1
|
Chen R, Wang H. Time-to-Event Endpoints in Imaging Biomarker Studies. J Magn Reson Imaging 2025; 61:561-567. [PMID: 38739014 PMCID: PMC11706316 DOI: 10.1002/jmri.29446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/01/2024] [Accepted: 05/01/2024] [Indexed: 05/14/2024] Open
Abstract
Time-to-event endpoints are widely used as measures of patients' well-being and indicators of prognosis. In imaging-based biomarker studies, there are increasingly more studies that focus on examining imaging biomarkers' prognostic or predictive utilities on those endpoints, whether in a trial or an observational study setting. In this educational review article, we briefly introduce some basic concepts of time-to-event endpoints and point out potential pitfalls in the context of imaging biomarker research in hope of improving radiologists' understanding of related subjects. Besides, we have included some review and discussions on the benefits of using time-to-event endpoints and considerations on selecting overall survival or progression-free survival for primary analysis. LEVEL OF EVIDENCE: 5 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Ruizhe Chen
- The Sidney Kimmel Comprehensive Cancer Center, Division of Quantitative Sciences, Department of OncologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Hao Wang
- The Sidney Kimmel Comprehensive Cancer Center, Division of Quantitative Sciences, Department of OncologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
2
|
Liu P, Huang J, Duan W, Song T, Wang J, Zhang C, Du Y, Chen Y, Fu R, Lu J, Chen Z. FET PET provides adjunctive value to FDG PET in distinction of spinal cord tumors. Heliyon 2024; 10:e33353. [PMID: 39040377 PMCID: PMC11261781 DOI: 10.1016/j.heliyon.2024.e33353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/23/2024] [Accepted: 06/19/2024] [Indexed: 07/24/2024] Open
Abstract
Objective This study aimed to compare the diagnostic efficacy of O-(2-18F-fluoroethyl)-l-tyrosine (18F-FET) PET and 2-deoxy-2-[18F]fluoro-d-deoxyglucose (18F-FDG) PET for spinal cord lesions. Materials and methods Paired preoperative 18F-FDG PET/MRI and 18F-FET PET/MRI scans were conducted on patients with suspected spinal cord tumors. Clinical manifestations and PET performance, including SUVmean, SUVmax, TBRmean, TBRmax, metabolic tumor volume (MTV), and total lesion metabolism (TLM), and tumor volume, were compared using group analysis and receiver operating characteristic (ROC) curves. Results Thirty-five patients were categorized into three groups based on their pathological diagnosis: high-grade tumors (HGTs, n = 6), low-grade tumors (LGTs, n = 19), and non-tumor diseases (NTDs, n = 10). The background SUVmean of 18F-FET PET was significantly lower than that of 18F-FDG PET (p < 0.0001), while the delineated tumor volumes showed no significant difference (p > 0.05). The mass SUVmean, SUVmax, MTV, and TLM values of both 18F-FDG PET and 18F-FET PET were statistically different between HGTs and LGTs (p < 0.05). Similarly, the mass SUVmax, TBRmax, MTV, and TLM values of both 18F-FDG PET and 18F-FET PET, as well as the mass SUVmean of 18F-FET PET, exhibited statistical differences between HGTs and NTDs (p < 0.05). But none were able to distinguish LGTs and NTDs (p > 0.05). Notably, 18F-FET PET provided valuable supporting diagnostic evidence in 1 case of mixed neuronal-glial tumor (MNGT) and 2 cases of intramedullary inflammatory lesions. Optimal cut-off values of all measured parameters for distinguishing tumors and NTDs were determined through ROC analysis. Conclusion 18F-FET PET presented comparable diagnostic performance to 18F-FDG PET in differentiating HGTs, LGTs, and NTDs, but exhibited particular utility in MNGT and inflammatory lesions.
Collapse
Affiliation(s)
- Penghao Liu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Jing Huang
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Wanru Duan
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Tianbin Song
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Jiyuan Wang
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Can Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yueqi Du
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Ye Chen
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Renkui Fu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Zan Chen
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| |
Collapse
|
3
|
Beuriat PA, Flaus A, Portefaix A, Szathmari A, Janier M, Hermier M, Lorthois-Ninou S, Scheiber C, Isal S, Costes N, Merida I, Lancelot S, Vasiljevic A, Leblond P, Faure Conter C, Saunier C, Kassai B, Vinchon M, Di Rocco F, Mottolese C. Preoperative 11 C-Methionine PET-MRI in Pediatric Infratentorial Tumors. Clin Nucl Med 2024; 49:381-386. [PMID: 38498623 DOI: 10.1097/rlu.0000000000005174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
PURPOSE MRI is the main imaging modality for pediatric brain tumors, but amino acid PET can provide additional information. Simultaneous PET-MRI acquisition allows to fully assess the tumor and lower the radiation exposure. Although symptomatic posterior fossa tumors are typically resected, the patient management is evolving and will benefit from an improved preoperative tumor characterization. We aimed to explore, in children with newly diagnosed posterior fossa tumor, the complementarity of the information provided by amino acid PET and MRI parameters and the correlation to histopathological results. PATIENTS AND METHODS Children with a newly diagnosed posterior fossa tumor prospectively underwent a preoperative 11 C-methionine (MET) PET-MRI. Images were assessed visually and semiquantitatively. Using correlation, minimum apparent diffusion coefficient (ADC min ) and contrast enhancement were compared with MET SUV max . The diameter of the enhancing lesions was compared with metabolic tumoral volume. Lesions were classified according to the 2021 World Health Organization (WHO) classification. RESULTS Ten children were included 4 pilocytic astrocytomas, 2 medulloblastomas, 1 ganglioglioma, 1 central nervous system embryonal tumor, and 1 schwannoma. All lesions showed visually increased MET uptake. A negative moderate correlation was found between ADC min and SUV max values ( r = -0.39). Mean SUV max was 3.8 (range, 3.3-4.2) in WHO grade 4 versus 2.5 (range, 1.7-3.0) in WHO grade 1 lesions. A positive moderate correlation was found between metabolic tumoral volume and diameter values ( r = 0.34). There was no correlation between SUV max and contrast enhancement intensity ( r = -0.15). CONCLUSIONS Preoperative 11 C-MET PET and MRI could provide complementary information to characterize pediatric infratentorial tumors.
Collapse
Affiliation(s)
| | | | | | - Alexandru Szathmari
- From the Department of Pediatric Neurosurgery, Hôpital Femme Mère Enfant, Hospices Civils de Lyon
| | | | - Marc Hermier
- Department of Neuroradiology, Hôpital Neurologique et Neurochirurgical P. Wertheimer, Hospices Civils de Lyon
| | - Sylvie Lorthois-Ninou
- Department of Pediatric Radiology, Hôpital Femme Mère Enfant, Hospices Civils de Lyon
| | | | - Sibel Isal
- Department of Nuclear Medicine, Hospices Civils de Lyon
| | | | | | | | | | - Pierre Leblond
- Institut d'Hématologie et d'Oncologie Pédiatrique (IHOPe), Centre Léon Bérard, Lyon, France
| | - Cécile Faure Conter
- Institut d'Hématologie et d'Oncologie Pédiatrique (IHOPe), Centre Léon Bérard, Lyon, France
| | - Clarisse Saunier
- EPICIME-CIC 1407 de Lyon, Inserm, Département d'Épidémiologie Clinique, Hospices Civils de Lyon
| | | | - Matthieu Vinchon
- From the Department of Pediatric Neurosurgery, Hôpital Femme Mère Enfant, Hospices Civils de Lyon
| | | | - Carmine Mottolese
- From the Department of Pediatric Neurosurgery, Hôpital Femme Mère Enfant, Hospices Civils de Lyon
| |
Collapse
|
4
|
Chou SC, Chen YN, Huang HY, Kuo MF, Wong TT, Kuo SH, Yang SH. Contemporary Management of Pediatric Brainstem Tumors. Adv Tech Stand Neurosurg 2024; 49:231-254. [PMID: 38700687 DOI: 10.1007/978-3-031-42398-7_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2025]
Abstract
Brain tumors are the second most common malignancy in childhood. Around 15-20% of pediatric brain tumors occur in the brainstem. The most common type of brainstem tumor are diffuse tumors in the ventral pons, whereas focal tumors tend to arise from the midbrain, medulla, and dorsal pons. Glioma is the most common pathological entity. Contemporary management consists of surgery, radiotherapy, chemotherapy, and other adjuvant treatment. Surgical options range from biopsy to radical excision. Biopsy can be performed for diagnostic and prognostic purposes, or in the setting of clinical trials, mainly for diffuse intrinsic pontine gliomas. For focal tumors, surgeons need to carefully balance clinical outcomes against possible neurological sequelae in order to achieve maximal safe resection. Radiotherapy is essential for control of high-grade tumors and may be applied to residual or recurrent low-grade tumors. Proton therapy may provide similar efficacy and less neurotoxicity in comparison to conventional photon therapy. Oncological treatment continues to evolve from conventional chemotherapy to targeted therapy, immunotherapy, and other novel treatment methods and holds great potential as adjuvant therapy for pediatric brainstem tumors.
Collapse
Affiliation(s)
- Sheng-Che Chou
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Traumatology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Ning Chen
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu County, Taiwan
| | - Hsin-Yi Huang
- Department of Pathology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Meng-Fai Kuo
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Tai-Tong Wong
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Sung-Hsin Kuo
- Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Shih-Hung Yang
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
5
|
Spencer D, Bonner ER, Tor-Díez C, Liu X, Bougher K, Prasad R, Gordish-Dressman H, Eze A, Packer RJ, Nazarian J, Linguraru MG, Bornhorst M. Tumor volume features predict survival outcomes for patients diagnosed with diffuse intrinsic pontine glioma. Neurooncol Adv 2024; 6:vdae151. [PMID: 39434924 PMCID: PMC11492488 DOI: 10.1093/noajnl/vdae151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024] Open
Abstract
Background Diffuse intrinsic pontine glioma (DIPG) is a fatal childhood central nervous system tumor. Diagnosis and monitoring of tumor response to therapy is based on magnetic resonance imaging (MRI). MRI-based analyses of tumor volume and appearance may aid in the prediction of patient overall survival (OS). Methods Contrast-enhanced T1- and FLAIR/T2-weighted MR images were retrospectively collected from children with classical DIPG diagnosed by imaging (n = 43 patients). MRI features were evaluated at diagnosis (n = 43 patients) and post-radiation (n = 40 patients) to determine OS outcome predictors. Features included 3D tumor volume (Twv), contrast-enhancing tumor core volume (Tc), Tc relative to Twv (TC/Twv), and Twv relative to whole brain volume. Support vector machine (SVM) learning was used to identify feature combinations that predicted OS outcome (defined as OS shorter or longer than 12 months from diagnosis). Results Features associated with poor OS outcome included the presence of contrast-enhancing tumor at diagnosis, >15% Tc/Twv post-radiation therapy (RT), and >20% ∆Tc/Twv post-RT. Consistently, SVM learning identified Tc/Twv at diagnosis (prediction accuracy of 74%) and ∆Tc/Twv at <2 months post-RT (accuracy = 75%) as primary features of poor survival. Conclusions This study demonstrates that tumor imaging features at diagnosis and within 4 months of RT can predict differential OS outcomes in DIPG. These findings provide a framework for incorporating tumor volume-based predictive analyses into the clinical setting, with the potential for treatment customization based on tumor risk characteristics and future applications of machine-learning-based analysis.
