51
|
Pigeon H, Pérès EA, Truillet C, Jego B, Boumezbeur F, Caillé F, Zinnhardt B, Jacobs AH, Le Bihan D, Winkeler A. TSPO-PET and diffusion-weighted MRI for imaging a mouse model of infiltrative human glioma. Neuro Oncol 2020; 21:755-764. [PMID: 30721979 DOI: 10.1093/neuonc/noz029] [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: 12/31/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most devastating brain tumor. Despite the use of multimodal treatments, most patients relapse, often due to the highly invasive nature of gliomas. However, the detection of glioma infiltration remains challenging. The aim of this study was to assess advanced PET and MRI techniques for visualizing biological activity and infiltration of the tumor. METHODS Using multimodality imaging, we investigated [18F]DPA-714, a radiotracer targeting the 18 kDa translocator protein (TSPO), [18F]FET PET, non-Gaussian diffusion MRI (apparent diffusion coefficient, kurtosis), and the S-index, a composite diffusion metric, to detect tumor infiltration in a human invasive glioma model. In vivo imaging findings were confirmed by autoradiography and immunofluorescence. RESULTS Increased tumor-to-contralateral [18F]DPA-714 uptake ratios (1.49 ± 0.11) were found starting 7 weeks after glioma cell implantation. TSPO-PET allowed visualization of glioma infiltration into the contralateral hemisphere 2 weeks earlier compared with the clinically relevant biomarker for biological glioma activity [18F]FET. Diffusion-weighted imaging (DWI), in particular kurtosis, was more sensitive than standard T2-weighted MRI to detect differences between the glioma-bearing and the contralateral hemisphere at 5 weeks. Immunofluorescence data reflect in vivo findings. Interestingly, labeling for tumoral and stromal TSPO indicates a predominant expression of TSPO by tumor cells. CONCLUSION These results suggest that advanced PET and MRI methods, such as [18F]DPA-714 and DWI, may be superior to standard imaging methods to visualize glioma growth and infiltration at an early stage.
Collapse
Affiliation(s)
- Hayet Pigeon
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Elodie A Pérès
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France.,NeuroSpin, CEA/Université Paris-Saclay, Gif sur Yvette, France.,Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, Caen, France
| | - Charles Truillet
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Benoit Jego
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | | | - Fabien Caillé
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Bastian Zinnhardt
- EIMI and Department of Nuclear Medicine, University Hospital Münster, Westfälische Wilhelms University Münster, Münster, Germany
| | - Andreas H Jacobs
- EIMI and Department of Nuclear Medicine, University Hospital Münster, Westfälische Wilhelms University Münster, Münster, Germany.,Department of Geriatrics, Johanniter Hospital, Evangelische Kliniken, Bonn, Germany
| | - Denis Le Bihan
- NeuroSpin, CEA/Université Paris-Saclay, Gif sur Yvette, France
| | - Alexandra Winkeler
- UMR 1023, IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| |
Collapse
|
52
|
Chavda V, Patel V, Yadav D, Shah J, Patel S, Jin JO. Therapeutics and Research Related to Glioblastoma: Advancements and Future Targets. Curr Drug Metab 2020; 21:186-198. [DOI: 10.2174/1389200221666200408083950] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 11/28/2019] [Accepted: 03/27/2020] [Indexed: 12/19/2022]
Abstract
Glioblastoma, the most common primary brain tumor, has been recognized as one of the most lethal and
fatal human tumors. It has a dismal prognosis, and survival after diagnosis is less than 15 months. Surgery and radiotherapy
are the only available treatment options at present. However, numerous approaches have been made to upgrade
in vivo and in vitro models with the primary goal of assessing abnormal molecular pathways that would be
suitable targets for novel therapeutic approaches. Novel drugs, delivery systems, and immunotherapy strategies to
establish new multimodal therapies that target the molecular pathways involved in tumor initiation and progression in
glioblastoma are being studied. The goal of this review was to describe the pathophysiology, neurodegeneration
mechanisms, signaling pathways, and future therapeutic targets associated with glioblastomas. The key features have
been detailed to provide an up-to-date summary of the advancement required in current diagnosis and therapeutics
for glioblastoma. The role of nanoparticulate system graphene quantum dots as suitable therapy for glioblastoma has
also been discussed.
Collapse
Affiliation(s)
- Vishal Chavda
- Department of Pharmacology, Nirma University, Ahmadabad, Gujarat, 382481, India
| | - Vimal Patel
- Department of Pharmaceutics, Nirma University, Ahmadabad, Gujarat, 382481, India
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 712-749, Korea
| | - Jigar Shah
- Department of Pharmaceutics, Nirma University, Ahmadabad, Gujarat, 382481, India
| | - Snehal Patel
- Department of Pharmacology, Nirma University, Ahmadabad, Gujarat, 382481, India
| | - Jun-O Jin
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 712-749, Korea
| |
Collapse
|
53
|
Banati RB, Wilcox P, Xu R, Yin G, Si E, Son ET, Shimizu M, Holsinger RMD, Parmar A, Zahra D, Arthur A, Middleton RJ, Liu GJ, Charil A, Graeber MB. Selective, high-contrast detection of syngeneic glioblastoma in vivo. Sci Rep 2020; 10:9968. [PMID: 32561881 PMCID: PMC7305160 DOI: 10.1038/s41598-020-67036-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 05/19/2020] [Indexed: 01/14/2023] Open
Abstract
Glioblastoma is a highly malignant, largely therapy-resistant brain tumour. Deep infiltration of brain tissue by neoplastic cells represents the key problem of diffuse glioma. Much current research focuses on the molecular makeup of the visible tumour mass rather than the cellular interactions in the surrounding brain tissue infiltrated by the invasive glioma cells that cause the tumour’s ultimately lethal outcome. Diagnostic neuroimaging that enables the direct in vivo observation of the tumour infiltration zone and the local host tissue responses at a preclinical stage are important for the development of more effective glioma treatments. Here, we report an animal model that allows high-contrast imaging of wild-type glioma cells by positron emission tomography (PET) using [18 F]PBR111, a selective radioligand for the mitochondrial 18 kDa Translocator Protein (TSPO), in the Tspo−/− mouse strain (C57BL/6-Tspotm1GuMu(GuwiyangWurra)). The high selectivity of [18 F]PBR111 for the TSPO combined with the exclusive expression of TSPO in glioma cells infiltrating into null-background host tissue free of any TSPO expression, makes it possible, for the first time, to unequivocally and with uniquely high biological contrast identify peri-tumoral glioma cell invasion at preclinical stages in vivo. Comparison of the in vivo imaging signal from wild-type glioma cells in a null background with the signal in a wild-type host tissue, where the tumour induces the expected TSPO expression in the host’s glial cells, illustrates the substantial extent of the peritumoral host response to the growing tumour. The syngeneic tumour (TSPO+/+) in null background (TSPO−/−) model is thus well suited to study the interaction of the tumour front with the peri-tumoral tissue, and the experimental evaluation of new therapeutic approaches targeting the invasive behaviour of glioblastoma.
Collapse
Affiliation(s)
- Richard B Banati
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia. .,Medical Imaging, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia.
| | - Paul Wilcox
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Ran Xu
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Grace Yin
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Emily Si
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Eric Taeyoung Son
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Mauricio Shimizu
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - R M Damian Holsinger
- Molecular Neuroscience, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Arvind Parmar
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia
| | - David Zahra
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia
| | - Andrew Arthur
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia
| | - Ryan J Middleton
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia
| | - Guo-Jun Liu
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia.,Medical Imaging, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia
| | - Arnaud Charil
- Australian Nuclear Science and Technology Organization, Locked Bag 2001, Kirrawee DC, NSW, 2232, Australia
| | - Manuel B Graeber
- Brain Tumour Research, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett Street, Camperdown, NSW, 2050, Australia.
| |
Collapse
|
54
|
Belykh E, Shaffer KV, Lin C, Byvaltsev VA, Preul MC, Chen L. Blood-Brain Barrier, Blood-Brain Tumor Barrier, and Fluorescence-Guided Neurosurgical Oncology: Delivering Optical Labels to Brain Tumors. Front Oncol 2020; 10:739. [PMID: 32582530 PMCID: PMC7290051 DOI: 10.3389/fonc.2020.00739] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 04/17/2020] [Indexed: 12/17/2022] Open
Abstract
Recent advances in maximum safe glioma resection have included the introduction of a host of visualization techniques to complement intraoperative white-light imaging of tumors. However, barriers to the effective use of these techniques within the central nervous system remain. In the healthy brain, the blood-brain barrier ensures the stability of the sensitive internal environment of the brain by protecting the active functions of the central nervous system and preventing the invasion of microorganisms and toxins. Brain tumors, however, often cause degradation and dysfunction of this barrier, resulting in a heterogeneous increase in vascular permeability throughout the tumor mass and outside it. Thus, the characteristics of both the blood-brain and blood-brain tumor barriers hinder the vascular delivery of a variety of therapeutic substances to brain tumors. Recent developments in fluorescent visualization of brain tumors offer improvements in the extent of maximal safe resection, but many of these fluorescent agents must reach the tumor via the vasculature. As a result, these fluorescence-guided resection techniques are often limited by the extent of vascular permeability in tumor regions and by the failure to stain the full volume of tumor tissue. In this review, we describe the structure and function of both the blood-brain and blood-brain tumor barriers in the context of the current state of fluorescence-guided imaging of brain tumors. We discuss features of currently used techniques for fluorescence-guided brain tumor resection, with an emphasis on their interactions with the blood-brain and blood-tumor barriers. Finally, we discuss a selection of novel preclinical techniques that have the potential to enhance the delivery of therapeutics to brain tumors in spite of the barrier properties of the brain.
Collapse
Affiliation(s)
- Evgenii Belykh
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Kurt V. Shaffer
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Chaoqun Lin
- Department of Neurosurgery, School of Medicine, Southeast University, Nanjing, China
| | - Vadim A. Byvaltsev
- Department of Neurosurgery, Irkutsk State Medical University, Irkutsk, Russia
| | - Mark C. Preul
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Lukui Chen
- Department of Neurosurgery, Neuroscience Center, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
55
|
Role of 68Ga-Prostate-Specific Membrane Antigen PET/CT in Disease Assessment in Glioblastoma Within 48 Hours of Surgery. Clin Nucl Med 2020; 45:204-205. [PMID: 31977462 DOI: 10.1097/rlu.0000000000002893] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Within 48 hours after surgery, disease assessment in glioblastoma is a challenge for both the clinician and the radiologist. Certain technical and logistical issues prevail in this period. Ga-prostate-specific membrane antigen PET/CT is a known molecular imaging marker in prostate cancer. Its role in high-grade gliomas has been recently discussed. We present a case of a 39-year-old man with recurrence glioblastoma of the right frontal lobe and underwent resurgery. After surgery, Ga-prostate-specific membrane antigen PET/CT showed residual disease along the posterior and inferior margin of the postoperative cavity.
Collapse
|
56
|
Ruff M, Kizilbash S, Buckner J. Further understanding of glioma mechanisms of pathogenesis: implications for therapeutic development. Expert Rev Anticancer Ther 2020; 20:355-363. [PMID: 32301635 DOI: 10.1080/14737140.2020.1757440] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Recent discoveries in the molecular makeup of gliomas, the relationship of certain molecular drivers, and the patient's response to therapy and overall prognosis have resulted in a paradigm shift and redefined our understanding of glioma and revealed potential vulnerabilities within this recalcitrant and lethal disease.Areas covered: We summarize the current classification of malignant glioma in the context of the historical background, current data-driven treatment strategies, and recent discoveries of the mechanisms of pathogenesis of this disease which recapitulates the developing brain. We describe the relationship to common genetic alterations found in glioma, and possible avenues to exploit these newly revealed mechanisms.Expert opinion: Improved understanding of the molecular underpinnings of this disease has been directly translated into treatment decisions and an improved ability to counsel patients regarding their prognosis. We are beginning to see the first glimmer of a return on the investment in regard to immunotherapy in malignant glioma, with further anticipated successful exploitations of the unique pathophysiology of glioma.
