1
|
Haley MJ, Bere L, Minshull J, Georgaka S, Garcia-Martin N, Howell G, Coope DJ, Roncaroli F, King A, Wedge DC, Allan SM, Pathmanaban ON, Brough D, Couper KN. Hypoxia coordinates the spatial landscape of myeloid cells within glioblastoma to affect survival. SCIENCE ADVANCES 2024; 10:eadj3301. [PMID: 38758780 PMCID: PMC11100569 DOI: 10.1126/sciadv.adj3301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 04/15/2024] [Indexed: 05/19/2024]
Abstract
Myeloid cells are highly prevalent in glioblastoma (GBM), existing in a spectrum of phenotypic and activation states. We now have limited knowledge of the tumor microenvironment (TME) determinants that influence the localization and the functions of the diverse myeloid cell populations in GBM. Here, we have utilized orthogonal imaging mass cytometry with single-cell and spatial transcriptomic approaches to identify and map the various myeloid populations in the human GBM tumor microenvironment (TME). Our results show that different myeloid populations have distinct and reproducible compartmentalization patterns in the GBM TME that is driven by tissue hypoxia, regional chemokine signaling, and varied homotypic and heterotypic cellular interactions. We subsequently identified specific tumor subregions in GBM, based on composition of identified myeloid cell populations, that were linked to patient survival. Our results provide insight into the spatial organization of myeloid cell subpopulations in GBM, and how this is predictive of clinical outcome.
Collapse
Affiliation(s)
- Michael J. Haley
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Lydia Becker Institute of Inflammation and Immunology, University of Manchester, Manchester, UK
| | - Leoma Bere
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Lydia Becker Institute of Inflammation and Immunology, University of Manchester, Manchester, UK
| | - James Minshull
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Division of Neuroscience, University of Manchester, Manchester, UK
| | - Sokratia Georgaka
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK
| | | | - Gareth Howell
- Flow Cytometry Core Research Facility, University of Manchester, Manchester, UK
| | - David J. Coope
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Division of Neuroscience, University of Manchester, Manchester, UK
- Manchester Centre for Clinical Neurosciences, Manchester, UK
| | - Federico Roncaroli
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Division of Neuroscience, University of Manchester, Manchester, UK
- Manchester Centre for Clinical Neurosciences, Manchester, UK
| | - Andrew King
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Manchester Centre for Clinical Neurosciences, Manchester, UK
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - David C. Wedge
- Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | - Stuart M. Allan
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Division of Neuroscience, University of Manchester, Manchester, UK
| | - Omar N. Pathmanaban
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Division of Neuroscience, University of Manchester, Manchester, UK
- Manchester Centre for Clinical Neurosciences, Manchester, UK
| | - David Brough
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Lydia Becker Institute of Inflammation and Immunology, University of Manchester, Manchester, UK
- Division of Neuroscience, University of Manchester, Manchester, UK
| | - Kevin N. Couper
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Manchester, UK
- Lydia Becker Institute of Inflammation and Immunology, University of Manchester, Manchester, UK
| |
Collapse
|
2
|
Im C, Ahn JH, Farag AK, Kim S, Kim JY, Lee YJ, Park JA, Kang CM. Porphyrin-Based Brain Tumor-Targeting Agents: [ 64Cu]Cu-porphyrin and [ 64Cu]Cu-TDAP. Mol Pharm 2023; 20:5856-5864. [PMID: 37851927 DOI: 10.1021/acs.molpharmaceut.3c00704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
The aim of this study is to evaluate a radioactive metal complex platform for brain tumor targeting. Herein, we introduce a new porphyrin derivative, 5,10,15,20-(tetra-N,N-dimethyl-4-aminophenyl)porphyrin (TDAP), in which four N,N-dimethyl-4-p-phenylenediamine (DMPD) moieties are conjugated to the porphyrin labeled with the radiometal 64Cu. DMPD affected the pharmacokinetics of porphyrin in terms of retention time in vivo and tumor-targeting ability relative to those of unmodified porphyrin. [64Cu]Cu-TDAP showed stronger enhancement than [64Cu]Cu-porphyrin in U87MG glioblastoma cells, especially in the cytoplasm and nucleus, indicating its tumor-targeting properties and potential use as a therapeutic agent. In the subcutaneous and orthotopic models of brain-tumor-bearing mice, [64Cu]Cu-TDAP was clearly visualized in the tumor site via positron emission tomography imaging and showed a tumor-to-brain ratio as high as 13. [64Cu]Cu-TDAP deserves attention as a new diagnostic agent that is suitable for the early diagnosis and treatment of brain tumors.
Collapse
Affiliation(s)
- Changkeun Im
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 01812, Korea
- Radiological and Medico-Oncological Sciences, University of Science and Technology (UST), Seoul 01812, Korea
| | - Jae Hun Ahn
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 01812, Korea
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Ahmed K Farag
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 01812, Korea
- CDN isotopes, Toronto Research Chemicals, Montreal, Quebec H9R 1H1, Canada
| | - Soyeon Kim
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 01812, Korea
| | - Jung Young Kim
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 01812, Korea
| | - Yong Jin Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 01812, Korea
| | - Ji-Ae Park
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 01812, Korea
- Radiological and Medico-Oncological Sciences, University of Science and Technology (UST), Seoul 01812, Korea
| | - Choong Mo Kang
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 01812, Korea
- Radiological and Medico-Oncological Sciences, University of Science and Technology (UST), Seoul 01812, Korea
| |
Collapse
|
3
|
Dadgar H, Jokar N, Nemati R, Larvie M, Assadi M. PET tracers in glioblastoma: Toward neurotheranostics as an individualized medicine approach. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2023; 3:1103262. [PMID: 39355049 PMCID: PMC11440984 DOI: 10.3389/fnume.2023.1103262] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/23/2023] [Indexed: 10/03/2024]
Abstract
Over the past decade, theragnostic radiopharmaceuticals have been used in nuclear medicine for both diagnosis and treatment of various tumors. In this review, we carried out a literature search to investigate and explain the role of radiotracers in the theragnostic approach to glioblastoma multiform (GBM). We primarily focused on basic and rather common positron emotion tomography (PET) radiotracers in these tumors. Subsequently, we introduced and evaluated the preclinical and clinical results of theranostic-based biomarkers including integrin receptor family, prostate-specific membrane antigen (PSMA), fibroblast activated protein (FAP), somatostatin receptors (SRS), and chemokine receptor-4 (CXCR4) for patients with GBM to confer the benefit of personalized therapy. Moreover, promising research opportunities that could have a profound impact on the treatment of GBM over the next decade are also highlighted. Preliminary results showed the potential feasibility of the theragnostic approach using theses biomarkers in GBM patients.
Collapse
Affiliation(s)
- Habibullah Dadgar
- Cancer Research Center, RAZAVI Hospital, Imam Reza International University, Mashhad, Iran
| | - Narges Jokar
- The Persian Gulf Nuclear Medicine Research Center, Department of Molecular Imaging and Theranostics, Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Reza Nemati
- Department of Neurology, Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mykol Larvie
- Department of Radiology, Cleveland Clinic, Cleveland, Ohio
| | - Majid Assadi
- The Persian Gulf Nuclear Medicine Research Center, Department of Molecular Imaging and Theranostics, Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
4
|
Dhawan A, Pifer PM, Sandulache VC, Skinner HD. Metabolic targeting, immunotherapy and radiation in locally advanced non-small cell lung cancer: Where do we go from here? Front Oncol 2022; 12:1016217. [PMID: 36591457 PMCID: PMC9794617 DOI: 10.3389/fonc.2022.1016217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/24/2022] [Indexed: 12/15/2022] Open
Abstract
In the US, there are ~250,000 new lung cancer diagnoses and ~130,000 deaths per year, and worldwide there are an estimated 1.6 million deaths per year from this deadly disease. Lung cancer is the most common cause of cancer death worldwide, and it accounts for roughly a quarter of all cancer deaths in the US. Non-small cell lung cancer (NSCLC) represents 80-85% of these cases. Due to an enormous tobacco cessation effort, NSCLC rates in the US are decreasing, and the implementation of lung cancer screening guidelines and other programs have resulted in a higher percentage of patients presenting with potentially curable locoregional disease, instead of distant disease. Exciting developments in molecular targeted therapy and immunotherapy have resulted in dramatic improvement in patients' survival, in combination with new surgical, pathological, radiographical, and radiation techniques. Concurrent platinum-based doublet chemoradiation therapy followed by immunotherapy has set the benchmark for survival in these patients. However, despite these advances, ~50% of patients diagnosed with locally advanced NSCLC (LA-NSCLC) survive long-term. In patients with local and/or locoregional disease, chemoradiation is a critical component of curative therapy. However, there remains a significant clinical gap in improving the efficacy of this combined therapy, and the development of non-overlapping treatment approaches to improve treatment outcomes is needed. One potential promising avenue of research is targeting cancer metabolism. In this review, we will initially provide a brief general overview of tumor metabolism as it relates to therapeutic targeting. We will then focus on the intersection of metabolism on both oxidative stress and anti-tumor immunity. This will be followed by discussion of both tumor- and patient-specific opportunities for metabolic targeting in NSCLC. We will then conclude with a discussion of additional agents currently in development that may be advantageous to combine with chemo-immuno-radiation in NSCLC.
Collapse
Affiliation(s)
- Annika Dhawan
- Department of Radiation Oncology, UPMC Hillman Cancer Center and University of Pittsburgh, Pittsburgh, PA, United States
| | - Phillip M. Pifer
- Department of Radiation Oncology, UPMC Hillman Cancer Center and University of Pittsburgh, Pittsburgh, PA, United States
| | - Vlad C. Sandulache
- Bobby R. Alford Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Heath D. Skinner
- Department of Radiation Oncology, UPMC Hillman Cancer Center and University of Pittsburgh, Pittsburgh, PA, United States,*Correspondence: Heath D. Skinner,
| |
Collapse
|
5
|
van Genugten EAJ, Weijers JAM, Heskamp S, Kneilling M, van den Heuvel MM, Piet B, Bussink J, Hendriks LEL, Aarntzen EHJG. Imaging the Rewired Metabolism in Lung Cancer in Relation to Immune Therapy. Front Oncol 2022; 11:786089. [PMID: 35070990 PMCID: PMC8779734 DOI: 10.3389/fonc.2021.786089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/10/2021] [Indexed: 12/14/2022] Open
Abstract
Metabolic reprogramming is recognized as one of the hallmarks of cancer. Alterations in the micro-environmental metabolic characteristics are recognized as important tools for cancer cells to interact with the resident and infiltrating T-cells within this tumor microenvironment. Cancer-induced metabolic changes in the micro-environment also affect treatment outcomes. In particular, immune therapy efficacy might be blunted because of somatic mutation-driven metabolic determinants of lung cancer such as acidity and oxygenation status. Based on these observations, new onco-immunological treatment strategies increasingly include drugs that interfere with metabolic pathways that consequently affect the composition of the lung cancer tumor microenvironment (TME). Positron emission tomography (PET) imaging has developed a wide array of tracers targeting metabolic pathways, originally intended to improve cancer detection and staging. Paralleling the developments in understanding metabolic reprogramming in cancer cells, as well as its effects on stromal, immune, and endothelial cells, a wave of studies with additional imaging tracers has been published. These tracers are yet underexploited in the perspective of immune therapy. In this review, we provide an overview of currently available PET tracers for clinical studies and discuss their potential roles in the development of effective immune therapeutic strategies, with a focus on lung cancer. We report on ongoing efforts that include PET/CT to understand the outcomes of interactions between cancer cells and T-cells in the lung cancer microenvironment, and we identify areas of research which are yet unchartered. Thereby, we aim to provide a starting point for molecular imaging driven studies to understand and exploit metabolic features of lung cancer to optimize immune therapy.
