer Institute, Detroit, Michigan Department of Neurosurgery, Wayne State University, Detroit, Michigan; and Department of Oncology, Wayne State University, Detroit, Michigan
| | - Csaba Juhász
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, Michigan Department of Pediatrics, Wayne State University, Detroit, Michigan Karmanos Cancer Institute, Detroit, Michigan Department of Neurology, Wayne State University, Detroit, Michigan
| |
Collapse
|
38
|
Yang M, Al Zaharna M, Chen YS, Li L, Cheung HY. In vitro antioxidant activities and anti-proliferative properties of the functional herb Abrus cantoniensis and its main alkaloid abrine. Food Funct 2014; 5:2268-77. [DOI: 10.1039/c4fo00217b] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
39
|
Kamson DO, Mittal S, Robinette NL, Muzik O, Kupsky WJ, Barger GR, Juhász C. Increased tryptophan uptake on PET has strong independent prognostic value in patients with a previously treated high-grade glioma. Neuro Oncol 2014; 16:1373-83. [PMID: 24670609 DOI: 10.1093/neuonc/nou042] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Previously, we demonstrated the high accuracy of alpha-[(11)C]methyl-L-tryptophan (AMT) PET for differentiating recurrent gliomas from radiation injury. The present study evaluated the prognostic value of increased AMT uptake in patients with previously treated high-grade glioma. METHODS AMT-PET was performed in 39 patients with suspected recurrence of World Health Organization grades III-IV glioma following surgical resection, radiation, and chemotherapy. Mean and maximum standardized uptake values (SUVs) and unidirectional AMT uptake (K) were measured in brain regions suspicious for tumor and compared with the contralateral cortex (ie, background). Optimal cutoff thresholds for 1-year survival prediction were determined for each AMT parameter and used for calculating the prognostic value of high (above threshold) versus low (below threshold) values for post-PET overall survival (OS). RESULTS In univariate analyses, 1-year survival was strongly associated with 3 AMT parameters (SUVmax, SUVmean, and tumor-to-background K-ratio; odds ratios: 21.3-25.6; P ≤ .001) and with recent change in MRI contrast enhancement (odds ratio: 14.7; P = .02). Median OS was 876 days in the low- versus 177 days in the high-AMT groups (log-rank P < .001). In multivariate analyses, all 3 AMT parameters remained strong predictors of survival: high AMT values were associated with unfavorable 1-year survival (binary regression P ≤ .003) and shorter overall survival in the whole group (Cox regression hazard ratios: 5.3-10.0) and in patients with recent enhancement change on MRI as well (hazard ratios: 7.0-9.3; P ≤ .001). CONCLUSION Increased AMT uptake on PET is highly prognostic for 1-year and overall survival, independent of MRI contrast enhancement and other prognostic factors in patients with a previously treated high-grade glioma.
Collapse
Affiliation(s)
- David O Kamson
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit Medical Center, Detroit, MI (D.O.K., O.M., C.J.); Department of Neurology, Wayne State University, Detroit, Michigan (G.R.B., C.J.); Department of Neurosurgery, Wayne State University, Detroit, Michigan (S.M.); Department of Oncology, Wayne State University, Detroit, Michigan (S.M.); Department of Pathology, Wayne State University, Detroit, Michigan (W.J.K.); Department of Pediatrics, Wayne State University, Detroit, Michigan (O.M., C.J.); Department of Radiology, Wayne State University, Detroit, Michigan (N.L.R., O.M.); Karmanos Cancer Institute, Detroit, Michigan (S.M., N.L.R., W.J.K., G.R.B., C.J.)
| | - Sandeep Mittal
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit Medical Center, Detroit, MI (D.O.K., O.M., C.J.); Department of Neurology, Wayne State University, Detroit, Michigan (G.R.B., C.J.); Department of Neurosurgery, Wayne State University, Detroit, Michigan (S.M.); Department of Oncology, Wayne State University, Detroit, Michigan (S.M.); Department of Pathology, Wayne State University, Detroit, Michigan (W.J.K.); Department of Pediatrics, Wayne State University, Detroit, Michigan (O.M., C.J.); Department of Radiology, Wayne State University, Detroit, Michigan (N.L.R., O.M.); Karmanos Cancer Institute, Detroit, Michigan (S.M., N.L.R., W.J.K., G.R.B., C.J.)
