1
|
Mueller S, Kline C, Franson A, van der Lugt J, Prados M, Waszak SM, Plasschaert SLA, Molinaro AM, Koschmann C, Nazarian J. Rational combination platform trial design for children and young adults with diffuse midline glioma: A report from PNOC. Neuro Oncol 2024; 26:S125-S135. [PMID: 38124481 PMCID: PMC11066905 DOI: 10.1093/neuonc/noad181] [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: 05/23/2023] [Indexed: 12/23/2023] Open
Abstract
Background Diffuse midline glioma (DMG) is a devastating pediatric brain tumor unresponsive to hundreds of clinical trials. Approximately 80% of DMGs harbor H3K27M oncohistones, which reprogram the epigenome to increase the metabolic profile of the tumor cells. Methods We have previously shown preclinical efficacy of targeting both oxidative phosphorylation and glycolysis through treatment with ONC201, which activates the mitochondrial protease ClpP, and paxalisib, which inhibits PI3K/mTOR, respectively. Results ONC201 and paxalisib combination treatment aimed at inducing metabolic distress led to the design of the first DMG-specific platform trial PNOC022 (NCT05009992). Conclusions Here, we expand on the PNOC022 rationale and discuss various considerations, including liquid biome, microbiome, and genomic biomarkers, quality-of-life endpoints, and novel imaging modalities, such that we offer direction on future clinical trials in DMG.
Collapse
Affiliation(s)
- Sabine Mueller
- Department of Neurology, Neurosurgery and Pediatrics, University of California, San Francisco, California, USA
| | - Cassie Kline
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Andrea Franson
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Michael Prados
- Department of Neurosurgery and Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | - Sebastian M Waszak
- Department of Neurology, University of California, San Francisco, San Francisco, California, USA
- Laboratory of Computational Neuro-Oncology, Swiss Institute for Experimental Cancer Research, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | | - Annette M Molinaro
- Division of Biomedical Statistics and Informatics, Department of Neurosurgery, University of California, San Francisco, San Francisco, California, USA
| | - Carl Koschmann
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Javad Nazarian
- Research Center for Genetic Medicine, Children’s National Health System, Washington, District of Columbia, USA
- Brain Tumor Institute, Children’s National Health System, Washington, District of Columbia, USA
- DMG Research Center, Department of Pediatrics, University Children’s Hospital, University of Zurich, Zürich, Switzerland
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Panigrahy A, Jakacki RI, Pollack IF, Ceschin R, Okada H, Nelson MD, Kohanbash G, Dhall G, Bluml S. Magnetic Resonance Spectroscopy Metabolites as Biomarkers of Disease Status in Pediatric Diffuse Intrinsic Pontine Gliomas (DIPG) Treated with Glioma-Associated Antigen Peptide Vaccines. Cancers (Basel) 2022; 14:5995. [PMID: 36497477 PMCID: PMC9739009 DOI: 10.3390/cancers14235995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/09/2022] [Accepted: 11/25/2022] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Diffuse intrinsic pontine gliomas (DIPG) are highly aggressive tumors with no currently available curative therapy. This study evaluated whether measurements of in vivo cell metabolites using magnetic resonance spectroscopy (MRS) may serve as biomarkers of response to therapy, including progression. METHODS Single-voxel MR spectra were serially acquired in two cohorts of patients with DIPG treated with radiation therapy (RT) with or without concurrent chemotherapy and prior to progression: 14 participants were enrolled in a clinical trial of adjuvant glioma-associated antigen peptide vaccines and 32 patients were enrolled who did not receive adjuvant vaccine therapy. Spearman correlations measured overall survival associations with absolute metabolite concentrations of myo-inositol (mI), creatine (Cr), and n-acetyl-aspartate (NAA) and their ratios relative to choline (Cho) during three specified time periods following completion of RT. Linear mixed-effects regression models evaluated the longitudinal associations between metabolite ratios and time from death (terminal decline). RESULTS Overall survival was not associated with metabolite ratios obtained shortly after RT (1.9-3.8 months post-diagnosis) in either cohort. In the vaccine cohort, an elevated mI/Cho ratio after 2-3 doses (3.9-5.2 months post-diagnosis) was associated with longer survival (rho = 0.92, 95% CI 0.67-0.98). Scans performed up to 6 months before death showed a terminal decline in the mI/Cho ratio, with an average of 0.37 ratio/month in vaccine patients (95% CI 0.11-0.63) and 0.26 (0.04-0.48) in the non-vaccine cohort. CONCLUSION Higher mI/Cho ratios following RT, consistent with less proliferate tumors and decreased cell turnover, were associated with longer survival, suggesting that this ratio can serve as a biomarker of prognosis following RT. This finding was seen in both cohorts, although the association with OS was detected earlier in the vaccine cohort. Increased mI/Cho (possibly reflecting immune-effector cell influx into the tumor as a mechanism of tumor response) requires further study.