Collapse
Affiliation(s)
- Evelien A J van Genugten
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Jetty A M Weijers
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Manfred Kneilling
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University, Tuebingen, Germany.,Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | | | - Berber Piet
- Department of Respiratory Diseases, Radboudumc, Nijmegen, Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Netherlands
| | - Lizza E L Hendriks
- Department of Pulmonary Diseases, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre (UMC), Maastricht, Netherlands
| | - Erik H J G Aarntzen
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| |
Collapse
|
6
|
Diagnosis of Glioblastoma by Immuno-Positron Emission Tomography. Cancers (Basel) 2021; 14:cancers14010074. [PMID: 35008238 PMCID: PMC8750680 DOI: 10.3390/cancers14010074] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/16/2021] [Accepted: 12/21/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Neuroimaging has transformed the way brain tumors are diagnosed and treated. Although different non-invasive modalities provide very helpful information, in some situations, they present a limited value. By merging the specificity of antibodies with the resolution, sensitivity, and quantitative capabilities of positron emission tomography (PET), “Immuno-PET” allows us to conduct the non-invasive diagnosis and monitoring of patients over time using antibody-based probes as an in vivo, integrated, quantifiable, 3D, full-body “immunohistochemistry”, like a “virtual biopsy”. This review provides and focuses on immuno-PET applications and future perspectives of this promising imaging approach for glioblastoma. Abstract Neuroimaging has transformed neuro-oncology and the way that glioblastoma is diagnosed and treated. Magnetic Resonance Imaging (MRI) is the most widely used non-invasive technique in the primary diagnosis of glioblastoma. Although MRI provides very powerful anatomical information, it has proven to be of limited value for diagnosing glioblastomas in some situations. The final diagnosis requires a brain biopsy that may not depict the high intratumoral heterogeneity present in this tumor type. The revolution in “cancer-omics” is transforming the molecular classification of gliomas. However, many of the clinically relevant alterations revealed by these studies have not yet been integrated into the clinical management of patients, in part due to the lack of non-invasive biomarker-based imaging tools. An innovative option for biomarker identification in vivo is termed “immunotargeted imaging”. By merging the high target specificity of antibodies with the high spatial resolution, sensitivity, and quantitative capabilities of positron emission tomography (PET), “Immuno-PET” allows us to conduct the non-invasive diagnosis and monitoring of patients over time using antibody-based probes as an in vivo, integrated, quantifiable, 3D, full-body “immunohistochemistry” in patients. This review provides the state of the art of immuno-PET applications and future perspectives on this imaging approach for glioblastoma.
Collapse
|
7
|
Liu Z, Lian X, Zhang X, Zhu Y, Zhang W, Wang J, Wang H, Liu B, Ren Z, Zhang M, Liu M, Gao Y. ESPL1 Is a Novel Prognostic Biomarker Associated With the Malignant Features of Glioma. Front Genet 2021; 12:666106. [PMID: 34512713 PMCID: PMC8428966 DOI: 10.3389/fgene.2021.666106] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022] Open
Abstract
Research has confirmed that extra spindle pole bodies-like 1 (ESPL1), an etiological factor, promotes the malignant progression of cancers. However, the relationship between ESPL1 and glioma has not yet been demonstrated. The purpose of this study was to reveal the potential mechanisms of ESPL1-mediated malignant glioma progression. Gene expression data and detailed clinical information of glioma cases were obtained from multiple public databases. Subsequently, a series of bioinformatics analyses were used to elucidate the effects of ESPL1 on glioma. The results demonstrated that the mRNA and protein levels of ESPL1 in glioma were higher than those in normal brain tissues. In addition, ESPL1 expression was considerably associated with the clinical and pathological features of gliomas, such as World Health Organization grade, histology, and 1p19q co-deletion status. Importantly, ESPL1 reduced the overall survival (OS) of glioma patients and had prognostic value for gliomas. Gene set enrichment analysis (GSEA) indirectly revealed that ESPL1 regulates the activation of cancer-related pathways, such as the cell cycle and base excision repair pathways. In addition, we used the Connectivity Map (CMap) database to screen three molecular drugs that inhibit ESPL1: thioguanosine, antimycin A, and zidovudine. Finally, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to detect the expression levels of ESPL1 in glioma cell lines. This study plays an important role in revealing the etiology of glioma by revealing the function of ESPL1, providing a potential molecular marker for the diagnosis and treatment of glioma, especially low-grade glioma.
Collapse
Affiliation(s)
- Zhendong Liu
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan International Joint Laboratory of Intelligentized Orthopedics Innovation and Transformation, Henan Key Laboratory for Intelligent Precision Orthopedics, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China.,Department of Surgery of Spine and Spinal Cord, Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Xiaoyu Lian
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Xiuru Zhang
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan International Joint Laboratory of Intelligentized Orthopedics Innovation and Transformation, Henan Key Laboratory for Intelligent Precision Orthopedics, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Yongjie Zhu
- Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Wang Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jialin Wang
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Hongbo Wang
- Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Binfeng Liu
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhishuai Ren
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Mengjun Zhang
- Harbin Medical University Cancer Hospital, Heilongjiang Provincial Cancer Hospital, Harbin, China
| | - Mingyang Liu
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan International Joint Laboratory of Intelligentized Orthopedics Innovation and Transformation, Henan Key Laboratory for Intelligent Precision Orthopedics, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Yanzheng Gao
- Department of Surgery of Spine and Spinal Cord, Henan Provincial People's Hospital, Henan International Joint Laboratory of Intelligentized Orthopedics Innovation and Transformation, Henan Key Laboratory for Intelligent Precision Orthopedics, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China.,Department of Surgery of Spine and Spinal Cord, Microbiome Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| |
Collapse
|
8
|
Beg U, Snyder BM, Madhani SI, Hamidi N, Padmanaban V, Tuanquin LC, Kruser TJ, Connor J, Mansouri A. Current Landscape and Future Prospects of Radiation Sensitizers for Malignant Brain Tumors: A Systematic Review. World Neurosurg 2021; 151:e839-e856. [PMID: 33974987 DOI: 10.1016/j.wneu.2021.04.134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Radiation therapy (RT) is the cornerstone of management of malignant brain tumors, but its efficacy is limited in hypoxic tumors. Although numerous radiosensitizer compounds have been developed to enhance the effect of RT, progress has been stagnant. Through this systematic review, we provide an overview of radiosensitizers developed for malignant brain tumors, summarize their safety and efficacy, and evaluate areas for possible improvement. METHODS Following PRISMA guidelines, PubMed, EMBASE, Cochrane, and Web of Science were searched using terminology pertaining to radiosensitizers for brain tumor RT. Articles reporting clinical evidence of nonantineoplastic radiosensitizers with RT for malignant central nervous system tumors were included. Data of interest were presumed mechanism of action, median overall survival (OS), progression-free survival (PFS), and adverse events. RESULTS Twenty-two unique radiosensitizers were identified. Only 2/22 agents (fluosol with oxygen, and efaproxiral) showed improvement in OS in patients with glioblastoma and brain metastasis, respectively. A larger study was not able to confirm the latter. Improved PFS was reported with use of metronidazole, sodium glycididazole, and chloroquine. There was a wide range of toxicities, which prompted change of schedule or complete discontinuation of 9 agents. CONCLUSIONS Progress in radiosensitizers for malignant CNS tumors has been limited. Only 2 radiosensitizers have shown limited improvement in survival. Alternative strategies such as synthetic drug design, based on a mechanism of action that is independent of crossing the blood-brain barrier, may be necessary. Use of drug development strategies using new technologies to overcome past challenges is necessary.
Collapse
Affiliation(s)
- Usman Beg
- Midwestern University Arizona College of Osteopathic Medicine, Glendale, Arizona, USA
| | | | | | - Nima Hamidi
- Midwestern University Arizona College of Osteopathic Medicine, Glendale, Arizona, USA
| | - Varun Padmanaban
- Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | | | | | - James Connor
- Penn State Cancer Institute, Hershey, Pennsylvania, USA; Penn State Department of Neurosurgery, Hershey, Pennsylvania, USA
| | - Alireza Mansouri
- Penn State Cancer Institute, Hershey, Pennsylvania, USA; Penn State Department of Neurosurgery, Hershey, Pennsylvania, USA.
| |
Collapse
|
9
|
Scarpelli ML, Healey DR, Fuentes A, Kodibagkar VD, Quarles CC. Correlation of Tumor Hypoxia Metrics Derived from 18F-Fluoromisonidazole Positron Emission Tomography and Pimonidazole Fluorescence Images of Optically Cleared Brain Tissue. Tomography 2020; 6:379-388. [PMID: 33364428 PMCID: PMC7744194 DOI: 10.18383/j.tom.2020.00046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
18F-fluoromisonidazole (FMISO) positron emission tomography (PET) is a widely used noninvasive imaging modality for assessing hypoxia. We describe the first spatial comparison of FMISO PET with an ex vivo reference standard for hypoxia across whole tumor volumes. Eighteen rats were orthotopically implanted with C6 or 9L brain tumors and made to undergo FMISO PET scanning. Whole brains were excised, sliced into 1-mm-thick sections, optically cleared, and fluorescently imaged for pimonidazole using an in vivo imaging system. FMISO maximum tumor uptake, maximum tumor-to-cerebellar uptake (TCmax), and hypoxic fraction (extracted 110 minutes after FMISO injection) were correlated with analogous metrics derived from pimonidazole fluorescence images. FMISO SUVmax was not significantly different between C6 and 9L brain tumors (P = .70), whereas FMISO TCmax and hypoxic fraction were significantly greater for C6 tumors (P < .01). FMISO TCmax was significantly correlated with the maximum tumor pimonidazole intensity (ρ = 0.76, P < .01), whereas FMISO SUVmax was not. FMISO tumor hypoxic fraction was significantly correlated with the pimonidazole-derived hypoxic fraction (ρ = 0.78, P < .01). Given that FMISO TCmax and tumor hypoxic fraction had strong correlations with the pimonidazole reference standard, these metrics may offer more reliable measures of tumor hypoxia than conventional PET uptake metrics (SUVmax). The voxel-wise correlation between FMISO uptake and pimonidazole intensity for a given tumor was strongly dependent on the tumor's TCmax (ρ = 0.81, P < .01) and hypoxic fraction (ρ = 0.85, P < .01), indicating PET measurements within individual voxels showed greater correlation with pimonidazole reference standard in tumors with greater hypoxia.
Collapse
Affiliation(s)
- Matthew L. Scarpelli
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ; and
| | - Debbie R. Healey
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ; and
| | - Alberto Fuentes
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ; and
| | - Vikram D. Kodibagkar
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ; and
| | - C. Chad Quarles
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ; and
| |
Collapse
|
10
|
Bhat K, Saki M, Vlashi E, Cheng F, Duhachek-Muggy S, Alli C, Yu G, Medina P, He L, Damoiseaux R, Pellegrini M, Zemke NR, Nghiemphu PL, Cloughesy TF, Liau LM, Kornblum HI, Pajonk F. The dopamine receptor antagonist trifluoperazine prevents phenotype conversion and improves survival in mouse models of glioblastoma. Proc Natl Acad Sci U S A 2020; 117:11085-11096. [PMID: 32358191 PMCID: PMC7245100 DOI: 10.1073/pnas.1920154117] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the deadliest adult brain cancer, and all patients ultimately succumb to the disease. Radiation therapy (RT) provides survival benefit of 6 mo over surgery alone, but these results have not improved in decades. We report that radiation induces a glioma-initiating cell phenotype, and we have identified trifluoperazine (TFP) as a compound that interferes with this phenotype conversion. TFP causes loss of radiation-induced Nanog mRNA expression, and activation of GSK3 with consecutive posttranslational reduction in p-Akt, Sox2, and β-catenin protein levels. TFP did not alter the intrinsic radiation sensitivity of glioma-initiating cells (GICs). Continuous treatment with TFP and a single dose of radiation reduced the number of GICs in vivo and prolonged survival in syngeneic and patient-derived orthotopic xenograft (PDOX) mouse models of GBM. Our findings suggest that the combination of a dopamine receptor antagonist with radiation enhances the efficacy of RT in GBM by preventing radiation-induced phenotype conversion of radiosensitive non-GICs into treatment-resistant, induced GICs (iGICs).