Collapse
Affiliation(s)
- D’Andre Spencer
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, District of Columbia, USA
- Institute for Clinical and Translational Science, University of California, Irvine, California, USA
| | - Erin R Bonner
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, District of Columbia, USA
| | - Carlos Tor-Díez
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, District of Columbia, USA
| | - Xinyang Liu
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, District of Columbia, USA
| | - Kristen Bougher
- School of Medicine and Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Rachna Prasad
- Department of Oncology, University Children’s Hospital Zürich, Zürich, Switzerland
| | - Heather Gordish-Dressman
- Department of Biostatistics, Children’s National Hospital, Washington, District of Columbia, USA
| | - Augustine Eze
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, District of Columbia, USA
| | - Roger J Packer
- Brain Tumor Institute, Children’s National Hospital, Washington, District of Columbia, USA
| | - Javad Nazarian
- Brain Tumor Institute, Children’s National Hospital, Washington, District of Columbia, USA
- School of Medicine and Health Sciences, The George Washington University, Washington, District of Columbia, USA
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, District of Columbia, USA
| | - Marius George Linguraru
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, District of Columbia, USA
| | - Miriam Bornhorst
- Stanley Manne Children’s Research Institute at Lurie Children’s, Chicago, Illinois, USA
- Department of Hematology, Oncology, Neuro-oncology and Stem Cell Transplant, Ann & Robert H. Lurie Children’s Hospital of Chicago, Illinois, USA
- Brain Tumor Institute, Children’s National Hospital, Washington, District of Columbia, USA
- School of Medicine and Health Sciences, The George Washington University, Washington, District of Columbia, USA
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, District of Columbia, USA
| |
Collapse
|
6
|
Lovibond S, Gewirtz AN, Pasquini L, Krebs S, Graham MS. The promise of metabolic imaging in diffuse midline glioma. Neoplasia 2023; 39:100896. [PMID: 36944297 PMCID: PMC10036941 DOI: 10.1016/j.neo.2023.100896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 02/10/2023] [Accepted: 03/13/2023] [Indexed: 03/23/2023]
Abstract
Recent insights into histopathological and molecular subgroups of glioma have revolutionized the field of neuro-oncology by refining diagnostic categories. An emblematic example in pediatric neuro-oncology is the newly defined diffuse midline glioma (DMG), H3 K27-altered. DMG represents a rare tumor with a dismal prognosis. The diagnosis of DMG is largely based on clinical presentation and characteristic features on conventional magnetic resonance imaging (MRI), with biopsy limited by its delicate neuroanatomic location. Standard MRI remains limited in its ability to characterize tumor biology. Advanced MRI and positron emission tomography (PET) imaging offer additional value as they enable non-invasive evaluation of molecular and metabolic features of brain tumors. These techniques have been widely used for tumor detection, metabolic characterization and treatment response monitoring of brain tumors. However, their role in the realm of pediatric DMG is nascent. By summarizing DMG metabolic pathways in conjunction with their imaging surrogates, we aim to elucidate the untapped potential of such imaging techniques in this devastating disease.
Collapse
Affiliation(s)
- Samantha Lovibond
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexandra N Gewirtz
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Luca Pasquini
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Simone Krebs
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Radiochemistry and Imaging Sciences Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Maya S Graham
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
7
|
11C-methionine PET imaging characteristics in children with diffuse intrinsic pontine gliomas and relationship to survival and H3 K27M mutation status. Eur J Nucl Med Mol Imaging 2023; 50:1709-1719. [PMID: 36697961 DOI: 10.1007/s00259-022-06105-z] [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: 09/19/2022] [Accepted: 12/30/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE This study aimed to describe 11C-methionine (11C-MET) PET imaging characteristics in patients with paediatric diffuse intrinsic pontine glioma (DIPG) and correlate them with survival and H3 K27M mutation status. METHODS We retrospectively analysed 98 children newly diagnosed with DIPG who underwent 11C-MET PET. PET imaging characteristics evaluated included uptake intensity, uniformity, metabolic tumour volume (MTV), and total lesion methionine uptake (TLMU). The maximum, mean, and peak of the tumour-to-background ratio (TBR), calculated as the corresponding standardised uptake values (SUV) divided by the mean reference value, were also recorded. The associations between the PET imaging characteristics and clinical outcomes in terms of progression-free survival (PFS) and overall survival (OS) and H3 K27M mutation status were assessed, respectively. RESULTS In univariate analysis, imaging characteristics significantly associated with shorter PFS and OS included a higher uniformity grade, higher TBRs, larger MTV, and higher TLMU. In multivariate analysis, larger MTV at diagnosis, shorter symptom duration, and no treatment were significantly correlated with shorter PFS and OS. The PET imaging features were not correlated with H3 K27M mutation status. CONCLUSION Although several imaging features were significantly associated with PFS and OS, only MTV, indicating the size of the active tumour, was identified as a strong independent prognostic factor.
Collapse
|
8
|
Zhao B, Sun K, Zhang Z, Xu T, Zhao L, Liu C, Xiao Y. A rare presentation of primary lateral ventricle H3 K27-altered diffuse midline glioma in a 14-year-old girl: a case description. Quant Imaging Med Surg 2022; 12:5288-5295. [PMID: 36330184 PMCID: PMC9622459 DOI: 10.21037/qims-22-339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/11/2022] [Indexed: 07/25/2023]
Affiliation(s)
- Baolian Zhao
- Department of Radiology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ke Sun
- Department of Radiology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhengwei Zhang
- Department of Pathology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Tao Xu
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Liang Zhao
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chen Liu
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yi Xiao
- Department of Radiology, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
9
|
TSPO PET Imaging as a Potent Non-Invasive Biomarker for Diffuse Intrinsic Pontine Glioma in a Patient-Derived Orthotopic Rat Model. Int J Mol Sci 2022; 23:ijms232012476. [PMID: 36293329 PMCID: PMC9603997 DOI: 10.3390/ijms232012476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/07/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPG), the first cause of cerebral pediatric cancer death, will greatly benefit from specific and non-invasive biomarkers for patient follow-up and monitoring of drug efficacy. Since biopsies are challenging for brain tumors, molecular imaging may be a technique of choice to target and follow tumor evolution. So far, MR remains the imaging technique of reference for DIPG, although it often fails to define the extent of tumors, an essential parameter for therapeutic efficacy assessment. Thanks to its high sensitivity, positron emission tomography (PET) offers a unique way to target specific biomarkers in vivo. We demonstrated in a patient-derived orthotopic xenograft (PDOX) model in the rat that the translocator protein of 18 kDa (TSPO) may be a promising biomarker for monitoring DIPG tumors. We studied the distribution of 18F-DPA-714, a TSPO radioligand, in rats inoculated with HSJD-DIPG-007 cells. The primary DIPG human cell line HSJD-DIPG-007 highly represents this pediatric tumor, displaying the most prevalent DIPG mutations, H3F3A (K27M) and ACVR1 (R206H). Kinetic modeling and parametric imaging using the brain 18F-DPA-714 PET data enabled specific delineation of the DIPG tumor area, which is crucial for radiotherapy dose management.
Collapse
|
10
|
Xu C, Li C, Xing C, Li J, Jiang X. Efficacy of MR diffusion kurtosis imaging for differentiating low-grade from high-grade glioma before surgery: A systematic review and meta-analysis. Clin Neurol Neurosurg 2022; 220:107373. [PMID: 35878557 DOI: 10.1016/j.clineuro.2022.107373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 07/09/2022] [Indexed: 01/16/2023]
Abstract
BACKGROUND Accurate discrimination and diagnosis of low-grade glioma (LGG) and high-grade glioma (HGG) before surgery is clinically important because it affects the patient's outcome and guides the clinicians to select appropriate management. The aim of this study was to evaluate the diagnostic performance of diffusion kurtosis imaging (DKI) for differentiating LGG from HGG. METHODS A literature search of the PubMed, Web of Science, Cochrane Library and EMBASE databases was conducted up to December 15, 2020. Studies that evaluated the diagnostic performance of DKI for differentiating LGG from HGG were selected. Retrieved hits were evaluated by the Quality Assessment of Diagnostic Accuracy Studies-2 tool. Summary sensitivity and specificity were determined, and the data analysis was performed using Stata 14.0 and Review Manager 5.3. RESULTS Thirteen studies with 705 patients were included. The individual sensitivity and specificity of the 13 studies varied from 71% to 100% for sensitivity and 73-100% for specificity. The pooled sensitivity of DKI was 88% (95% confidence interval [CI], 83-91%), and the pooled specificity was 91% (95% CI, 86-95%). The area under the summary receiver operating characteristic curve was 0.93 (95% CI, 0.90-0.95). The pooled diagnostic odds ratio of DKI was 64.85 (95% CI 38.52-109.19). The levels of heterogeneity for sensitivity and specificity across the included studies were high (I2 =66%) and mild (I2 =47.04%), respectively. The multiple subgroup analyses were driven by DKI technique and study region. CONCLUSIONS DKI demonstrated a high diagnostic performance for differentiation of LGG from HGG.