| | - Natasha L Robinette
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit Medical Center, Detroit, MI (D.O.K., O.M., C.J.); Department of Neurology, Wayne State University, Detroit, Michigan (G.R.B., C.J.); Department of Neurosurgery, Wayne State University, Detroit, Michigan (S.M.); Department of Oncology, Wayne State University, Detroit, Michigan (S.M.); Department of Pathology, Wayne State University, Detroit, Michigan (W.J.K.); Department of Pediatrics, Wayne State University, Detroit, Michigan (O.M., C.J.); Department of Radiology, Wayne State University, Detroit, Michigan (N.L.R., O.M.); Karmanos Cancer Institute, Detroit, Michigan (S.M., N.L.R., W.J.K., G.R.B., C.J.)
| | - Otto Muzik
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit Medical Center, Detroit, MI (D.O.K., O.M., C.J.); Department of Neurology, Wayne State University, Detroit, Michigan (G.R.B., C.J.); Department of Neurosurgery, Wayne State University, Detroit, Michigan (S.M.); Department of Oncology, Wayne State University, Detroit, Michigan (S.M.); Department of Pathology, Wayne State University, Detroit, Michigan (W.J.K.); Department of Pediatrics, Wayne State University, Detroit, Michigan (O.M., C.J.); Department of Radiology, Wayne State University, Detroit, Michigan (N.L.R., O.M.); Karmanos Cancer Institute, Detroit, Michigan (S.M., N.L.R., W.J.K., G.R.B., C.J.)
| | - William J Kupsky
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit Medical Center, Detroit, MI (D.O.K., O.M., C.J.); Department of Neurology, Wayne State University, Detroit, Michigan (G.R.B., C.J.); Department of Neurosurgery, Wayne State University, Detroit, Michigan (S.M.); Department of Oncology, Wayne State University, Detroit, Michigan (S.M.); Department of Pathology, Wayne State University, Detroit, Michigan (W.J.K.); Department of Pediatrics, Wayne State University, Detroit, Michigan (O.M., C.J.); Department of Radiology, Wayne State University, Detroit, Michigan (N.L.R., O.M.); Karmanos Cancer Institute, Detroit, Michigan (S.M., N.L.R., W.J.K., G.R.B., C.J.)
| | - Geoffrey R Barger
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit Medical Center, Detroit, MI (D.O.K., O.M., C.J.); Department of Neurology, Wayne State University, Detroit, Michigan (G.R.B., C.J.); Department of Neurosurgery, Wayne State University, Detroit, Michigan (S.M.); Department of Oncology, Wayne State University, Detroit, Michigan (S.M.); Department of Pathology, Wayne State University, Detroit, Michigan (W.J.K.); Department of Pediatrics, Wayne State University, Detroit, Michigan (O.M., C.J.); Department of Radiology, Wayne State University, Detroit, Michigan (N.L.R., O.M.); Karmanos Cancer Institute, Detroit, Michigan (S.M., N.L.R., W.J.K., G.R.B., C.J.)
| | - Csaba Juhász
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit Medical Center, Detroit, MI (D.O.K., O.M., C.J.); Department of Neurology, Wayne State University, Detroit, Michigan (G.R.B., C.J.); Department of Neurosurgery, Wayne State University, Detroit, Michigan (S.M.); Department of Oncology, Wayne State University, Detroit, Michigan (S.M.); Department of Pathology, Wayne State University, Detroit, Michigan (W.J.K.); Department of Pediatrics, Wayne State University, Detroit, Michigan (O.M., C.J.); Department of Radiology, Wayne State University, Detroit, Michigan (N.L.R., O.M.); Karmanos Cancer Institute, Detroit, Michigan (S.M., N.L.R., W.J.K., G.R.B., C.J.)