Collapse
Affiliation(s)
- Kruttika Bhat
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Mohammad Saki
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Erina Vlashi
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
| | - Fei Cheng
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Sara Duhachek-Muggy
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Claudia Alli
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Garrett Yu
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Paul Medina
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Ling He
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Robert Damoiseaux
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
- Molecular Screening Shared Resource, University of California, Los Angeles, CA 90095
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095
| | - Nathan R Zemke
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095
| | - Phioanh Leia Nghiemphu
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
- Department of Neurology, University of California, Los Angeles, CA 90095
| | - Timothy F Cloughesy
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
- Department of Neurology, University of California, Los Angeles, CA 90095
| | - Linda M Liau
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
- Department of Neurosurgery, University of California, Los Angeles, CA 90095
| | - Harley I Kornblum
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
- Neuropsychiatric Institute-Semel Institute for Neuroscience & Human Behavior, University of California, Los Angeles, CA 90095
| | - Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095;
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
| |
Collapse
|
11
|
Chen A, Zhong L, Ju K, Lu T, Lv J, Cao H. Plasmatic circRNA Predicting the Occurrence of Human Glioblastoma. Cancer Manag Res 2020; 12:2917-2923. [PMID: 32425605 PMCID: PMC7196774 DOI: 10.2147/cmar.s248621] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/09/2020] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common primary malignant tumor in adult central nervous system and results in disappointing survival outcomes. Although the diagnosis and therapy approach have been developed recently, the prognosis of GBM remains poor. A novel, minimally invasive biomarker for GBM is necessary for early diagnosis or prognosis prediction. METHODS All circRNAs were detected by qRT-PCR in GBM samples including training and validation sets. We used the risk score analysis to assume the diagnosis ability for GBM. The receiver operating characteristic curve was also employed. RESULTS Among the 14 candidates, circRNA, circNT5E, circFOXO3, circ_0001946, circ_0029426, circ-SHPRH, and circMMP9 were detected with increased levels in the training set. Further investigation in the validation set indicated that circFOXO3, circ_0029426, and circ-SHPRH might be the fingerprints for GBM compared with controls. The risk score analysis revealed that the combination of three circRNAs could distinguish the GBM from healthy control with the area under curve value of 0.980 and 0.906, respectively. CONCLUSION The three circRNAs might be novel fingerprints for predicting the occurrence of GBM.
Collapse
Affiliation(s)
- Ainian Chen
- Department of Neurology, The Affiliated Huai’an No.1 People’s Hospital of Nanjing Medical University, Huai’an, Jiangsu223300, People’s Republic of China
| | - Lingling Zhong
- Department of Neurology, The Affiliated Huai’an No.1 People’s Hospital of Nanjing Medical University, Huai’an, Jiangsu223300, People’s Republic of China
| | - Keju Ju
- Department of Neurology, The Affiliated Huai’an No.1 People’s Hospital of Nanjing Medical University, Huai’an, Jiangsu223300, People’s Republic of China
| | - Ting Lu
- Department of Neurology, The Affiliated Huai’an No.1 People’s Hospital of Nanjing Medical University, Huai’an, Jiangsu223300, People’s Republic of China
| | - Jia Lv
- Department of Neurosurgery, The Affiliated Huai’an No.1 People’s Hospital of Nanjing Medical University, Huai’an, Jiangsu223300, People’s Republic of China
| | - Hua Cao
- Department of Neurology, The Affiliated Huai’an No.1 People’s Hospital of Nanjing Medical University, Huai’an, Jiangsu223300, People’s Republic of China
| |
Collapse
|
12
|
Goos JACM, Davydova M, Lengkeek N, Greguric I, Whittaker MR, Quinn JF, Baell JB, Lewis JS, Davis TP. pH-Responsive Polymers for Improving the Signal-to-Noise Ratio of Hypoxia PET Imaging with [ 18 F]Fluoromisonidazole. Macromol Rapid Commun 2020; 41:e2000061. [PMID: 32250004 DOI: 10.1002/marc.202000061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/16/2020] [Accepted: 03/19/2020] [Indexed: 01/09/2023]
Abstract
To improve the signal-to-noise ratio of hypoxia positron emission tomography (PET) imaging, stimuli-responsive polymers are designed for the delivery of the hypoxia PET tracer fluorine-18 labeled fluoromisonidazole ([18 F]FMISO). Linear poly(N-(2-(hydroxypropyl)methacrylamide)) polymers are functionalized with hydrazide linkers that form pH-sensitive acyl hydrazone bonds after their conjugation to an [18 F]FMISO ketone analogue. The release of the [18 F]FMISO ketone analogue from the polymers is considerably faster at a lower pH and its uptake is significantly higher in cancer cells growing under acidic conditions. Additionally, the retention of the PET tracer is significantly higher in hypoxic cells compared to normoxic cells. The delivery of a PET tracer using stimuli-responsive polymers may be an attractive strategy to improve signal-to-noise ratios in PET imaging.
Collapse
Affiliation(s)
- Jeroen A C M Goos
- Department of Clinical Neuroscience and MedTechLabs, Karolinska Institute, Solna, 17177, Sweden
| | - Maria Davydova
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Nigel Lengkeek
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, 2234, Australia
| | - Ivan Greguric
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, 2234, Australia
| | - Michael R Whittaker
- Department of Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia
| | - John F Quinn
- Department of Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia
| | - Jonathan B Baell
- Department of Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Thomas P Davis
- Department of Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Parkville, VIC, 3052, Australia
| |
Collapse
|
13
|
Werner JM, Lohmann P, Fink GR, Langen KJ, Galldiks N. Current Landscape and Emerging Fields of PET Imaging in Patients with Brain Tumors. Molecules 2020; 25:E1471. [PMID: 32213992 PMCID: PMC7146177 DOI: 10.3390/molecules25061471] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/17/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Abstract
The number of positron-emission tomography (PET) tracers used to evaluate patients with brain tumors has increased substantially over the last years. For the management of patients with brain tumors, the most important indications are the delineation of tumor extent (e.g., for planning of resection or radiotherapy), the assessment of treatment response to systemic treatment options such as alkylating chemotherapy, and the differentiation of treatment-related changes (e.g., pseudoprogression or radiation necrosis) from tumor progression. Furthermore, newer PET imaging approaches aim to address the need for noninvasive assessment of tumoral immune cell infiltration and response to immunotherapies (e.g., T-cell imaging). This review summarizes the clinical value of the landscape of tracers that have been used in recent years for the above-mentioned indications and also provides an overview of promising newer tracers for this group of patients.
Collapse
Affiliation(s)
- Jan-Michael Werner
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937 Cologne, Germany; (J.-M.W.); (G.R.F.)
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Leo-Brandt-St., 52425 Juelich, Germany; (P.L.); (K.-J.L.)
| | - Gereon R. Fink
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937 Cologne, Germany; (J.-M.W.); (G.R.F.)
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Leo-Brandt-St., 52425 Juelich, Germany; (P.L.); (K.-J.L.)
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Leo-Brandt-St., 52425 Juelich, Germany; (P.L.); (K.-J.L.)
- Department of Nuclear Medicine, University Hospital Aachen, 52074 Aachen, Germany
| | - Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937 Cologne, Germany; (J.-M.W.); (G.R.F.)
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Leo-Brandt-St., 52425 Juelich, Germany; (P.L.); (K.-J.L.)
| |
Collapse
|
14
|
Speed Switch in Glioblastoma Growth Rate due to Enhanced Hypoxia-Induced Migration. Bull Math Biol 2020; 82:43. [PMID: 32180054 DOI: 10.1007/s11538-020-00718-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 03/04/2020] [Indexed: 10/24/2022]
Abstract
We analyze the wave speed of the Proliferation Invasion Hypoxia Necrosis Angiogenesis (PIHNA) model that was previously created and applied to simulate the growth and spread of glioblastoma (GBM), a particularly aggressive primary brain tumor. We extend the PIHNA model by allowing for different hypoxic and normoxic cell migration rates and study the impact of these differences on the wave-speed dynamics. Through this analysis, we find key variables that drive the outward growth of the simulated GBM. We find a minimum tumor wave-speed for the model; this depends on the migration and proliferation rates of the normoxic cells and is achieved under certain conditions on the migration rates of the normoxic and hypoxic cells. If the hypoxic cell migration rate is greater than the normoxic cell migration rate above a threshold, the wave speed increases above the predicted minimum. This increase in wave speed is explored through an eigenvalue and eigenvector analysis of the linearized PIHNA model, which yields an expression for this threshold. The PIHNA model suggests that an inherently faster-diffusing hypoxic cell population can drive the outward growth of a GBM as a whole, and that this effect is more prominent for faster-proliferating tumors that recover relatively slowly from a hypoxic phenotype. The findings presented here act as a first step in enabling patient-specific calibration of the PIHNA model.
Collapse
|
15
|
Hypoxia PET imaging beyond 18F-FMISO in patients with high-grade glioma: 18F-FAZA and other hypoxia radiotracers. Clin Transl Imaging 2020. [DOI: 10.1007/s40336-020-00358-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
16
|
Spatial and quantitative mapping of glycolysis and hypoxia in glioblastoma as a predictor of radiotherapy response and sites of relapse. Eur J Nucl Med Mol Imaging 2020; 47:1476-1485. [PMID: 32025750 DOI: 10.1007/s00259-020-04706-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/21/2020] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Tumor hypoxia is a centerpiece of disease progression mechanisms such as neoangiogenesis or aggressive hypoxia-resistant malignant cells selection that impacts on radiotherapy strategies. Early identification of regions at risk for recurrence and prognostic-based classification of patients is a necessity to devise tailored therapeutic strategies. We developed an image-based algorithm to spatially map areas of aerobic and anaerobic glycolysis (Glyoxia). METHODS 18F-FDG and 18F-FMISO PET studies were used in the algorithm to produce DICOM-co-registered representations and maximum intensity projections combined with quantitative analysis of hypoxic volume (HV), hypoxic glycolytic volume (HGV), and anaerobic glycolytic volume (AGV) with CT/MRI co-registration. This was applied to a prospective clinical trial of 10 glioblastoma patients with post-operative, pre-radiotherapy, and early post-radiotherapy 18F-FDG and 18F-FMISO PET and MRI studies. RESULTS In the 10 glioblastoma patients (5M:5F; age range 51-69 years), 14/18 18F-FMISO PET studies showed detectable hypoxia. Seven patients survived to complete post-radiotherapy studies. The patient with the longest overall survival showed non-detectable hypoxia in both pre-radiotherapy and post-radiotherapy 18F-FMISO PET. The three patients with increased HV, HGV, and AGV volumes after radiotherapy showed 2.8 months mean progression-free interval vs. 5.9 months for the other 4 patients. These parameters correlated at that time point with progression-free interval. Parameters combining hypoxia and glycolytic information (i.e., HGV and AGV) showed more prominent variation than hypoxia-based information alone (HV). Glyoxia-generated images were consistent with disease relapse topology; in particular, one patient had distant relapse anticipated by HV, HGV, and AGV maps. CONCLUSION Spatial mapping of aerobic and anaerobic glycolysis allows unique information on tumor metabolism and hypoxia to be evaluated with PET, providing a greater understanding of tumor biology and potential response to therapy.