Collapse
Affiliation(s)
- Chang Xu
- Department of Radiology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Chenglong Li
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Chengyan Xing
- Department of Radiology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jun Li
- Department of Radiology,Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Xingyue Jiang
- Department of Radiology, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
11
|
Schelhaas S, Frohwein LJ, Wachsmuth L, Hermann S, Faber C, Schäfers KP, Jacobs AH. Voxel-Based Analysis of the Relation of 3'-Deoxy-3'-[ 18F]fluorothymidine ([ 18F]FLT) PET and Diffusion-Weighted (DW) MR Signals in Subcutaneous Tumor Xenografts Does Not Reveal a Direct Spatial Relation of These Two Parameters. Mol Imaging Biol 2022; 24:359-364. [PMID: 34755247 PMCID: PMC9085704 DOI: 10.1007/s11307-021-01673-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 10/26/2022]
Abstract
PURPOSE Multimodal molecular imaging allows a direct coregistration of different images, facilitating analysis of the spatial relation of various imaging parameters. Here, we further explored the relation of proliferation, as measured by [18F]FLT PET, and water diffusion, as an indicator of cellular density and cell death, as measured by diffusion-weighted (DW) MRI, in preclinical tumor models. We expected these parameters to be negatively related, as highly proliferative tissue should have a higher density of cells, hampering free water diffusion. PROCEDURES Nude mice subcutaneously inoculated with either lung cancer cells (n = 11 A549 tumors, n = 20 H1975 tumors) or colorectal cancer cells (n = 13 Colo205 tumors) were imaged with [18F]FLT PET and DW-MRI using a multimodal bed, which was transferred from one instrument to the other within the same imaging session. Fiducial markers allowed coregistration of the images. An automatic post-processing was developed in MATLAB handling the spatial registration of DW-MRI (measured as apparent diffusion coefficient, ADC) and [18F]FLT image data and subsequent voxel-wise analysis of regions of interest (ROIs) in the tumor. RESULTS Analyses were conducted on a total of 76 datasets, comprising a median of 2890 data points (ranging from 81 to 13,597). Scatterplots showing [18F]FLT vs. ADC values displayed various grades of relations (Pearson correlation coefficient (PCC) varied from - 0.58 to 0.49, median: -0.07). When relating PCC to tumor volume (median: 46 mm3, range: 3 mm3 to 584 mm3), lung tumors tended to have a more pronounced negative spatial relation of [18F]FLT and ADC with increasing tumor size. However, due to the low number of large tumors (> ~ 200 mm3), this conclusion has to be treated with caution. CONCLUSIONS A spatial relation of water diffusion, as measured by DW-MRI, and cellular proliferation, as measured by [18F]FLT PET, cannot be detected in the experimental datasets investigated in this study.
Collapse
Affiliation(s)
- Sonja Schelhaas
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität Münster, Waldeyerstr. 15, 48149, Münster, Germany.
| | - Lynn Johann Frohwein
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität Münster, Waldeyerstr. 15, 48149, Münster, Germany
| | - Lydia Wachsmuth
- Translational Research Imaging Center, Clinic of Radiology, University Hospital of Münster, Münster, Germany
| | - Sven Hermann
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität Münster, Waldeyerstr. 15, 48149, Münster, Germany
| | - Cornelius Faber
- Translational Research Imaging Center, Clinic of Radiology, University Hospital of Münster, Münster, Germany
| | - Klaus P Schäfers
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität Münster, Waldeyerstr. 15, 48149, Münster, Germany
| | - Andreas H Jacobs
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität Münster, Waldeyerstr. 15, 48149, Münster, Germany
- Department of Geriatric Medicine, Johanniter Hospital, Bonn, Germany
| |
Collapse
|
12
|
Duran-Peña A, Ducray F, Ramirez C, Bauchet L, Constans JM, Grand S, Guillamo JS, Larrieu-Ciron D, Frappaz D, Pyatigorskaya N, Savatovsky J, Loiseau H, Duverneuil NM, Laigle-Donadey F. Adult brainstem glioma differential diagnoses: an MRI-based approach in a series of 68 patients. J Neurol 2022; 269:4349-4362. [PMID: 35441889 DOI: 10.1007/s00415-022-11070-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/24/2022] [Accepted: 03/06/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Brainstem gliomas are rare in adults. The diagnosis is often difficult, as some teams still consider brainstem biopsies dangerous and often avoid this procedure. The aim of this study was to describe differential diagnoses that can mimic brainstem glioma, to help clinicians avoid diagnostic and therapeutic mistakes, and to propose a diagnostic algorithm according to radiological presentations. METHODS The French network of adult brainstem gliomas (GLITRAD) retrospectively collected all reported cases of differential diagnoses between 2006 and 2017. The inclusion criteria were as follows: age over 18 years, lesion epicenter in the brainstem, radiological pattern suggestive of a glioma and diagnostic confirmation (histopathological or not, depending on the disease). RESULTS We identified a total of 68 cases. Most cases (58/68, 85%) presented as contrast-enhancing lesions. The most frequent final diagnosis in this group was metastases in 24/58 (41%), followed by central nervous system lymphoma in 8/58 (14%). Conversely, MRI findings revealed 10/68 nonenhancing lesions. The most frequent diagnosis in this group was demyelinating disease (3/10, 30%). CONCLUSION The risk of diagnostic mistakes illustrates the need to consider the more systematic use of a brainstem biopsy when reasonably possible. However, we propose an MRI-based approach to the differential diagnosis of gliomas to limit the risk of misdiagnosis in cases where a biopsy is not a reasonable option.
Collapse
Affiliation(s)
- Alberto Duran-Peña
- Service de Neurologie 2, Division Mazarin CHU Pitié-Salpêtrière, Paris, France
| | - François Ducray
- Service de Neuro-Oncologie, Hôpital Pierre Wertheimer, CHU Lyon, Lyon, France
| | - Carole Ramirez
- Service de Neurochirurgie, Hôpital Roger Salengro, CHU Lille, Lille, France
| | - Luc Bauchet
- Service de Neurochirurgie, CHU Montpellier, Montpellier, France
| | | | - Sylvie Grand
- Service de Neuroradiologie, CHU Grenoble Alpes, Grenoble, France
| | | | | | | | | | - Julien Savatovsky
- Service de Neuroradiologie, Fondation Ophtalmologique Rothschild, Paris, France
| | - Hugues Loiseau
- Service de Neurochirurgie, CHU Bordeaux, Bordeaux, France
| | | | | |
Collapse
|
13
|
Emfietzoglou M, Terentes-Printzios D, Kotronias RA, Marin F, Montalto C, De Maria GL, Banning AP. The spectrum and systemic associations of microvascular dysfunction in the heart and other organs. NATURE CARDIOVASCULAR RESEARCH 2022; 1:298-311. [PMID: 39196132 DOI: 10.1038/s44161-022-00045-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 03/02/2022] [Indexed: 08/29/2024]
Abstract
Microvascular dysfunction (MVD) contributes to several conditions that increase morbidity and mortality, including ischemic heart disease, heart failure, dementia, chronic kidney disease and hypertension. Consequently, MVD imposes a substantial burden on healthcare systems worldwide. In comparison to macrovascular dysfunction, MVD has been incompletely investigated, and it remains uncertain whether MVD in an organ constitutes a distinct pathology or a manifestation of a systemic disorder. Here, we summarize and appraise the techniques that are used to diagnose MVD. We review the disorders of the heart, brain and kidneys in which the role of MVD has been highlighted and summarize evidence hinting at a systemic or multi-organ nature of MVD. Finally, we discuss the benefits and limitations of implementing MVD testing in clinical practice with a focus on new interventions that are beginning to emerge.
Collapse
Affiliation(s)
| | | | | | - Federico Marin
- Oxford Heart Centre, Oxford University Hospitals, Oxford, UK
| | | | | | | |
Collapse
|
14
|
Cistaro A, Albano D, Alongi P, Laudicella R, Pizzuto DA, Formica G, Romagnolo C, Stracuzzi F, Frantellizzi V, Piccardo A, Quartuccio N. The Role of PET in Supratentorial and Infratentorial Pediatric Brain Tumors. Curr Oncol 2021; 28:2481-2495. [PMID: 34287265 PMCID: PMC8293135 DOI: 10.3390/curroncol28040226] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE This review aims to provide a summary of the clinical indications and limitations of PET imaging with different radiotracers, including 18F-fluorodeoxyglucose (18F-FDG) and other radiopharmaceuticals, in pediatric neuro-oncology, discussing both supratentorial and infratentorial tumors, based on recent literature (from 2010 to present). METHODS A literature search of the PubMed/MEDLINE database was carried out searching for articles on the use of PET in pediatric brain tumors. The search was updated until December 2020 and limited to original studies published in English after 1 January 2010. RESULTS 18F-FDG PET continues to be successfully employed in different settings in pediatric neuro-oncology, including diagnosis, grading and delineation of the target for stereotactic biopsy, estimation of prognosis, evaluation of recurrence, treatment planning and assessment of treatment response. Nevertheless, non-18F-FDG tracers, especially amino acid analogues seem to show a better performance in each clinical setting. CONCLUSIONS PET imaging adds important information in the diagnostic work-up of pediatric brain tumors. International or national multicentric studies are encouraged in order to collect larger amount of data.
Collapse
Affiliation(s)
- Angelina Cistaro
- Nuclear Medicine Department, Ospedali Galliera, 16128 Genova, Italy; (A.C.); (A.P.)
- AIMN Pediatric Study Group, 20159 Milan, Italy;
| | - Domenico Albano
- Department of Nuclear Medicine, University of Brescia and Spedali Civili Brescia, 25123 Brescia, Italy;
| | - Pierpaolo Alongi
- Unit of Nuclear Medicine, Fondazione Istituto G. Giglio, 90015 Cefalù, Italy
| | - Riccardo Laudicella
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, A.O.U. Policlinico G. Martino, University of Messina, 98125 Messina, Italy; (R.L.); (G.F.); (F.S.)
| | | | - Giuseppe Formica
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, A.O.U. Policlinico G. Martino, University of Messina, 98125 Messina, Italy; (R.L.); (G.F.); (F.S.)
| | - Cinzia Romagnolo
- Nuclear Medicine Unit, Ospedali Riuniti, Torrette di Ancona, 60126 Ancona, Italy;
| | - Federica Stracuzzi
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, A.O.U. Policlinico G. Martino, University of Messina, 98125 Messina, Italy; (R.L.); (G.F.); (F.S.)
| | - Viviana Frantellizzi
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, 00161 Rome, Italy;
| | - Arnoldo Piccardo
- Nuclear Medicine Department, Ospedali Galliera, 16128 Genova, Italy; (A.C.); (A.P.)