Collapse
Affiliation(s)
- Ashok Panigrahy
- Department of Radiology, UPMC Children’s Hospital of Pittsburgh, 4401 Penn Ave Floor 2, Pittsburgh, PA 15224, USA
| | - Regina I. Jakacki
- Department of Hematology Oncology, UPMC Children’s Hospital of Pittsburgh, 4401 Penn Ave Floor 9, Pittsburgh, PA 15224, USA
| | - Ian F. Pollack
- Department of Neurosurgery, UPMC Children’s Hospital of Pittsburgh, 4401 Penn Ave Floor 2, Pittsburgh, PA 15224, USA
| | - Rafael Ceschin
- Department of Radiology, UPMC Children’s Hospital of Pittsburgh, 4401 Penn Ave Floor 2, Pittsburgh, PA 15224, USA
| | - Hideho Okada
- Department of Neurological Surgery, Box 0112 505 Parnassus Ave, University of California San Francisco, Room M779, San Francisco, CA 94143, USA
- Cancer Immunotherapy Program, Helen Diller Family Comprehensive Cancer Center, Box 0981 UCSF, San Francisco, CA 94143-0981, USA
| | - Marvin D. Nelson
- Department of Radiology, Children’s Hospital Los Angeles, 4650 Sunset Blvd, Los Angeles, CA 90027, USA
- Keck School of Medicine, University of Southern California, 1441 Eastlake Ave # 2315, Los Angeles, CA 90089, USA
| | - Gary Kohanbash
- Department of Neurosurgery, UPMC Children’s Hospital of Pittsburgh, 4401 Penn Ave Floor 2, Pittsburgh, PA 15224, USA
| | - Girish Dhall
- Department of Pediatrics, University of Alabama at Birmingham, 1600 7 th Ave S, Birmingham, AL 35233, USA
| | - Stefan Bluml
- Keck School of Medicine, University of Southern California, 1441 Eastlake Ave # 2315, Los Angeles, CA 90089, USA
| |
Collapse
|
5
|
Di Ruscio V, Del Baldo G, Fabozzi F, Vinci M, Cacchione A, de Billy E, Megaro G, Carai A, Mastronuzzi A. Pediatric Diffuse Midline Gliomas: An Unfinished Puzzle. Diagnostics (Basel) 2022; 12:2064. [PMID: 36140466 PMCID: PMC9497626 DOI: 10.3390/diagnostics12092064] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/15/2022] Open
Abstract
Diffuse midline glioma (DMG) is a heterogeneous group of aggressive pediatric brain tumors with a fatal prognosis. The biological hallmark in the major part of the cases is H3K27 alteration. Prognosis remains poor, with median survival ranging from 9 to 12 months from diagnosis. Clinical and radiological prognostic factors only partially change the progression-free survival but they do not improve the overall survival. Despite efforts, there is currently no curative therapy for DMG. Radiotherapy remains the standard treatment with only transitory benefits. No chemotherapeutic regimens were found to significantly improve the prognosis. In the new era of a deeper integration between histological and molecular findings, potential new approaches are currently under investigation. The entire international scientific community is trying to target DMG on different aspects. The therapeutic strategies involve targeting epigenetic alterations, such as methylation and acetylation status, as well as identifying new molecular pathways that regulate oncogenic proliferation; immunotherapy approaches too are an interesting point of research in the oncology field, and the possibility of driving the immune system against tumor cells has currently been evaluated in several clinical trials, with promising preliminary results. Moreover, thanks to nanotechnology amelioration, the development of innovative delivery approaches to overcross a hostile tumor microenvironment and an almost intact blood-brain barrier could potentially change tumor responses to different treatments. In this review, we provide a comprehensive overview of available and potential new treatments that are worldwide under investigation, with the intent that patient- and tumor-specific treatment could change the biological inauspicious history of this disease.
Collapse
Affiliation(s)
- Valentina Di Ruscio
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Giada Del Baldo
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Francesco Fabozzi
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
- Department of Pediatrics, University of Rome Tor Vergata, 00165 Rome, Italy
| | - Maria Vinci
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Antonella Cacchione
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Emmanuel de Billy
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Giacomina Megaro
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Andrea Carai
- Neurosurgery Unit, Department of Neurosciences, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Angela Mastronuzzi
- Department of Onco-Hematology, Cell and Gene Therapies, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
- Faculty of Medicine and Surgery, Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| |
Collapse
|
6
|
Bronk JK, Hou P, Amsbaugh MJ, Khatua S, Mahajan A, Ketonen L, McGovern SL. Sequential Diffusion Tensor Imaging and Magnetic Resonance Spectroscopy in Patients Undergoing Reirradiation for Progressive Diffuse Intrinsic Pontine Glioma. Adv Radiat Oncol 2022; 7:100847. [PMID: 35071836 PMCID: PMC8763636 DOI: 10.1016/j.adro.2021.100847] [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: 12/07/2020] [Accepted: 10/31/2021] [Indexed: 11/29/2022] Open
Abstract
Purpose Diffusion tensor imaging for evaluation of white matter tracts is used with magnetic resonance spectroscopy (MRS) to improve management of diffuse intrinsic pontine glioma (DIPG). Changes in the apparent diffusion coefficient (ADC), fractional anisotropy (FA), and tumor metabolite ratios have been reported after initial radiation for DIPG, but these markers have not been studied sequentially in patients undergoing reirradiation for progressive DIPG. Here, we report a case series of 4 patients who received reirradiation for progressive DIPG on a prospective clinical trial in which we evaluated quantitative changes in FA, ADC, and tumor metabolites and qualitative changes in white matter tracts. Methods and Materials The median reirradiation dose was 25.2 Gy (24-30.8 Gy). Fiber tracking was performed using standard tractography analysis. The FA and ADC values for the corticospinal and medial lemniscus tracts were calculated before and after reirradiation. Multivoxel MRS was performed. Findings were correlated with clinical features and conventional MRI of tumors. Results All patients had an initial response to reirradiation as shown by a decrease in tumor size. In general, FA increased with disease response and decreased with progression, whereas ADC decreased with disease response and increased with progression. At second progression, the FA fold change relative to values during disease response decreased in both patients with available imaging at second progression. Visualization of tracts demonstrated robust reconstitution of previously disrupted paths during tumor response; conversely, there was increased fiber tract disruption and infiltration during tumor progression. The MRS analysis revealed a decrease in choline:creatinine and choline:N-acetylaspartate ratios during tumor response and increase during progression. Conclusions Distinct changes in white matter tracts and tumor metabolism were observed in patients with DIPG undergoing reirradiation on a prospective clinical trial. Changes related to tumor response and progression were observed after 24 to 30.8 Gy reirradiation.