Collapse
|
17
|
Pruis IJ, van Dongen GAMS, Veldhuijzen van Zanten SEM. The Added Value of Diagnostic and Theranostic PET Imaging for the Treatment of CNS Tumors. Int J Mol Sci 2020; 21:E1029. [PMID: 32033160 PMCID: PMC7037158 DOI: 10.3390/ijms21031029] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 01/27/2020] [Accepted: 02/01/2020] [Indexed: 12/11/2022] Open
Abstract
This review highlights the added value of PET imaging in Central Nervous System (CNS) tumors, which is a tool that has rapidly evolved from a merely diagnostic setting to multimodal molecular diagnostics and the guidance of targeted therapy. PET is the method of choice for studying target expression and target binding behind the assumedly intact blood-brain barrier. Today, a variety of diagnostic PET tracers can be used for the primary staging of CNS tumors and to determine the effect of therapy. Additionally, theranostic PET tracers are increasingly used in the context of pharmaceutical and radiopharmaceutical drug development and application. In this approach, a single targeted drug is used for PET diagnosis, upon the coupling of a PET radionuclide, as well as for targeted (nuclide) therapy. Theranostic PET tracers have the potential to serve as a non-invasive whole body navigator in the selection of the most effective drug candidates and their most optimal dose and administration route, together with the potential to serve as a predictive biomarker in the selection of patients who are most likely to benefit from treatment. PET imaging supports the transition from trial and error medicine to predictive, preventive, and personalized medicine, hopefully leading to improved quality of life for patients and more cost-effective care.
Collapse
Affiliation(s)
- Ilanah J. Pruis
- Department of Radiology & Nuclear Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands;
| | - Guus A. M. S. van Dongen
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Sophie E. M. Veldhuijzen van Zanten
- Department of Radiology & Nuclear Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands;
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Department of Pediatrics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
18
|
Drake LR, Hillmer AT, Cai Z. Approaches to PET Imaging of Glioblastoma. Molecules 2020; 25:E568. [PMID: 32012954 PMCID: PMC7037643 DOI: 10.3390/molecules25030568] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the deadliest type of brain tumor, affecting approximately three in 100,000 adults annually. Positron emission tomography (PET) imaging provides an important non-invasive method of measuring biochemically specific targets at GBM lesions. These powerful data can characterize tumors, predict treatment effectiveness, and monitor treatment. This review will discuss the PET imaging agents that have already been evaluated in GBM patients so far, and new imaging targets with promise for future use. Previously used PET imaging agents include the tracers for markers of proliferation ([11C]methionine; [18F]fluoro-ethyl-L-tyrosine, [18F]Fluorodopa,[18F]fluoro-thymidine, and [18F]clofarabine), hypoxia sensing ([18F]FMISO, [18F]FET-NIM, [18F]EF5, [18F]HX4, and [64Cu]ATSM), and ligands for inflammation. As cancer therapeutics evolve toward personalized medicine and therapies centered on tumor biomarkers, the development of complimentary selective PET agents can dramatically enhance these efforts. Newer biomarkers for GBM PET imaging are discussed, with some already in use for PET imaging other cancers and neurological disorders. These targets include Sigma 1, Sigma 2, programmed death ligand 1, poly-ADP-ribose polymerase, and isocitrate dehydrogenase. For GBM, these imaging agents come with additional considerations such as blood-brain barrier penetration, quantitative modeling approaches, and nonspecific binding.
Collapse
Affiliation(s)
- Lindsey R. Drake
- Yale PET Center, Yale University School of Medicine, New Haven, CT 06511, USA; (A.T.H.); (Z.C.)
- Department of Radiology and Bioimaging Sciences, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Ansel T. Hillmer
- Yale PET Center, Yale University School of Medicine, New Haven, CT 06511, USA; (A.T.H.); (Z.C.)
- Department of Radiology and Bioimaging Sciences, Yale University School of Medicine, New Haven, CT 06511, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, CT 06511, USA
| | - Zhengxin Cai
- Yale PET Center, Yale University School of Medicine, New Haven, CT 06511, USA; (A.T.H.); (Z.C.)
- Department of Radiology and Bioimaging Sciences, Yale University School of Medicine, New Haven, CT 06511, USA
| |
Collapse
|
19
|
Riccardo L, Natale Q, Pierpaolo A, Domenico A, Maria G, Rexhep D, Francesco B, Sergio B. 18F-FMISO PET imaging: insights over MRI in patients with glioma. Clin Transl Imaging 2020. [DOI: 10.1007/s40336-019-00353-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
20
|
Cellular and Extracellular Components in Tumor Microenvironment and Their Application in Early Diagnosis of Cancers. Anal Cell Pathol (Amst) 2020; 2020:6283796. [PMID: 32377504 PMCID: PMC7199555 DOI: 10.1155/2020/6283796] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/11/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022] Open
Abstract
Tumors are surrounded by complex environmental components, including blood and lymph vessels, fibroblasts, endothelial cells, immune cells, cytokines, extracellular vesicles, and extracellular matrix. All the stromal components together with the tumor cells form the tumor microenvironment (TME). In addition, extracellular physical and chemical factors, including extracellular pH, hypoxia, elevated interstitial fluid pressure, and fibrosis, are closely associated with tumor progression, metastasis, immunosuppression, and drug resistance. Cellular and extracellular components in TME contribute to nearly all procedures of carcinogenesis. By summarizing the recent work in this field, we make a comprehensive review on the role of cellular and extracellular components in the process of carcinogenesis and their potential application in early diagnosis of cancer. We hope that a systematic review of the diverse aspects of TME will help both research scientists and clinicians in this field.
Collapse
|
21
|
Ermert J, Benešová M, Hugenberg V, Gupta V, Spahn I, Pietzsch HJ, Liolios C, Kopka K. Radiopharmaceutical Sciences. Clin Nucl Med 2020. [DOI: 10.1007/978-3-030-39457-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
22
|
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]
|
23
|
Shimizu Y, Kudo K, Kameda H, Harada T, Fujima N, Toyonaga T, Tha KK, Shirato H. Prediction of Hypoxia in Brain Tumors Using a Multivariate Model Built from MR Imaging and 18F-Fluorodeoxyglucose Accumulation Data. Magn Reson Med Sci 2019; 19:227-234. [PMID: 31611541 PMCID: PMC7553805 DOI: 10.2463/mrms.mp.2019-0049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Purpose: The aim of this study was to generate a multivariate model using various MRI markers of blood flow and vascular permeability and accumulation of 18F-fluorodeoxyglucose (FDG) to predict the extent of hypoxia in an 18F-fluoromisonidazole (FMISO)-positive region. Methods: Fifteen patients aged 27–74 years with brain tumors (glioma, n = 13; lymphoma, n = 1; germinoma, n = 1) were included. MRI scans were performed using a 3T scanner, and dynamic contrast-enhanced (DCE) perfusion and arterial spin labeling images were obtained. Ktrans and Vp maps were generated using the DCE images. FDG and FMISO positron emission tomography scans were also obtained. A model for predicting FMISO positivity was generated on a voxel-by-voxel basis by a multivariate logistic regression model using all the MRI parameters with and without FDG. Receiver-operating characteristic curve analysis was used to detect FMISO positivity with multivariate and univariate analysis of each parameter. Cross-validation was performed using the leave-one-out method. Results: The area under the curve (AUC) was highest for the multivariate prediction model with FDG (0.892) followed by the multivariate model without FDG and univariate analysis with FDG and Ktrans (0.844 for all). In cross-validation, the multivariate model with FDG had the highest AUC (0.857 ± 0.08) followed by the multivariate model without FDG (0.834 ± 0.119). Conclusion: A multivariate prediction model created using blood flow, vascular permeability, and glycometabolism parameters can predict the extent of hypoxia in FMISO-positive areas in patients with brain tumors.
Collapse
Affiliation(s)
- Yukie Shimizu
- Department of Radiation Medicine, Hokkaido University Graduate School of Medicine
| | - Kohsuke Kudo
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital.,Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education, Hokkaido University
| | - Hiroyuki Kameda
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital
| | - Taisuke Harada
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital
| | - Noriyuki Fujima
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital
| | - Takuya Toyonaga
- Department of Nuclear Medicine, Hokkaido University Graduate School of Medicine
| | - Khin Khin Tha
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital.,Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education, Hokkaido University
| | - Hiroki Shirato
- Department of Radiation Medicine, Hokkaido University Graduate School of Medicine.,Global Station for Quantum Medical Science and Engineering, Global Institution for Collaborative Research and Education, Hokkaido University
| |
Collapse
|
24
|
Libby CJ, McConathy J, Darley-Usmar V, Hjelmeland AB. The Role of Metabolic Plasticity in Blood and Brain Stem Cell Pathophysiology. Cancer Res 2019; 80:5-16. [PMID: 31575548 DOI: 10.1158/0008-5472.can-19-1169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 08/04/2019] [Accepted: 09/18/2019] [Indexed: 02/06/2023]
Abstract
Our understanding of intratumoral heterogeneity in cancer continues to evolve, with current models incorporating single-cell signatures to explore cell-cell interactions and differentiation state. The transition between stem and differentiation states in nonneoplastic cells requires metabolic plasticity, and this plasticity is increasingly recognized to play a central role in cancer biology. The insights from hematopoietic and neural stem cell differentiation pathways were used to identify cancer stem cells in leukemia and gliomas. Similarly, defining metabolic heterogeneity and fuel-switching signals in nonneoplastic stem cells may also give important insights into the corresponding molecular mechanisms controlling metabolic plasticity in cancer. These advances are important, because metabolic adaptation to anticancer therapeutics is rooted in this inherent metabolic plasticity and is a therapeutic challenge to be overcome.
Collapse
Affiliation(s)
- Catherine J Libby
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jonathan McConathy
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Victor Darley-Usmar
- Mitochondrial Medicine Laboratory, Center for Free Radical Biology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Anita B Hjelmeland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
25
|
Chen A, Zhong L, Lv J. FOXL1 overexpression is associated with poor outcome in patients with glioma. Oncol Lett 2019; 18:751-757. [PMID: 31289550 PMCID: PMC6540307 DOI: 10.3892/ol.2019.10351] [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] [Received: 12/19/2018] [Accepted: 04/19/2019] [Indexed: 11/21/2022] Open
Abstract
Gliomas are the most common primary tumors in adult central nervous system and result in disappointing survival outcomes. FOXL1, as a transcription factor, plays an important role in regulating the expression of genes involved in cell metabolism, proliferation and differentiation. In this study, we investigated the relationship between FOXL1 expression and prognosis of patients with glioma. We selected 611 glioma patients from The Cancer Genome Atlas (TCGA) database and 132 glioma patients from Huai'an First People's Hospital (PFHH). The prognostic values of FOXL1 in glioma were analyzed in both cohorts. In TCGA cohort, the median (10.2389) was used as the cut-off value of FOXL1 mRNA levels in tumor tissue. Kaplan-Meier analysis showed that higher WHO glioma grade (P<0.001) and expression of FOXL1 (P<0.001) were associated with worse overall survival (OS). The univariate Cox regression model revealed that age (P<0.001), WHO grade (P<0.001), histological type (P<0.001) and FOXL1 expression (P<0.001) were associated with prognosis of glioma patients. In PFHH cohort, expression of FOXL1 in tumor cells was detected by immunohistochemistry (IHC) staining based on a tissue microarray (TMA) sample. Kaplan-Meier analysis also showed that WHO glioma grade (P<0.001) and expression of FOXL1 (P=0.012) were associated with OS in glioma patients. The univariate Cox regression showed that WHO grade (P=0.001), histological type (P<0.001) and FOXL1 expression (P=0.013) were associated with prognosis of glioma patients. In both cohorts Kaplan-Meier subgroup analyses showed FOXL expression correlated with OS in high WHO grade subgroup, while low grade subgroup showed no such correlation. This study showed that higher expression of FOXL1 is associated with poor OS of glioma patients in TCGA and PFHH cohorts. Especially, FOXL1 overexpression is associated with worse outcomes in high WHO grade subgroup. Our findings suggest that FOXL1 expression is a candidate predictor of clinical outcome in glioma patients and may act as an effective molecular marker for immunotherapeutic strategies of glioma patients in clinical practice.