- AIMN Pediatric Study Group, 20159 Milan, Italy;
| | - Natale Quartuccio
- AIMN Pediatric Study Group, 20159 Milan, Italy;
- Nuclear Medicine Unit, A.R.N.A.S. Ospedali Civico, Di Cristina e Benfratelli, 90127 Palermo, Italy
| |
Collapse
|
15
|
Su C, Li S, Chen X, Liu C, Shaghaghi M, Jiang J, Zhang S, Qin Y, Cai K. Predicting cancer malignancy and proliferation in glioma patients: intra-subject inter-metabolite correlation analyses using MRI and MRSI contrast scans. Quant Imaging Med Surg 2021; 11:2721-2732. [PMID: 34079736 DOI: 10.21037/qims-20-1163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background The non-invasive characterization of glioma metabolites would greatly assist the management of glioma patients in the clinical setting. This study investigated the applicability of intra-subject inter-metabolite correlation analyses for differentiating glioma malignancy and proliferation. Methods A total of 17 negative controls (NCs), 39 low-grade gliomas (LGGs) patients, and 25 high-grade gliomas (HGGs) subjects were included in this retrospective study. Amide proton transfer (APT) and magnetization transfer contrast (MTC) imaging contrasts, as well as total choline/total creatine (tCho/tCr) and total N-acetylaspartate/total creatine (tNAA/tCr) ratios quantified from magnetic resonance spectroscopic imaging (MRSI) were co-registered voxel-wise and used to produce three intra-subject inter-metabolite correlation coefficients (IMCCs), namely, RAPT vs . MTC, RAPT vs . tCho/tCr, and RMTC vs . tNAA/tCr. The correlation between the IMCCs and tumor grade and Ki-67 labeling index (LI) for tumor proliferation were explored. The differences in the IMCCs between the three groups were compared with one-way analysis of variance (ANOVA). Finally, regression analysis was used to build a combined model with multiple IMCCs to improve the diagnostic performance for tumor grades based on receiver operator characteristic curves. Results Compared with the NCs, gliomas showed stronger inter-metabolic correlations. RAPT vs . MTC was significantly different among the three groups (NC vs. LGGs vs. HGGs: -0.18±0.38 vs. -0.40±0.34 vs. -0.70±0.29, P<0.0001). No significant differences were detected in RMTC vs . tNAA/tCr among the three groups. RAPT vs . MTC and RAPT vs . tCho/tCr correlated significantly with tumor grade (R=-0.41, P=0.001 and R=0.448, P<0.001, respectively). However, only RAPT vs . MTC was mildly correlated with Ki-67 (R=-0.33, P=0.02). RAPT vs . MTC and RAPT vs . tCho/tCr achieved areas under the curve (AUCs) of 0.754 and 0.71, respectively, for differentiating NCs from gliomas; and 0.77 and 0.78, respectively, for differentiating LGGs from HGGs. The combined multi-IMCCs model improved the correlation with the Ki-67 LI (R=0.46, P=0.0008) and the tumor-grade stratification with AUC increased to 0.85 (sensitivity: 80.0%, specificity: 79.5%). Conclusions This study demonstrated that glioma patients showed stronger inter-metabolite correlations than control subjects, and the IMCCs were significantly correlated with glioma grade and proliferation. The multi-IMCCs combined model further improved the performance of clinical diagnosis.
Collapse
Affiliation(s)
- Changliang Su
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Shihui Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaowei Chen
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengxia Liu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mehran Shaghaghi
- Department of Radiology, the University of Illinois at Chicago, Chicago, Illinois, USA
| | - Jingjing Jiang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shun Zhang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanyuan Qin
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kejia Cai
- Department of Radiology, the University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
16
|
Leach JL, Roebker J, Schafer A, Baugh J, Chaney B, Fuller C, Fouladi M, Lane A, Doughman R, Drissi R, DeWire-Schottmiller M, Ziegler DS, Minturn JE, Hansford JR, Wang SS, Monje-Deisseroth M, Fisher PG, Gottardo NG, Dholaria H, Packer R, Warren K, Leary SES, Goldman S, Bartels U, Hawkins C, Jones BV. MR imaging features of diffuse intrinsic pontine glioma and relationship to overall survival: report from the International DIPG Registry. Neuro Oncol 2021; 22:1647-1657. [PMID: 32506137 DOI: 10.1093/neuonc/noaa140] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND This study describes imaging features of diffuse intrinsic pontine glioma (DIPG) and correlates with overall survival (OS) and histone mutation status in the International DIPG Registry (IDIPGR). METHODS Four hundred cases submitted to the IDIPGR with a local diagnosis of DIPG and baseline MRI were evaluated by consensus review of 2 neuroradiologists; 43 cases were excluded (inadequate imaging or alternative diagnoses). Agreement between reviewers, association with histone status, and univariable and multivariable analyses relative to OS were assessed. RESULTS On univariable analysis imaging features significantly associated with worse OS included: extrapontine extension, larger size, enhancement, necrosis, diffusion restriction, and distant disease. On central review, 9.5% of patients were considered not to have DIPG. There was moderate mean agreement of MRI features between reviewers. On multivariable analysis, chemotherapy, age, and distant disease were predictors of OS. There was no difference in OS between wild-type and H3 mutated cases. The only imaging feature associated with histone status was the presence of ill-defined signal infiltrating pontine fibers. CONCLUSIONS Baseline imaging features are assessed in the IDIPGR. There was a 9.5% discordance in DIPG diagnosis between local and central review, demonstrating need for central imaging confirmation for prospective trials. Although several imaging features were significantly associated with OS (univariable), only age and distant disease were significant on multivariable analyses. There was limited association of imaging features with histone mutation status, although numbers are small and evaluation exploratory.
Collapse
Affiliation(s)
- James L Leach
- Department of Radiology and Medical Imaging, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - James Roebker
- Department of Radiology and Medical Imaging, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Austin Schafer
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Joshua Baugh
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Neuro-oncology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Brooklyn Chaney
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Christine Fuller
- Department of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Maryam Fouladi
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Adam Lane
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Renee Doughman
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Rachid Drissi
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | | | - Jane E Minturn
- Division of Oncology, Children's Hospital of Philadelphia, Pennsylvania
| | - Jordan R Hansford
- Children's Cancer Centre, Royal Children's Hospital; Murdoch Children's Research Institute; University of Melbourne, Melbourne, Australia
| | - Stacie S Wang
- Children's Cancer Centre, Royal Children's Hospital; Murdoch Children's Research Institute; University of Melbourne, Melbourne, Australia
| | | | - Paul G Fisher
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, California
| | | | - Hetal Dholaria
- Department of Oncology, Perth Children's Hospital, Perth, AU
| | - Roger Packer
- Division of Oncology, Children's National Medical Center, Washington, DC
| | - Katherine Warren
- Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Harvard Cancer Center, Boston Massachusetts
| | - Sarah E S Leary
- Cancer and Blood Disorders Center, Seattle Children's, Seattle, Washington
| | - Stewart Goldman
- Division of Oncology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Ute Bartels
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, CA
| | - Cynthia Hawkins
- Division of Pathology, The Hospital for Sick Children, Toronto, CA
| | - Blaise V Jones
- Department of Radiology and Medical Imaging, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
17
|
Morana G, Tortora D, Bottoni G, Puntoni M, Piatelli G, Garibotto F, Barra S, Giannelli F, Cistaro A, Severino M, Verrico A, Milanaccio C, Massimino M, Garrè ML, Rossi A, Piccardo A. Correlation of multimodal 18F-DOPA PET and conventional MRI with treatment response and survival in children with diffuse intrinsic pontine gliomas. Am J Cancer Res 2020; 10:11881-11891. [PMID: 33204317 PMCID: PMC7667677 DOI: 10.7150/thno.50598] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 10/03/2020] [Indexed: 01/29/2023] Open
Abstract
To evaluate the contribution of 18F-dihydroxyphenylalanine (DOPA) PET in association with conventional MRI in predicting treatment response and survival outcome of pediatric patients with diffuse intrinsic pontine gliomas (DIPGs). Methods: We retrospectively analyzed 19 children with newly diagnosed DIPGs who underwent 18F-DOPA PET/CT and conventional MRI within one week of each other at admission and subsequent MRI follow-up. Following co-registration and fusion of PET and MRI, 18F-DOPA uptake avidity and extent (PET tumor volume and uniformity) at admission, along with MRI indices including presence of ring contrast-enhancement, tumor volume at admission and at maximum response following first-line treatment, were evaluated and correlated with overall survival (OS). The association between 18F-DOPA uptake tumor volume at admission and MRI tumor volume following treatment was evaluated. Statistics included Wilcoxon signed-rank and Mann-Whitney U tests, Kaplan-Meier OS curve and Cox analysis. Results: DIPGs with a 18F-DOPA uptake Tumor/Striatum (T/S) ratio >1 presented an OS ≤ 12 months and lower degree of tumor volume reduction following treatment (p = 0.001). On multivariate analysis, T/S (p = 0.001), ring enhancement (p = 0.01) and the degree of MRI tumor volume reduction (p = 0.01) independently correlated with OS. In all patients, areas of increased 18F-DOPA uptake overlapped with regions demonstrating more prominent residual components/lack of response following treatment. Conclusions:18F-DOPA PET provides useful information for evaluating the metabolism of DIPGs. T/S ratio is an independent predictor of outcome. 18F-DOPA uptake extent delineates tumoral regions with a more aggressive biological behaviour, less sensitive to first line treatment.
Collapse
|
18
|
Felker J, Broniscer A. Improving long-term survival in diffuse intrinsic pontine glioma. Expert Rev Neurother 2020; 20:647-658. [PMID: 32543245 DOI: 10.1080/14737175.2020.1775584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Diffuse intrinsic pontine glioma (DIPG) is an almost universally fatal pediatric brain cancer. There has been no improvement in event-free survival (EFS) or overall survival (OS) despite immense effort through a multitude of clinical trials to find a cure. Recently, there has been a surge in the knowledge of DIPG biology, including the discovery of a recurrent H3F3A mutation in over 80% of these tumors. AREAS COVERED The authors review the most recent approaches to diagnosis and treatment of DIPG including chemotherapy, biologics, surgical approaches, and immunotherapy. EXPERT OPINION The authors propose four main opportunities to improve long-term survival. First, patients should be enrolled in scientifically sound clinical trials that include molecularly profiling either via stereotactic biopsy or liquid biopsy. Second, clinical trials should include more innovative endpoints other than traditional EFS and OS such as MRI/PET imaging findings combined with surrogates of activity (e.g. serial liquid biopsies) to better ascertain biologically active treatments. Third, innovative clinical trial approaches are needed to help allow for the rapid development of combination therapies to be tested. Finally, effort should be concentrated on reversing the effects of the histone mutation, as this malfunctioning development program seems to be key to DIPG relentlessness.