Collapse
Affiliation(s)
- Michael Ruff
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.,Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Sani Kizilbash
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Jan Buckner
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
57
|
Rudà R, Angileri FF, Ius T, Silvani A, Sarubbo S, Solari A, Castellano A, Falini A, Pollo B, Del Basso De Caro M, Papagno C, Minniti G, De Paula U, Navarria P, Nicolato A, Salmaggi A, Pace A, Fabi A, Caffo M, Lombardi G, Carapella CM, Spena G, Iacoangeli M, Fontanella M, Germanò AF, Olivi A, Bello L, Esposito V, Skrap M, Soffietti R. Italian consensus and recommendations on diagnosis and treatment of low-grade gliomas. An intersociety (SINch/AINO/SIN) document. J Neurosurg Sci 2020; 64:313-334. [PMID: 32347684 DOI: 10.23736/s0390-5616.20.04982-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In 2018, the SINch (Italian Society of Neurosurgery) Neuro-Oncology Section, AINO (Italian Association of Neuro-Oncology) and SIN (Italian Association of Neurology) Neuro-Oncology Section formed a collaborative Task Force to look at the diagnosis and treatment of low-grade gliomas (LGGs). The Task Force included neurologists, neurosurgeons, neuro-oncologists, pathologists, radiologists, radiation oncologists, medical oncologists, a neuropsychologist and a methodologist. For operational purposes, the Task Force was divided into five Working Groups: diagnosis, surgical treatment, adjuvant treatments, supportive therapies, and follow-up. The resulting guidance document is based on the available evidence and provides recommendations on diagnosis and treatment of LGG patients, considering all aspects of patient care along their disease trajectory.
Collapse
Affiliation(s)
- Roberta Rudà
- Department of Neuro-Oncology, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Filippo F Angileri
- Section of Neurosurgery, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy -
| | - Tamara Ius
- Neurosurgery Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Antonio Silvani
- Department of Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Silvio Sarubbo
- Department of Neurosurgery, Structural and Functional Connectivity Lab Project, "S. Chiara" Hospital, Trento, Italy
| | - Alessandra Solari
- Unit of Neuroepidemiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Antonella Castellano
- Neuroradiology Unit, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milan, Italy
| | - Andrea Falini
- Neuroradiology Unit, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milan, Italy
| | - Bianca Pollo
- Section of Oncologic Neuropathology, Division of Neurology V - Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | - Costanza Papagno
- Center of Neurocognitive Rehabilitation (CeRiN), Interdepartmental Center of Mind/Brain, University of Trento, Trento, Italy.,Department of Psychology, University of Milan-Bicocca, Milan, Italy
| | - Giuseppe Minniti
- Radiation Oncology Unit, Department of Medicine, Surgery and Neurosciences, Policlinico Le Scotte, University of Siena, Siena, Italy
| | - Ugo De Paula
- Unit of Radiotherapy, San Giovanni-Addolorata Hospital, Rome, Italy
| | - Pierina Navarria
- Department of Radiotherapy and Radiosurgery, Humanitas Cancer Center and Research Hospital, Rozzano, Milan, Italy
| | - Antonio Nicolato
- Unit of Stereotaxic Neurosurgery, Department of Neurosciences, Hospital Trust of Verona, Verona, Italy
| | - Andrea Salmaggi
- Neurology Unit, Department of Neurosciences, A. Manzoni Hospital, Lecco, Italy
| | - Andrea Pace
- IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Alessandra Fabi
- Division of Medical Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Maria Caffo
- Section of Neurosurgery, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Giuseppe Lombardi
- Unit of Oncology 1, Department of Oncology, Veneto Institute of Oncology-IRCCS, Padua, Italy
| | | | - Giannantonio Spena
- Neurosurgery Unit, Department of Neurosciences, A. Manzoni Hospital, Lecco, Italy
| | - Maurizio Iacoangeli
- Department of Neurosurgery, Marche Polytechnic University, Umberto I General University Hospital, Ancona, Italy
| | - Marco Fontanella
- Division of Neurosurgery, Department of Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Antonino F Germanò
- Section of Neurosurgery, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Alessandro Olivi
- Neurosurgery Unit, Department of Neurosciences, Università Cattolica del Sacro Cuore, Fondazione Policlinico "A. Gemelli", Rome, Italy
| | - Lorenzo Bello
- Unit of Oncologic Neurosurgery, Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Vincenzo Esposito
- Sapienza University, Rome, Italy.,Giampaolo Cantore Department of Neurosurgery, IRCCS Neuromed, Pozzilli, Isernia, Italy
| | - Miran Skrap
- Neurosurgery Unit, Department of Neurosciences, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Riccardo Soffietti
- Department of Neuro-Oncology, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | | |
Collapse
|
58
|
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
|
59
|
Unterrainer M, Eze C, Ilhan H, Marschner S, Roengvoraphoj O, Schmidt-Hegemann NS, Walter F, Kunz WG, Rosenschöld PMA, Jeraj R, Albert NL, Grosu AL, Niyazi M, Bartenstein P, Belka C. Recent advances of PET imaging in clinical radiation oncology. Radiat Oncol 2020; 15:88. [PMID: 32317029 PMCID: PMC7171749 DOI: 10.1186/s13014-020-01519-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/19/2020] [Indexed: 12/25/2022] Open
Abstract
Radiotherapy and radiation oncology play a key role in the clinical management of patients suffering from oncological diseases. In clinical routine, anatomic imaging such as contrast-enhanced CT and MRI are widely available and are usually used to improve the target volume delineation for subsequent radiotherapy. Moreover, these modalities are also used for treatment monitoring after radiotherapy. However, some diagnostic questions cannot be sufficiently addressed by the mere use standard morphological imaging. Therefore, positron emission tomography (PET) imaging gains increasing clinical significance in the management of oncological patients undergoing radiotherapy, as PET allows the visualization and quantification of tumoral features on a molecular level beyond the mere morphological extent shown by conventional imaging, such as tumor metabolism or receptor expression. The tumor metabolism or receptor expression information derived from PET can be used as tool for visualization of tumor extent, for assessing response during and after therapy, for prediction of patterns of failure and for definition of the volume in need of dose-escalation. This review focuses on recent and current advances of PET imaging within the field of clinical radiotherapy / radiation oncology in several oncological entities (neuro-oncology, head & neck cancer, lung cancer, gastrointestinal tumors and prostate cancer) with particular emphasis on radiotherapy planning, response assessment after radiotherapy and prognostication.
Collapse
Affiliation(s)
- M Unterrainer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany. .,Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany. .,German Cancer Consortium (DKTK), partner site Munich; and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - C Eze
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - H Ilhan
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - S Marschner
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - O Roengvoraphoj
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - N S Schmidt-Hegemann
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - F Walter
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - W G Kunz
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - P Munck Af Rosenschöld
- Radiation Physics, Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, and Lund University, Lund, Sweden
| | - R Jeraj
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, USA
| | - N L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK), partner site Munich; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - A L Grosu
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), partner Site Freiburg, Freiburg, Germany
| | - M Niyazi
- German Cancer Consortium (DKTK), partner site Munich; and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - P Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK), partner site Munich; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - C Belka
- German Cancer Consortium (DKTK), partner site Munich; and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
60
|
Fuenfgeld B, Mächler P, Fischer DR, Esposito G, Rushing EJ, Kaufmann PA, Stolzmann P, Huellner MW. Reference values of physiological 18F-FET uptake: Implications for brain tumor discrimination. PLoS One 2020; 15:e0230618. [PMID: 32302317 PMCID: PMC7164612 DOI: 10.1371/journal.pone.0230618] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 02/28/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose The aim of this study was to derive reference values of 18F-fluoro-ethyl-L-tyrosine positron emission tomography (18F-FET-PET) uptake in normal brain and head structures to allow for differentiation from tumor tissue. Materials and methods We examined the datasets of 70 patients (median age 53 years, range 15–79), whose dynamic 18F-FET-PET was acquired between January 2016 and October 2017. Maximum standardized uptake value (SUVmax), target-to-background standardized uptake value ratio (TBR), and time activity curve (TAC) of the 18F-FET-PET were assessed in tumor tissue and in eight normal anatomic structures and compared using the t-test and Mann-Whitney U-test. Correlation analyses were performed using Pearson or Spearman coefficients, and comparisons between several variables with Pearson’s chi-squared tests and Kruskal-Wallis tests as well as the Benjamini-Hochberg correction. Results All analyzed structures showed an 18F-FET uptake higher than background (threshold: TBR > 1.5). The venous sinuses and cranial muscles exhibited a TBR of 2.03±0.46 (confidence interval (CI) 1.92–2.14), higher than the uptake of caudate nucleus, pineal gland, putamen, and thalamus (TBR 1.42±0.17, CI 1.38–1.47). SUVmax, TBR, and TAC showed no difference in the analyzed structures between subjects with high-grade gliomas and subjects with low-grade gliomas, except the SUVmax of the pineal gland (t-tests of the pineal gland: SUVmax: p = 0.022; TBR: p = 0.411). No significant differences were found for gender and age. Conclusion Normal brain tissue demonstrates increased 18F-FET uptake compared to background tissue. Two distinct clusters have been identified, comprising venous structures and gray matter with a reference uptake of up to SUVmax of 2.99 and 2.33, respectively.
Collapse
Affiliation(s)
- Brigitte Fuenfgeld
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Philipp Mächler
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Dorothee R. Fischer
- Department of Radiology and Nuclear Medicine, Hospital St. Anna, Lucerne, Switzerland
| | - Giuseppe Esposito
- Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | - Elisabeth Jane Rushing
- University of Zurich, Zurich, Switzerland
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Philipp A. Kaufmann
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Paul Stolzmann
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Martin W. Huellner
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
61
|
Villena Martín M, Pena Pardo FJ, Jiménez Aragón F, Borras Moreno JM, García Vicente AM. Metabolic targeting can improve the efficiency of brain tumor biopsies. Semin Oncol 2020; 47:148-154. [DOI: 10.1053/j.seminoncol.2020.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 12/27/2022]
|
62
|
Rich LJ, Bagga P, Wilson NE, Schnall MD, Detre JA, Haris M, Reddy R. 1H magnetic resonance spectroscopy of 2H-to- 1H exchange quantifies the dynamics of cellular metabolism in vivo. Nat Biomed Eng 2020; 4:335-342. [PMID: 31988460 PMCID: PMC7071956 DOI: 10.1038/s41551-019-0499-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/27/2019] [Indexed: 11/09/2022]
Abstract
The quantitative mapping of the in vivo dynamics of cellular metabolism via non-invasive imaging contributes to the understanding of the initiation and progression of diseases associated with dysregulated metabolic processes. Current methods for imaging cellular metabolism are limited by low sensitivities, by costs, or by the use of specialized hardware. Here, we introduce a method that captures the turnover of cellular metabolites by quantifying signal reductions in proton magnetic resonance spectroscopy (MRS) resulting from the replacement of 1H with 2H. The method, which we termed quantitative exchanged-label turnover MRS, only requires deuterium-labelled glucose and standard MRI scanners, and with a single acquisition provides steady-state information and metabolic rates for several metabolites. We used the method to monitor glutamate, glutamine, γ-aminobutyric acid and lactate in the brains of normal and glioma-bearing rats following the administration of 2H2-labelled glucose and 2H3-labelled acetate. Quantitative exchanged-label turnover MRS should broaden the applications of routine 1H MRS.