Collapse
Affiliation(s)
- Julianna K. Bronk
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ping Hou
- Departments of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark J. Amsbaugh
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Soumen Khatua
- Departments of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anita Mahajan
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Leena Ketonen
- Departments of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Susan L. McGovern
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Corresponding author: Susan L. McGovern, MD, PhD
| |
Collapse
|
7
|
McGee KP, Hwang K, Sullivan DC, Kurhanewicz J, Hu Y, Wang J, Li W, Debbins J, Paulson E, Olsen JR, Hua C, Warner L, Ma D, Moros E, Tyagi N, Chung C. Magnetic resonance biomarkers in radiation oncology: The report of AAPM Task Group 294. Med Phys 2021; 48:e697-e732. [PMID: 33864283 PMCID: PMC8361924 DOI: 10.1002/mp.14884] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/24/2021] [Accepted: 03/28/2021] [Indexed: 12/16/2022] Open
Abstract
A magnetic resonance (MR) biologic marker (biomarker) is a measurable quantitative characteristic that is an indicator of normal biological and pathogenetic processes or a response to therapeutic intervention derived from the MR imaging process. There is significant potential for MR biomarkers to facilitate personalized approaches to cancer care through more precise disease targeting by quantifying normal versus pathologic tissue function as well as toxicity to both radiation and chemotherapy. Both of which have the potential to increase the therapeutic ratio and provide earlier, more accurate monitoring of treatment response. The ongoing integration of MR into routine clinical radiation therapy (RT) planning and the development of MR guided radiation therapy systems is providing new opportunities for MR biomarkers to personalize and improve clinical outcomes. Their appropriate use, however, must be based on knowledge of the physical origin of the biomarker signal, the relationship to the underlying biological processes, and their strengths and limitations. The purpose of this report is to provide an educational resource describing MR biomarkers, the techniques used to quantify them, their strengths and weakness within the context of their application to radiation oncology so as to ensure their appropriate use and application within this field.
Collapse
Affiliation(s)
| | - Ken‐Pin Hwang
- Department of Imaging PhysicsDivision of Diagnostic ImagingMD Anderson Cancer CenterUniversity of TexasHoustonTexasUSA
| | | | - John Kurhanewicz
- Department of RadiologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Yanle Hu
- Department of Radiation OncologyMayo ClinicScottsdaleArizonaUSA
| | - Jihong Wang
- Department of Radiation OncologyMD Anderson Cancer CenterUniversity of TexasHoustonTexasUSA
| | - Wen Li
- Department of Radiation OncologyUniversity of ArizonaTucsonArizonaUSA
| | - Josef Debbins
- Department of RadiologyBarrow Neurologic InstitutePhoenixArizonaUSA
| | - Eric Paulson
- Department of Radiation OncologyMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Jeffrey R. Olsen
- Department of Radiation OncologyUniversity of Colorado Denver ‐ Anschutz Medical CampusDenverColoradoUSA
| | - Chia‐ho Hua
- Department of Radiation OncologySt. Jude Children’s Research HospitalMemphisTennesseeUSA
| | | | - Daniel Ma
- Department of Radiation OncologyMayo ClinicRochesterMinnesotaUSA
| | - Eduardo Moros
- Department of Radiation OncologyMoffitt Cancer CenterTampaFloridaUSA
| | - Neelam Tyagi
- Department of Medical PhysicsMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Caroline Chung
- Department of Radiation OncologyMD Anderson Cancer CenterUniversity of TexasHoustonTexasUSA
| |
Collapse
|
8
|
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
|
9
|
Fudaba H, Momii Y, Matsuta H, Onishi K, Kawasaki Y, Sugita K, Shimomura T, Fujiki M. Perfusion Parameter Obtained on 3-Tesla Magnetic Resonance Imaging and the Ki-67 Labeling Index Predict the Overall Survival of Glioblastoma. World Neurosurg 2021; 149:e469-e480. [PMID: 33567368 DOI: 10.1016/j.wneu.2021.02.002] [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: 10/24/2020] [Revised: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Pulsed arterial spin-labeling, diffusion tensor imaging (DTI), and magnetic resonance spectroscopy (MRS) are useful for predicting glioma survival. We performed a comparative review of multiple parameters obtained using these pulse sequences on 3-Tesla magnetic resonance imaging (MRI) including the molecular status and Ki-67 labeling index in newly diagnosed supratentorial glioblastomas. METHODS A total of 35 patients with glioblastomas underwent pulsed arterial spin-labeling, DTI, and MRS studies using 3-Tesla MRI preoperatively. The isocitrate dehydrogenase (IDH) mutation status, methylguanine-DNA methyltransferase methylation status, and Ki-67 labeling index were calculated from the tumor specimen. Cutoff values were identified by analyzing a receiver operating characteristic curve, and the multivariate survival statistical technique was performed to determine the significant and independent parameters for predicting overall survival. RESULTS The multivariate Cox analysis showed that the maximum/mean relative cerebral blood flow (rCBF) ratio and the Ki-67 labeling index were significant and independent predictive parameters with a cutoff value of 1.589 for the maximum rCBF ratio, 1.286 for the mean rCBF ratio, and 19% for the Ki-67 labeling index and hazard ratios of 6.132 and 5.119, respectively. The Kaplan-Meier survival curves showed that patients with higher rCBF ratios and Ki-67 labeling indices had a shorter overall survival than others, with median overall survival durations of 479 (95% CI, 370-559) and 1243 (95% CI, 666-NA) days, respectively (P = 0.000167). CONCLUSIONS Our findings indicate that the preoperative rCBF ratio and Ki-67 labeling index are useful parameters for predicting the overall survival of cerebral glioblastomas.