Collapse
Affiliation(s)
- James Felker
- Department of Pediatrics, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA.,Pediatric Neuro-Oncology, UPMC Children's Hospital of Pittsburgh , Pittsburgh, PA, USA
| | - Alberto Broniscer
- Department of Pediatrics, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA.,Pediatric Neuro-Oncology, UPMC Children's Hospital of Pittsburgh , Pittsburgh, PA, USA
| |
Collapse
|
19
|
Jaimes C, Vajapeyam S, Brown D, Kao PC, Ma C, Greenspan L, Gupta N, Goumnerova L, Bandopahayay P, Dubois F, Greenwald NF, Zack T, Shapira O, Beroukhim R, Ligon KL, Chi S, Kieran MW, Wright KD, Poussaint TY. MR Imaging Correlates for Molecular and Mutational Analyses in Children with Diffuse Intrinsic Pontine Glioma. AJNR Am J Neuroradiol 2020; 41:874-881. [PMID: 32381545 DOI: 10.3174/ajnr.a6546] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/16/2020] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND PURPOSE Recent advances in molecular techniques have characterized distinct subtypes of diffuse intrinsic pontine gliomas. Our aim was the identification of MR imaging correlates of these subtypes. MATERIALS AND METHODS Initial MRIs from subjects with diffuse intrinsic pontine gliomas recruited for a prospective clinical trial before treatment were analyzed. Retrospective imaging analyses included FLAIR/T2 tumor volume, tumor volume enhancing, the presence of cyst and/or necrosis, median, mean, mode, skewness, kurtosis of ADC tumor volume based on FLAIR, and enhancement at baseline. Molecular subgroups based on EGFR and MGMT mutations were established. Histone mutations were also determined (H3F3A, HIST1H3B, HIST1H3C). Univariate Cox proportional hazards regression was used to test the association of imaging predictors with overall and progression-free survival. Wilcoxon rank sum, Kruskal-Wallis, and Fisher exact tests were used to compare imaging measures among groups. RESULTS Fifty patients had biopsy and MR imaging. The median age at trial registration was 6 years (range, 3.3-17.5 years); 52% were female. On the basis of immunohistochemical results, 48 patients were assigned to 1 of 4 subgroups: 28 in MGMT-/epidermal growth factor receptor (EGFR)-, 14 in MGMT-/EGFR+, 3 in MGMT+/EGFR-, and 3 in MGMT+/EGFR+. Twenty-three patients had histone mutations in H3F3A, 8 in HIST1H3B, and 3 in HIST1H3C. Enhancing tumor volume was near-significantly different across molecular subgroups (P = .04), after accounting for the false discovery rate. Tumor volume enhancing, median, mode, skewness, and kurtosis ADC T2-FLAIR/T2 were significantly different (P ≤ .048) between patients with H3F3A and HIST1H3B/C mutations. CONCLUSIONS MR imaging features including enhancement and ADC histogram parameters are correlated with molecular subgroups and mutations in children with diffuse intrinsic pontine gliomas.
Collapse
Affiliation(s)
- C Jaimes
- From the Departments of Radiology (C.J., S.V., T.Y.P.).,Fetal-Neonatal Neuroimaging and Developmental Science Center (C.J.), Division of Newborn Medicine; Boston Children's Hospital, Boston, Massachusetts.,Harvard Medical School (C.J., S.V., C.M., P.B., F.D., R.B., K.L.L., S.C., K.D.W., T.Y.P.), Boston, Massachusetts
| | - S Vajapeyam
- From the Departments of Radiology (C.J., S.V., T.Y.P.).,Harvard Medical School (C.J., S.V., C.M., P.B., F.D., R.B., K.L.L., S.C., K.D.W., T.Y.P.), Boston, Massachusetts
| | - D Brown
- Tumor Imaging Metrics Core (D.B.), Massachusetts General Hospital, Boston, Massachusetts
| | - P-C Kao
- Pediatrics, Division of Oncology (P.-C.K., C.M., L.G., P.B., R.B., S.C., K.D.W.).,Dana Farber Cancer Institute (P.-C.K., C.M., L.G., P.B., F.D., O.S., R.B., K.L.L., S.C., K.D.W.), Boston, Massachusetts
| | - C Ma
- Pediatrics, Division of Oncology (P.-C.K., C.M., L.G., P.B., R.B., S.C., K.D.W.).,Dana Farber Cancer Institute (P.-C.K., C.M., L.G., P.B., F.D., O.S., R.B., K.L.L., S.C., K.D.W.), Boston, Massachusetts.,Harvard Medical School (C.J., S.V., C.M., P.B., F.D., R.B., K.L.L., S.C., K.D.W., T.Y.P.), Boston, Massachusetts
| | - L Greenspan
- Pediatrics, Division of Oncology (P.-C.K., C.M., L.G., P.B., R.B., S.C., K.D.W.).,Dana Farber Cancer Institute (P.-C.K., C.M., L.G., P.B., F.D., O.S., R.B., K.L.L., S.C., K.D.W.), Boston, Massachusetts
| | - N Gupta
- Department of Pediatric Neurosurgery (N.G.), University of California San Francisco Benioff Children's Hospital, San Francisco, California.,University of California San Francisco School of Medicine (N.G., T.Z.), San Francisco, California
| | | | - P Bandopahayay
- Pediatrics, Division of Oncology (P.-C.K., C.M., L.G., P.B., R.B., S.C., K.D.W.).,Dana Farber Cancer Institute (P.-C.K., C.M., L.G., P.B., F.D., O.S., R.B., K.L.L., S.C., K.D.W.), Boston, Massachusetts.,Harvard Medical School (C.J., S.V., C.M., P.B., F.D., R.B., K.L.L., S.C., K.D.W., T.Y.P.), Boston, Massachusetts
| | - F Dubois
- Dana Farber Cancer Institute (P.-C.K., C.M., L.G., P.B., F.D., O.S., R.B., K.L.L., S.C., K.D.W.), Boston, Massachusetts.,Harvard Medical School (C.J., S.V., C.M., P.B., F.D., R.B., K.L.L., S.C., K.D.W., T.Y.P.), Boston, Massachusetts
| | - N F Greenwald
- Stanford University School of Medicine (N.F.G.), Palo Alto, California
| | - T Zack
- University of California San Francisco School of Medicine (N.G., T.Z.), San Francisco, California
| | - O Shapira
- Dana Farber Cancer Institute (P.-C.K., C.M., L.G., P.B., F.D., O.S., R.B., K.L.L., S.C., K.D.W.), Boston, Massachusetts.,Broad Institute of Massachusetts Institute of Technology and Harvard University (O.S.), Cambridge, Massachusetts
| | - R Beroukhim
- Pediatrics, Division of Oncology (P.-C.K., C.M., L.G., P.B., R.B., S.C., K.D.W.).,Dana Farber Cancer Institute (P.-C.K., C.M., L.G., P.B., F.D., O.S., R.B., K.L.L., S.C., K.D.W.), Boston, Massachusetts.,Harvard Medical School (C.J., S.V., C.M., P.B., F.D., R.B., K.L.L., S.C., K.D.W., T.Y.P.), Boston, Massachusetts
| | - K L Ligon
- Dana Farber Cancer Institute (P.-C.K., C.M., L.G., P.B., F.D., O.S., R.B., K.L.L., S.C., K.D.W.), Boston, Massachusetts.,Department of Pathology (K.L.L.), Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School (C.J., S.V., C.M., P.B., F.D., R.B., K.L.L., S.C., K.D.W., T.Y.P.), Boston, Massachusetts
| | - S Chi
- Pediatrics, Division of Oncology (P.-C.K., C.M., L.G., P.B., R.B., S.C., K.D.W.).,Dana Farber Cancer Institute (P.-C.K., C.M., L.G., P.B., F.D., O.S., R.B., K.L.L., S.C., K.D.W.), Boston, Massachusetts.,Harvard Medical School (C.J., S.V., C.M., P.B., F.D., R.B., K.L.L., S.C., K.D.W., T.Y.P.), Boston, Massachusetts
| | - M W Kieran
- Clinical Trials Division (M.W.K.), Bristol-Myers-Squibb, New York, New York
| | - K D Wright
- Pediatrics, Division of Oncology (P.-C.K., C.M., L.G., P.B., R.B., S.C., K.D.W.).,Dana Farber Cancer Institute (P.-C.K., C.M., L.G., P.B., F.D., O.S., R.B., K.L.L., S.C., K.D.W.), Boston, Massachusetts.,Harvard Medical School (C.J., S.V., C.M., P.B., F.D., R.B., K.L.L., S.C., K.D.W., T.Y.P.), Boston, Massachusetts
| | - T Y Poussaint
- From the Departments of Radiology (C.J., S.V., T.Y.P.) .,Harvard Medical School (C.J., S.V., C.M., P.B., F.D., R.B., K.L.L., S.C., K.D.W., T.Y.P.), Boston, Massachusetts
| |
Collapse
|
20
|
Fuchs Q, Pierrevelcin M, Messe M, Lhermitte B, Blandin AF, Papin C, Coca A, Dontenwill M, Entz-Werlé N. Hypoxia Inducible Factors' Signaling in Pediatric High-Grade Gliomas: Role, Modelization and Innovative Targeted Approaches. Cancers (Basel) 2020; 12:cancers12040979. [PMID: 32326644 PMCID: PMC7226233 DOI: 10.3390/cancers12040979] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/04/2020] [Accepted: 04/07/2020] [Indexed: 12/15/2022] Open
Abstract
The brain tumor microenvironment has recently become a major challenge in all pediatric cancers, but especially in brain tumors like high-grade gliomas. Hypoxia is one of the extrinsic tumor features that interacts with tumor cells, but also with the blood-brain barrier and all normal brain cells. It is the result of a dramatic proliferation and expansion of tumor cells that deprive the tissues of oxygen inflow. However, cancer cells, especially tumor stem cells, can endure extreme hypoxic conditions by rescheduling various genes' expression involved in cell proliferation, metabolism and angiogenesis and thus, promote tumor expansion, therapeutic resistance and metabolic adaptation. This cellular adaptation implies Hypoxia-Inducible Factors (HIF), namely HIF-1α and HIF-2α. In pediatric high-grade gliomas (pHGGs), several questions remained open on hypoxia-specific role in normal brain during gliomagenesis and pHGG progression, as well how to model it in preclinical studies and how it might be counteracted with targeted therapies. Therefore, this review aims to gather various data about this key extrinsic tumor factor in pHGGs.