Collapse
Affiliation(s)
- Ainian Chen
- Department of Neurology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Lingling Zhong
- Department of Neurology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Jia Lv
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| |
Collapse
|
26
|
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
|
27
|
Abstract
PET holds potential to provide additional information about tumour metabolic processes, which could aid brain tumour differential diagnosis, grading, molecular subtyping and/or the distinction of therapy effects from disease recurrence. This review discusses PET techniques currently in use for untreated and treated glioma characterization and aims to critically assess the evidence for different tracers ([F]Fluorodeoxyglucose, choline and amino acid tracers) in this context.
Collapse
|
28
|
Choudhary G, Langen KJ, Galldiks N, McConathy J. Investigational PET tracers for high-grade gliomas. 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... 2018; 62:281-294. [PMID: 29869489 DOI: 10.23736/s1824-4785.18.03105-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
High-grade gliomas (HGGs) are the most common primary malignant tumors of the brain, with glioblastoma (GBM) constituting over 50% of all the gliomas in adults. The disease carries very high mortality, and even with optimal treatment, the median survival is 2-5 years for anaplastic tumors and 1-2 years for GBMs. Neuroimaging is critical to managing patients with HGG for diagnosis, treatment planning, response assessment, and detecting recurrent disease. Magnetic resonance imaging (MRI) is the cornerstone of imaging in neuro-oncology, but molecular imaging with positron emission tomography (PET) can overcome some of the inherent limitations of MRI. Additionally, PET has the potential to target metabolic and molecular alterations in HGGs relevant to prognosis and therapy that cannot be assessed with anatomic imaging. Many classes of PET tracers have been evaluated in HGG including agents that target cell membrane biosynthesis, protein synthesis, amino acid transport, DNA synthesis, the tricarboxylic acid (TCA) cycle, hypoxic environments, cell surface receptors, blood flow, vascular endothelial growth factor (VEGF), epidermal growth factor (EGFR), and the 18-kDa translocator protein (TSPO), among others. This chapter will provide an overview of PET tracers for HGG that have been evaluated in human subjects with a focus on tracers that are not yet in widespread use for neuro-oncology.
Collapse
Affiliation(s)
- Gagandeep Choudhary
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4), Jülich Research Center, Jülich, Germany.,Department of Nuclear Medicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-3, -4), Jülich Research Center, Jülich, Germany.,Department of Neurology, University of Cologne, Cologne, Germany.,Center of Integrated Oncology (CIO), Universities of Cologne and Bonn, Cologne, Germany
| | - Jonathan McConathy
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA -
| |
Collapse
|
29
|
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
|
30
|
Wada Y, Hirose K, Harada T, Sato M, Watanabe T, Anbai A, Hashimoto M, Takai Y. Impact of oxygen status on 10B-BPA uptake into human glioblastoma cells, referring to significance in boron neutron capture therapy. JOURNAL OF RADIATION RESEARCH 2018; 59:122-128. [PMID: 29315429 PMCID: PMC5950927 DOI: 10.1093/jrr/rrx080] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 10/30/2017] [Indexed: 06/01/2023]
Abstract
Boron neutron capture therapy (BNCT) can potentially deliver high linear energy transfer particles to tumor cells without causing severe damage to surrounding normal tissue, and may thus be beneficial for cases with characteristics of infiltrative growth, which need a wider irradiation field, such as glioblastoma multiforme. Hypoxia is an important factor contributing to resistance to anticancer therapies such as radiotherapy and chemotherapy. In this study, we investigated the impact of oxygen status on 10B uptake in glioblastoma cells in vitro in order to evaluate the potential impact of local hypoxia on BNCT. T98G and A172 glioblastoma cells were used in the present study, and we examined the influence of oxygen concentration on cell viability, mRNA expression of L-amino acid transporter 1 (LAT1), and the uptake amount of 10B-BPA. T98G and A172 glioblastoma cells became quiescent after 72 h under 1% hypoxia but remained viable. Uptake of 10B-BPA, which is one of the agents for BNCT in clinical use, decreased linearly as oxygen levels were reduced from 20% through to 10%, 3% and 1%. Hypoxia with <10% O2 significantly decreased mRNA expression of LAT1 in both cell lines, indicating that reduced uptake of 10B-BPA in glioblastoma in hypoxic conditions may be due to reduced expression of this important transporter protein. Hypoxia inhibits 10B-BPA uptake in glioblastoma cells in a linear fashion, meaning that approaches to overcoming local tumor hypoxia may be an effective method of improving the success of BNCT treatment.
Collapse
Affiliation(s)
- Yuki Wada
- Department of Radiation Oncology, Southern Tohoku BNCT Research Center, 7-10 Yatsuyamada, Koriyama, Fukushima 963-8052, Japan
- Department of Radiology and Radiation Oncology, Akita University Graduate School of Medicine, Hospital, 1-1-1 Hondo, Akita, Akita 010-8543, Japan
| | - Katsumi Hirose
- Department of Radiation Oncology, Southern Tohoku BNCT Research Center, 7-10 Yatsuyamada, Koriyama, Fukushima 963-8052, Japan
- Department of Radiology and Radiation Oncology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Takaomi Harada
- Department of Radiation Oncology, Southern Tohoku BNCT Research Center, 7-10 Yatsuyamada, Koriyama, Fukushima 963-8052, Japan
| | - Mariko Sato
- Department of Radiology and Radiation Oncology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Tsubasa Watanabe
- Particle Radiation Oncology Research Center, Kyoto University Research Reactor Institute, 2 Asashiro-nisi, Sennan-gun, Osaka 590-0494, Japan
| | - Akira Anbai
- Department of Radiology and Radiation Oncology, Akita University Graduate School of Medicine, Hospital, 1-1-1 Hondo, Akita, Akita 010-8543, Japan
| | - Manabu Hashimoto
- Department of Radiology and Radiation Oncology, Akita University Graduate School of Medicine, Hospital, 1-1-1 Hondo, Akita, Akita 010-8543, Japan
| | - Yoshihiro Takai
- Department of Radiation Oncology, Southern Tohoku BNCT Research Center, 7-10 Yatsuyamada, Koriyama, Fukushima 963-8052, Japan
- Department of Radiology and Radiation Oncology, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| |
Collapse
|
31
|
Preibisch C, Shi K, Kluge A, Lukas M, Wiestler B, Göttler J, Gempt J, Ringel F, Al Jaberi M, Schlegel J, Meyer B, Zimmer C, Pyka T, Förster S. Characterizing hypoxia in human glioma: A simultaneous multimodal MRI and PET study. NMR IN BIOMEDICINE 2017; 30:e3775. [PMID: 28805936 DOI: 10.1002/nbm.3775] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 06/19/2017] [Accepted: 06/29/2017] [Indexed: 06/07/2023]
Abstract
Hypoxia plays an important role for the prognosis and therapy response of cancer. Thus, hypoxia imaging would be a valuable tool for pre-therapeutic assessment of tumor malignancy. However, there is no standard validated technique for clinical application available yet. Therefore, we performed a study in 12 patients with high-grade glioma, where we directly compared the two currently most promising techniques, namely the MR-based relative oxygen extraction fraction (MR-rOEF) and the PET hypoxia marker H-1-(3-[18 F]-fluoro-2-hydroxypropyl)-2-nitroimidazole ([18 F]-FMISO). MR-rOEF was determined from separate measurements of T2 , T2 * and relative cerebral blood volume (rCBV) employing a multi-parametric approach for quantification of the blood-oxygenation-level-dependent (BOLD) effect. With respect to [18 F]-FMISO-PET, besides the commonly used late uptake between 120 and 130 min ([18 F]-FMISO120-130 min ), we also analyzed the hypoxia specific uptake rate [18 F]-FMISO-k3 , as obtained by pharmacokinetic modeling of dynamic uptake data. Since pharmacokinetic modeling of partially acquired dynamic [18 F]-FMISO data was sensitive to a low signal-to-noise-ratio, analysis was restricted to high-uptake tumor regions. Individual spatial analyses of deoxygenation and hypoxia-related parameter maps revealed that high MR-rOEF values clustered in (edematous) peritumoral tissue, while areas with high [18 F]-FMISO120-130 min concentrated in and around active tumor with disrupted blood-brain barrier, i.e. contrast enhancement in T1 -weighted MRI. Volume-of-interest-based correlations between MR-rOEF and [18 F]-FMISO120-130 min as well as [18 F]-FMISO-k3 , and voxel-wise analyses in individual patients, yielded limited correlations, supporting the notion that [18 F]-FMISO uptake, even after 2 h, might still be influenced by perfusion while [18 F]-FMISO-k3 was severely hampered by noise. According to these results, vascular deoxygenation, as measured by MR-rOEF, and severe tissue hypoxia, as measured by [18 F]-FMISO, show a poor spatial correspondence. Overall, the two methods appear to rather provide complementary than redundant information about high-grade glioma biology.
Collapse
Affiliation(s)
- Christine Preibisch
- Department of Diagnostic and Interventional Neuroradiology, Technische Universität München, Munich, Germany
- Clinic for Neurology, Technische Universität München, Munich, Germany
| | - Kuangyu Shi
- Clinic for Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Anne Kluge
- Department of Diagnostic and Interventional Neuroradiology, Technische Universität München, Munich, Germany
| | - Mathias Lukas
- Clinic for Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Benedikt Wiestler
- Department of Diagnostic and Interventional Neuroradiology, Technische Universität München, Munich, Germany
| | - Jens Göttler
- Department of Diagnostic and Interventional Neuroradiology, Technische Universität München, Munich, Germany
| | - Jens Gempt
- Department of Neurosurgery, Technische Universität München, Munich, Germany
| | - Florian Ringel
- Department of Neurosurgery, Technische Universität München, Munich, Germany
| | - Mohamed Al Jaberi
- Department of Neuropathology, Technische Universität München, Munich, Germany
| | - Jürgen Schlegel
- Department of Neuropathology, Technische Universität München, Munich, Germany
| | - Bernhard Meyer
- Department of Neurosurgery, Technische Universität München, Munich, Germany
| | - Claus Zimmer
- Department of Diagnostic and Interventional Neuroradiology, Technische Universität München, Munich, Germany
| | - Thomas Pyka
- Clinic for Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Stefan Förster
- Clinic for Nuclear Medicine, Technische Universität München, Munich, Germany
| |
Collapse
|
32
|
Bell C, Puttick S, Rose S, Smith J, Thomas P, Dowson N. Design and utilisation of protocols to characterise dynamic PET uptake of two tracers using basis pursuit. Phys Med Biol 2017; 62:4897-4916. [DOI: 10.1088/1361-6560/aa6b44] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
33
|
Vats K, Mallia MB, Mathur A, Sarma HD, Banerjee S. ‘4+1’ Mixed Ligand Strategy for the Preparation of 99m
Tc-Radiopharmaceuticals for Hypoxia Detecting Applications. ChemistrySelect 2017. [DOI: 10.1002/slct.201700150] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Kusum Vats
- Radiopharmaceuticals Division; Bhabha Atomic Research Centre; Mumbai- 400085
- Homi Bhabha National Institute; Anushakti Nagar Mumbai- 400094 India
| | - Madhava B. Mallia
- Radiopharmaceuticals Division; Bhabha Atomic Research Centre; Mumbai- 400085
| | - Anupam Mathur
- Radiopharmaceuticals Program; Board of Radiation and Isotope Technology; Mumbai- 400705 India
| | - Haladhar D. Sarma
- Radiation Biology and Health Science Division; Bhabha Atomic Research Centre; Mumbai- 400085
| | - Sharmila Banerjee
- Radiation Medicine Centre; Parel Mumbai- 400012 India
- Homi Bhabha National Institute; Anushakti Nagar Mumbai- 400094 India
| |
Collapse
|
34
|
Bekaert L, Valable S, Lechapt-Zalcman E, Ponte K, Collet S, Constans JM, Levallet G, Bordji K, Petit E, Branger P, Emery E, Manrique A, Barré L, Bernaudin M, Guillamo JS. [18F]-FMISO PET study of hypoxia in gliomas before surgery: correlation with molecular markers of hypoxia and angiogenesis. Eur J Nucl Med Mol Imaging 2017; 44:1383-1392. [PMID: 28315948 DOI: 10.1007/s00259-017-3677-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 03/09/2017] [Indexed: 12/12/2022]
Abstract
PURPOSE Hypoxia in gliomas is associated with tumor resistance to radio- and chemotherapy. However, positron emission tomography (PET) imaging of hypoxia remains challenging, and the validation of biological markers is, therefore, of great importance. We investigated the relationship between uptake of the PET hypoxia tracer [18F]-FMISO and other markers of hypoxia and angiogenesis and with patient survival. PATIENTS AND METHODS In this prospective single center clinical study, 33 glioma patients (grade IV: n = 24, III: n = 3, and II: n = 6) underwent [18F]-FMISO PET and MRI including relative cerebral blood volume (rCBV) maps before surgery. Maximum standardized uptake values (SUVmax) and hypoxic volume were calculated, defining two groups of patients based on the presence or absence of [18F]-FMISO uptake. After surgery, molecular quantification of CAIX, VEGF, Ang2 (rt-qPCR), and HIF-1α (immunohistochemistry) were performed on tumor specimens. RESULTS [18F]-FMISO PET uptake was closely linked to tumor grade, with high uptake in glioblastomas (GB, grade IV). Expression of biomarkers of hypoxia (CAIX, HIF-1α), and angiogenesis markers (VEGF, Ang2, rCBV) were significantly higher in the [18F]-FMISO uptake group. We found correlations between the degree of hypoxia (hypoxic volume and SUVmax) and expression of HIF-1α, CAIX, VEGF, Ang2, and rCBV (p < 0.01). Patients without [18F]-FMISO uptake had a longer survival time than uptake positive patients (log-rank, p < 0.005). CONCLUSIONS Tumor hypoxia as evaluated by [18F]-FMISO PET is associated with the expression of hypoxia markers on a molecular level and is related to angiogenesis. [18F]-FMISO uptake is a mark of an aggressive tumor, almost always a glioblastoma. Our results underline that [18F]-FMISO PET could be useful to guide glioma treatment, and in particular radiotherapy, since hypoxia is a well-known factor of resistance.