Collapse
Affiliation(s)
- Hirotaka Fudaba
- Department of Neurosurgery, Oita University Faculty of Medicine, Oita, Japan.
| | - Yasutomo Momii
- Department of Neurosurgery, Oita University Faculty of Medicine, Oita, Japan
| | - Hiroyuki Matsuta
- Department of Neurosurgery, Oita University Faculty of Medicine, Oita, Japan
| | - Kouhei Onishi
- Department of Neurosurgery, Oita University Faculty of Medicine, Oita, Japan
| | - Yukari Kawasaki
- Department of Neurosurgery, Oita University Faculty of Medicine, Oita, Japan
| | - Kenji Sugita
- Department of Neurosurgery, Oita University Faculty of Medicine, Oita, Japan
| | - Tsuyoshi Shimomura
- Department of Medical Informatics, Oita University Faculty of Medicine, Oita, Japan
| | - Minoru Fujiki
- Department of Neurosurgery, Oita University Faculty of Medicine, Oita, Japan
| |
Collapse
|
10
|
Laino ME, Young R, Beal K, Haque S, Mazaheri Y, Corrias G, Bitencourt AG, Karimi S, Thakur SB. Magnetic resonance spectroscopic imaging in gliomas: clinical diagnosis and radiotherapy planning. BJR Open 2020; 2:20190026. [PMID: 33178960 PMCID: PMC7594883 DOI: 10.1259/bjro.20190026] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 01/13/2020] [Accepted: 03/18/2020] [Indexed: 12/23/2022] Open
Abstract
The reprogramming of cellular metabolism is a hallmark of cancer diagnosis and prognosis. Proton magnetic resonance spectroscopic imaging (MRSI) is a non-invasive diagnostic technique for investigating brain metabolism to establish cancer diagnosis and IDH gene mutation diagnosis as well as facilitate pre-operative planning and treatment response monitoring. By allowing tissue metabolism to be quantified, MRSI provides added value to conventional MRI. MRSI can generate metabolite maps from a single volume or multiple volume elements within the whole brain. Metabolites such as NAA, Cho and Cr, as well as their ratios Cho:NAA ratio and Cho:Cr ratio, have been used to provide tumor diagnosis and aid in radiation therapy planning as well as treatment assessment. In addition to these common metabolites, 2-hydroxygluterate (2HG) has also been quantified using MRSI following the recent discovery of IDH mutations in gliomas. This has opened up targeted drug development to inhibit the mutant IDH pathway. This review provides guidance on MRSI in brain gliomas, including its acquisition, analysis methods, and evolving clinical applications.
Collapse
Affiliation(s)
| | - Robert Young
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | - Kathryn Beal
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | - Sofia Haque
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | | | - Giuseppe Corrias
- Department of Radiology, University of Cagliari, 40 Via Università, 09124 Cagliari, Italy
| | | | - Sasan Karimi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | | |
Collapse
|
11
|
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
|
12
|
Take Advantage of Glutamine Anaplerosis, the Kernel of the Metabolic Rewiring in Malignant Gliomas. Biomolecules 2020; 10:biom10101370. [PMID: 32993063 PMCID: PMC7599606 DOI: 10.3390/biom10101370] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/18/2020] [Accepted: 09/24/2020] [Indexed: 12/11/2022] Open
Abstract
Glutamine is a non-essential amino acid that plays a key role in the metabolism of proliferating cells including neoplastic cells. In the central nervous system (CNS), glutamine metabolism is particularly relevant, because the glutamine-glutamate cycle is a way of controlling the production of glutamate-derived neurotransmitters by tightly regulating the bioavailability of the amino acids in a neuron-astrocyte metabolic symbiosis-dependent manner. Glutamine-related metabolic adjustments have been reported in several CNS malignancies including malignant gliomas that are considered ‘glutamine addicted’. In these tumors, glutamine becomes an essential amino acid preferentially used in energy and biomass production including glutathione (GSH) generation, which is crucial in oxidative stress control. Therefore, in this review, we will highlight the metabolic remodeling that gliomas undergo, focusing on glutamine metabolism. We will address some therapeutic regimens including novel research attempts to target glutamine metabolism and a brief update of diagnosis strategies that take advantage of this altered profile. A better understanding of malignant glioma cell metabolism will help in the identification of new molecular targets and the design of new therapies.
Collapse
|
13
|
Gilligan LA, DeWire-Schottmiller MD, Fouladi M, DeBlank P, Leach JL. Tumor Response Assessment in Diffuse Intrinsic Pontine Glioma: Comparison of Semiautomated Volumetric, Semiautomated Linear, and Manual Linear Tumor Measurement Strategies. AJNR Am J Neuroradiol 2020; 41:866-873. [PMID: 32354716 DOI: 10.3174/ajnr.a6555] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/26/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND PURPOSE 2D measurements of diffuse intrinsic pontine gliomas are limited by variability, and volumetric response criteria are poorly defined. Semiautomated 2D measurements may improve consistency; however, the impact on tumor response assessments is unknown. The purpose of this study was to compare manual 2D, semiautomated 2D, and volumetric measurement strategies for diffuse intrinsic pontine gliomas. MATERIALS AND METHODS This study evaluated patients with diffuse intrinsic pontine gliomas through a Phase I/II trial (NCT02607124). Clinical 2D cross-product values were derived from manual linear measurements (cross-product = long axis × short axis). By means of dedicated software (mint Lesion), tumor margins were traced and maximum cross-product and tumor volume were automatically derived. Correlation and bias between methods were assessed, and response assessment per measurement strategy was reported. RESULTS Ten patients (median age, 7.6 years) underwent 58 MR imaging examinations. Correlation and mean bias (95% limits) of percentage change in tumor size from prior examinations were the following: clinical and semiautomated cross-product, r = 0.36, -1.5% (-59.9%, 56.8%); clinical cross-product and volume, r = 0.61, -2.1% (-52.0%, 47.8%); and semiautomated cross-product and volume, r = 0.79, 0.6% (-39.3%, 38.1%). Stable disease, progressive disease, and partial response rates per measurement strategy were the following: clinical cross-product, 82%, 18%, 0%; semiautomated cross-product, 54%, 42%, 4%; and volume, 50%, 46%, 4%, respectively. CONCLUSIONS Manual 2D cross-product measurements may underestimate tumor size and disease progression compared with semiautomated 2D and volumetric measurements.