| |
Collapse
|
40
|
Rheims S, Rubi S, Bouvard S, Bernard E, Streichenberger N, Guenot M, Le Bars D, Hammers A, Ryvlin P. Accuracy of distinguishing between dysembryoplastic neuroepithelial tumors and other epileptogenic brain neoplasms with [¹¹C]methionine PET. Neuro Oncol 2014; 16:1417-26. [PMID: 24598358 DOI: 10.1093/neuonc/nou022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Dysembryoplastic neuroepithelial tumors (DNTs) represent a prevalent cause of epileptogenic brain tumors, the natural evolution of which is much more benign than that of most gliomas. Previous studies have suggested that [(11)C]methionine positron emission tomography (MET-PET) could help to distinguish DNTs from other epileptogenic brain tumors, and hence optimize the management of patients. Here, we reassessed the diagnostic accuracy of MET-PET for the differentiation between DNT and other epileptogenic brain neoplasms in a larger population. METHODS We conducted a retrospective study of 77 patients with focal epilepsy related to a nonrapidly progressing brain tumor on MRI who underwent MET-PET, including 52 with a definite histopathology. MET-PET data were assessed by a structured visual analysis that distinguished normal, moderately abnormal, and markedly abnormal tumor methionine uptake and by semiquantitative ratio measurements. RESULTS Pathology showed 21 DNTs (40%), 10 gangliogliomas (19%), 19 low-grade gliomas (37%), and 2 high-grade gliomas (4%). MET-PET visual findings significantly differed among the various tumor types (P < .001), as confirmed by semiquantitative analyses (P < .001 for all calculated ratios), regardless of gadolinium enhancement on MRI. All gliomas and gangliogliomas were associated with moderately or markedly increased tumor methionine uptake, whereas 9/21 DNTs had normal methionine uptake. Receiver operating characteristics analysis of the semiquantitative ratios showed an optimal cutoff threshold that distinguished DNTs from other tumor types with 90% specificity and 89% sensitivity. CONCLUSIONS Normal MET-PET findings in patients with an epileptogenic nonrapidly progressing brain tumor are highly suggestive of DNT, whereas a markedly increased tumor methionine uptake makes this diagnosis unlikely.
Collapse
Affiliation(s)
- Sylvain Rheims
- Department of Functional Neurology and Epileptology and Institute of Epilepsies (IDEE) (S.Rh., E.B., P.R.); Department of Pathology (N.S.); Department of Functional Neurosurgery (M.G.); Hospices Civils de Lyon, Lyon, France; Lyon Neuroscience Research Center, INSERM U1028/CNRS UMR5292, Lyon, France (S.Rh., S.B., P.R.); CERMEP-Imagerie du Vivant, Lyon, France (S.B., D.L.B.); Neurodis Foundation, CERMEP-Imagerie du Vivant, Lyon, France (A.H.); Hospital Clinic de Barcelona, Barcelona, Spain (S.Ru.)
| | - Sebastià Rubi
- Department of Functional Neurology and Epileptology and Institute of Epilepsies (IDEE) (S.Rh., E.B., P.R.); Department of Pathology (N.S.); Department of Functional Neurosurgery (M.G.); Hospices Civils de Lyon, Lyon, France; Lyon Neuroscience Research Center, INSERM U1028/CNRS UMR5292, Lyon, France (S.Rh., S.B., P.R.); CERMEP-Imagerie du Vivant, Lyon, France (S.B., D.L.B.); Neurodis Foundation, CERMEP-Imagerie du Vivant, Lyon, France (A.H.); Hospital Clinic de Barcelona, Barcelona, Spain (S.Ru.)
| | - Sandrine Bouvard
- Department of Functional Neurology and Epileptology and Institute of Epilepsies (IDEE) (S.Rh., E.B., P.R.); Department of Pathology (N.S.); Department of Functional Neurosurgery (M.G.); Hospices Civils de Lyon, Lyon, France; Lyon Neuroscience Research Center, INSERM U1028/CNRS UMR5292, Lyon, France (S.Rh., S.B., P.R.); CERMEP-Imagerie du Vivant, Lyon, France (S.B., D.L.B.); Neurodis Foundation, CERMEP-Imagerie du Vivant, Lyon, France (A.H.); Hospital Clinic de Barcelona, Barcelona, Spain (S.Ru.)
| | - Emilien Bernard
- Department of Functional Neurology and Epileptology and Institute of Epilepsies (IDEE) (S.Rh., E.B., P.R.); Department of Pathology (N.S.); Department of Functional Neurosurgery (M.G.); Hospices Civils de Lyon, Lyon, France; Lyon Neuroscience Research Center, INSERM U1028/CNRS UMR5292, Lyon, France (S.Rh., S.B., P.R.); CERMEP-Imagerie du Vivant, Lyon, France (S.B., D.L.B.); Neurodis Foundation, CERMEP-Imagerie du Vivant, Lyon, France (A.H.); Hospital Clinic de Barcelona, Barcelona, Spain (S.Ru.)
| | - Nathalie Streichenberger
- Department of Functional Neurology and Epileptology and Institute of Epilepsies (IDEE) (S.Rh., E.B., P.R.); Department of Pathology (N.S.); Department of Functional Neurosurgery (M.G.); Hospices Civils de Lyon, Lyon, France; Lyon Neuroscience Research Center, INSERM U1028/CNRS UMR5292, Lyon, France (S.Rh., S.B., P.R.); CERMEP-Imagerie du Vivant, Lyon, France (S.B., D.L.B.); Neurodis Foundation, CERMEP-Imagerie du Vivant, Lyon, France (A.H.); Hospital Clinic de Barcelona, Barcelona, Spain (S.Ru.)