Collapse
Affiliation(s)
- Quentin Fuchs
- UMR CNRS 7021, Laboratory Bioimaging and Pathologies, Tumoral Signaling and Therapeutic Targets team, Faculty of Pharmacy, 74 route du Rhin, 67405 Illkirch, France; (Q.F.); (M.P.); (M.M.); (B.L.); (M.D.)
| | - Marina Pierrevelcin
- UMR CNRS 7021, Laboratory Bioimaging and Pathologies, Tumoral Signaling and Therapeutic Targets team, Faculty of Pharmacy, 74 route du Rhin, 67405 Illkirch, France; (Q.F.); (M.P.); (M.M.); (B.L.); (M.D.)
| | - Melissa Messe
- UMR CNRS 7021, Laboratory Bioimaging and Pathologies, Tumoral Signaling and Therapeutic Targets team, Faculty of Pharmacy, 74 route du Rhin, 67405 Illkirch, France; (Q.F.); (M.P.); (M.M.); (B.L.); (M.D.)
| | - Benoit Lhermitte
- UMR CNRS 7021, Laboratory Bioimaging and Pathologies, Tumoral Signaling and Therapeutic Targets team, Faculty of Pharmacy, 74 route du Rhin, 67405 Illkirch, France; (Q.F.); (M.P.); (M.M.); (B.L.); (M.D.)
- Pathology Department, University Hospital of Strasbourg, 1 avenue Molière, 67098 Strasbourg, France
| | | | - Christophe Papin
- Inserm U1258, UMR CNRS 7104, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, 67400 Illkirch, France;
| | - Andres Coca
- Neurosurgery, University Hospital of Strasbourg, 1 avenue Molière, 67098 Strasbourg, France;
| | - Monique Dontenwill
- UMR CNRS 7021, Laboratory Bioimaging and Pathologies, Tumoral Signaling and Therapeutic Targets team, Faculty of Pharmacy, 74 route du Rhin, 67405 Illkirch, France; (Q.F.); (M.P.); (M.M.); (B.L.); (M.D.)
| | - Natacha Entz-Werlé
- UMR CNRS 7021, Laboratory Bioimaging and Pathologies, Tumoral Signaling and Therapeutic Targets team, Faculty of Pharmacy, 74 route du Rhin, 67405 Illkirch, France; (Q.F.); (M.P.); (M.M.); (B.L.); (M.D.)
- Pediatric Onco-Hematology Department, Pediatrics, University hospital of Strasbourg, 1 avenue Molière, 67098 Strasbourg, France
- Correspondence: ; Tel.: +33-388128396; Fax: +33-388128092
| |
Collapse
|
21
|
Vajapeyam S, Brown D, Billups C, Patay Z, Vezina G, Shiroishi MS, Law M, Baxter P, Onar-Thomas A, Fangusaro JR, Dunkel IJ, Poussaint TY. Advanced ADC Histogram, Perfusion, and Permeability Metrics Show an Association with Survival and Pseudoprogression in Newly Diagnosed Diffuse Intrinsic Pontine Glioma: A Report from the Pediatric Brain Tumor Consortium. AJNR Am J Neuroradiol 2020; 41:718-724. [PMID: 32241771 DOI: 10.3174/ajnr.a6499] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/10/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND PURPOSE Diffuse intrinsic pontine glioma is a lethal childhood brain cancer with dismal prognosis and MR imaging is the primary methodology used for diagnosis and monitoring. Our aim was to determine whether advanced diffusion, perfusion, and permeability MR imaging metrics predict survival and pseudoprogression in children with newly diagnosed diffuse intrinsic pontine glioma. MATERIALS AND METHODS A clinical trial using the poly (adenosine diphosphate ribose) polymerase (PARP) inhibitor veliparib concurrently with radiation therapy, followed by maintenance therapy with veliparib + temozolomide, in children with diffuse intrinsic pontine glioma was conducted by the Pediatric Brain Tumor Consortium. Standard MR imaging, DWI, dynamic contrast-enhanced perfusion, and DSC perfusion were performed at baseline and approximately every 2 months throughout treatment. ADC histogram metrics of T2-weighted FLAIR and enhancing tumor volume, dynamic contrast-enhanced permeability metrics for enhancing tumors, and tumor relative CBV from DSC perfusion MR imaging were calculated. Baseline values, post-radiation therapy changes, and longitudinal trends for all metrics were evaluated for associations with survival and pseudoprogression. RESULTS Fifty children were evaluable for survival analyses. Higher baseline relative CBV was associated with shorter progression-free survival (P = .02, Q = 0.089) and overall survival (P = .006, Q = 0.055). Associations of higher baseline mean transfer constant from the blood plasma into the extravascular extracellular space with shorter progression-free survival (P = .03, Q = 0.105) and overall survival (P = .03, Q = 0.102) trended toward significance. An increase in relative CBV with time was associated with shorter progression-free survival (P < .001, Q < 0.001) and overall survival (P = .004, Q = 0.043). Associations of longitudinal mean extravascular extracellular volume fraction with progression-free survival (P = .03, Q = 0.104) and overall survival (P = .03, Q = 0.105) and maximum transfer constant from the blood plasma into the extravascular extracellular space with progression-free survival (P = .03, Q = 0.102) trended toward significance. Greater increases with time were associated with worse outcomes. True radiologic progression showed greater post-radiation therapy decreases in mode_ADC_FLAIR compared with pseudoprogression (means, -268.15 versus -26.11, P = .01.) CONCLUSIONS: ADC histogram, perfusion, and permeability MR imaging metrics in diffuse intrinsic pontine glioma are useful in predicting survival and pseudoprogression.
Collapse
Affiliation(s)
- S Vajapeyam
- From the Radiology (S.V., T.Y.P.), Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - D Brown
- DF/HCC Tumor Imaging Metrics Core (D.B.), Massachusetts General Hospital, Boston, Massachusetts
| | | | - Z Patay
- Diagnostic Imaging (Z.P.), St. Jude Children's Research Hospital, Memphis, Tennessee
| | - G Vezina
- Radiology (G.V.), Children's National Medical Center, Washington, DC
| | - M S Shiroishi
- Radiology (M.S.S.), Keck Medical Center of USC, Los Angeles, California
| | - M Law
- Neuroscience (M.L.), Monash University, Melbourne, Australia
| | - P Baxter
- Cancer and Hematology Center (P.B.), Texas Children's Hospital, Houston, Texas
| | | | - J R Fangusaro
- Aflac Cancer and Blood Disorders Center (J.R.F.), Children's Healthcare of Atlanta, Atlanta, Georgia
| | - I J Dunkel
- Pediatrics (I.J.D.), Memorial Sloan Kettering Cancer Center, New York, New York
| | - T Y Poussaint
- From the Radiology (S.V., T.Y.P.), Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
22
|
Sun SL, Shu YG, Tao MY. LncRNA CCAT2 promotes angiogenesis in glioma through activation of VEGFA signalling by sponging miR-424. Mol Cell Biochem 2020; 468:69-82. [PMID: 32236863 DOI: 10.1007/s11010-020-03712-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 03/06/2020] [Indexed: 12/17/2022]
Abstract
Glioma is characterized by high morbidity, high mortality and poor prognosis. Recent studies exhibited that lncRNA CCAT2 is overexpressed in glioma and promotes glioma progression, but the specific molecular biological mechanism remains to be determined. We performed qRT-PCR to evaluate the expression of related genes, Western blotting analysis to measure protein levels, colony formation assay to detect the proliferative ability of glioma cells, flow cytometry to measure cell apoptosis, bioinformatics analysis and dual luciferase assay to verify the binding sites and the targeted regulatory relationship in A172 and U251 cell lines and tube formation assay to determine endothelial angiogenesis. LncRNA CCAT2 and VEGFA were highly expressed, while miR-424 was expressed at low levels in NHA cells. Furthermore, knockdown of lncRNA CCAT2 decreased cell proliferation, increased cell apoptosis and inhibited endothelial angiogenesis in glioma. Moreover, lncRNA CCAT2 shared a complementary sequence with miR-424 which in turn directly bound to the 3'-UTR of VEGFA. Further investigation indicated that lncRNA CCAT2 promoted cell proliferation and endothelial angiogenesis by inducing the PI3K/AKT signalling pathway in glioma. The oncogenic lncRNA CCAT2 is highly associated with the development of glioma and exerts its function by upregulating VEGFA via miR-424.
Collapse
Affiliation(s)
- Sheng-Li Sun
- Department of Neurosurgery, Hunan Provincial People's Hospital, No.61, Jiefang West Road, Changsha, 410005, Hunan, People's Republic of China
| | - Yu-Gao Shu
- Department of Neurosurgery, Hunan Provincial People's Hospital, No.61, Jiefang West Road, Changsha, 410005, Hunan, People's Republic of China
| | - Mei-Yi Tao
- Department of Neurosurgery, Hunan Provincial People's Hospital, No.61, Jiefang West Road, Changsha, 410005, Hunan, People's Republic of China.
| |
Collapse
|
23
|
Gao J, Huang X, Meng H, Zhang M, Zhang X, Lin X, Li B. Performance of Multiparametric Functional Imaging and Texture Analysis in Predicting Synchronous Metastatic Disease in Pancreatic Ductal Adenocarcinoma Patients by Hybrid PET/MR: Initial Experience. Front Oncol 2020; 10:198. [PMID: 32158690 PMCID: PMC7052324 DOI: 10.3389/fonc.2020.00198] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 02/05/2020] [Indexed: 12/16/2022] Open
Abstract
Objectives: To assess the imaging biomarkers of glucose metabolic activity and diffusion-weighted imaging (DWI) derived from pretreatment integrated 18F-fluorodeoxyglucose positron emission tomography-magnetic resonance (18F-FDG PET/MR) imaging as potential predictive factors of metastasis in patients with pancreatic ductal adenocarcinoma (PDAC). Patients and Methods: We retrospectively included 17 consecutive patients with pathologically confirmed PDAC by pretreatment 18F-FDG PET/MR. The study subjects were divided into a non-metastatic group (M0, six cases) and a metastatic group (M1, 11 cases). The 18F-FDG PET/MR images were reviewed independently by two board certificated nuclear medicine physicians and one radiologist. Conventional characteristics and quantitative parameters from both PET and apparent diffusion coefficient (ADC) were assessed. The texture features were extracted from LIFEx packages (www.lifexsoft.org), and a 3D tumor volume of interest was manually drawn on fused PET/ADC images. Chi-square tests, independent-samples t-tests and Mann-Whitney U-tests were used to compare the differences in single parameters between the two groups. A logistic regression analysis was performed to determine independent predictors. A receiver operating characteristic (ROC) curve analysis was performed to assess the discriminatory power of the selected parameters. Correlations between metabolic parameters and ADC features were calculated with Spearman's rank correlation coefficient test. Results: For conventional parameters, univariable analysis demonstrated that the M1 group had a significantly larger size and a higher peak of standardized uptake value (SUVpeak), metabolic tumor volume (MTV), and total lesion glycolysis (TLG) than those of the M0 group (p < 0.05 for all). TLG remained significant predictor in the multivariable analysis, but there were no significant differences for the area under the ROC curve (AUC) among the four conventional features in differential diagnoses (p > 0.05 for all). For the texture features, there were four features from the PET image and 13 from the ADC map that showed significant differences between the two groups. Multivariate analysis indicated that one feature from PET and three from the ADC were significant predictors. TLG was associated with ADC-GLRLM_GLNU (r = 0.659), ADC-GLRLM_LRHGE (r = 0.762), and PET-GLRLM_LRHGE (r = 0.806). Conclusions: Multiple parameters and texture features of primary tumors from 18F-FDG PET/MR images maybe reliable biomarkers to predict synchronous metastatic disease for the pretreatment PDAC.