Collapse
Affiliation(s)
- L A Gilligan
- From the Departments of Radiology (L.A.G., J.L.L.).,Department of Graduate Medical Education (L.A.G., M.D.D.-S.), Mount Carmel Health System, Columbus, Ohio
| | - M D DeWire-Schottmiller
- and Cancer and Blood Diseases Institute (M.D.D.-S., M.F.), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Graduate Medical Education (L.A.G., M.D.D.-S.), Mount Carmel Health System, Columbus, Ohio
| | - M Fouladi
- and Cancer and Blood Diseases Institute (M.D.D.-S., M.F.), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,and Departments of Pediatrics (M.F., P.D.)
| | - P DeBlank
- and Departments of Pediatrics (M.F., P.D.)
| | - J L Leach
- From the Departments of Radiology (L.A.G., J.L.L.) .,Radiology (J.L.L.), University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
14
|
Akagi Y, Noguchi N, Hata N, Hatae R, Michiwaki Y, Sangatsuda Y, Amemiya T, Kuga D, Yamashita K, Togao O, Hiwatashi A, Yoshimoto K, Mizoguchi M, Iihara K. Correlation between prognosis of glioblastoma and choline/N-acetyl aspartate ratio in MR spectroscopy. INTERDISCIPLINARY NEUROSURGERY 2019. [DOI: 10.1016/j.inat.2019.100498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
15
|
Gandía-González ML, Cerdán S, Barrios L, López-Larrubia P, Feijoó PG, Palpan A, Roda JM, Solivera J. Assessment of Overall Survival in Glioma Patients as Predicted by Metabolomic Criteria. Front Oncol 2019; 9:328. [PMID: 31134147 PMCID: PMC6524167 DOI: 10.3389/fonc.2019.00328] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/11/2019] [Indexed: 11/17/2022] Open
Abstract
Objective: We assess the efficacy of the metabolomic profile from glioma biopsies in providing estimates of postsurgical Overall Survival in glioma patients. Methods: Tumor biopsies from 46 patients bearing gliomas, obtained neurosurgically in the period 1992–1998, were analyzed by high resolution 1H magnetic resonance spectroscopy (HR- 1H MRS), following retrospectively individual postsurgical Overall Survival up to 720 weeks. Results: The Overall Survival profile could be resolved in three groups; Short (shorter than 52 weeks, n = 19), Intermediate (between 53 and 364 weeks, n = 19) or Long (longer than 365 weeks, n = 8), respectively. Classical histopathological analysis assigned WHO grades II–IV to every biopsy but notably, some patients with low grade glioma depicted unexpectedly Short Overall Survival, while some patients with high grade glioma, presented unpredictably Long Overall Survival. To explore the reasons underlying these different responses, we analyzed HR-1H MRS spectra from acid extracts of the same biopsies, to characterize the metabolite patterns associated to OS predictions. Poor prognosis was found in biopsies with higher contents of alanine, acetate, glutamate, total choline, phosphorylcholine, and glycine, while more favorable prognosis was achieved in biopsies with larger contents of total creatine, glycerol-phosphorylcholine, and myo-inositol. We then implemented a multivariate analysis to identify hierarchically the influence of metabolomic biomarkers on OS predictions, using a Classification Regression Tree (CRT) approach. The CRT based in metabolomic biomarkers grew up to three branches and split into eight nodes, predicting correctly the outcome of 94.7% of the patients in the Short Overall Survival group, 78.9% of the patients in the Intermediate Overall Survival group, and 75% of the patients in the Long Overall Survival group, respectively. Conclusion: Present results indicate that metabolic profiling by HR-1H MRS improves the Overall Survival predictions derived exclusively from classical histopathological gradings, thus favoring more precise therapeutic decisions.