| | - Marc Guenot
- Department of Functional Neurology and Epileptology and Institute of Epilepsies (IDEE) (S.Rh., E.B., P.R.); Department of Pathology (N.S.); Department of Functional Neurosurgery (M.G.); Hospices Civils de Lyon, Lyon, France; Lyon Neuroscience Research Center, INSERM U1028/CNRS UMR5292, Lyon, France (S.Rh., S.B., P.R.); CERMEP-Imagerie du Vivant, Lyon, France (S.B., D.L.B.); Neurodis Foundation, CERMEP-Imagerie du Vivant, Lyon, France (A.H.); Hospital Clinic de Barcelona, Barcelona, Spain (S.Ru.)
| | - Didier Le Bars
- Department of Functional Neurology and Epileptology and Institute of Epilepsies (IDEE) (S.Rh., E.B., P.R.); Department of Pathology (N.S.); Department of Functional Neurosurgery (M.G.); Hospices Civils de Lyon, Lyon, France; Lyon Neuroscience Research Center, INSERM U1028/CNRS UMR5292, Lyon, France (S.Rh., S.B., P.R.); CERMEP-Imagerie du Vivant, Lyon, France (S.B., D.L.B.); Neurodis Foundation, CERMEP-Imagerie du Vivant, Lyon, France (A.H.); Hospital Clinic de Barcelona, Barcelona, Spain (S.Ru.)
| | - Alexander Hammers
- Department of Functional Neurology and Epileptology and Institute of Epilepsies (IDEE) (S.Rh., E.B., P.R.); Department of Pathology (N.S.); Department of Functional Neurosurgery (M.G.); Hospices Civils de Lyon, Lyon, France; Lyon Neuroscience Research Center, INSERM U1028/CNRS UMR5292, Lyon, France (S.Rh., S.B., P.R.); CERMEP-Imagerie du Vivant, Lyon, France (S.B., D.L.B.); Neurodis Foundation, CERMEP-Imagerie du Vivant, Lyon, France (A.H.); Hospital Clinic de Barcelona, Barcelona, Spain (S.Ru.)
| | - Philippe Ryvlin
- Department of Functional Neurology and Epileptology and Institute of Epilepsies (IDEE) (S.Rh., E.B., P.R.); Department of Pathology (N.S.); Department of Functional Neurosurgery (M.G.); Hospices Civils de Lyon, Lyon, France; Lyon Neuroscience Research Center, INSERM U1028/CNRS UMR5292, Lyon, France (S.Rh., S.B., P.R.); CERMEP-Imagerie du Vivant, Lyon, France (S.B., D.L.B.); Neurodis Foundation, CERMEP-Imagerie du Vivant, Lyon, France (A.H.); Hospital Clinic de Barcelona, Barcelona, Spain (S.Ru.)
| |
Collapse
|
41
|
Abstract
The management of epilepsy is an essential clinical issue in many patients with brain tumors. Tumoral epilepsy is often drug resistant and is associated with poor quality of life. Surgery represents a key therapeutic option in the management of patients with refractory tumoral epilepsy, with high rates of postoperative seizure freedom, especially when gross total resection can be performed. The selection of surgical candidates first requires extrapolation of the presumed underlying pathology and its potential for malignant transformation from clinical and imaging data, especially MRI characteristics. These data determine the decision for surgery, as well as its timing and technical aspects in relation to the risk of postoperative deficit. In glioneuronal tumors, where seizures are often drug-resistant and risk of malignant transformation is very low, epilepsy surgery is usually recommended to alleviate disabling seizures and side effects of antiepileptic drugs. However, the risk of postoperative deficit may outweigh potential benefits of surgery in tumors located within eloquent cortex. This issue is particularly relevant for glioneuronal tumors located within the dominant mesial temporal structures in patients in whom seizure control might require additional hippocampectomy, associated with a high risk of memory decline. In contrast, in patients with low-grade gliomas or aggressive brain neoplasms, both the decision to perform surgery and selection of the best surgical approach primarily rely on the oncologic imperative rather than epileptologic considerations. In these patients, the extent of tumor resection correlates with improved survival, progression-free survival, as well as with the chances of postoperative seizure control.