Collapse
Affiliation(s)
- Laurie J Rich
- Center for Magnetic Resonance and Optical Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Puneet Bagga
- Center for Magnetic Resonance and Optical Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Neil E Wilson
- Center for Magnetic Resonance and Optical Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mitchell D Schnall
- Center for Magnetic Resonance and Optical Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John A Detre
- Center for Magnetic Resonance and Optical Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohammad Haris
- Center for Magnetic Resonance and Optical Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Research Branch, Sidra Medicine, Doha, Qatar.,Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Ravinder Reddy
- Center for Magnetic Resonance and Optical Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
63
|
Bonm AV, Ritterbusch R, Throckmorton P, Graber JJ. Clinical Imaging for Diagnostic Challenges in the Management of Gliomas: A Review. J Neuroimaging 2020; 30:139-145. [PMID: 31925884 DOI: 10.1111/jon.12687] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/03/2020] [Accepted: 01/03/2020] [Indexed: 02/06/2023] Open
Abstract
Neuroimaging plays a critical role in the management of patients with gliomas. While conventional magnetic resonance imaging (MRI) remains the standard imaging modality, it is frequently insufficient to inform clinical decision-making. There is a need for noninvasive strategies for reliably distinguishing low-grade from high-grade gliomas, identifying important molecular features of glioma, choosing an appropriate target for biopsy, delineating target area for surgery or radiosurgery, and distinguishing tumor progression (TP) from pseudoprogression (PsP). One recent advance is the identification of the T2/fluid-attenuated inversion recovery mismatch sign on standard MRI to identify isocitrate dehydrogenase mutant astrocytomas. However, to meet other challenges, neuro-oncologists are increasingly turning to advanced imaging modalities. Diffusion-weighted imaging modalities including diffusion tensor imaging and diffusion kurtosis imaging can be helpful in delineating tumor margins and better visualization of tissue architecture. Perfusion imaging including dynamic contrast-enhanced MRI using gadolinium or ferumoxytol contrast agents can be helpful for grading as well as distinguishing TP from PsP. Positron emission tomography is useful for measuring tumor metabolism, which correlates with grade and can distinguish TP/PsP in the right setting. Magnetic resonance spectroscopy can identify tissue by its chemical composition, can distinguish TP/PsP, and can identify molecular features like 2-hydroxyglutarate. Finally, amide proton transfer imaging measures intracellular protein content, which can be used to identify tumor grade/progression and distinguish TP/PsP.
Collapse
Affiliation(s)
- Alipi V Bonm
- Department of Neurology, University of Washington, Seattle, WA
| | | | | | - Jerome J Graber
- Department of Neurology, University of Washington, Seattle, WA.,Departments of Neurology and Neurosurgery, Alvord Brain Tumor Center, University of Washington, Seattle, WA
| |
Collapse
|
64
|
Fleischmann DF, Unterrainer M, Schön R, Corradini S, Maihöfer C, Bartenstein P, Belka C, Albert NL, Niyazi M. Margin reduction in radiotherapy for glioblastoma through 18F-fluoroethyltyrosine PET? - A recurrence pattern analysis. Radiother Oncol 2020; 145:49-55. [PMID: 31923709 DOI: 10.1016/j.radonc.2019.12.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 12/09/2019] [Accepted: 12/11/2019] [Indexed: 11/18/2022]
Abstract
BACKGROUND AND PURPOSE 18F-fluoroethyltyrosine (18F-FET) PET is increasingly used in radiation treatment planning for the primary treatment of glioblastoma (GBM) patients additionally to contrast-enhanced MRI. To answer the question, whether a margin reduction in the primary treatment setting could be achieved through 18F-FET PET imaging, a recurrence pattern analysis was performed. PATIENTS AND METHODS GBM patients undergoing 18F-FET PET examination before primary radiochemotherapy from 05/2009 to 11/2014 were included into the recurrence pattern analysis. Biological tumour volumes were semi-automatically created and fused with MR-based gross tumour volumes (MRGTVs). The pattern of recurrence was examined for MRGTVs and for PET-MRGTVs. The minimal margin including all recurrent tumour sites was assessed by gradual expansion of the PET-MRGTVs and MRGTVs until inclusion of all contrast-enhancing areas at recurrence. RESULTS 36 GBM patients were included to the analysis. The minimal margin including all contrast enhancing tumour at recurrence was significantly smaller for the PET-MRGTVs compared to the MRGTVs (median 12.5 mm vs. 16.5 mm; p < 0.001, Wilcoxon-Test). PET-MRGTVs with 15 mm CTV margins were significantly smaller than MRGTVs with 20 mm CTV margins (median volume 255.92 vs. 258.35 cm3; p = 0.020, Wilcoxon-Test; excluding 3 cases with large non-contrast enhancing tumours). The pattern of recurrence of PET-MRGTVs with 15 mm CTV margins was comparable to MRGTVs with 20 mm CTV margins (32 vs. 30 central, 2 vs. 4 in-field, 2 vs. 2 ex-field and no marginal recurrences). CONCLUSION Target volume delineation of GBM patients can be improved through 18F-FET PET imaging prior to primary radiation treatment, since vital tumour can be detected more accurately. Furthermore, the results suggest that CTV margins could be reduced through 18F-FET PET imaging prior to primary RT of GBM.
Collapse
Affiliation(s)
- Daniel F Fleischmann
- Department of Radiation Oncology, University Hospital, LMU Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Marcus Unterrainer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Germany.
| | - Rudolph Schön
- Department of Radiation Oncology, University Hospital, LMU Munich, Germany.
| | - Stefanie Corradini
- Department of Radiation Oncology, University Hospital, LMU Munich, Germany.
| | - Cornelius Maihöfer
- Department of Radiation Oncology, University Hospital, LMU Munich, Germany.
| | - Peter Bartenstein
- German Cancer Consortium (DKTK), Partner Site Munich, Germany; Department of Nuclear Medicine, University Hospital, LMU Munich, Germany.
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, Germany.
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Germany.
| | - Maximilian Niyazi
- Department of Radiation Oncology, University Hospital, LMU Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, Germany.
| |
Collapse
|
65
|
Leao DJ, Craig PG, Godoy LF, Leite CC, Policeni B. Response Assessment in Neuro-Oncology Criteria for Gliomas: Practical Approach Using Conventional and Advanced Techniques. AJNR Am J Neuroradiol 2020; 41:10-20. [PMID: 31857322 PMCID: PMC6975322 DOI: 10.3174/ajnr.a6358] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/29/2019] [Indexed: 01/08/2023]
Abstract
The Response Assessment in Neuro-Oncology criteria were developed as an objective tool for radiologic assessment of treatment response in high-grade gliomas. Imaging plays a critical role in the management of the patient with glioma, from initial diagnosis to posttreatment follow-up, which can be particularly challenging for radiologists. Interpreting findings after surgery, radiation, and chemotherapy requires profound knowledge about the tumor biology, as well as the peculiar changes expected to ensue as a consequence of each treatment technique. In this article, we discuss the imaging findings associated with tumor progression, tumor response, pseudoprogression, and pseudoresponse according to the Response Assessment in Neuro-Oncology criteria for high-grade and lower-grade gliomas. We describe relevant practical issues when evaluating patients with glioma, such as the need for imaging in the first 48 hours, the radiation therapy planning and isodose curves, the significance of T2/FLAIR hyperintense lesions, the impact of the timing for the evaluation after radiation therapy, and the definition of progressive disease on the histologic specimen. We also illustrate the correlation among the findings on conventional MR imaging with advanced techniques, such as perfusion, diffusion-weighted imaging, spectroscopy, and amino acid PET. Because many of the new lesions represent a mixture of tumor cells and tissue with radiation injury, the radiologist aims to identify the predominant component of the lesion and categorize the findings according to Response Assessment in Neuro-Oncology criteria so that the patient can receive the best treatment.
Collapse
Affiliation(s)
- D J Leao
- From the Cancer Hospital of Federal University of Uberlandia (D.J.L.), Uberlandia, Brazil
| | - P G Craig
- Department of Radiology, (P.G.C., B.P.), University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - L F Godoy
- Department of Diagnostic Radiology (L.F.G.), Hospital Sirio-Libanes, Sao Paulo, Brazil
- Department of Neuroradiology (L.F.G., C.C.L.), Faculdade de Medicina Instituto de Radiologia, Universidade de Sao Paulo Neuroradiology, Sao Paulo, Brazil
| | - C C Leite
- Department of Neuroradiology (L.F.G., C.C.L.), Faculdade de Medicina Instituto de Radiologia, Universidade de Sao Paulo Neuroradiology, Sao Paulo, Brazil
| | - B Policeni
- Department of Radiology, (P.G.C., B.P.), University of Iowa Hospitals and Clinics, Iowa City, Iowa
| |
Collapse
|
66
|
Bi J, Chowdhry S, Wu S, Zhang W, Masui K, Mischel PS. Altered cellular metabolism in gliomas - an emerging landscape of actionable co-dependency targets. Nat Rev Cancer 2020; 20:57-70. [PMID: 31806884 DOI: 10.1038/s41568-019-0226-5] [Citation(s) in RCA: 177] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/31/2019] [Indexed: 12/18/2022]
Abstract
Altered cellular metabolism is a hallmark of gliomas. Propelled by a set of recent technological advances, new insights into the molecular mechanisms underlying glioma metabolism are rapidly emerging. In this Review, we focus on the dynamic nature of glioma metabolism and how it is shaped by the interaction between tumour genotype and brain microenvironment. Recent advances integrating metabolomics with genomics are discussed, yielding new insight into the mechanisms that drive glioma pathogenesis. Studies that shed light on interactions between the tumour microenvironment and tumour genotype are highlighted, providing important clues as to how gliomas respond to and adapt to their changing tissue and biochemical contexts. Finally, a road map for the discovery of potential new glioma drug targets is suggested, with the goal of translating these new insights about glioma metabolism into clinical benefits for patients.
Collapse
Affiliation(s)
- Junfeng Bi
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Sudhir Chowdhry
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Sihan Wu
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Wenjing Zhang
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA
| | - Kenta Masui
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Paul S Mischel
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, USA.
- Department of Pathology, UCSD School of Medicine, La Jolla, CA, USA.
- Moores Cancer Center, UCSD School of Medicine, La Jolla, CA, USA.
| |
Collapse
|
67
|
Dissaux G, Basse V, Schick U, EL Kabbaj O, Auberger B, Magro E, Kassoul A, Abgral R, Salaun PY, Bourhis D, Querellou S. Prognostic value of 18F-FET PET/CT in newly diagnosed WHO 2016 high-grade glioma. Medicine (Baltimore) 2020; 99:e19017. [PMID: 32000446 PMCID: PMC7004648 DOI: 10.1097/md.0000000000019017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
O-(2-[F]fluoroethyl)-L-tyrosine positron-emission tomography/computed tomography (F-FET PET/CT) is well known in brain tumor management. Our study aimed to identify the prognostic value of F-FET PET/CT in high-grade gliomas (HGG) according the current 2016 World Health Organization (WHO) classification.Patients with histologically proven WHO 2016 HGG were prospectively included. A dynamic F-FET PET/CT was performed allowing to obtain 2 static PET frames (static frame 1: 20-40 minutes and static frame 2: 2-22 minutes). We analyzed static parameters (standard uptake value [SUV]max, SUVmean, SUVpeak, TBRmax, TBRmean, tumoral lesion glycolysis, and metabolic tumoral volume) for various isocontours (from 10% to 90%). PET parameters, clinical features, and molecular biomarkers were compared with progression-free survival (PFS) and overall survival (OS) in univariate and multivariate analysis.Twenty-nine patients were included (grade III n = 3, grade IV n = 26). Mean PFS and OS were, respectively, 8.8 and 13.9 months. According to univariate analysis, SUVmean, SUVpeak, TBRmax, and TBRmean were significantly correlated with OS. In static 1 analysis, TBRmax seemed to be the best OS prognostic parameter (P = .004). In static 2 analysis, TBRmean was the best parameter (P = .01). In static 1 analysis, only SUVpeak was significant (P = .05) for PFS. Good performance status (PS < 2; P < .0001) and extent of resection (P = .019) identified the subgroup of patients with the best OS. Only TBRmax (P = .026) and extent of resection (P = .025) remained significant parameters in multivariate analysis.Our data suggested that high TBRmax seemed to be the most significant OS independent prognostic factor in patients with newly diagnosed HGG.