Collapse
Affiliation(s)
| | - Sebastián Cerdán
- Institute of Biomedical Research "Alberto Sols" CSIC/UAM, Madrid, Spain
| | | | | | - Pablo G Feijoó
- Department of Neurosurgery, Hospital Universitario La Paz, Madrid, Spain
| | - Alexis Palpan
- Department of Neurosurgery, Hospital Universitario La Paz, Madrid, Spain
| | - José M Roda
- Department of Neurosurgery, Hospital Universitario La Paz, Madrid, Spain
| | - Juan Solivera
- Department of Neurosurgery, University Hospital Reina Sofía, Córdoba, Spain
| |
Collapse
|
16
|
Colafati GS, Voicu IP, Carducci C, Caulo M, Vinci M, Diomedi-Camassei F, Merli P, Carai A, Miele E, Cacchione A, Tomà P, Locatelli F, Mastronuzzi A. Direct Involvement of Cranial Nerve V at Diagnosis in Patients With Diffuse Intrinsic Pontine Glioma: A Potential Magnetic Resonance Predictor of Short-Term Survival. Front Oncol 2019; 9:204. [PMID: 31019890 PMCID: PMC6458256 DOI: 10.3389/fonc.2019.00204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 03/11/2019] [Indexed: 12/05/2022] Open
Abstract
Background: Diffuse intrinsic pontine glioma (DIPG) has a dismal prognosis. Magnetic resonance imaging (MRI) remains the gold standard for non-invasive DIPG diagnosis. MRI features have been tested as surrogate biomarkers. We investigated the direct involvement of cranial nerve V (CN V) in DIPG at diagnosis and its utility as predictor of poor overall survival. Materials and Methods: We examined MRI scans of 35 consecutive patients with radiological diagnosis of DIPG. Direct involvement of CN V was assessed on the diagnostic scans. Differences in overall survival (OS) and time to progression (TTP) were analyzed for involvement of CN V, sex, age, tumor size, ring enhancement, and treatment regimen. Correlations between involvement of CN V and disease dissemination, magnet strength and slice thickness were analyzed. Statistical analyses included Kaplan-Meier curves, log-rank test and Spearman's Rho. Results: After excluding six long-term survivors, 29 patients were examined (15 M, 14 F). Four patients presented direct involvement of CN V. Histological data were available in 12 patients. Median OS was 11 months (range 3-23 months). Significant differences in OS were found for direct involvement of CN V (median OS: 7 months, 95% CI 1.1-12.9 months for involvement of CN V vs. 13 months, 95% CI 10.2-15.7 for lack of involvement of CN V, respectively, p < 0.049). Significant differences in TTP were found for the two treatment regimens (median TTP: 4 months, 95% CI 2.6-5.3 vs. 7 months, 95% CI 5.9-8.1, respectively, p < 0.027). No significant correlation was found between involvement of CN V and magnet strength or slice thickness (r = -0.201; p = NS). A trend toward positive correlation was found between direct involvement of CN V at diagnosis and dissemination of disease at follow-up (r = 0.347; p < 0.065). Conclusions: In our cohort, direct involvement of CN V correlated with poor prognosis. Based on our data, we suggest that in DIPG direct involvement of CN V should be routinely evaluated on diagnostic scans.
Collapse
Affiliation(s)
| | - Ioan Paul Voicu
- Department of Imaging, Neuroradiology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Chiara Carducci
- Department of Imaging, Neuroradiology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Massimo Caulo
- Department of Neuroscience and Imaging, ITAB-Institute of Advanced Biomedical Technologies, University G. d'Annunzio, Chieti, Italy
| | - Maria Vinci
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Pietro Merli
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Andrea Carai
- Neurosurgery Unit, Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Evelina Miele
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Antonella Cacchione
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Paolo Tomà
- Radiology Unit, Department of Imaging, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Franco Locatelli
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Angela Mastronuzzi
- Department of Onco-haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
17
|
Role of Radiation Therapy in the Management of Diffuse Intrinsic Pontine Glioma: A Systematic Review. Adv Radiat Oncol 2019; 4:520-531. [PMID: 31360809 PMCID: PMC6639749 DOI: 10.1016/j.adro.2019.03.009] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 03/20/2019] [Indexed: 01/05/2023] Open
Abstract
Purpose Diffuse intrinsic pontine glioma (DIPG) is the most aggressive primary pediatric brain tumor, with <10% of children surviving 2 years. Radiation therapy (RT) remains the mainstay of treatment, but there is a great clinical need for improvements and advancements in treatment strategies. The aim of this systematic review was to identify all available studies in which RT was used to treat patients with DIPG. Methods and Materials A literature search for studies published up to March 10, 2018 was conducted using the PubMed database. We identified 384 articles using search items “diffuse intrinsic pontine glioma” and 221 articles using search items “diffuse brainstem glioma radiotherapy.” Included studies were prospective and retrospective series that reported outcomes of DIPG treatment with RT. Results We identified 49 studies (1286 patients) using upfront conventionally fractionated RT, 5 studies (92 patients) using hypofractionated RT, and 8 studies (348 patients) using hyperfractionated RT. The mean median overall survival (OS) was 12.0 months, 10.2 months, and 7.9 months in patients who received conventional, hyperfractionated, and hypofractionated RT regimens, respectively. Patients undergoing radiosensitizing therapy had a mean median OS of 11.5 months, and patients who did not receive concomitant systemic therapy had an OS of 9.4 months. In patients who received salvage RT, the mean median OS from initial diagnosis was 16.3 months. Conclusions As one of the largest systematic reviews examining RT for DIPG, this report may serve as a useful tool to help clinicians choose the most appropriate treatment approach, while also providing a platform for future investigations into the utility of RT and systemic therapy.