Collapse
Affiliation(s)
- Sylvain Rheims
- Department of Functional Neurology and Epileptology, Hospices Civils de Lyon, Lyon, France; INSERM U1028/CNRS UMR5292, Translational and Integrative Group in Epilepsy Research, Lyon Neuroscience Research Center, Lyon, France
| | | | | |
Collapse
|
42
|
Juhász C, Buth A, Chugani DC, Kupsky WJ, Chugani HT, Shah AK, Mittal S. Successful surgical treatment of an inflammatory lesion associated with new-onset refractory status epilepticus. Neurosurg Focus 2014; 34:E5. [PMID: 23724839 DOI: 10.3171/2013.3.focus1336] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
New-onset refractory status epilepticus (NORSE) has high morbidity and mortality. The authors describe the successful surgical treatment of a 56-year-old man presenting with NORSE. Magnetic resonance imaging showed a left temporal lobe lesion suspicious for a low-grade tumor, while PET imaging with the alpha[(11)C]methyl-L-tryptophan (AMT) radiotracer showed increased cortical uptake extending beyond this lesion and partly overlapping with epileptogenic cortex mapped by chronic intracranial electroencephalographic monitoring. Resection of the epileptic focus resulted in long-term seizure freedom, and the nonresected portion of the PET-documented abnormality normalized. Histopathology showed reactive gliosis and inflammatory markers in the AMT-PET-positive cortex. Molecular imaging of neuroinflammation can be instrumental in the management of NORSE by guiding placement of intracranial electrodes or assessing the extent and severity of inflammation for antiinflammatory interventions.
Collapse
Affiliation(s)
- Csaba Juhász
- Department of Pediatrics, Wayne State University; Detroit, MI 48201, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Christensen M, Kamson DO, Snyder M, Kim H, Robinette NL, Mittal S, Juhász C. Tryptophan PET-defined gross tumor volume offers better coverage of initial progression than standard MRI-based planning in glioblastoma patients. ACTA ACUST UNITED AC 2013; 3:131-138. [PMID: 25414765 DOI: 10.1007/s13566-013-0132-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
OBJECTIVE Glioblastoma is an infiltrative malignancy that tends to extend beyond the MRI-defined tumor volume. We utilized positron emission tomography (PET) imaging with the radiotracer alpha-[11C]methyl-L -tryptophan (AMT) to develop a reliable high-risk gross tumor volume (HR-GTV) method for delineation of glioblastoma. AMT can detect solid tumor mass and tumoral brain infiltration by increased tumoral tryptophan transport and metabolism via the immunosuppressive kynurenine pathway. METHODS We reviewed all patients in our database with histologically proven glioblastoma who underwent preoperative AMT-PET scan prior to surgery and chemoradiation. Treated radiotherapy volumes were derived from the simulation CT with MRI fusion. High-GTV with contrast enhanced T1-weighted MRI alone (GTVMRI) was defined as the postoperative cavity plus any residual area of enhancement on postcontrast T1-weighted images. AMT-PET images were retrospectively fused to the simulation CT, and a high-risk GTVs generated by both AMT-PET alone (GTVAMT) was defined using a threshold previously established to distinguish tumor tissue from peritumoral edema. A composite volume of MRI and AMT tumor volume was also created (combination of MRI fused with AMT-PET data; GTVMRI+AMT). In patients with definitive radiographic progression, follow-up MRI demonstrating initial tumor progression was fused with the pretreatment images and a progression volume was contoured. The coverage of the progression volume by GTVMRI, GTVAMT, and GTVMRI+AMT was determined and compared using the Wilcoxon's signed-rank test. RESULTS Eleven patients completed presurgical AMT-PET scan, seven of whom had progressive disease after initial therapy. GTVMRI (mean, 50.2 cm3) and GTVAMT (mean, 48.9 cm3) were not significantly different. Mean concordance index of the volumes was 39±15 %. Coverage of the initial recurrence volume by HR-GTVMRI (mean, 52 %) was inferior to both GTVAMT (mean, 68 %; p =0.028) and GTVMRI+AMT (mean 73 %; p =0.018). The AMT-PET-exclusive coverage was up to 41 % of the recurrent volume. There was a tendency towards better recurrence coverage with GTVMRI+AMT than with GTVAMT alone (p =0.068). Addition of 5 mm concentric margin around GTVMRI, GTVAMT, and GTVMRI+AMT would have completely covered the initial progression volume in 14, 57, and 71 % of the patients, respectively. CONCLUSION We found that a GTV defined by AMT-PET produced similar volume, but superior recurrence coverage than the treated standard MRI-determined volume. A prospective study is necessary to fully determine the usefulness of AMT-PET for volume definition in glioblastoma radiotherapy planning.