Collapse
Affiliation(s)
| | - Victor Basse
- Oncology Department, University Hospital Morvan, Brest Cedex
| | | | | | | | - Elsa Magro
- Neurosurgery Department University Hospital Cavale Blanche
| | - Aboubakr Kassoul
- Nuclear Medicine Department, University Hospitam Morvan, Brest cedex
| | - Ronan Abgral
- Nuclear Medicine Department, University Hospitam Morvan, Brest cedex
- EA 3878 GETBO IFR 148
- University of Bretagne Occidentale, Brest, France
| | - Pierre-Yves Salaun
- Nuclear Medicine Department, University Hospitam Morvan, Brest cedex
- EA 3878 GETBO IFR 148
- University of Bretagne Occidentale, Brest, France
| | - David Bourhis
- Nuclear Medicine Department, University Hospitam Morvan, Brest cedex
- EA 3878 GETBO IFR 148
- University of Bretagne Occidentale, Brest, France
| | - Solène Querellou
- Nuclear Medicine Department, University Hospitam Morvan, Brest cedex
- EA 3878 GETBO IFR 148
- University of Bretagne Occidentale, Brest, France
| |
Collapse
|
68
|
Histogram analysis of 11C-methionine integrated PET/MRI may facilitate to determine the O6-methylguanylmethyltransferase methylation status in gliomas. Nucl Med Commun 2019; 40:850-856. [PMID: 31135693 DOI: 10.1097/mnm.0000000000001039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE We evaluate the O6-methylguanylmethyltransferase (MGMT) methylation status noninvasively by analyzing radiomics features of C-methionine (MET) PET images, which may reflect the detailed biological properties of gliomas. PATIENTS AND METHODS Fifty-seven patients with histopathologically confirmed gliomas, who were initially examined with C-MET PET/MR were retrospectively enrolled. Quantitative uptake of MET was assessed using conventional, histogram and texture features. These features were compared between the two groups classified by MGMT promoter methylation status. RESULTS The histogram features (Skewness and Kurtosis) of the MGMT methylated group were significantly higher than those of the MGMT unmethylated group (Skewness: 0.90 ± 0.71 vs. 0.49 ± 0.45; P = 0.01) (Kurtosis: 1.36 ± 2.30 vs. 0.08 ± 0.65; P = 0.003), but there were no significant differences in Skewness or Kurtosis between the groups in glioma-grade-matched subgroup analysis. Moreover, there was no significant difference in other features between the methylated group and unmethylated group. CONCLUSION The histogram features (Skewness and Kurtosis) of MET PET/MRI may be two key indicators to detect MGMT methylation status in gliomas and valuable predictors for the clinical responses of patients scheduled to receive temozolomide chemotherapeutics.
Collapse
|
69
|
Ebrahimpour-Koujan S, Shayanfar M, Benisi-Kohansal S, Mohammad-Shirazi M, Sharifi G, Esmaillzadeh A. Adherence to low carbohydrate diet in relation to glioma: A case-control study. Clin Nutr 2019; 38:2690-2695. [DOI: 10.1016/j.clnu.2018.11.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/24/2018] [Accepted: 11/15/2018] [Indexed: 01/04/2023]
|
70
|
Nosrati S, Molavipordanjani S, Emami S, Abedi SM, Talebpour Amiri F, Hosseinimehr SJ. 99mTc-radiolabeled imidazo[2,1-b]benzothiazole derivatives as potential radiotracers for glioblastoma. J Radioanal Nucl Chem 2019. [DOI: 10.1007/s10967-019-06945-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
71
|
Targeting MMP-14 for dual PET and fluorescence imaging of glioma in preclinical models. Eur J Nucl Med Mol Imaging 2019; 47:1412-1426. [PMID: 31773232 DOI: 10.1007/s00259-019-04607-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/07/2019] [Indexed: 02/08/2023]
Abstract
PURPOSE There is a clinical need for agents that target glioma cells for non-invasive and intraoperative imaging to guide therapeutic intervention and improve the prognosis of glioma. Matrix metalloproteinase (MMP)-14 is overexpressed in glioma with negligible expression in normal brain, presenting MMP-14 as an attractive biomarker for imaging glioma. In this study, we designed a peptide probe containing a near-infrared fluorescence (NIRF) dye/quencher pair, a positron emission tomography (PET) radionuclide, and a moiety with high affinity to MMP-14. This novel substrate-binding peptide allows dual modality imaging of glioma only after cleavage by MMP-14 to activate the quenched NIRF signal, enhancing probe specificity and imaging contrast. METHODS MMP-14 expression and activity in human glioma tissues and cells were measured in vitro by immunofluorescence and gel zymography. Cleavage of the novel substrate and substrate-binding peptides by glioma cells in vitro and glioma xenograft tumors in vivo was determined by NIRF imaging. Biodistribution of the radiolabeled MMP-14-binding peptide or substrate-binding peptide was determined in mice bearing orthotopic patient-derived xenograft (PDX) glioma tumors by PET imaging. RESULTS Glioma cells with MMP-14 activity showed activation and retention of NIRF signal from the cleaved peptides. Resected mouse brains with PDX glioma tumors showed tumor-to-background NIRF ratios of 7.6-11.1 at 4 h after i.v. injection of the peptides. PET/CT images showed localization of activity in orthotopic PDX tumors after i.v. injection of 68Ga-binding peptide or 64Cu-substrate-binding peptide; uptake of the radiolabeled peptides in tumors was significantly reduced (p < 0.05) by blocking with the non-labeled-binding peptide. PET and NIRF signals correlated linearly in the orthotopic PDX tumors. Immunohistochemistry showed co-localization of MMP-14 expression and NIRF signal in the resected tumors. CONCLUSIONS The novel MMP-14 substrate-binding peptide enabled PET/NIRF imaging of glioma models in mice, warranting future image-guided resection studies with the probe in preclinical glioma models.
Collapse
|
72
|
Puranik AD, Boon M, Purandare N, Rangarajan V, Gupta T, Moiyadi A, Shetty P, Sridhar E, Agrawal A, Dev I, Shah S. Utility of FET-PET in detecting high-grade gliomas presenting with equivocal MR imaging features. World J Nucl Med 2019; 18:266-272. [PMID: 31516370 PMCID: PMC6714153 DOI: 10.4103/wjnm.wjnm_89_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
High-grade gliomas, metastases, and primary central nervous system lymphoma (PCNSL) are common high-grade brain lesions, which may have overlapping features on magnetic resonance (MR) imaging. Our objective was to assess the utility of 18-fluoride-fluoro-ethyl-tyrosine positron emission tomography (FET-PET) in reliably differentiating between these lesions, by studying their metabolic characteristics. Patients with high-grade brain lesions suspicious for glioma, with overlapping features for metastases and PCNSL were referred for FET-PET by Neuroradiologists from Multidisciplinary Neuro-Oncology Joint Clinic. Tumor-to-contralateral white mater ratio (T/Wm) at 5 and 20 min was derived and compared to histopathology. Receiver operating characteristic curve analysis was used to find the optimal T/Wm cutoff to differentiate between the tumor types. T/Wm was higher for glial tumors compared to nonglial tumors (metastases, PCNSL, tuberculoma, and anaplastic meningioma). A cutoff of 1.9 was derived to reliably diagnose a tumor of glial origin with a sensitivity and specificity of 93.8% and 91%, respectively. FET-PET can be used to diagnose glial tumors presenting as high-grade brain lesions when MR findings show overlapping features for other common high-grade lesions.
Collapse
Affiliation(s)
- Ameya D Puranik
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Homi Bhabha National University, Mumbai, Maharashtra, India
| | - Mathew Boon
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Homi Bhabha National University, Mumbai, Maharashtra, India
| | - Nilendu Purandare
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Homi Bhabha National University, Mumbai, Maharashtra, India
| | - Venkatesh Rangarajan
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Homi Bhabha National University, Mumbai, Maharashtra, India
| | - Tejpal Gupta
- Department of Radiation Oncology, Tata Memorial Hospital, Homi Bhabha National University, Mumbai, Maharashtra, India
| | - Aliasgar Moiyadi
- Department of Neurosurgery, Tata Memorial Hospital, Homi Bhabha National University, Mumbai, Maharashtra, India
| | - Prakash Shetty
- Department of Neurosurgery, Tata Memorial Hospital, Homi Bhabha National University, Mumbai, Maharashtra, India
| | - Epari Sridhar
- Department of Pathology, Tata Memorial Hospital, Homi Bhabha National University, Mumbai, Maharashtra, India
| | - Archi Agrawal
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Homi Bhabha National University, Mumbai, Maharashtra, India
| | - Indraja Dev
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Homi Bhabha National University, Mumbai, Maharashtra, India
| | - Sneha Shah
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital, Homi Bhabha National University, Mumbai, Maharashtra, India
| |
Collapse
|
73
|
Maurer GD, Brucker DP, Stoffels G, Filipski K, Filss CP, Mottaghy FM, Galldiks N, Steinbach JP, Hattingen E, Langen KJ. 18F-FET PET Imaging in Differentiating Glioma Progression from Treatment-Related Changes: A Single-Center Experience. J Nucl Med 2019; 61:505-511. [PMID: 31519802 DOI: 10.2967/jnumed.119.234757] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 09/06/2019] [Indexed: 11/16/2022] Open
Abstract
In glioma patients, differentiation between tumor progression (TP) and treatment-related changes (TRCs) remains challenging. Difficulties in classifying imaging alterations may result in a delay or an unnecessary discontinuation of treatment. PET using O-(2-18F-fluoroethyl)-l-tyrosine (18F-FET) has been shown to be a useful tool for detecting TP and TRCs. Methods: We retrospectively evaluated 127 consecutive patients with World Health Organization grade II-IV glioma who underwent 18F-FET PET imaging to distinguish between TP and TRCs. 18F-FET PET findings were verified by neuropathology (40 patients) or clinicoradiologic follow-up (87 patients). Maximum tumor-to-brain ratios (TBRmax) of 18F-FET uptake and the slope of the time-activity curves (20-50 min after injection) were determined. The diagnostic accuracy of 18F-FET PET parameters was evaluated by receiver-operating-characteristic analysis and χ2 testing. The prognostic value of 18F-FET PET was estimated using the Kaplan-Meier method. Results: TP was diagnosed in 94 patients (74%) and TRCs in 33 (26%). For differentiating TP from TRCs, receiver-operating-characteristic analysis yielded an optimal 18F-FET TBRmax cutoff of 1.95 (sensitivity, 70%; specificity, 71%; accuracy, 70%; area under the curve, 0.75 ± 0.05). The highest accuracy was achieved by a combination of TBRmax and slope (sensitivity, 86%; specificity, 67%; accuracy, 81%). However, accuracy was poorer when tumors harbored isocitrate dehydrogenase (IDH) mutations (91% in IDH-wild-type tumors, 67% in IDH-mutant tumors, P < 0.001). 18F-FET PET results correlated with overall survival (P < 0.001). Conclusion: In our neurooncology department, the diagnostic performance of 18F-FET PET was convincing but slightly inferior to that of previous reports.