Collapse
|
18
|
Ter Voert EEGW, Heijmen L, van Asten JJA, Wright AJ, Nagtegaal ID, Punt CJA, de Wilt JHW, van Laarhoven HWM, Heerschap A. Levels of choline-containing compounds in normal liver and liver metastases of colorectal cancer as recorded by 1 H MRS. NMR IN BIOMEDICINE 2019; 32:e4035. [PMID: 30457686 DOI: 10.1002/nbm.4035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 09/07/2018] [Accepted: 09/28/2018] [Indexed: 06/09/2023]
Abstract
PURPOSE A relatively high signal for choline-containing compounds (total choline, tCho) is commonly found in 1 H MR spectra of malignant tumors, but it is unclear if this also occurs in tumors in the liver. We evaluated the potential of the tCho signal in single voxel 1 H MR spectra of the human liver to assess metastases of colorectal cancers. EXPERIMENT MR spectra of an 8 cm3 PRESS-localized voxel were obtained at 3 T from the livers of 12 healthy volunteers and from metastatic lesions in 20 patients in two different sessions. To correct for motion artifacts, sequentially recorded spectra were individually phased and frequency aligned before averaging. Spectra were analyzed using LCModel and tissue levels estimated by water referencing. Repeatability was assessed with Bland-Altman analyses. To estimate tumor necrosis, diffusion-weighted imaging of the liver was performed. High resolution magic angle spinning (HRMAS) spectra of tumor and normal liver samples were obtained at 11.7 T. RESULTS With increasing tumor volumes, tCho levels decreased, indicating a partial volume effect. Mean tCho content in tumors larger than the PRESS voxel (>8 cm3 ) was significantly lower (p < 0.01) than for normal liver: 1.6 (range 0.0-3.4) versus 6.9 (range 4.9-11.1) mmol/kg wet weight, while it was comparable for tumors smaller than 8 cm3 : 7.0 (range 3.8-9.3) mmol/kg. The higher 90th percentile apparent diffusion coefficient value in the larger lesions indicates more necrosis. Measurement repeatability was average in normal livers and poor in tumors. HRMAS did not show substantial differences in choline-containing compounds between normal liver and metastasis. CONCLUSION An increased tCho content was not observed in 1 H MR spectra of liver metastasis of colorectal cancer, compared with normal liver. This may be due to the background of a high tCho signal in spectra of normal liver or to an intrinsic lower tCho content in these tumors, but is most likely the result of necrosis in metastatic tumor tissue.
Collapse
Affiliation(s)
- Edwin E G W Ter Voert
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Linda Heijmen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jack J A van Asten
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alan J Wright
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Iris D Nagtegaal
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cornelis J A Punt
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Oncology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Johannes H W de Wilt
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hanneke W M van Laarhoven
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Oncology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Arend Heerschap
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
19
|
Cohen KJ, Jabado N, Grill J. Diffuse intrinsic pontine gliomas-current management and new biologic insights. Is there a glimmer of hope? Neuro Oncol 2018; 19:1025-1034. [PMID: 28371920 DOI: 10.1093/neuonc/nox021] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) has proven to be one of the most challenging of all pediatric cancers. Owing to a historical reticence to obtain tumor tissue for study, and based on an erroneous assumption that the biology of DIPG would mirror that of supratentorial high-grade astrocytomas, innumerable studies have been undertaken-all of which have had a negligible impact on the natural history of this disease. More recently, improvements in neurosurgical techniques have allowed for the safe upfront biopsy of DIPG, which, together with a wider use of autopsy tissue, has led to an evolving understanding of the biology of this tumor. The discovery of a recurrent somatic gain-of-function mutation leading to lysine 27 to methionine (p.Lys27Met, K27M) substitution in histone 3 variants characterizes more than 85% of DIPG, suggesting for the first time the role of the epigenome and histones in the pathogenesis of this disease, and more unified diagnostic criteria. Along with further molecular insights into the pathogenesis of DIPG, rational targets are being identified and studied in the hopes of improving the otherwise dismal outcome for children with DIPG.
Collapse
Affiliation(s)
- Kenneth J Cohen
- Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland; Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Université Paris-Saclay & Gustave Roussy Unité Mixte de Recherche 8203 du Centre National de la Recherche Scientifique & Departement de Cancerologie de l'Enfant et de l'Adolescent, Villejuif, France
| | - Nada Jabado
- Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland; Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Université Paris-Saclay & Gustave Roussy Unité Mixte de Recherche 8203 du Centre National de la Recherche Scientifique & Departement de Cancerologie de l'Enfant et de l'Adolescent, Villejuif, France
| | - Jacques Grill
- Pediatric Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland; Department of Pediatrics, McGill University, Montreal, Quebec, Canada; Université Paris-Saclay & Gustave Roussy Unité Mixte de Recherche 8203 du Centre National de la Recherche Scientifique & Departement de Cancerologie de l'Enfant et de l'Adolescent, Villejuif, France
| |
Collapse
|
20
|
Chen F, Li Z, Weng C, Li P, Tu L, Chen L, Xie W, Li L. Progressive multifocal exophytic pontine glioblastoma: a case report with literature review. CHINESE JOURNAL OF CANCER 2017; 36:34. [PMID: 28347331 PMCID: PMC5369214 DOI: 10.1186/s40880-017-0201-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 01/03/2017] [Indexed: 11/10/2022]
Abstract
Multifocal pontine glioblastoma exhibiting an exophytic growth pattern in the cerebello-pontine angle (CPA) is rare. We present a case of a 5-year-old girl with consecutive neurological imaging and other clinical findings indicating progressive multifocal exophytic pontine glioblastoma. Three lesions were reported, of which two were initially presented, and one was developed 2 months later. One lesion demonstrated a progressing exophytic extension in the cistern of the left side of the CPA. The other two lesions were located and confined within the pons. Initial magnetic resonance imaging and positron emission tomography–computed tomography indicated low-grade glioma or inflammatory disease. However, 2 and 3 months later, subsequent magnetic resonance spectroscopy (MRS) displayed elevated choline and depressed N-acetyl aspartate peaks compared with the peaks on the initial MRS, indicating a high-grade glioma. Subtotal resection was performed for the CPA lesion. Histopathologic examination showed discrepant features of different parts of the CPA lesion. The patient received no further chemotherapy or radiotherapy and died 2 months after surgery. The multifocal and exophytic features of this case and the heterogeneous manifestations on neurological images were rare and confusing for both diagnosis and surgical decision-making. Our case report may contribute knowledge and helpful guidance for other medical doctors.