Collapse
Affiliation(s)
- Lien Bekaert
- Department of Neurology, CHU de Caen, Caen, France. .,Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France. .,Department of Neurosurgery, CHU de Caen, Caen, France. .,Service de Neurochirurgie, CHU de Caen, Avenue de la Côte de Nacre, 14000, Caen, France.
| | - Samuel Valable
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France
| | - Emmanuèle Lechapt-Zalcman
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France.,Department of Pathology, CHU de Caen, Caen, France
| | - Keven Ponte
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France.,Department of Neurosurgery, CHU de Caen, Caen, France
| | - Solène Collet
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France
| | - Jean-Marc Constans
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France.,Department of Neuroradiology, CHU de Caen, Caen, France
| | | | - Karim Bordji
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France
| | - Edwige Petit
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France
| | | | - Evelyne Emery
- Department of Neurosurgery, CHU de Caen, Caen, France
| | - Alain Manrique
- Department of Nuclear Medicine, CHU de Caen, Caen, France
| | - Louisa Barré
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/LDM-TEP group, 14000, Caen, France
| | - Myriam Bernaudin
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France
| | - Jean-Sébastien Guillamo
- Department of Neurology, CHU de Caen, Caen, France. .,Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, 14000, Caen, France. .,Department of Neurology, CHU de Nimes, Place du Professeur Robert Debre, 30029, Nimes cedex 9, France.
| |
Collapse
|
35
|
Cao J, Liu Y, Zhang L, Du F, Ci Y, Zhang Y, Xiao H, Yao X, Shi S, Zhu L, Kung HF, Qiao J. Synthesis of novel PEG-modified nitroimidazole derivatives via “hot-click” reaction and their biological evaluation as potential PET imaging agent for tumors. J Radioanal Nucl Chem 2017. [DOI: 10.1007/s10967-017-5210-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
36
|
Ferda J, Ferdová E, Hes O, Mraček J, Kreuzberg B, Baxa J. PET/MRI: Multiparametric imaging of brain tumors. Eur J Radiol 2017; 94:A14-A25. [PMID: 28283219 DOI: 10.1016/j.ejrad.2017.02.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 02/19/2017] [Accepted: 02/20/2017] [Indexed: 12/01/2022]
Abstract
A combination of morphological imaging of the brain with microstructural and functional imaging provides a comprehensive overview of the properties of individual tissues. While diffusion weighted imaging provides information about tissue cellularity, spectroscopic imaging allows us to evaluate the integrity of neurons and possible anaerobic glycolysis during tumor hypoxia, in addition to the presence of accelerated synthesis or degradation of cellular membranes; on the other hand, PET metabolic imaging is used to evaluate major metabolic pathways, determining the overall extent of the tumor (18F-FET, 18F-FDOPA, 18F-FCH) or the degree of differentiation (18F-FDG, 18F-FLT, 18F-FDOPA and 18F-FET). Multi-parameter analysis of tissue characteristics and determination of the phenotype of the tumor tissue is a natural advantage of PET/MRI scanning. The disadvantages are higher cost and limited availability in all centers with neuro-oncology surgery. PET/MRI scanning of brain tumors is one of the most promising indications since the earliest experiments with integrated PET/MRI imaging systems, and along with hybrid imaging of neurodegenerative diseases, represent a new direction in the development of neuroradiology on the path towards comprehensive imaging at the molecular level.
Collapse
Affiliation(s)
- Jiří Ferda
- Clinic of the Imaging Methods, University Hospital Plzen, Alej Svobody 80, 304 60 Plzeň, Czech Republic.
| | - Eva Ferdová
- Clinic of the Imaging Methods, University Hospital Plzen, Alej Svobody 80, 304 60 Plzeň, Czech Republic.
| | - Ondřej Hes
- Sikl's Institute of Pathological Anatomy, University Hospital Plzen, Alej Svobody 80;304 60 Plzeň, Czech Republic.
| | - Jan Mraček
- Clinic of the Neurosurgery, University Hospital Plzen, Alej Svobody 80, 304 60 Plzeň, Czech Republic.
| | - Boris Kreuzberg
- Clinic of the Imaging Methods, University Hospital Plzen, Alej Svobody 80, 304 60 Plzeň, Czech Republic.
| | - Jan Baxa
- Clinic of the Imaging Methods, University Hospital Plzen, Alej Svobody 80, 304 60 Plzeň, Czech Republic.
| |
Collapse
|
37
|
Baran N, Konopleva M. Molecular Pathways: Hypoxia-Activated Prodrugs in Cancer Therapy. Clin Cancer Res 2017; 23:2382-2390. [PMID: 28137923 DOI: 10.1158/1078-0432.ccr-16-0895] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/16/2016] [Accepted: 12/19/2016] [Indexed: 12/11/2022]
Abstract
Hypoxia is a known feature of aggressive solid tumors as well as a critical hallmark of the niche in aggressive hematologic malignances. Hypoxia is associated with insufficient response to standard therapy, resulting in disease progression and curtailed patients' survival through maintenance of noncycling cancer stem-like cells. A better understanding of the mechanisms and signaling pathways induced by hypoxia is essential to overcoming these effects. Recent findings demonstrate that bone marrow in the setting of hematologic malignancies is highly hypoxic, and that progression of the disease is associated with expansion of hypoxic niches and stabilization of the oncogenic hypoxia-inducible factor-1alpha (HIF1α). Solid tumors have also been shown to harbor hypoxic areas, maintaining survival of cancer cells via the HIF1α pathway. Developing new strategies for targeting hypoxia has become a crucial approach in modern cancer therapy. The number of preclinical and clinical trials targeting low-oxygen tumor compartments or the hypoxic bone marrow niche via hypoxia-activated prodrugs is increasing. This review discusses the development of the hypoxia-activated prodrugs and their applicability in treating both hematologic malignancies and solid tumors. Clin Cancer Res; 23(10); 2382-90. ©2017 AACR.
Collapse
Affiliation(s)
- Natalia Baran
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marina Konopleva
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
38
|
Abadjian MCZ, Edwards WB, Anderson CJ. Imaging the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1036:229-257. [PMID: 29275475 DOI: 10.1007/978-3-319-67577-0_15] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The tumor microenvironment consists of tumor, stromal, and immune cells, as well as extracellular milieu. Changes in numbers of these cell types and their environments have an impact on cancer growth and metastasis. Non-invasive imaging of aspects of the tumor microenvironment can provide important information on the aggressiveness of the cancer, whether or not it is metastatic, and can also help to determine early response to treatment. This chapter provides an overview on non-invasive in vivo imaging in humans and mouse models of various cell types and physiological parameters that are unique to the tumor microenvironment. Current clinical imaging and research investigation are in the areas of nuclear imaging (positron emission tomography (PET) and single photon emission computed tomography (SPECT)), magnetic resonance imaging (MRI) and optical (near infrared (NIR) fluorescence) imaging. Aspects of the tumor microenvironment that have been imaged by PET, MRI and/or optical imaging are tumor associated inflammation (primarily macrophages and T cells), hypoxia, pH changes, as well as enzymes and integrins that are highly prevalent in tumors, stroma and immune cells. Many imaging agents and strategies are currently available for cancer patients; however, the investigation of novel avenues for targeting aspects of the tumor microenvironment in pre-clinical models of cancer provides the cancer researcher with a means to monitor changes and evaluate novel treatments that can be translated into the clinic.
Collapse
Affiliation(s)
| | - W Barry Edwards
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Carolyn J Anderson
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
39
|
Change in 18F-Fluoromisonidazole PET Is an Early Predictor of the Prognosis in the Patients with Recurrent High-Grade Glioma Receiving Bevacizumab Treatment. PLoS One 2016; 11:e0167917. [PMID: 27936194 PMCID: PMC5148016 DOI: 10.1371/journal.pone.0167917] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 11/22/2016] [Indexed: 11/19/2022] Open
Abstract
Background Bevacizumab (BEV), a humanized monoclonal antibody, become a currently important chemotherapeutic option for the patients with recurrent glioma. The aim of this retrospective study is to investigate whether 18F-Fluoromisonidazole (FMISO) PET have the potential to detect BEV-resistant gliomas in the early-stage. Methods We reviewed the FMISO PET and MRI appearances before and 3 to 4 courses after BEV treatment on 18 recurrent glioma patients. FMISO accumulation was assessed by visual inspection and semi-quantitative values which were tumor-to-normal (T/N) ratio and hypoxic volume. MRI responses were evaluated based on RANO (Response Assessment in Neuro-Oncology) criteria. The prognostic analysis was performed in relation to the response assessment by FMISO PET and MRI using overall survival (OS) after BEV application. Results After BEV application, MRI revealed partial response in 14 of 18 patients (78%), of which 9 patients also demonstrated decreased FMISO accumulation. These 9 patients (50%) were classified as “MRI-FMISO double responder”. As for the other 5 patients (28%), FMISO accumulation volumes increased or remained stable after BEV treatment although partial responses were achieved on MRI. Therefore, these cases were classified as “MRI-only responder”. The remaining 4 patients (22%) did not show treatment response on FMISO PET or MRI (“non-responder”). MRI-FMISO double responders showed significantly longer OS than that in other groups (median 12.4 vs 5.7 months; P < 0.001), whereas there were no overall survival difference between MRI-only responders and non-responders (median OS, 5.7 and 4.8 months; P = 0.58). Among the pre-treatment clinical factors, high FMISO T/N ratio was a significant prognostic factor of overall survival in these patients under the assessment of Cox proportional hazard model. Conclusions Recurrent gliomas with decreasing FMISO accumulation after short-term BEV application could derive a survival benefit from BEV treatment. Change in FMISO PET appearance can identify BEV-resistant gliomas in early-stage regardless of MRI findings in a comprehensible way.