Collapse
Affiliation(s)
- Michael Christensen
- Department of Radiation Oncology, Barbara Ann, Karmanos Cancer Center, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI 48201, USA. Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
| | - David Olayinka Kamson
- Departments of Pediatrics and Neurology, Wayne State University School of Medicine, Detroit, MI, USA. PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, USA
| | - Michael Snyder
- Department of Radiation Oncology, Barbara Ann, Karmanos Cancer Center, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI 48201, USA. Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
| | - Harold Kim
- Department of Radiation Oncology, Barbara Ann, Karmanos Cancer Center, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI 48201, USA. Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
| | - Natasha L Robinette
- Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA. Department of Radiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Sandeep Mittal
- Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA. Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Csaba Juhász
- Departments of Pediatrics and Neurology, Wayne State University School of Medicine, Detroit, MI, USA. Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA. PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, USA
| |
Collapse
|
44
|
Kamson DO, Mittal S, Buth A, Muzik O, Kupsky WJ, Robinette NL, Barger GR, Juhász C. Differentiation of Glioblastomas from Metastatic Brain Tumors by Tryptophan Uptake and Kinetic Analysis: A Positron Emission Tomographic Study with Magnetic Resonance Imaging Comparison. Mol Imaging 2013. [DOI: 10.2310/7290.2013.00048] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- David O. Kamson
- From the PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, and the Departments of Neurosurgery, Pediatrics, Radiology, Pathology, and Neurology and The Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Sandeep Mittal
- From the PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, and the Departments of Neurosurgery, Pediatrics, Radiology, Pathology, and Neurology and The Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Amy Buth
- From the PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, and the Departments of Neurosurgery, Pediatrics, Radiology, Pathology, and Neurology and The Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Otto Muzik
- From the PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, and the Departments of Neurosurgery, Pediatrics, Radiology, Pathology, and Neurology and The Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - William J. Kupsky
- From the PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, and the Departments of Neurosurgery, Pediatrics, Radiology, Pathology, and Neurology and The Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Natasha L. Robinette
- From the PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, and the Departments of Neurosurgery, Pediatrics, Radiology, Pathology, and Neurology and The Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Geoffrey R. Barger
- From the PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, and the Departments of Neurosurgery, Pediatrics, Radiology, Pathology, and Neurology and The Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Csaba Juhász
- From the PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, and the Departments of Neurosurgery, Pediatrics, Radiology, Pathology, and Neurology and The Karmanos Cancer Institute, Wayne State University, Detroit, MI
| |
Collapse
|
45
|
Zitron IM, Kamson DO, Kiousis S, Juhász C, Mittal S. In vivo metabolism of tryptophan in meningiomas is mediated by indoleamine 2,3-dioxygenase 1. Cancer Biol Ther 2013; 14:333-9. [PMID: 23358471 DOI: 10.4161/cbt.23624] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Expression and activity of indoleamine 2,3-dioxygenase (IDO), the first and rate-limiting step of the kynurenine pathway of tryptophan catabolism, can enable tumor cells to effectively evade the host's immune response. The potential role of this system was investigated in meningiomas. Surgical specimens from 22 patients with meningiomas were used for cellular, immunological and molecular techniques (immunofluorescence, western blotting, RT-PCR and biochemical assay of enzyme activity) to investigate the expression and activity of IDO. In addition, PET imaging was obtained preoperatively in 10 patients using the tracer α-[ ( 11) C]methyl-L-tryptophan (AMT) which interrogates the uptake and metabolism of tryptophan. Strong AMT accumulation was noted in all meningiomas by PET imaging indicating in vivo tryptophan uptake. Freshly-resected meningiomas expressed both LAT1, the tryptophan transporter system and IDO, demonstrating an active kynurenine pathway. Dissociated meningioma cells lost IDO expression. Following exposure to interferon-γ (IFNγ), IDO expression was reinduced and could be blocked by a selective IDO1 inhibitor. IDO activity may represent an element of local self-protection by meningiomas and could be targeted by emerging IDO1 inhibitors.