Collapse
Affiliation(s)
- Gabriele D Maurer
- Dr. Senckenberg Institute of Neurooncology, Goethe University Hospital, Frankfurt am Main, Germany .,University Cancer Center Frankfurt, Goethe University Hospital, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Heidelberg, Germany
| | - Daniel P Brucker
- University Cancer Center Frankfurt, Goethe University Hospital, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Heidelberg, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-3 and INM-4), Research Center Juelich, Juelich, Germany
| | - Katharina Filipski
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Neurology (Edinger Institute), Goethe University Hospital, Frankfurt am Main, Germany
| | - Christian P Filss
- Institute of Neuroscience and Medicine (INM-3 and INM-4), Research Center Juelich, Juelich, Germany.,Department of Nuclear Medicine, RWTH University Hospital, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, RWTH University Hospital, Aachen, Germany.,Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands.,Center of Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3 and INM-4), Research Center Juelich, Juelich, Germany.,Center of Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany.,Department of Neurology, University Hospital Cologne, Cologne, Germany; and
| | - Joachim P Steinbach
- Dr. Senckenberg Institute of Neurooncology, Goethe University Hospital, Frankfurt am Main, Germany.,University Cancer Center Frankfurt, Goethe University Hospital, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Heidelberg, Germany
| | - Elke Hattingen
- Institute of Neuroradiology, Goethe University Hospital, Frankfurt am Main, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3 and INM-4), Research Center Juelich, Juelich, Germany.,Department of Nuclear Medicine, RWTH University Hospital, Aachen, Germany.,Center of Integrated Oncology, Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany
| |
Collapse
|
74
|
Abstract
PURPOSE This pilot study aimed to evaluate the amino acid tracer F-FACBC with simultaneous PET/MRI in diagnostic assessment and neurosurgery of gliomas. MATERIALS AND METHODS Eleven patients with suspected primary or recurrent low- or high-grade glioma received an F-FACBC PET/MRI examination before surgery. PET and MRI were used for diagnostic assessment, and for guiding tumor resection and histopathological tissue sampling. PET uptake, tumor-to-background ratios (TBRs), time-activity curves, as well as PET and MRI tumor volumes were evaluated. The sensitivities of lesion detection and to detect glioma tissue were calculated for PET, MRI, and combined PET/MRI with histopathology (biopsies for final diagnosis and additional image-localized biopsies) as reference. RESULTS Overall sensitivity for lesion detection was 54.5% (95% confidence interval [CI], 23.4-83.3) for PET, 45.5% (95% CI, 16.7-76.6) for contrast-enhanced MRI (MRICE), and 100% (95% CI, 71.5-100.0) for combined PET/MRI, with a significant difference between MRICE and combined PET/MRI (P = 0.031). TBRs increased with tumor grade (P = 0.004) and were stable from 10 minutes post injection. PET tumor volumes enclosed most of the MRICE volumes (>98%) and were generally larger (1.5-2.8 times) than the MRICE volumes. Based on image-localized biopsies, combined PET/MRI demonstrated higher concurrence with malignant findings at histopathology (89.5%) than MRICE (26.3%). CONCLUSIONS Low- versus high-grade glioma differentiation may be possible with F-FACBC using TBR. F-FACBC PET/MRI outperformed MRICE in lesion detection and in detection of glioma tissue. More research is required to evaluate F-FACBC properties, especially in grade II and III tumors, and for different subtypes of gliomas.
Collapse
|
75
|
Xiao J, Jin Y, Nie J, Chen F, Ma X. Diagnostic and grading accuracy of 18F-FDOPA PET and PET/CT in patients with gliomas: a systematic review and meta-analysis. BMC Cancer 2019; 19:767. [PMID: 31382920 PMCID: PMC6683403 DOI: 10.1186/s12885-019-5938-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 07/15/2019] [Indexed: 02/05/2023] Open
Abstract
Background Positron emission tomography (PET) and PET/computed tomography (PET/CT) imaging with 3,4-dihydroxy-6-[18F] fluoro-L-phenylalanine (18F-FDOPA) has been used in the evaluation of gliomas. We performed a meta-analysis to obtain the diagnostic and grading accuracy of 18F-FDOPA PET and PET/CT in patients with gliomas. Methods PubMed, Embase, Cochrane Library and Web of Science were searched through 13 May 2019. We included studies reporting the diagnostic performance of 18F-FDOPA PET or PET/CT in glioma patients. Pooled sensitivity, specificity, and area under the summary receiver operating characteristic (SROC) curve were calculated from eligible studies on a per-lesion basis. Results Eventually, 19 studies were included. Across 13 studies (370 patients) for glioma diagnosis, the pooled sensitivity and specificity of 18F-FDOPA PET and PET/CT were 0.90 (95%CI: 0.86–0.93) and 0.75 (95%CI: 0.65–0.83). Across 7 studies (219 patients) for glioma grading, 18F-FDOPA PET and PET/CT showed a pooled sensitivity of 0.88 (95%CI: 0.81–0.93) and a pooled specificity of 0.73 (95%CI: 0.64–0.81). Conclusions 18F-FDOPA PET and PET/CT demonstrated good performance for diagnosing gliomas and differentiating high-grade gliomas (HGGs) from low-grade gliomas (LGGs). Further studies implementing standardized PET protocols and investigating the grading parameters are needed.
Collapse
Affiliation(s)
- Jiarui Xiao
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No.37, Guoxue Alley, Chengdu, 610041, Sichuan, China.,Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Yizi Jin
- Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Ji Nie
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No.37, Guoxue Alley, Chengdu, 610041, Sichuan, China.,West China School of Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Fukun Chen
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, Yunnan, China.
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No.37, Guoxue Alley, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
76
|
Kasten BB, Udayakumar N, Leavenworth JW, Wu AM, Lapi SE, McConathy JE, Sorace AG, Bag AK, Markert JM, Warram JM. Current and Future Imaging Methods for Evaluating Response to Immunotherapy in Neuro-Oncology. Theranostics 2019; 9:5085-5104. [PMID: 31410203 PMCID: PMC6691392 DOI: 10.7150/thno.34415] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 04/20/2019] [Indexed: 12/28/2022] Open
Abstract
Imaging plays a central role in evaluating responses to therapy in neuro-oncology patients. The advancing clinical use of immunotherapies has demonstrated that treatment-related inflammatory responses mimic tumor growth via conventional imaging, thus spurring the development of new imaging approaches to adequately distinguish between pseudoprogression and progressive disease. To this end, an increasing number of advanced imaging techniques are being evaluated in preclinical and clinical studies. These novel molecular imaging approaches will serve to complement conventional response assessments during immunotherapy. The goal of these techniques is to provide definitive metrics of tumor response at earlier time points to inform treatment decisions, which has the potential to improve patient outcomes. This review summarizes the available immunotherapy regimens, clinical response criteria, current state-of-the-art imaging approaches, and groundbreaking strategies for future implementation to evaluate the anti-tumor and immune responses to immunotherapy in neuro-oncology applications.
Collapse
Affiliation(s)
- Benjamin B. Kasten
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Neha Udayakumar
- School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jianmei W. Leavenworth
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Anna M. Wu
- Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, United States
| | - Suzanne E. Lapi
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jonathan E. McConathy
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Anna G. Sorace
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Asim K. Bag
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - James M. Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jason M. Warram
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
77
|
The Continuing Evolution of Molecular Functional Imaging in Clinical Oncology: The Road to Precision Medicine and Radiogenomics (Part I). Mol Diagn Ther 2019; 23:1-26. [PMID: 30411216 DOI: 10.1007/s40291-018-0366-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The present era of precision medicine sees 'cancer' as a consequence of molecular derangements occurring at the commencement of the disease process, with morphologic changes happening much later in the process of tumorigenesis. Conventional imaging techniques, such as computed tomography (CT), ultrasound, and magnetic resonance imaging (MRI), play an integral role in the detection of disease at a macroscopic level. However, molecular functional imaging (MFI) techniques entail the visualisation and quantification of biochemical and physiological processes occurring during tumorigenesis, and thus has the potential to play a key role in heralding the transition from the concept of 'one size fits all' to 'precision medicine'. Integration of MFI with other fields of tumour biology such as genomics has spawned a novel concept called 'radiogenomics', which could serve as an indispensable tool in translational cancer research. With recent advances in medical image processing, such as texture analysis, deep learning, and artificial intelligence (AI), the future seems promising; however, their clinical utility remains unproven at present. Despite the emergence of novel imaging biomarkers, a majority of these require validation before clinical translation is possible. In this two-part review, we discuss the systematic collaboration across structural, anatomical, and molecular imaging techniques that constitute MFI. Part I reviews positron emission tomography, radiogenomics, AI, and optical imaging, while part II reviews MRI, CT and ultrasound, their current status, and recent advances in the field of precision oncology.
Collapse
|
78
|
|
79
|
Yang Y, Chen J, Yang Y, Xie Z, Song L, Zhang P, Liu C, Liu J. A 1064 nm excitable semiconducting polymer nanoparticle for photoacoustic imaging of gliomas. NANOSCALE 2019; 11:7754-7760. [PMID: 30949651 DOI: 10.1039/c9nr00552h] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Photoacoustic (PA) imaging in the second near-infrared (NIR-II) window (especially at 1064 nm) has the benefits of low background signal, high spatial resolution and deep tissue penetration. Here we report a semiconducting polymer nanoparticle (PDPPTBZ NP) and demonstrate its potential as a contrast agent for PA imaging of orthotopic brain tumors, using a 1064 nm pulsed laser as a light source. PDPPTBZ NPs have maximum absorption at 1064 nm with a mass extinction coefficient of 43 mL mg-1 cm-1, which is the highest value reported so far in this region. The high photothermal conversion efficiency (67%) and near non-fluorescence impart PDPPTBZ NPs with excellent PA properties. We used PDPPTBZ NP-containing agar gel phantoms even at a low concentration (50 μg mL-1) to successfully image to a depth of 4 cm (of chicken-breast tissue), with an ultralow power fluence (4 mJ cm-2). Furthermore, we could clearly visualize a glioma tumor in a mouse at a depth of 3.8 mm below the skull. This study demonstrates that PDPPTBZ NPs display great potential as a NIR-II PA contrast agent for high quality deep tissue imaging.
Collapse
Affiliation(s)
- Yanqing Yang
- Key Laboratory of Flexible Electronics (KLOFE) Institute of Advanced Materials (IAM), Nanjing Tech University (Nianjing Tech), 30 South Puzhu Road, Nanjing 211800, China.
| | | | | | | | | | | | | | | |
Collapse
|
80
|
Zhang Q, Gao X, Wei G, Qiu C, Qu H, Zhou X. Prognostic Value of MTV, SUVmax and the T/N Ratio of PET/CT in Patients with Glioma: A Systematic Review and Meta-Analysis. J Cancer 2019; 10:1707-1716. [PMID: 31205526 PMCID: PMC6548003 DOI: 10.7150/jca.28605] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 01/13/2019] [Indexed: 12/11/2022] Open
Abstract
Background: In the past decade, positron emission tomography/computed tomography (PET/CT) has become an important imaging tool for clinical assessment of tumor patients. Our meta-analysis aimed to compare the predictive value of PET/CT parameters regard to overall survival (OS) and progression-free survival (PFS) outcomes in glioma. Methods: Relevant articles were systematically searched in PMC, PubMed, EMBASE and WEB of science. Studies involving the prognostic roles of PET/CT parameters with OS and PFS in glioma patients were evaluated. The impact of metabolic tumor volume (MTV), maximal standard uptake value (SUVmax), and the ratio of uptake in tumor to normal (T/N ratio) on survival was measured by calculating combined hazard ratios (HRs) and 95% confidence intervals (CIs). Results: A total of 32 articles with 1715 patients were included. The combined HRs of higher MTV, higher SUVmax and higher T/N ratio for OS were 1.14 (95% CI: 0.98-1.32, P heterogeneity<0.001), 1.69 (95% CI: 1.18-2.41, P heterogeneity<0.001) and 1.68 (95% CI: 1.40-2.01, P heterogeneity< 0.001), respectively. Regarding PFS, the combined HRs were 1.04 (95% CI: 0.97-1.11, P heterogeneity=0.002) with higher MTV, 1.45 (95% CI: 1.11-1.90, P heterogeneity<0.001) with higher SUVmax and 2.07 (95% CI: 1.45-2.95, P heterogeneity<0.001) with higher T/N ratio. Results remained similar in the sub-group analyses. Conclusion: PET/CT parameters T/N ratio may be a significant prognostic factor in patients with glioma. Evidence of SUVmax and MTV needed more large-scale studies performed to validate. PET/CT scan could be a promising technique to provide prognostic information for these patients.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Neurosurgery, Xinghua People's Hospital, Xinghua 225700, Jiangsu, P.R. China
| | - Xian Gao
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Guohua Wei
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Cheng Qiu
- Department of Neurosurgery, Second Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, P.R. China
| | - Hongyi Qu
- Department of Neurosurgery, Xinghua People's Hospital, Xinghua 225700, Jiangsu, P.R. China
| | - Xin Zhou
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
81
|
Thon N, Tonn JC, Kreth FW. The surgical perspective in precision treatment of diffuse gliomas. Onco Targets Ther 2019; 12:1497-1508. [PMID: 30863116 PMCID: PMC6390867 DOI: 10.2147/ott.s174316] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Over the last decade, advances in molecular and imaging-based biomarkers have induced a more versatile diagnostic classification and prognostic evaluation of glioma patients. This, in combination with a growing therapeutic armamentarium, enables increasingly individualized, risk-benefit-optimized treatment strategies. This path to precision medicine in glioma patients requires surgical procedures to be reassessed within multidimensional management considerations. This article attempts to integrate the surgical intervention into a dynamic network of versatile diagnostic characterization, prognostic assessment, and multimodal treatment options in the light of the latest 2016 World Health Organization (WHO) classification of diffuse brain tumors, WHO grade II, III, and IV. Special focus is set on surgical aspects such as resectability, extent of resection, and targeted surgical strategies including minimal invasive stereotactic biopsy procedures, convection enhanced delivery, and photodynamic therapy. Moreover, the influence of recent advances in radiomics/radiogenimics on the process of surgical decision-making will be touched.