Collapse
|
40
|
|
41
|
Toyonaga T, Yamaguchi S, Hirata K, Kobayashi K, Manabe O, Watanabe S, Terasaka S, Kobayashi H, Hattori N, Shiga T, Kuge Y, Tanaka S, Ito YM, Tamaki N. Hypoxic glucose metabolism in glioblastoma as a potential prognostic factor. Eur J Nucl Med Mol Imaging 2016; 44:611-619. [PMID: 27752745 DOI: 10.1007/s00259-016-3541-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 09/29/2016] [Indexed: 02/05/2023]
Abstract
PURPOSE Metabolic activity and hypoxia are both important factors characterizing tumor aggressiveness. Here, we used F-18 fluoromisonidazole (FMISO) and F-18 fluorodeoxyglucose (FDG) positron emission tomography (PET) to define metabolically active hypoxic volume, and investigate its clinical significance in relation to progression free survival (PFS) and overall survival (OS) in glioblastoma patients. EXPERIMENTAL DESIGN Glioblastoma patients (n = 32) underwent FMISO PET, FDG PET, and magnetic resonance imaging (MRI) before surgical intervention. FDG and FMISO PET images were coregistered with gadolinium-enhanced T1-weighted MR images. Volume of interest (VOI) of gross tumor volume (GTV) was manually created to enclose the entire gadolinium-positive areas. The FMISO tumor-to-normal region ratio (TNR) and FDG TNR were calculated in a voxel-by-voxel manner. For calculating TNR, standardized uptake value (SUV) was divided by averaged SUV of normal references. Contralateral frontal and parietal cortices were used as the reference region for FDG, whereas the cerebellar cortex was used as the reference region for FMISO. FDG-positive was defined as the FDG TNR ≥1.0, and FMISO-positive was defined as FMISO TNR ≥1.3. Hypoxia volume (HV) was defined as the volume of FMISO-positive and metabolic tumor volume in hypoxia (hMTV) was the volume of FMISO/FDG double-positive. The total lesion glycolysis in hypoxia (hTLG) was hMTV × FDG SUVmean. The extent of resection (EOR) involving cytoreduction surgery was volumetric change based on planimetry methods using MRI. These factors were tested for correlation with patient prognosis. RESULTS All tumor lesions were FMISO-positive and FDG-positive. Univariate analysis indicated that hMTV, hTLG, and EOR were significantly correlated with PFS (p = 0.007, p = 0.04, and p = 0.01, respectively) and that hMTV, hTLG, and EOR were also significantly correlated with OS (p = 0.0028, p = 0.037, and p = 0.014, respectively). In contrast, none of FDG TNR, FMISO TNR, GTV, HV, patients' age, or Karnofsky performance scale (KPS) was significantly correlated with PSF or OS. The hMTV and hTLG were found to be independent factors affecting PFS and OS on multivariate analysis. CONCLUSIONS We introduced hMTV and hTLG using FDG and FMISO PET to define metabolically active hypoxic volume. Univariate and multivariate analyses demonstrated that both hMTV and hTLG are significant predictors for PFS and OS in glioblastoma patients.
Collapse
Affiliation(s)
- Takuya Toyonaga
- Department of Nuclear Medicine, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Shigeru Yamaguchi
- Department of Nuclear Medicine, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan.,Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kenji Hirata
- Department of Nuclear Medicine, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan.
| | - Kentaro Kobayashi
- Department of Nuclear Medicine, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Osamu Manabe
- Department of Nuclear Medicine, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Shiro Watanabe
- Department of Nuclear Medicine, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Shunsuke Terasaka
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hiroyuki Kobayashi
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Naoya Hattori
- Department of Nuclear Medicine, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Tohru Shiga
- Department of Nuclear Medicine, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Yuji Kuge
- Central Institute of Isotope Science, Hokkaido University, Sapporo, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yoichi M Ito
- Department of Biostatistics, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Nagara Tamaki
- Department of Nuclear Medicine, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, Hokkaido, 060-8638, Japan
| |
Collapse
|
42
|
Development of a mathematical model to estimate intra-tumor oxygen concentrations through multi-parametric imaging. Biomed Eng Online 2016; 15:114. [PMID: 27733170 PMCID: PMC5062945 DOI: 10.1186/s12938-016-0235-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Accepted: 10/04/2016] [Indexed: 01/22/2023] Open
Abstract
Background Tumor hypoxia is involved in every stage of solid tumor development: formation, progression, metastasis, and apoptosis. Two types of hypoxia exist in tumors—chronic hypoxia and acute hypoxia. Recent studies indicate that the regional hypoxia kinetics is closely linked to metastasis and therapeutic responses, but regional hypoxia kinetics is hard to measure. We propose a novel approach to determine the local pO2 by fusing the parameters obtained from in vivo functional imaging through the use of a modified multivariate Krogh model. Methods To test our idea and its potential to translate into an in vivo setting through the use of existing imaging techniques, simulation studies were performed comparing the local partial oxygen pressure (pO2) from the proposed multivariate image fusion model to the referenced pO2 derived by Green’s function, which considers the contribution from every vessel segment of an entire three-dimensional tumor vasculature to profile tumor oxygen with high spatial resolution. Results pO2 derived from our fusion approach were close to the referenced pO2 with regression slope near 1.0 and an r2 higher than 0.8 if the voxel size (or the spatial resolution set by functional imaging modality) was less than 200 μm. The simulation also showed that the metabolic rate, blood perfusion, and hemoglobin concentration were dominant factors in tissue oxygenation. The impact of the measurement error of functional imaging to the pO2 precision and accuracy was simulated. A Gaussian error function with FWHM equal to 20 % of blood perfusion or fractional vascular volume measurement contributed to average 7 % statistical error in pO2. Conclusion The simulation results indicate that the fusion of multiple parametric maps through the biophysically derived mathematical models can monitor the intra-tumor spatial variations of hypoxia in tumors with existing imaging methods, and the potential to further investigate different forms of hypoxia, such as chronic and acute hypoxia, in response to cancer therapies. Electronic supplementary material The online version of this article (doi:10.1186/s12938-016-0235-5) contains supplementary material, which is available to authorized users.
Collapse
|
43
|
Abstract
A previous review published in 2012 demonstrated the role of clinical PET for diagnosis and management of brain tumors using mainly FDG, amino acid tracers, and 18F-fluorothymidine. This review provides an update on clinical PET studies, most of which are motivated by prediction of prognosis and planning and monitoring of therapy in gliomas. For FDG, there has been additional evidence supporting late scanning, and combination with 13N ammonia has yielded some promising results. Large neutral amino acid tracers have found widespread applications mostly based on 18F-labeled compounds fluoroethyltyrosine and fluorodopa for targeting biopsies, therapy planning and monitoring, and as outcome markers in clinical trials. 11C-alpha-methyltryptophan (AMT) has been proposed as an alternative to 11C-methionine, and there may also be a role for cyclic amino acid tracers. 18F-fluorothymidine has shown strengths for tumor grading and as an outcome marker. Studies using 18F-fluorocholine (FCH) and 68Ga-labeled compounds are promising but have not yet clearly defined their role. Studies on radiotherapy planning have explored the use of large neutral amino acid tracers to improve the delineation of tumor volume for irradiation and the use of hypoxia markers, in particular 18F-fluoromisonidazole. Many studies employed the combination of PET with advanced multimodal MR imaging methods, mostly demonstrating complementarity and some potential benefits of hybrid PET/MR.
Collapse
Affiliation(s)
- Karl Herholz
- The University of Manchester, Division of Neuroscience and Experimental Psychology Wolfson Molecular Imaging Centre, Manchester, England, United Kingdom.
| |
Collapse
|
44
|
Tong X, Srivatsan A, Jacobson O, Wang Y, Wang Z, Yang X, Niu G, Kiesewetter DO, Zheng H, Chen X. Monitoring Tumor Hypoxia Using (18)F-FMISO PET and Pharmacokinetics Modeling after Photodynamic Therapy. Sci Rep 2016; 6:31551. [PMID: 27546160 PMCID: PMC4992876 DOI: 10.1038/srep31551] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 07/13/2016] [Indexed: 11/09/2022] Open
Abstract
Photodynamic therapy (PDT) is an efficacious treatment for some types of cancers. However, PDT-induced tumor hypoxia as a result of oxygen consumption and vascular damage can reduce the efficacy of this therapy. Measuring and monitoring intrinsic and PDT-induced tumor hypoxia in vivo during PDT is of high interest for prognostic and treatment evaluation. In the present study, static and dynamic (18)F-FMISO PET were performed with mice bearing either U87MG or MDA-MB-435 tumor xenografts immediately before and after PDT at different time points. Significant difference in tumor hypoxia in response to PDT over time was found between the U87MG and MDA-MB-435 tumors in both static and dynamic PET. Dynamic PET with pharmacokinetics modeling further monitored the kinetics of (18)F-FMISO retention to hypoxic sites after treatment. The Ki and k3 parametric analysis provided information on tumor hypoxia by distinction of the specific tracer retention in hypoxic sites from its non-specific distribution in tumor. Dynamic (18)F-FMISO PET with pharmacokinetics modeling, complementary to static PET analysis, provides a potential imaging tool for more detailed and more accurate quantification of tumor hypoxia during PDT.
Collapse
Affiliation(s)
- Xiao Tong
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States.,Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Avinash Srivatsan
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Yu Wang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Zhantong Wang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Xiangyu Yang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Dale O Kiesewetter
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
45
|
Schweifer A, Maier F, Ehrlichmann W, Lamparter D, Kneilling M, Pichler BJ, Hammerschmidt F, Reischl G. [ 18F]Fluoro-azomycin-2´-deoxy-β-d-ribofuranoside - A new imaging agent for tumor hypoxia in comparison with [ 18F]FAZA. Nucl Med Biol 2016; 43:759-769. [PMID: 27693670 DOI: 10.1016/j.nucmedbio.2016.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/25/2016] [Accepted: 08/07/2016] [Indexed: 01/30/2023]
Abstract
INTRODUCTION Radiolabeled 2-nitroimidazoles (azomycins) are a prominent class of biomarkers for PET imaging of hypoxia. [18F]Fluoro-azomycin-α-arabinoside ([18F]FAZA) - already in clinical use - may be seen as α-configuration nucleoside, but enters cells only via diffusion and is not transported by cellular nucleoside transporters. To enhance image contrast in comparison to [18F]FAZA our objective was to 18F-radiolabel an azomycin-2´-deoxyriboside with β-configuration ([18F]FAZDR, [18F]-β-8) to mimic nucleosides more closely and comparatively evaluate it versus [18F]FAZA. METHODS Precursor and cold standards for [18F]FAZDR were synthesized from methyl 2-deoxy-d-ribofuranosides α- and β-1 in 6 steps yielding precursors α- and β-5. β-5 was radiolabeled in a GE TRACERlab FXF-N synthesizer in DMSO and deprotected with NH4OH to give [18F]FAZDR ([18F]-β-8). [18F]FAZA or [18F]FAZDR was injected in BALB/c mice bearing CT26 colon carcinoma xenografts, PET scans (10min) were performed after 1, 2 and 3h post injection (p.i.). On a subset of mice injected with [18F]FAZDR, we analyzed biodistribution. RESULTS [18F]FAZDR was obtained in non-corrected yields of 10.9±2.4% (9.1±2.2GBq, n=4) 60min EOB, with radiochemical purity >98% and specific activity >50GBq/μmol. Small animal PET imaging showed a decrease in uptake over time for both [18F]FAZDR (1h p.i.: 0.56±0.22% ID/cc, 3h: 0.17±0.08% ID/cc, n=9) and [18F]FAZA (1h: 1.95±0.59% ID/cc, 3h: 0.87±0.55% ID/cc), whereas T/M ratios were significantly higher for [18F]FAZDR at 1h (2.76) compared to [18F]FAZA (1.69, P<0.001), 3h p.i. ratios showed no significant difference. Moreover, [18F]FAZDR showed an inverse correlation between tracer uptake in carcinomas and oxygen breathing, while muscle tissue uptake was not affected by switching from air to oxygen. CONCLUSIONS First PET imaging results with [18F]FAZDR showed advantages over [18F]FAZA regarding higher tumor contrast at earlier time points p.i. Availability of precursor and cold fluoro standard together with high output radiosynthesis will allow for a more detailed quantitative evaluation of [18F]FAZDR, especially with regard to mechanistic studies whether active transport processes are involved, compared to passive diffusion as observed for [18F]FAZA.