Collapse
Affiliation(s)
- Jing Gao
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyun Huang
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongping Meng
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Miao Zhang
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaozhe Zhang
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaozhu Lin
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Biao Li
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
24
|
Comparison between MRI-derived ADC maps and 18FLT-PET in pre-operative glioblastoma. J Neuroradiol 2019; 46:359-366. [PMID: 31229576 DOI: 10.1016/j.neurad.2019.05.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 05/02/2019] [Accepted: 05/23/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND PURPOSE Among principal MRI sequences used for a better pre-therapeutic characterization of glioblastoma (GBM), DWI-derived ADC is expected to be a good parameter for the evaluation of cellularity, due to restricted water diffusivity. We aimed here to compare ADC maps to 18FLT-PET, a proliferation tracer, in GBM cases. MATERIALS AND METHODS Patients underwent 18FLT-PET, followed by multiparametric magnetic resonance imaging (MRI) just prior to surgery. We analysed in this study twenty GBM confirmed patients. The 5th percentile (5p) of the ADC values were thresholded to define the ADCmin ROI, while the 95th percentile (95p) of the SUV FLT values were used to define the FLTmax ROI. The statistical and spatial correlations between these two groups of ROIs were analyzed. RESULTS We did not observe any significant correlations between ADCmin and FLTmax cut-off values (R2=0.0285), neither between ADCmin and FLTmax ROIs (mean Dice=0.09±0.12). Mean ADC values in the FLTmax defined ROI were significantly higher than the values in the ADCmin ROI (P<0.001). Mean FLT values in the FLTmax ROI were significantly higher than the values in the ADCmin ROI (P<0.001). CONCLUSIONS When comparing ADC maps to 18FLT uptake, we did not observe significant anatomical overlap nor correlation, between the regions of low ADC and high FLT disabling to clearly link ADC values to cellular proliferation. The exact significance of ADC maps in GBM has yet to be elaborated.
Collapse
|
25
|
Ceschin R, Kocak M, Vajapeyam S, Pollack IF, Onar-Thomas A, Dunkel IJ, Poussaint TY, Panigrahy A. Quantifying radiation therapy response using apparent diffusion coefficient (ADC) parametric mapping of pediatric diffuse intrinsic pontine glioma: a report from the pediatric brain tumor consortium. J Neurooncol 2019; 143:79-86. [PMID: 30810873 DOI: 10.1007/s11060-019-03133-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/22/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND PURPOSE Baseline diffusion or apparent diffusion coefficient (ADC) characteristics have been shown to predict outcome related to DIPG, but the predictive value of post-radiation ADC is less well understood. ADC parametric mapping (FDM) was used to measure radiation-related changes in ADC and compared these metrics to baseline ADC in predicting progression-free survival and overall survival using a large multi-center cohort of DIPG patients (Pediatric Brain Tumor Consortium-PBTC). MATERIALS AND METHODS MR studies at baseline and post-RT in 95 DIPG patients were obtained and serial quantitative ADC parametric maps were generated from diffusion-weighted imaging based on T2/FLAIR and enhancement regions of interest (ROIs). Metrics assessed included total voxels with: increase in ADC (iADC); decrease in ADC (dADC), no change in ADC (nADC), fraction of voxels with increased ADC (fiADC), fraction of voxels with decreased ADC (fdADC), and the ratio of fiADC and fdADC (fDM Ratio). RESULTS A total of 72 patients were included in the final analysis. Tumors with higher fiADC between baseline and the first RT time point showed a trend toward shorter PFS with a hazard ratio of 6.44 (CI 0.79, 52.79, p = 0.083). In contrast, tumors with higher log mean ADC at baseline had longer PFS, with a hazard ratio of 0.27 (CI 0.09, 0.82, p = 0.022). There was no significant association between fDM derived metrics and overall survival. CONCLUSIONS Baseline ADC values are a stronger predictor of outcome compared to radiation related ADC changes in pediatric DIPG. We show the feasibility of employing parametric mapping techniques in multi-center studies to quantitate spatially heterogeneous treatment response in pediatric tumors, including DIPG.
Collapse
Affiliation(s)
- Rafael Ceschin
- Department of Radiology, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, 4401 Penn Avenue, Suite 2464, Pittsburgh, PA, 15201, USA.
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Pediatric Imaging Research Center, Department of Pediatric Radiology, UPMC Children's Hospital of Pittsburgh, 45th Street and Penn Avenue, Pittsburgh, PA, 15224, USA.
| | - Mehmet Kocak
- Department of Biostatistics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Sridhar Vajapeyam
- Department of Preventive Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Radiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Ian F Pollack
- Department of Neurosurgery, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Arzu Onar-Thomas
- Department of Biostatistics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Ira J Dunkel
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tina Young Poussaint
- Department of Preventive Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Radiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Ashok Panigrahy
- Department of Radiology, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, 4401 Penn Avenue, Suite 2464, Pittsburgh, PA, 15201, USA
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
26
|
Huang R, Chen Y, Li W, Zhang X. An evidence-based approach to assess the accuracy of diffusion kurtosis imaging in characterization of gliomas. Medicine (Baltimore) 2018; 97:e13068. [PMID: 30383687 PMCID: PMC6221635 DOI: 10.1097/md.0000000000013068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE Accurate and noninvasive pathologic grading of glioma patients before surgery was crucial to guiding clinicians to select appropriate treatment and improve patient prognosis. This study was performed to investigate the potential diagnostic value of diffusion kurtosis imaging (DKI) to distinguish high-grade gliomas (HGGs) from low-grade gliomas (LGGs) based on an evidence-based approach. METHODS Relevant articles that used DKI to distinguish HGG from LGG in Embase, PubMed, China Knowledge Resource Integrated database (CNKI), Web of Knowledge, and Cochrane Libraries databases were electronically searched to April 31, 2018 by 2 reviewers. All analysis was performed by using Meta-disc1.4 and Stata. Influence factors on the diagnostic accuracy were evaluated using meta-regression analysis. RESULTS Five eligible studies were included in this meta-analysis. The pooled sensitivity (SEN) and specificity (SPE) was 91% (confidence interval [CI]: 0.78-0.96; P = .02) and 91% (CI: 0.80-0.97; P = .01). The pooled data showed that diagnostic odds ratio (DOR) of DKI was 79.75 (CI: 31.57-201.45). The area under the curve (AUC) of summary receiver operating characteristic curve was 0.96. There is no evidence that our research has a threshold effect (Spearman correlation coefficient: 0.300, P = .624) and publication bias. Meta regression analysis identified that country, language, field strength, and parameter of magnetic resonance imaging had no significant effect on diagnostic performance. CONCLUSION The present meta-analysis shows that the mean kurtosis values derived from DKI may be useful in characterization of gliomas with high sensitivity and specificity. Taken into consideration the small sample of this study, we need to be cautious when interpreting the results of this study.
Collapse
Affiliation(s)
- Ruiyu Huang
- Department of MRI, The Second Affiliated Hospital of Shaanxi University of Chinese Medicine
| | - Yanni Chen
- Department of Radiology, XianYang Rainbow Hospital, XianYang, Shaanxi
| | - Wenfei Li
- Department of Radiology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, China
| | - Xvfeng Zhang
- Department of Radiology, XianYang Rainbow Hospital, XianYang, Shaanxi
| |
Collapse
|
27
|
Tinkle CL, Duncan EC, Doubrovin M, Han Y, Li Y, Kim H, Broniscer A, Snyder SE, Merchant TE, Shulkin BL. Evaluation of 11C-Methionine PET and Anatomic MRI Associations in Diffuse Intrinsic Pontine Glioma. J Nucl Med 2018; 60:312-319. [PMID: 30072503 DOI: 10.2967/jnumed.118.212514] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/13/2018] [Indexed: 11/16/2022] Open
Abstract
The role of metabolic imaging in the diagnosis, treatment, and response assessment of diffuse intrinsic pontine glioma (DIPG) is poorly defined. We investigated the uptake of 11C-methionine in pediatric patients with newly diagnosed DIPG and evaluated the associations of 11C-methionine PET metrics with conventional MRI indices and survival outcomes. Methods: Twenty-two patients with newly diagnosed DIPG were prospectively enrolled on an institutional review board-approved investigational study of 11C-methionine PET. All patients underwent baseline 11C-methionine PET/CT, and initial treatment-response scans after chemotherapy or radiation therapy were obtained for 17 patients. Typical and atypical DIPGs were assessed clinically and radiographically and defined by multidisciplinary consensus. Three-dimensional regions of interest, reviewed by consensus between a nuclear medicine physician and a radiation oncologist, were delineated after coregistration of PET and MR images. Associations of 11C-methionine uptake intensity and uniformity with survival, along with associations between 11C-methionine uptake and conventional MRI tumor indices over time, were evaluated. 11C-methionine PET voxel values within regions of interest were assessed as threshold values across proportions of the study population, and 11C-methionine uptake at baseline was assessed relative to MRI-defined tumor progression. Results: 11C-methionine uptake above that of uninvolved brain tissue was observed in 18 of 22 baseline scans (82%) and 15 of 17 initial response scans (88%). 11C-methionine avidity within MRI-defined tumor was limited in extent, with 11 of 18 positive baseline 11C-methionine PET scans (61%) showing less than 25% 11C-methionine-avid tumor. The increase in total tumor volume with 11C-methionine PET was relatively limited (17.2%; interquartile range, 6.53%-38.90%), as was the extent of 11C-methionine uptake beyond the MRI-defined tumor (2.2%; interquartile range, 0.55%-10.88%). Although baseline 11C-methionine PET intensity and uniformity metrics did not correlate with survival outcomes, initial 11C-methionine avidity overlapped with recurrent tumor in 100% of cases. A clinical diagnosis of atypical DIPG was associated with borderline significantly prolonged progression-free survival (P = 0.07), yet 11C-methionine PET indices at diagnosis did not differ significantly between atypical and typical DIPGs. Conclusion: Most newly diagnosed DIPGs are successfully visualized by 11C-methionine PET. Baseline 11C-methionine uptake delineates regions at increased risk for recurrence, yet intensity and uniformity metrics did not correlate with treatment outcomes in children with DIPG in this study.