Collapse
Affiliation(s)
- Niklas Thon
- Department of Neurosurgery, Ludwig-Maximilians-University Munich, Munich, Germany,
| | - Joerg-Christian Tonn
- Department of Neurosurgery, Ludwig-Maximilians-University Munich, Munich, Germany,
| | | |
Collapse
|
82
|
Chambers G, Frood R, Patel C, Scarsbrook A. 18F-FDG PET-CT in paediatric oncology: established and emerging applications. Br J Radiol 2019; 92:20180584. [PMID: 30383441 PMCID: PMC6404840 DOI: 10.1259/bjr.20180584] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 10/01/2018] [Accepted: 10/27/2018] [Indexed: 12/11/2022] Open
Abstract
Accurate staging and response assessment is vital in the management of childhood malignancies. Fluorine-18 fluorodeoxyglucose positron emission tomography/CT (FDG PET-CT) provides complimentary anatomical and functional information. Oncological applications of FDG PET-CT are not as well-established within the paediatric population compared to adults. This article will comprehensively review established oncological PET-CT applications in paediatric oncology and provide an overview of emerging and future developments in this domain.
Collapse
Affiliation(s)
- Greg Chambers
- Department of Nuclear Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Russell Frood
- Department of Nuclear Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Chirag Patel
- Department of Nuclear Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | | |
Collapse
|
83
|
Khangembam BC, Singhal A, Kumar R, Bal C. Tc-99m Glucoheptonate Single Photon Emission Computed Tomography-Computed Tomography for Detection of Recurrent Glioma: A Prospective Comparison with N-13 Ammonia Positron Emission Tomography-Computed Tomography. Indian J Nucl Med 2019; 34:107-117. [PMID: 31040521 PMCID: PMC6481207 DOI: 10.4103/ijnm.ijnm_164_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose of the Study: To assess the efficacies of Tc-99m glucoheptonate single photon emission computed tomography-computed tomography (Tc-99m GHA SPECT-CT) and N-13 ammonia positron emission tomography-computed tomography (N-13 NH3 PET-CT) in detecting recurrent glioma. Materials and Methods: Fifty-five consecutive, histologically proven, and previously treated glioma patients (age, 38.9 ± 12.2 years; 61.8% males) presenting with clinical suspicion of recurrence were evaluated with Tc-99m GHA SPECT-CT and N-13 NH3 PET-CT. Images were evaluated both qualitatively and semiquantitatively. A combination of clinicoradiological follow-up, repeat imaging, and/or biopsy (when available) was considered as the reference standard. Results: Based on the reference standard, 28/55 (50.9%) patients had recurrence. Sensitivity, specificity, positive predictive value, negative predictive value, accuracy of Tc-99m GHA SPECT-CT, and N-13 NH3 PET-CT were 85.7%, 85.2%, 85.7%, 85.2%, 85.5% and 78.6%, 88.9%, 88.0%, 80.0%, 83.6%, respectively (concordant findings in 46 patients). The performances of the two modalities were equivalent both in overall and subgroup McNemar analyses (P = 0.508, overall; P = 0.687, low grade; P = 1.000, high grade). Conclusion: Tc-99m GHA SPECT-CT is an alternative imaging modality equally efficacious as N-13 NH3 PET-CT in detecting recurrent glioma.
Collapse
Affiliation(s)
- Bangkim Chandra Khangembam
- Department of Nuclear Medicine, Institute of Liver and Biliary Sciences, New Delhi, India.,Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Abhinav Singhal
- Department of Nuclear Medicine, Institute of Liver and Biliary Sciences, New Delhi, India.,Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Rajeev Kumar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Chandrasekhar Bal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
84
|
|
85
|
Wright E, Amankwah EK, Winesett SP, Tuite GF, Jallo G, Carey C, Rodriguez LF, Stapleton S. Incidentally found brain tumors in the pediatric population: a case series and proposed treatment algorithm. J Neurooncol 2018; 141:355-361. [DOI: 10.1007/s11060-018-03039-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 10/27/2018] [Indexed: 12/18/2022]
|
86
|
Maralani PJ, Das S, Mainprize T, Phan N, Bharatha A, Keith J, Munoz DG, Sahgal A, Symons S, Ironside S, Faraji-Dana Z, Eilaghi A, Chan A, Alcaide-Leon P, Shearkhani O, Jakubovic R, Atenafu EG, Zaharchuk G, Mikulis D. Hypoxia Detection in Infiltrative Astrocytoma: Ferumoxytol-based Quantitative BOLD MRI with Intraoperative and Histologic Validation. Radiology 2018; 288:821-829. [DOI: 10.1148/radiol.2018172601] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
87
|
99mTc-Methionine Hybrid SPECT/CT for Detection of Recurrent Glioma: Comparison With 18F-FDG PET/CT and Contrast-Enhanced MRI. Clin Nucl Med 2018. [PMID: 29517540 DOI: 10.1097/rlu.0000000000002036] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Posttherapy changes in treated glioma patients cannot be reliably differentiated from tumor recurrence. We evaluated the role of Tc-methionine SPECT/CT for the detection of recurrent glioma and compared the same with F-FDG PET/CT and contrast-enhanced MRI (CeMRI). METHODS Forty-four patients with histologically proven, previously treated glioma and clinical suspicion of recurrence were prospectively enrolled in the study. Of these 44 patients, 39 (28 male and 11 female subjects; age, 38.05 ± 9.7 years) underwent Tc-methionine SPECT/CT, F-FDG PET/CT, and CeMRI of the brain and were included for final analysis. Combination of repeat imaging, biopsy, and/or clinical follow-up (6-36 months) was taken as reference standard. Sensitivity, specificity, positive predictive value, negative predictive value, and accuracy were calculated. Diagnostic values among modalities were compared. RESULTS Positive predictive value and negative predictive value for Tc-methionine SPECT/CT, F-FDG PET/CT, and CeMRI were 95.6% and 56.2%, 92.3% and 61.5%, and 79.4% and 42.9%, respectively. Sensitivity and specificity for the 3 modalities were 75.9% and 90%, 82.8% and 80%, and 87.1% and 30%. Specificity of Tc-methionine SPECT/CT was significantly higher than that of CeMRI (P < 0.0001) but not of F-FDG PET/CT (P = 0.36). No significant difference was seen between the modalities for sensitivity and accuracy. CONCLUSIONS Tc-methionine is a promising tracer for detection of recurrent glioma. Diagnostic values of Tc-methionine SPECT/CT are similar to F-FDG, although it is more specific than CeMRI. So it may be used as a cost-effective alternative and also where PET/CT is not available.
Collapse
|
88
|
Alfonso JCL, Talkenberger K, Seifert M, Klink B, Hawkins-Daarud A, Swanson KR, Hatzikirou H, Deutsch A. The biology and mathematical modelling of glioma invasion: a review. J R Soc Interface 2018; 14:rsif.2017.0490. [PMID: 29118112 DOI: 10.1098/rsif.2017.0490] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/17/2017] [Indexed: 12/13/2022] Open
Abstract
Adult gliomas are aggressive brain tumours associated with low patient survival rates and limited life expectancy. The most important hallmark of this type of tumour is its invasive behaviour, characterized by a markedly phenotypic plasticity, infiltrative tumour morphologies and the ability of malignant progression from low- to high-grade tumour types. Indeed, the widespread infiltration of healthy brain tissue by glioma cells is largely responsible for poor prognosis and the difficulty of finding curative therapies. Meanwhile, mathematical models have been established to analyse potential mechanisms of glioma invasion. In this review, we start with a brief introduction to current biological knowledge about glioma invasion, and then critically review and highlight future challenges for mathematical models of glioma invasion.
Collapse
Affiliation(s)
- J C L Alfonso
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Centre for Information Services and High Performance Computing, Technische Universität Dresden, Germany
| | - K Talkenberger
- Centre for Information Services and High Performance Computing, Technische Universität Dresden, Germany
| | - M Seifert
- Institute for Medical Informatics and Biometry, Technische Universität Dresden, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany
| | - B Klink
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany.,German Cancer Consortium (DKTK), partner site, Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - A Hawkins-Daarud
- Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, USA
| | - K R Swanson
- Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, USA
| | - H Hatzikirou
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Centre for Information Services and High Performance Computing, Technische Universität Dresden, Germany
| | - A Deutsch
- Centre for Information Services and High Performance Computing, Technische Universität Dresden, Germany
| |
Collapse
|
89
|
Blanc-Durand P, Van Der Gucht A, Verger A, Langen KJ, Dunet V, Bloch J, Brouland JP, Nicod-Lalonde M, Schaefer N, Prior JO. Voxel-based 18F-FET PET segmentation and automatic clustering of tumor voxels: A significant association with IDH1 mutation status and survival in patients with gliomas. PLoS One 2018; 13:e0199379. [PMID: 29953478 PMCID: PMC6023198 DOI: 10.1371/journal.pone.0199379] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 06/06/2018] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION Aim was to develop a full automatic clustering approach of the time-activity curves (TAC) from dynamic 18F-FET PET and evaluate its association with IDH1 mutation status and survival in patients with gliomas. METHODS Thirty-seven patients (mean age: 45±13 y) with newly diagnosed gliomas and dynamic 18F-FET PET before any histopathologic investigation or treatment were retrospectively included. Each dynamic 18F-FET PET was realigned to the first image and spatially normalized in the Montreal Neurological Institute template. A tumor mask was semi-automatically generated from Z-score maps. Each brain tumor voxel was clustered in one of the 3 following centroids using dynamic time warping and k-means clustering (centroid #1: slowly increasing slope; centroid #2: rapidly increasing followed by slowly decreasing slope; and centroid #3: rapidly increasing followed by rapidly decreasing slope). The percentage of each dynamic 18F-FET TAC within tumors and other conventional 18F-FET PET parameters (maximum and mean tumor-to-brain ratios [TBRmax and TBRmean], time-to-peak [TTP] and slope) was compared between wild-type and IDH1 mutant tumors. Their prognostic value was assessed in terms of progression free-survival (PFS) and overall survival (OS) by Kaplan-Meier estimates. RESULTS Twenty patients were IDH1 wild-type and 17 IDH1 mutant. Higher percentage of centroid #1 and centroid #3 within tumors were positively (P = 0.016) and negatively (P = 0.01) correlated with IDH1 mutated status. Also, TBRmax, TBRmean, TTP, and slope discriminated significantly between tumors with and without IDH1 mutation (P range 0.01 to 0.04). Progression occurred in 22 patients (59%) at a median of 13.1 months (7.6-37.6 months) and 13 patients (35%) died from tumor progression. Patients with a percentage of centroid #1 > 90% had a longer survival compared with those with a percentage of centroid #1 < 90% (P = 0.003 for PFS and P = 0.028 for OS). This remained significant after stratification on IDH1 mutation status (P = 0.029 for PFS and P = 0.034 for OS). Compared to other conventional 18F-FET PET parameters, TTP and slope were associated with PFS and OS (P range 0.009 to 0.04). CONCLUSIONS Based on dynamic 18F-FET PET acquisition, we developed a full automatic clustering approach of TAC which appears to be a valuable noninvasive diagnostic and prognostic marker in patients with gliomas.