Collapse
Affiliation(s)
- Ian M Zitron
- Department of Neurosurgery, Wayne State University, Detroit, MI, USA
| | | | | | | | | |
Collapse
|
46
|
Tryptophan PET in pretreatment delineation of newly-diagnosed gliomas: MRI and histopathologic correlates. J Neurooncol 2013; 112:121-32. [PMID: 23299463 DOI: 10.1007/s11060-013-1043-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 01/02/2013] [Indexed: 10/27/2022]
Abstract
Pretreatment delineation of infiltrating glioma volume remains suboptimal with current neuroimaging techniques. Gadolinium-enhanced T1-weighted (T1-Gad) MR images often underestimate the true extent of the tumor, while T2-weighted images preferentially highlight peritumoral edema. Accumulation of α-[(11)C]methyl-L-tryptophan (AMT) on positron emission tomography (PET) has been shown in gliomas. To determine whether increased uptake on AMT-PET would detect tumor-infiltrated brain tissue outside the contrast-enhancing region and differentiate it from peritumoral vasogenic edema, volumes and spatial concordance of T1-Gad and T2 MRI abnormalities as well as AMT-PET abnormalities were analyzed in 28 patients with newly-diagnosed WHO grade II-IV gliomas. AMT-accumulating grade I meningiomas were used to define an AMT uptake cutoff threshold that detects the tumor but excludes peri-meningioma vasogenic edema. Tumor infiltration in AMT-accumulating areas was studied in stereotactically-resected specimens from patients with glioblastoma. In the 28 gliomas, mean AMT-PET-defined tumor volumes were greater than the contrast-enhancing volume, but smaller than T2 abnormalities. Volume of AMT-accumulating tissue outside MRI abnormalities increased with higher tumor proliferative index and was the largest in glioblastomas. Tumor infiltration was confirmed by histopathology from AMT-positive regions outside contrast-enhancing glioblastoma mass, while no or minimal tumor cells were found in AMT-negative specimens. These results demonstrate that increased AMT accumulation on PET detects glioma-infiltrated brain tissue extending beyond the contrast-enhanced tumor mass. While tryptophan uptake is low in peritumoral vasogenic edema, AMT-PET can detect tumor-infiltrated brain outside T2-lesions. Thus, AMT-PET may assist pretreatment delineation of tumor infiltration, particularly in high-grade gliomas.
Collapse
|
47
|
Quantitative PET imaging of tryptophan accumulation in gliomas and remote cortex: correlation with tumor proliferative activity. Clin Nucl Med 2012; 37:838-42. [PMID: 22889771 DOI: 10.1097/rlu.0b013e318251e458] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE PET studies with α[C-11]methyl-L-tryptophan (AMT) have shown decreased serotonin synthesis based on a decrease of the unidirectional uptake rate (K-complex) in neuropsychiatric conditions such as autism and depression. Increased AMT K-complex in tumors can indicate increased tryptophan metabolism via the immunosuppressive kynurenine pathway. Moreover, apparent AMT volume of distribution (VD') reflects net tryptophan transport from blood to tissue. We evaluated if kinetic parameters (K-complex, VD') of AMT, measured by PET, can predict the proliferative activity of glioma, and if these AMT parameters are altered in the remote cortex. METHODS We evaluated dynamic AMT PET images of 30 adult patients with grade 2 to 4 gliomas according to the World Health Organization's classification to determine tumoral AMT VD' and K-complex values, which were correlated with tumor proliferative activity as assessed by the Ki-67 labeling index in resected tumor specimens. We also compared cortical VD' and K-complex values between patients with glioma and healthy controls. RESULTS Both VD' and K-complex values were significantly higher in gliomas than in the contralateral cortex (VD', P < 0.001; K-complex, P < 0.001). Tumoral VD' values and tumor/cortex VD' ratios, but not the K-complex, showed strong positive correlations with the proliferative activity of glioma (P ≤ 0.001). The contralateral frontal cortex showed decreased AMT VD' and K-complex in patients with glioma compared with those in controls (P ≤ 0.01). CONCLUSIONS Increased net amino acid transport into tumor tissue, quantified by PET, can serve as an imaging marker of the proliferative activity of glioma. The data also suggest a glioma-induced down-regulation of cortical serotonin synthesis, likely mediated by shunting of tryptophan from serotonin synthesis to kynurenine metabolism.