Collapse
Affiliation(s)
- Anna Schweifer
- Institute of Organic Chemistry, University of Vienna, Vienna, Austria
| | - Florian Maier
- Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Walter Ehrlichmann
- Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Denis Lamparter
- Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Manfred Kneilling
- Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany; Department of Dermatology, University of Tübingen, Tübingen, Germany
| | - Bernd J Pichler
- Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | | | - Gerald Reischl
- Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
46
|
Luderer MJ, Muz B, de la Puente P, Chavalmane S, Kapoor V, Marcelo R, Biswas P, Thotala D, Rogers B, Azab AK. A Hypoxia-Targeted Boron Neutron Capture Therapy Agent for the Treatment of Glioma. Pharm Res 2016; 33:2530-9. [PMID: 27401411 DOI: 10.1007/s11095-016-1977-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 06/20/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE Boron neutron capture therapy (BNCT) has the potential to become a viable cancer treatment modality, but its clinical translation has been limited by the poor tumor selectivity of agents. To address this unmet need, a boronated 2-nitroimidazole derivative (B-381) was synthesized and evaluated for its capability of targeting hypoxic glioma cells. METHODS B-381 has been synthesized from a 1-step reaction. Using D54 and U87 glioma cell lines, the in vitro cytotoxicity and cellular accumulation of B-381 has been evaluated under normoxic and hypoxic conditions compared to L-boronophenylalanine (BPA). Furthermore, tumor retention of B-381 was evaluated in vivo. RESULTS B-381 had low cytotoxicity in normal and cancer cells. Unlike BPA, B-381 illustrated preferential retention in hypoxic glioma cells compared to normoxic glioma cells and normal tissues in vitro. In vivo, B-381 illustrated significantly higher long-term tumor retention compared to BPA, with 9.5-fold and 6.5-fold higher boron levels at 24 and 48 h, respectively. CONCLUSIONS B-381 represents a new class of BNCT agents in which their selectivity to tumors is based on hypoxic tumor metabolism. Further studies are warranted to evaluate B-381 and similar compounds as preclinical candidates for future BNCT clinical trials for the treatment of glioma.
Collapse
Affiliation(s)
- Micah John Luderer
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, Missouri, 63108, USA
| | - Barbara Muz
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, Missouri, 63108, USA
| | - Pilar de la Puente
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, Missouri, 63108, USA
| | - Sanmathi Chavalmane
- Department of Energy, Environmental and Chemical Engineering, Washington University in St. Louis, St. Louis, Missouri, 63130, USA
| | - Vaishali Kapoor
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, Missouri, 63108, USA
| | - Raymundo Marcelo
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, Missouri, 63108, USA
| | - Pratim Biswas
- Department of Energy, Environmental and Chemical Engineering, Washington University in St. Louis, St. Louis, Missouri, 63130, USA
| | - Dinesh Thotala
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, Missouri, 63108, USA
| | - Buck Rogers
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, Missouri, 63108, USA
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave., Room 3103, St. Louis, Missouri, 63108, USA.
| |
Collapse
|
47
|
Panichelli P, Villano C, Cistaro A, Bruno A, Barbato F, Piccardo A, Duatti A. Imaging of Brain Tumors with Copper-64 Chloride: Early Experience and Results. Cancer Biother Radiopharm 2016; 31:159-67. [PMID: 27228278 DOI: 10.1089/cbr.2016.2028] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVES To conduct the first investigational study that is aimed at evaluating the ability of the simple salt (64)CuCl2 to diagnose cerebral tumors in patients affected by glioblastoma multiforme (GBM). METHODS Nineteen patients with a documented history and radiologic evidence of brain tumors were enrolled in the study. Eighteen patients were diagnosed with GBM, and one patient was diagnosed with grade II astrocytoma. After initial cerebral magnetic resonance imaging (MRI), patients were administered with (64)CuCl2 (13 MBq/kg) and brain positron emission tomography (PET)/computed tomography (CT) imaging was performed at 1, 3, and 24 hours after administration. Standardized uptake values (SUVs) were calculated and used to figure out the pharmacokinetic profile of the tracer. Absorbed radiation doses were estimated using OLINDA/EXM. RESULTS Copper-64 chloride clearly visualized brain cancerous lesions within 1 hour after injection, with stable retention of radioactivity at 3 and 24 hours. Excellent agreement was found between PET/CT and MRI. No uptake of the tracer was observed in low-grade astrocytoma. The agent cleared rapidly from the blood and was mostly excreted through the liver, without significant kidney washout. Analysis of time variation of SUVmax values showed persistent uptake in malignant tissues with a slight increase of radioactive concentration at 24 hours. CONCLUSIONS Copper-64 chloride has favorable biological properties for brain imaging and warrants further investigation as a diagnostic tracer for GBM.
Collapse
Affiliation(s)
| | | | - Angelina Cistaro
- 3 Positron Emission Tomography Centre, IRMET , Affidea, Turin, Italy
| | | | | | | | - Adriano Duatti
- 7 Department of Chemical and Pharmaceutical Sciences, University of Ferrara , Ferrara, Italy
| |
Collapse
|
48
|
Hawkins-Daarud A, Rockne R, Corwin D, Anderson ARA, Kinahan P, Swanson KR. In silico analysis suggests differential response to bevacizumab and radiation combination therapy in newly diagnosed glioblastoma. J R Soc Interface 2016. [PMID: 26202682 PMCID: PMC4535409 DOI: 10.1098/rsif.2015.0388] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Recently, two phase III studies of bevacizumab, an anti-angiogenic, for newly diagnosed glioblastoma (GBM) patients were released. While they were unable to statistically significantly demonstrate that bevacizumab in combination with other therapies increases the overall survival of GBM patients, there remains a question of potential benefits for subpopulations of patients. We use a mathematical model of GBM growth to investigate differential benefits of combining surgical resection, radiation and bevacizumab across observed tumour growth kinetics. The differential hypoxic burden after gross total resection (GTR) was assessed along with the change in radiation cell kill from bevacizumab-induced tissue re-normalization when starting therapy for tumours at different diagnostic sizes. Depending on the tumour size at the time of treatment, our model predicted that GTR would remove a variable portion of the hypoxic burden ranging from 11% to 99.99%. Further, our model predicted that the combination of bevacizumab with radiation resulted in an additional cell kill ranging from 2.6×107 to 1.1×1010 cells. By considering the outcomes given individual tumour kinetics, our results indicate that the subpopulation of patients who would receive the greatest benefit from bevacizumab and radiation combination therapy are those with large, aggressive tumours and who are not eligible for GTR.
Collapse
Affiliation(s)
| | - Russell Rockne
- Department of Neurological Surgery, Northwestern University, Chicago, IL 60611, USA
| | - David Corwin
- Department of Neurological Surgery, Northwestern University, Chicago, IL 60611, USA
| | | | - Paul Kinahan
- Department of Radiology, University of Washington, Seattle, WA 98195-7987, USA
| | - Kristin R Swanson
- Department of Neurological Surgery, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
49
|
Nedergaard MK, Michaelsen SR, Perryman L, Erler J, Poulsen HS, Stockhausen MT, Lassen U, Kjaer A. Comparison of 18F-FET and 18F-FLT small animal PET for the assessment of anti-VEGF treatment response in an orthotopic model of glioblastoma. Nucl Med Biol 2016; 43:198-205. [DOI: 10.1016/j.nucmedbio.2015.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 12/18/2015] [Accepted: 12/18/2015] [Indexed: 01/31/2023]
|
50
|
Toyonaga T, Hirata K, Yamaguchi S, Hatanaka KC, Yuzawa S, Manabe O, Kobayashi K, Watanabe S, Shiga T, Terasaka S, Kobayashi H, Kuge Y, Tamaki N. (18)F-fluoromisonidazole positron emission tomography can predict pathological necrosis of brain tumors. Eur J Nucl Med Mol Imaging 2016; 43:1469-76. [PMID: 26841941 DOI: 10.1007/s00259-016-3320-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 01/15/2016] [Indexed: 01/09/2023]
Abstract
PURPOSE Tumor necrosis is one of the indicators of tumor aggressiveness. (18)F-fluoromisonidazole (FMISO) is the most widely used positron emission tomography (PET) tracer to evaluate severe hypoxia in vivo. Because severe hypoxia causes necrosis, we hypothesized that intratumoral necrosis can be detected by FMISO PET in brain tumors regardless of their histopathology. We applied FMISO PET to various types of brain tumors before tumor resection and evaluated the correlation between histopathological necrosis and FMISO uptake. METHODS This study included 59 brain tumor patients who underwent FMISO PET/computed tomography before any treatments. According to the pathological diagnosis, the brain tumors were divided into three groups: astrocytomas (group 1), neuroepithelial tumors except for astrocytomas (group 2), and others (group 3). Two experienced neuropathologists evaluated the presence of necrosis in consensus. FMISO uptake in the tumor was evaluated visually and semi-quantitatively using the tumor-to-normal cerebellum ratio (TNR). RESULTS In visual analyses, 26/27 cases in the FMISO-positive group presented with necrosis, whereas 28/32 cases in the FMISO-negative group did not show necrosis. Mean TNRs with and without necrosis were 3.49 ± 0.97 and 1.43 ± 0.42 (p < 0.00001) in group 1, 2.91 ± 0.83 and 1.44 ± 0.20 (p < 0.005) in group 2, and 2.63 ± 1.16 and 1.35 ± 0.23 (p < 0.05) in group 3, respectively. Using a cut-off value of TNR = 1.67, which was calculated by normal reference regions of interest, we could predict necrosis with sensitivity, specificity, and accuracy of 96.7, 93.1, and 94.9 %, respectively. CONCLUSIONS FMISO uptake within the lesion indicated the presence of histological micro-necrosis. When we used a TNR of 1.67 as the cut-off value, intratumoral micro-necrosis was sufficiently predictable. Because the presence of necrosis implies a poor prognosis, our results suggest that FMISO PET could provide important information for treatment decisions or surgical strategies of any type of brain tumor.
Collapse
Affiliation(s)
- Takuya Toyonaga
- Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Kenji Hirata
- Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Shigeru Yamaguchi
- Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-Ku, Sapporo, 060-8638, Japan. .,Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo, Japan.
| | - Kanako C Hatanaka
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Sayaka Yuzawa
- Department of Cancer Pathology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Osamu Manabe
- Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Kentaro Kobayashi
- Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Shiro Watanabe
- Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Tohru Shiga
- Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Shunsuke Terasaka
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroyuki Kobayashi
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuji Kuge
- Central Institute of Isotope Science, Hokkaido University, Sapporo, Japan
| | - Nagara Tamaki
- Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-Ku, Sapporo, 060-8638, Japan
| |
Collapse
|