Collapse
Affiliation(s)
- Paul Blanc-Durand
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Axel Van Der Gucht
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Antoine Verger
- Department of Nuclear Medicine and Molecular Imaging, Nancy University Hospital, Nancy, France
| | - Karl-Josef Langen
- Department of Nuclear Medicine, University of Aachen, Aachen, Germany
| | - Vincent Dunet
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Jocelyne Bloch
- Department of Neurosurgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Jean-Philippe Brouland
- Department of Pathology and Laboratory Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Marie Nicod-Lalonde
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Niklaus Schaefer
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - John O. Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
90
|
Ly KI, Gerstner ER. The Role of Advanced Brain Tumor Imaging in the Care of Patients with Central Nervous System Malignancies. Curr Treat Options Oncol 2018; 19:40. [PMID: 29931476 DOI: 10.1007/s11864-018-0558-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OPINION STATEMENT T1-weighted post-contrast and T2-weighted fluid-attenuated inversion recovery (FLAIR) magnetic resonance imaging (MRI) constitute the gold standard for diagnosis and response assessment in neuro-oncologic patients but are limited in their ability to accurately reflect tumor biology and metabolism, particularly over the course of a patient's treatment. Advanced MR imaging methods are sensitized to different biophysical processes in tissue, including blood perfusion, tumor metabolism, and chemical composition of tissue, and provide more specific information on tissue physiology than standard MRI. This review provides an overview of the most common and emerging advanced imaging modalities in the field of brain tumor imaging and their applications in the care of neuro-oncologic patients.
Collapse
Affiliation(s)
- K Ina Ly
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, 55 Fruit Street, Yawkey 9E, Boston, MA, 02114, USA
| | - Elizabeth R Gerstner
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, 55 Fruit Street, Yawkey 9E, Boston, MA, 02114, USA.
| |
Collapse
|
91
|
Faria G, Silva E, Da Fonseca C, Quirico-Santos T. Circulating Cell-Free DNA as a Prognostic and Molecular Marker for Patients with Brain Tumors under Perillyl Alcohol-Based Therapy. Int J Mol Sci 2018; 19:ijms19061610. [PMID: 29848970 PMCID: PMC6032335 DOI: 10.3390/ijms19061610] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/16/2018] [Accepted: 05/23/2018] [Indexed: 12/28/2022] Open
Abstract
Tumor infiltration into brain tissue usually remains undetected even by high-resolution imaging. Molecular markers are used to increase diagnostic accuracy, but with limited continuous monitoring application. We evaluated the potential of circulating cell-free DNA (cfDNA) as a molecular indicator of the response to therapy by the intranasal administration (ITN) of perillyl alcohol (POH) in brain tumors. The cohort included 130 healthy subjects arranged as control-paired groups and patients at terminal stages with glioblastoma (GBM, n = 122) or brain metastasis (BM, n = 55) from stage IV adenocarcinomas. Serum cfDNA was isolated and quantified by fluorimetry. Compared with the controls (40 ng/mL), patients with brain tumors before ITN-POH treatment had increased (p < 0.0001) cfDNA median levels: GBM (286 ng/mL) and BM (588 ng/mL). ITN-POH treatment was significantly correlated (rho = −0.225; p = 0.024) with survival of >6 months at a concentration of 599 ± 221 ng/mL and of <6 months at 1626 ± 505 ng/mL, but a sharp and abrupt increase of cfDNA and tumor recurrence occurred after ITN-POH discontinuation. Patients under continuous ITN-POH treatment and checked with brain magnetic resonance imaging (MRI) compatible with complete response had cfDNA levels similar to the controls. cfDNA may be used as a noninvasive prognostic and molecular marker for POH-based therapy in brain tumors and as an accurate screening tool for the early detection of tumor progression.
Collapse
Affiliation(s)
- Giselle Faria
- Department of Cellular and Molecular Biology, Institute of Biology, Fluminense Federal University, Niteroi, Rio de Janeiro 24020-141, Brazil.
| | - Emanuelle Silva
- Department of Cellular and Molecular Biology, Institute of Biology, Fluminense Federal University, Niteroi, Rio de Janeiro 24020-141, Brazil.
| | - Clovis Da Fonseca
- Department of Specialized Medicine, School of Medicine, Fluminense Federal University, Niteroi, Rio de Janeiro 24020-141, Brazil.
| | - Thereza Quirico-Santos
- Department of Cellular and Molecular Biology, Institute of Biology, Fluminense Federal University, Niteroi, Rio de Janeiro 24020-141, Brazil.
| |
Collapse
|
92
|
Suchorska B, Albert NL, Bauer EK, Tonn JC, Galldiks N. The role of amino-acid PET in the light of the new WHO classification 2016 for brain tumors. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2018; 62:267-271. [PMID: 29696947 DOI: 10.23736/s1824-4785.18.03090-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Since its introduction in 2016, the revision of the World Health Organization (WHO) classification of central nervous system tumors has already changed the diagnostic and therapeutic approach in glial tumors. Blurring the lines between entities formerly labelled as "high-grade" or "low-grade", molecular markers define distinct biological subtypes with different clinical course. This new classification raises the demand for non-invasive imaging methods focusing on depicting metabolic processes. We performed a review of current literature on the use of amino-acid PET (AA-PET) for obtaining diagnostic or prognostic information on glioma in the setting of the current WHO 2016 classification. So far, only a few studies have focused on combining molecular genetic information and metabolic imaging using AA-PET. The current review summarizes the information available on "molecular grading" as well as prognostic information obtained from AA-PET and delivers an insight into a possible interrelation between metabolic imaging and glioma genetics. Within the framework of molecular characterization of gliomas, metabolic imaging using AA-PET is a promising tool for non-invasive characterization of molecular features and to provide additional prognostic information. Further studies incorporating molecular and metabolic features are necessary to improve the explanatory power of AA-PET in glial tumors.
Collapse
Affiliation(s)
- Bogdana Suchorska
- Department of Neurosurgery, Ludwig-Maximilians-University, Munich, Germany -
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Elena K Bauer
- Department of Neurology, University Hospital Cologne, Cologne, Germany
| | | | - Norbert Galldiks
- Department of Neurology, University Hospital Cologne, Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Juelich, Juelich, Germany.,Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Cologne, Germany
| |
Collapse
|
93
|
Blanc-Durand P, Van Der Gucht A, Schaefer N, Itti E, Prior JO. Automatic lesion detection and segmentation of 18F-FET PET in gliomas: A full 3D U-Net convolutional neural network study. PLoS One 2018; 13:e0195798. [PMID: 29652908 PMCID: PMC5898737 DOI: 10.1371/journal.pone.0195798] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/29/2018] [Indexed: 01/17/2023] Open
Abstract
INTRODUCTION Amino-acids positron emission tomography (PET) is increasingly used in the diagnostic workup of patients with gliomas, including differential diagnosis, evaluation of tumor extension, treatment planning and follow-up. Recently, progresses of computer vision and machine learning have been translated for medical imaging. Aim was to demonstrate the feasibility of an automated 18F-fluoro-ethyl-tyrosine (18F-FET) PET lesion detection and segmentation relying on a full 3D U-Net Convolutional Neural Network (CNN). METHODS All dynamic 18F-FET PET brain image volumes were temporally realigned to the first dynamic acquisition, coregistered and spatially normalized onto the Montreal Neurological Institute template. Ground truth segmentations were obtained using manual delineation and thresholding (1.3 x background). The volumetric CNN was implemented based on a modified Keras implementation of a U-Net library with 3 layers for the encoding and decoding paths. Dice similarity coefficient (DSC) was used as an accuracy measure of segmentation. RESULTS Thirty-seven patients were included (26 [70%] in the training set and 11 [30%] in the validation set). All 11 lesions were accurately detected with no false positive, resulting in a sensitivity and a specificity for the detection at the tumor level of 100%. After 150 epochs, DSC reached 0.7924 in the training set and 0.7911 in the validation set. After morphological dilatation and fixed thresholding of the predicted U-Net mask a substantial improvement of the DSC to 0.8231 (+ 4.1%) was noted. At the voxel level, this segmentation led to a 0.88 sensitivity [95% CI, 87.1 to, 88.2%] a 0.99 specificity [99.9 to 99.9%], a 0.78 positive predictive value: [76.9 to 78.3%], and a 0.99 negative predictive value [99.9 to 99.9%]. CONCLUSIONS With relatively high performance, it was proposed the first full 3D automated procedure for segmentation of 18F-FET PET brain images of patients with different gliomas using a U-Net CNN architecture.
Collapse
Affiliation(s)
- Paul Blanc-Durand
- Department of Nuclear Medicine, Henri Mondor University Hospital, Créteil, France
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
- * E-mail: (PBD); (AVDG)
| | - Axel Van Der Gucht
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
- * E-mail: (PBD); (AVDG)
| | - Niklaus Schaefer
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Emmanuel Itti
- Department of Nuclear Medicine, Henri Mondor University Hospital, Créteil, France
| | - John O. Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
94
|
Laukamp KR, Lindemann F, Weckesser M, Hesselmann V, Ligges S, Wölfer J, Jeibmann A, Zinnhardt B, Viel T, Schäfers M, Paulus W, Stummer W, Schober O, Jacobs AH. Multimodal Imaging of Patients With Gliomas Confirms 11C-MET PET as a Complementary Marker to MRI for Noninvasive Tumor Grading and Intraindividual Follow-Up After Therapy. Mol Imaging 2018; 16:1536012116687651. [PMID: 28654379 PMCID: PMC5470145 DOI: 10.1177/1536012116687651] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The value of combined L-( methyl-[11C]) methionine positron-emitting tomography (MET-PET) and magnetic resonance imaging (MRI) with regard to tumor extent, entity prediction, and therapy effects in clinical routine in patients with suspicion of a brain tumor was investigated. In n = 65 patients with histologically verified brain lesions n = 70 MET-PET and MRI (T1-weighted gadolinium-enhanced [T1w-Gd] and fluid-attenuated inversion recovery or T2-weighted [FLAIR/T2w]) examinations were performed. The computer software "visualization and analysis framework volume rendering engine (Voreen)" was used for analysis of extent and intersection of tumor compartments. Binary logistic regression models were developed to differentiate between World Health Organization (WHO) tumor types/grades. Tumor sizes as defined by thresholding based on tumor-to-background ratios were significantly different as determined by MET-PET (21.6 ± 36.8 cm3), T1w-Gd-MRI (3.9 ± 7.8 cm3), and FLAIR/T2-MRI (64.8 ± 60.4 cm3; P < .001). The MET-PET visualized tumor activity where MRI parameters were negative: PET positive tumor volume without Gd enhancement was 19.8 ± 35.0 cm3 and without changes in FLAIR/T2 10.3 ± 25.7 cm3. FLAIR/T2-MRI visualized greatest tumor extent with differences to MET-PET being greater in posttherapy (64.6 ± 62.7 cm3) than in newly diagnosed patients (20.5 ± 52.6 cm3). The binary logistic regression model differentiated between WHO tumor types (fibrillary astrocytoma II n = 10 from other gliomas n = 16) with an accuracy of 80.8% in patients at primary diagnosis. Combined PET and MRI improve the evaluation of tumor activity, extent, type/grade prediction, and therapy-induced changes in patients with glioma and serve information highly relevant for diagnosis and management.