Collapse
|
48
|
Adams S, Braidy N, Bessesde A, Brew BJ, Grant R, Teo C, Guillemin GJ. The Kynurenine Pathway in Brain Tumor Pathogenesis. Cancer Res 2012; 72:5649-57. [DOI: 10.1158/0008-5472.can-12-0549] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
49
|
Alkonyi B, Barger GR, Mittal S, Muzik O, Chugani DC, Bahl G, Robinette NL, Kupsky WJ, Chakraborty PK, Juhász C. Accurate differentiation of recurrent gliomas from radiation injury by kinetic analysis of α-11C-methyl-L-tryptophan PET. J Nucl Med 2012; 53:1058-64. [PMID: 22653792 DOI: 10.2967/jnumed.111.097881] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
UNLABELLED PET of amino acid transport and metabolism may be more accurate than conventional neuroimaging in differentiating recurrent gliomas from radiation-induced tissue changes. α-(11)C-methyl-l-tryptophan ((11)C-AMT) is an amino acid PET tracer that is not incorporated into proteins but accumulates in gliomas, mainly because of tumoral transport and metabolism via the immunomodulatory kynurenine pathway. The aim of this study was to evaluate the usefulness of (11)C-AMT PET supplemented by tracer kinetic analysis for distinguishing recurrent gliomas from radiation injury. METHODS Twenty-two (11)C-AMT PET scans were obtained in adult patients who presented with a lesion suggestive of tumor recurrence on conventional MRI 1-6 y (mean, 3 y) after resection and postsurgical radiation of a World Health Organization grade II-IV glioma. Lesional standardized uptake values were calculated, as well as lesion-to-contralateral cortex ratios and 2 kinetic (11)C-AMT PET parameters (volume of distribution [VD], characterizing tracer transport, and unidirectional uptake rate [K]). Tumor was differentiated from radiation-injured tissue by histopathology (n = 13) or 1-y clinical and MRI follow-up (n = 9). Accuracy of tumor detection by PET variables was assessed by receiver-operating-characteristic analysis. RESULTS All (11)C-AMT PET parameters were higher in tumors (n = 12) than in radiation injury (n = 10) (P ≤ 0.012 in all comparisons). The lesion-to-cortex K-ratio most accurately identified tumor recurrence, with highly significant differences both in the whole group (P < 0.0001) and in lesions with histologic verification (P = 0.006); the area under the receiver-operating-characteristic curve was 0.99. A lesion-to-cortex K-ratio threshold of 1.39 (i.e., a 39% increase) correctly differentiated tumors from radiation injury in all but 1 case (100% sensitivity and 91% specificity). In tumors that were high-grade initially (n = 15), a higher lesion-to-cortex K-ratio threshold completely separated recurrent tumors (all K-ratios ≥ 1.70) from radiation injury (all K-ratios < 1.50) (100% sensitivity and specificity). CONCLUSION Kinetic analysis of dynamic (11)C-AMT PET images may accurately differentiate between recurrent World Health Organization grade II-IV infiltrating gliomas and radiation injury. Separation of unidirectional uptake rates from transport can enhance the differentiating accuracy of (11)C-AMT PET. Applying the same approach to other amino acid PET tracers might also improve their ability to differentiate recurrent gliomas from radiation injury.
Collapse
Affiliation(s)
- Bálint Alkonyi
- PET Center, Children's Hospital of Michigan, Detroit, MI, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Juhász C, Nahleh Z, Zitron I, Chugani DC, Janabi MZ, Bandyopadhyay S, Ali-Fehmi R, Mangner TJ, Chakraborty PK, Mittal S, Muzik O. Tryptophan metabolism in breast cancers: molecular imaging and immunohistochemistry studies. Nucl Med Biol 2012; 39:926-32. [PMID: 22444239 DOI: 10.1016/j.nucmedbio.2012.01.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 01/25/2012] [Accepted: 01/28/2012] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Tryptophan oxidation via the kynurenine pathway is an important mechanism of tumoral immunoresistance. Increased tryptophan metabolism via the serotonin pathway has been linked to malignant progression in breast cancer. In this study, we combined quantitative positron emission tomography (PET) with tumor immunohistochemistry to analyze tryptophan transport and metabolism in breast cancer. METHODS Dynamic α-[(11)C]methyl-l-tryptophan (AMT) PET was performed in nine women with stage II-IV breast cancer. PET tracer kinetic modeling was performed in all tumors. Expression of L-type amino acid transporter 1 (LAT1), indoleamine 2,3-dioxygenase (IDO; the initial and rate-limiting enzyme of the kynurenine pathway) and tryptophan hydroxylase 1 (TPH1; the initial enzyme of the serotonin pathway) was assessed by immunostaining of resected tumor specimens. RESULTS Tumor AMT uptake peaked at 5-20 min postinjection in seven tumors; the other two cases showed protracted tracer accumulation. Tumor standardized uptake values (SUVs) varied widely (2.6-9.8) and showed a strong positive correlation with volume of distribution values derived from kinetic analysis (P<.01). Invasive ductal carcinomas (n=6) showed particularly high AMT SUVs (range, 4.7-9.8). Moderate to strong immunostaining for LAT1, IDO and TPH1 was detected in most tumor cells. CONCLUSIONS Breast cancers show differential tryptophan kinetics on dynamic PET. SUVs measured 5-20 min postinjection reflect reasonably the tracer's volume of distribution. Further studies are warranted to determine if in vivo AMT accumulation in these tumors is related to tryptophan metabolism via the kynurenine and serotonin pathways.
Collapse
Affiliation(s)
- Csaba Juhász
- PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI 48201, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|