Collapse
Affiliation(s)
- Christopher L Tinkle
- Department of Radiation Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Mikhail Doubrovin
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Yuanyuan Han
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Yimei Li
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Hyun Kim
- Department of Radiation Oncology, Washington University, St. Louis, Missouri; and
| | - Alberto Broniscer
- Department of Oncology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Scott E Snyder
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Thomas E Merchant
- Department of Radiation Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Barry L Shulkin
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
28
|
Cervical Cancer: Associations between Metabolic Parameters and Whole Lesion Histogram Analysis Derived from Simultaneous 18F-FDG-PET/MRI. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:5063285. [PMID: 30154687 PMCID: PMC6098855 DOI: 10.1155/2018/5063285] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/12/2018] [Accepted: 06/25/2018] [Indexed: 01/16/2023]
Abstract
Multimodal imaging has been increasingly used in oncology, especially in cervical cancer. By using a simultaneous positron emission (PET) and magnetic resonance imaging (MRI, PET/MRI) approach, PET and MRI can be obtained at the same time which minimizes motion artefacts and allows an exact imaging fusion, which is especially important in anatomically complex regions like the pelvis. The associations between functional parameters from MRI and 18F-FDG-PET reflecting different tumor aspects are complex with inconclusive results in cervical cancer. The present study correlates histogram analysis and 18F-FDG-PET parameters derived from simultaneous FDG-PET/MRI in cervical cancer. Overall, 18 female patients (age range: 32–79 years) with histopathologically confirmed squamous cell cervical carcinoma were retrospectively enrolled. All 18 patients underwent a whole-body simultaneous 18F-FDG-PET/MRI, including diffusion-weighted imaging (DWI) using b-values 0 and 1000 s/mm2. Apparent diffusion coefficient (ADC) histogram parameters included several percentiles, mean, min, max, mode, median, skewness, kurtosis, and entropy. Furthermore, mean and maximum standardized uptake values (SUVmean and SUVmax), metabolic tumor volume (MTV), and total lesion glycolysis (TLG) were estimated. No statistically significant correlations were observed between SUVmax or SUVmean and ADC histogram parameters. TLG correlated inversely with p25 (r=−0.486, P=0.041), p75 (r=−0.490, P=0.039), p90 (r=−0.513, P=0.029), ADC median (r=−0.497, P=0.036), and ADC mode (r=−0.546, P=0.019). MTV also showed significant correlations with several ADC parameters: mean (r=−0.546, P=0.019), p10 (r=−0.473, P=0.047), p25 (r=−0.569, P=0.014), p75 (r=−0.576, P=0.012), p90 (r=−0.585, P=0.011), ADC median (r=−0.577, P=0.012), and ADC mode (r=−0.597, P=0.009). ADC histogram analysis and volume-based metabolic 18F-FDG-PET parameters are related to each other in cervical cancer.
Collapse
|
29
|
Sasaki T, Moritani T, Belay A, Capizzano AA, Sato SP, Sato Y, Kirby P, Ishitoya S, Oya A, Toda M, Takahashi K. Role of the Apparent Diffusion Coefficient as a Predictor of Tumor Progression in Patients with Chordoma. AJNR Am J Neuroradiol 2018; 39:1316-1321. [PMID: 29724767 DOI: 10.3174/ajnr.a5664] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 03/10/2018] [Indexed: 01/15/2023]
Abstract
BACKGROUND AND PURPOSE Diffusion-weighted imaging may aid in distinguishing aggressive chordoma from nonaggressive chordoma. This study explores the prognostic role of the apparent diffusion coefficient in chordomas. MATERIALS AND METHODS Sixteen patients with residual or recurrent chordoma were divided postoperatively into those with an aggressive tumor, defined as a growing tumor having a doubling time of <1 year, and those with a nonaggressive tumor on follow-up MR images. The ability of the ADC to predict an aggressive tumor phenotype was investigated by receiver operating characteristic analysis. The prognostic role of ADC was assessed using a Kaplan-Meier curve with a log-rank test. RESULTS Seven patients died during a median follow-up of 48 months (range, 4-126 months). Five of these 7 patients were in the aggressive tumor group, and 2 were in the nonaggressive tumor group. The mean ADC was significantly lower in the aggressive tumor group than in the nonaggressive tumor group (P = .002). Receiver operating characteristic analysis showed that a cutoff ADC value of 1.494 × 10-3 × mm2/s could be used to diagnose aggressive tumors with an area under the curve of 0.983 (95% CI, 0.911-1.000), a sensitivity of 1.000 (95% CI, 0.541-1.000), and a specificity of 0.900 (95% CI, 0.555-0.998). Furthermore, a cutoff ADC of ≤1.494 × 10-3 × mm2/s was associated with a significantly worse prognosis (P = .006). CONCLUSIONS Lower ADC values could predict tumor progression in postoperative chordomas.
Collapse
Affiliation(s)
- T Sasaki
- From the Departments of Radiology (T.S., T.M., A.B., A.A.C., S.P.S., Y.S.)
- Asahikawa Medical University (T.S., S.I., A.O., M.T., K.T.), Asahikawa, Hokkaido, Japan
| | - T Moritani
- From the Departments of Radiology (T.S., T.M., A.B., A.A.C., S.P.S., Y.S.)
- Department of Radiology (T.M.), University of Michigan, Ann Arbor, Michigan
| | - A Belay
- From the Departments of Radiology (T.S., T.M., A.B., A.A.C., S.P.S., Y.S.)
| | - A A Capizzano
- From the Departments of Radiology (T.S., T.M., A.B., A.A.C., S.P.S., Y.S.)
| | - S P Sato
- From the Departments of Radiology (T.S., T.M., A.B., A.A.C., S.P.S., Y.S.)
| | - Y Sato
- From the Departments of Radiology (T.S., T.M., A.B., A.A.C., S.P.S., Y.S.)
| | - P Kirby
- Pathology (P.K.), University of Iowa, Iowa City, Iowa
| | - S Ishitoya
- Asahikawa Medical University (T.S., S.I., A.O., M.T., K.T.), Asahikawa, Hokkaido, Japan
| | - A Oya
- Asahikawa Medical University (T.S., S.I., A.O., M.T., K.T.), Asahikawa, Hokkaido, Japan
| | - M Toda
- Asahikawa Medical University (T.S., S.I., A.O., M.T., K.T.), Asahikawa, Hokkaido, Japan
| | - K Takahashi
- Asahikawa Medical University (T.S., S.I., A.O., M.T., K.T.), Asahikawa, Hokkaido, Japan
| |
Collapse
|
30
|
Grading and outcome prediction of pediatric diffuse astrocytic tumors with diffusion and arterial spin labeling perfusion MRI in comparison with 18F-DOPA PET. Eur J Nucl Med Mol Imaging 2017; 44:2084-2093. [PMID: 28752225 DOI: 10.1007/s00259-017-3777-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 07/10/2017] [Indexed: 01/02/2023]
Abstract
PURPOSE The aim of this study was to investigate MRI-derived diffusion weighted imaging (DWI) and arterial spin labeling (ASL) perfusion imaging in comparison with 18F-dihydroxyphenylalanine (DOPA) PET with respect to diagnostic performance in tumor grading and outcome prediction in pediatric patients with diffuse astrocytic tumors (DAT). METHODS We retrospectively analyzed 26 children with histologically proven treatment naïve low and high grade DAT who underwent ASL and DWI performed within 2 weeks of 18F-DOPA PET. Relative ASL-derived cerebral blood flow max (rCBF max) and DWI-derived minimum apparent diffusion coefficient (rADC min) were compared with 18F-DOPA uptake tumor/normal tissue (T/N) and tumor/striatum (T/S) ratios, and correlated with World Health Organization (WHO) tumor grade and progression-free survival (PFS). Statistics included Pearson's chi-square and Mann-Whitney U tests, Spearman's rank correlation, receiver operating characteristic (ROC) analysis, discriminant function analysis (DFA), Kaplan-Meier survival curve, and Cox analysis. RESULTS A significant correlation was demonstrated between rCBF max, rADC min, and 18F-DOPA PET data (p < 0.001). Significant differences in terms of rCBF max, rADC min, and 18F-DOPA uptake were found between low- and high-grade DAT (p ≤ 0.001). ROC analysis and DFA demonstrated that T/S and T/N values were the best parameters for predicting tumor progression (AUC 0.93, p < 0.001). On univariate analysis, all diagnostic tools correlated with PFS (p ≤ 0.001); however, on multivariate analysis, only 18F-DOPA uptake remained significantly associated with outcome (p ≤ 0.03), while a trend emerged for rCBF max (p = 0.09) and rADC min (p = 0.08). The combination of MRI and PET data increased the predictive power for prognosticating tumor progression (AUC 0.97, p < 0.001). CONCLUSIONS DWI, ASL and 18F-DOPA PET provide useful complementary information for pediatric DAT grading. 18F-DOPA uptake better correlates with PFS prediction. Combining MRI and PET data provides the highest predictive power for prognosticating tumor progression suggesting a synergistic role of these diagnostic tools.
Collapse
|
31
|
Gerstner ER. MRI and PET: Noninvasive Tools to Probe the Biology of Diffuse Intrinsic Pontine Glioma. J Nucl Med 2017; 58:1262-1263. [PMID: 28385794 DOI: 10.2967/jnumed.117.192526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 04/01/2017] [Indexed: 11/16/2022] Open
|