Collapse
Affiliation(s)
- Kai R Laukamp
- 1 European Institute for Molecular Imaging, Westfälische Wilhelms-Universität Münster, Munster, Germany.,2 Department of Radiology, University Hospital of Cologne, Cologne, Germany
| | - Florian Lindemann
- 3 Department of Computer Science, Visualization and Computer Graphics Research Group, Westfälische Wilhelms-Universität Münster, Munster, Germany
| | - Matthias Weckesser
- 4 Departments of Nuclear Medicine, Westfälische Wilhelms-Universität Münster, Munster, Germany
| | - Volker Hesselmann
- 5 Departments of Radiology, Westfälische Wilhelms-Universität Münster, Munster, Germany
| | - Sandra Ligges
- 6 Institute of Biostatistics and Clinical Research, Westfälische Wilhelms-Universität Münster, Munster, Germany
| | - Johannes Wölfer
- 7 Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Munster, Germany
| | - Astrid Jeibmann
- 8 Department of Neuropathology, Westfälische Wilhelms-Universität Münster, Munster, Germany
| | - Bastian Zinnhardt
- 1 European Institute for Molecular Imaging, Westfälische Wilhelms-Universität Münster, Munster, Germany
| | - Thomas Viel
- 1 European Institute for Molecular Imaging, Westfälische Wilhelms-Universität Münster, Munster, Germany
| | - Michael Schäfers
- 1 European Institute for Molecular Imaging, Westfälische Wilhelms-Universität Münster, Munster, Germany.,4 Departments of Nuclear Medicine, Westfälische Wilhelms-Universität Münster, Munster, Germany.,9 Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), Westfälische Wilhelms-Universität Münster, Munster, Germany
| | - Werner Paulus
- 8 Department of Neuropathology, Westfälische Wilhelms-Universität Münster, Munster, Germany.,9 Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), Westfälische Wilhelms-Universität Münster, Munster, Germany
| | - Walter Stummer
- 7 Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Munster, Germany.,9 Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), Westfälische Wilhelms-Universität Münster, Munster, Germany
| | - Otmar Schober
- 1 European Institute for Molecular Imaging, Westfälische Wilhelms-Universität Münster, Munster, Germany.,4 Departments of Nuclear Medicine, Westfälische Wilhelms-Universität Münster, Munster, Germany.,9 Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), Westfälische Wilhelms-Universität Münster, Munster, Germany
| | - Andreas H Jacobs
- 1 European Institute for Molecular Imaging, Westfälische Wilhelms-Universität Münster, Munster, Germany.,4 Departments of Nuclear Medicine, Westfälische Wilhelms-Universität Münster, Munster, Germany.,9 Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), Westfälische Wilhelms-Universität Münster, Munster, Germany.,10 Department of Geriatric Medicine, Johanniter Hospital, Evangelische Kliniken, Bonn, Germany
| |
Collapse
|
95
|
Gkika E, Oehlke O, Bunea H, Wiedenmann N, Adebahr S, Nestle U, Zamboglou C, Kirste S, Fennell J, Brunner T, Gainey M, Baltas D, Langer M, Urbach H, Bock M, Meyer PT, Grosu AL. Biological imaging for individualized therapy in radiation oncology: part II medical and clinical aspects. Future Oncol 2018. [DOI: 10.2217/fon-2017-0465] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Positron emission tomography and multiparametric MRI provide crucial information concerning tumor extent and normal tissue anatomy. Moreover, they are able to visualize biological characteristics of the tumor, which can be considered in the radiation treatment planning and monitoring. In this review we discuss the impact of biological imaging positron emission tomography and multiparametric MRI for radiation oncology, based on the data of the literature and on the experience of our own institution in this field.
Collapse
Affiliation(s)
- Eleni Gkika
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Oliver Oehlke
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Hatice Bunea
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Nicole Wiedenmann
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Sonja Adebahr
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Ursula Nestle
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Simon Kirste
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Jamina Fennell
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Thomas Brunner
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Mark Gainey
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Dimos Baltas
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Mathias Langer
- Department of Radiology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
| | - Horst Urbach
- Department of Neuroradiology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
| | - Michael Bock
- Department of Radiology – Medical Physics, Department of Radiology, Faculty of Medicine, Medical Center, University of Freiburg, D-79106, Germany
| | - Philipp T Meyer
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
- Department of Nuclear Medicine, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| |
Collapse
|
96
|
Trikalinos NA, Nihashi T, Evangelou E, Terasawa T. Positron emission tomography (PET) for prediction of glioma histology: protocol for an individual-level data meta-analysis of test performance. BMJ Open 2018; 8:e020187. [PMID: 29455169 PMCID: PMC5855451 DOI: 10.1136/bmjopen-2017-020187] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
INTRODUCTION Gliomas, the most commonly diagnosed primary brain tumours, are associated with varied survivals based, in part, on their histological subtype. Therefore, accurate pretreatment tumour grading is essential for patient care and clinical trial design. METHODS AND ANALYSIS We will perform an individual-level data meta-analysis of published studies to evaluate the ability of different types of positron emission tomography (PET) to differentiate high from low-grade gliomas. We will search PubMed and Scopus from inception through 30 July 2017 with no language restriction and full-text evaluation of potentially relevant articles. We will choose studies that assess PET using 18-Fludeoxyglucose (18F-FDG), l-[Methyl-()11C]Methionine (11C-MET), 18F-Fluoro-Ethyl-Tyrosine (18F-FET) or (18)F-Fluorothymidine (18F-FLT)for grading, verified with histological confirmation. We will include both prospective and retrospective studies. Bias will be assessed by two reviewers with the Quality Assessment of Diagnostic Accuracy Studies-2 tool and as per method described by Deeks et al. ETHICS AND DISSEMINATION Ethics approval was not applicable, as this is a meta-analytic study. Results of the analysis will be submitted for publication in a peer-reviewed journal. PROSPERO REGISTRATION NUMBER CRD42017078649.
Collapse
Affiliation(s)
- Nikolaos A Trikalinos
- Department of Oncology, Washington University in St Louis School of Medicine, St Louis, Missouri, USA
| | - Takashi Nihashi
- Department of Radiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of radiology, Komaki City Hospital, Jobushi, Japan
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Teruhiko Terasawa
- Section of General Internal Medicine, Department of Emergency and General Internal Medicine, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| |
Collapse
|
97
|
Inhibition of GLO1 in Glioblastoma Multiforme Increases DNA-AGEs, Stimulates RAGE Expression, and Inhibits Brain Tumor Growth in Orthotopic Mouse Models. Int J Mol Sci 2018; 19:ijms19020406. [PMID: 29385725 PMCID: PMC5855628 DOI: 10.3390/ijms19020406] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 01/22/2018] [Accepted: 01/26/2018] [Indexed: 12/22/2022] Open
Abstract
Cancers that exhibit the Warburg effect may elevate expression of glyoxylase 1 (GLO1) to detoxify the toxic glycolytic byproduct methylglyoxal (MG) and inhibit the formation of pro-apoptotic advanced glycation endproducts (AGEs). Inhibition of GLO1 in cancers that up-regulate glycolysis has been proposed as a therapeutic targeting strategy, but this approach has not been evaluated for glioblastoma multiforme (GBM), the most aggressive and difficult to treat malignancy of the brain. Elevated GLO1 expression in GBM was established in patient tumors and cell lines using bioinformatics tools and biochemical approaches. GLO1 inhibition in GBM cell lines and in an orthotopic xenograft GBM mouse model was examined using both small molecule and short hairpin RNA (shRNA) approaches. Inhibition of GLO1 with S-(p-bromobenzyl) glutathione dicyclopentyl ester (p-BrBzGSH(Cp)2) increased levels of the DNA-AGE N2-1-(carboxyethyl)-2′-deoxyguanosine (CEdG), a surrogate biomarker for nuclear MG exposure; substantially elevated expression of the immunoglobulin-like receptor for AGEs (RAGE); and induced apoptosis in GBM cell lines. Targeting GLO1 with shRNA similarly increased CEdG levels and RAGE expression, and was cytotoxic to glioma cells. Mice bearing orthotopic GBM xenografts treated systemically with p-BrBzGSH(Cp)2 exhibited tumor regression without significant off-target effects suggesting that GLO1 inhibition may have value in the therapeutic management of these drug-resistant tumors.
Collapse
|
98
|
Imaging Genetic Heterogeneity in Glioblastoma and Other Glial Tumors: Review of Current Methods and Future Directions. AJR Am J Roentgenol 2018; 210:30-38. [DOI: 10.2214/ajr.17.18754] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
99
|
Pronin AI, Dolgushin MB, Lyuosev AS, Odzharova AA, Nevzorov DI, Nechipay EA, Gasparyan TG. [Capabilities of 18F-FET PET/CT in a patient with brain glioma (a case report and literature review)]. ZHURNAL VOPROSY NEIROKHIRURGII IMENI N. N. BURDENKO 2018; 82:95-99. [PMID: 29795092 DOI: 10.17116/oftalma201882295-99] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Positron emission tomography combined with computed tomography (PET/CT) enables assessment of not only anatomical and structural but also metabolic changes in tumor mass. 18F-fluoroethyl tyrosine (18F-FET) PET/CT is based on evaluation of transport of 18F-labeled tyrosine in tissues. We present a clinical case of a patient with a newly diagnosed brain tumor, demonstrating the capabilities of 18F-FET PET/CT in assessing the reliable volume and degree of tumor anaplasia, which is important when choosing the treatment approach for a patient.
Collapse
Affiliation(s)
- A I Pronin
- Blokhin Russian Cancer Research Center, Kashirskoe Shosse, 23, Moscow, Russia, 115478
| | - M B Dolgushin
- Blokhin Russian Cancer Research Center, Kashirskoe Shosse, 23, Moscow, Russia, 115478
| | - A S Lyuosev
- Blokhin Russian Cancer Research Center, Kashirskoe Shosse, 23, Moscow, Russia, 115478
| | - A A Odzharova
- Blokhin Russian Cancer Research Center, Kashirskoe Shosse, 23, Moscow, Russia, 115478
| | - D I Nevzorov
- Blokhin Russian Cancer Research Center, Kashirskoe Shosse, 23, Moscow, Russia, 115478
| | - E A Nechipay
- Blokhin Russian Cancer Research Center, Kashirskoe Shosse, 23, Moscow, Russia, 115478
| | - T G Gasparyan
- Blokhin Russian Cancer Research Center, Kashirskoe Shosse, 23, Moscow, Russia, 115478
| |
Collapse
|
100
|
Jung JH, Ahn BC. Current Radiopharmaceuticals for Positron Emission Tomography of Brain Tumors. Brain Tumor Res Treat 2018; 6:47-53. [PMID: 30381916 PMCID: PMC6212689 DOI: 10.14791/btrt.2018.6.e13] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/17/2018] [Accepted: 09/19/2018] [Indexed: 12/20/2022] Open
Abstract
Brain tumors represent a diverse spectrum of histology, biology, prognosis, and treatment options. Although MRI remains the gold standard for morphological tumor characterization, positron emission tomography (PET) can play a critical role in evaluating disease status. This article focuses on the use of PET with radiolabeled glucose and amino acid analogs to aid in the diagnosis of tumors and differentiate between recurrent tumors and radiation necrosis. The most widely used tracer is ¹⁸F-fluorodeoxyglucose (FDG). Although the intensity of FDG uptake is clearly associated with tumor grade, the exact role of FDG PET imaging remains debatable. Additionally, high uptake of FDG in normal grey matter limits its use in some low-grade tumors that may not be visualized. Because of their potential to overcome the limitation of FDG PET of brain tumors, ¹¹C-methionine and ¹⁸F-3,4-dihydroxyphenylalanine (FDOPA) have been proposed. Low accumulation of amino acid tracers in normal brains allows the detection of low-grade gliomas and facilitates more precise tumor delineation. These amino acid tracers have higher sensitivity and specificity for detecting brain tumors and differentiating recurrent tumors from post-therapeutic changes. FDG and amino acid tracers may be complementary, and both may be required for assessment of an individual patient. Additional tracers for brain tumor imaging are currently under development. Combinations of different tracers might provide more in-depth information about tumor characteristics, and current limitations may thus be overcome in the near future. PET with various tracers including FDG, ¹¹C-methionine, and FDOPA has improved the management of patients with brain tumors. To evaluate the exact value of PET, however, additional prospective large sample studies are needed.
Collapse
Affiliation(s)
- Ji Hoon Jung
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Korea
| | - Byeong Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Korea.
| |
Collapse
|