Collapse
Affiliation(s)
- Fanfan Chen
- Neurosurgery Department, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, Guangdong, P. R. China
| | - Zongyang Li
- Neurosurgery Department, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, 518000, Guangdong, P. R. China
| | - Chengyin Weng
- Oncology Department, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, Guangdong, P. R. China
| | - Peng Li
- Neurosurgery Department, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, Guangdong, P. R. China
| | - Lanbo Tu
- Neurosurgery Department, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, Guangdong, P. R. China
| | - Lei Chen
- Neurosurgery Department, Shenzhen Second People's Hospital, Shenzhen University, Shenzhen, 518000, Guangdong, P. R. China
| | - Wei Xie
- Neurosurgery Department, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, Guangdong, P. R. China
| | - Ling Li
- Record Department, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, 510180, Guangdong, P. R. China.
| |
Collapse
|
21
|
Bredlau AL, Dixit S, Chen C, Broome AM. Nanotechnology Applications for Diffuse Intrinsic Pontine Glioma. Curr Neuropharmacol 2017; 15:104-115. [PMID: 26903150 PMCID: PMC5327462 DOI: 10.2174/1570159x14666160223121002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 10/12/2015] [Accepted: 01/30/2016] [Indexed: 12/19/2022] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are invariably fatal tumors found in the pons of elementary school aged children. These tumors are grade II-IV gliomas, with a median survival of less than 1 year from diagnosis when treated with standard of care (SOC) therapy. Nanotechnology may offer therapeutic options for the treatment of DIPGs. Multiple nanoparticle formulations are currently being investigated for the treatment of DIPGs. Nanoparticles based upon stable elements, polymer nanoparticles, and organic nanoparticles are under development for the treatment of brain tumors, including DIPGs. Targeting of nanoparticles is now possible as delivery techniques that address the difficulty in crossing the blood brain barrier (BBB) are developed. Theranostic nanoparticles, a combination of therapeutics and diagnostic nanoparticles, improve imaging of the cancerous tissue while delivering therapy to the local region. However, additional time and attention should be directed to developing a nanoparticle delivery system for treatment of the uniformly fatal pediatric disease of DIPG.
Collapse
Affiliation(s)
| | | | | | - Ann-Marie Broome
- Department of Radiology and Radiological Sciences, Medical University of South Carolina, 68 President Street, MSC 120/BEB 213, Charleston, SC 29425, USA
| |
Collapse
|
22
|
Tisnado J, Young R, Peck KK, Haque S. Conventional and Advanced Imaging of Diffuse Intrinsic Pontine Glioma. J Child Neurol 2016; 31:1386-93. [PMID: 27071471 PMCID: PMC5659185 DOI: 10.1177/0883073816634855] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 01/12/2016] [Indexed: 12/11/2022]
Abstract
Diffuse intrinsic pontine glioma is the most common brainstem tumor in pediatric patients. This tumor remains one of the most deadly pediatric brain tumors. The diagnosis primarily relies on clinical symptoms and imaging findings. Conventional MRI provides a noninvasive accurate method of diagnosis of these tumors. Advanced MRI techniques are becoming more widely used and studied as additional noninvasive methods to assist clinicians in initial diagnosis and staging, monitoring disease, as well as in surgical and radiation planning. This article will provide an overview of DIPG and describe the typical imaging findings with a focus on advanced imaging techniques.
Collapse
Affiliation(s)
- Jamie Tisnado
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert Young
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kyung K Peck
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sofia Haque
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
23
|
Hennika T, Becher OJ. Diffuse Intrinsic Pontine Glioma: Time for Cautious Optimism. J Child Neurol 2016; 31:1377-85. [PMID: 26374787 PMCID: PMC6025797 DOI: 10.1177/0883073815601495] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/20/2015] [Indexed: 01/03/2023]
Abstract
Diffuse intrinsic pontine glioma is a lethal brain cancer that arises in the pons of children. The median survival for children with diffuse intrinsic pontine glioma is less than 1 year from diagnosis, and no improvement in survival has been realized in more than 30 years. Currently, the standard of care for diffuse intrinsic pontine glioma is focal radiation therapy, which provides only temporary relief. Recent genomic analysis of tumors from biopsies and autopsies, have resulted in the discovery of K27M H3.3/H3.1 mutations in 80% and ACVR1 mutations in 25% of diffuse intrinsic pontine gliomas, providing renewed hope for future success in identifying effective therapies. In addition, as stereotactic tumor biopsies at diagnosis at specialized centers have been demonstrated to be safe, biopsies have now been incorporated into several prospective clinical trials. This article summarizes the epidemiology, clinical presentation, diagnosis, prognosis, molecular genetics, current treatment, and future therapeutic directions for diffuse intrinsic pontine glioma.
Collapse
Affiliation(s)
- Tammy Hennika
- Department of Pediatrics Duke University Medical Center, Durham, NC, USA Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA
| | - Oren J Becher
- Department of Pediatrics Duke University Medical Center, Durham, NC, USA Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, USA Department of Pathology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
24
|
Discrepant longitudinal volumetric and metabolic evolution of diffuse intrinsic Pontine gliomas during treatment: implications for current response assessment strategies. Neuroradiology 2016; 58:1027-1034. [PMID: 27438806 DOI: 10.1007/s00234-016-1724-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/04/2016] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Based on clinical observations, we hypothesized that in infiltrative high-grade brainstem neoplasms, such as diffuse intrinsic pontine glioma (DIPG), longitudinal metabolic evaluation of the tumor by magnetic resonance spectroscopy (MRS) may be more accurate than volumetric data for monitoring the tumor's biological evolution during standard treatment. METHODS We evaluated longitudinal MRS data and corresponding tumor volumes of 31 children with DIPG. We statistically analyzed correlations between tumor volume and ratios of Cho/NAA, Cho/Cr, and NAA/Cr at key time points during the course of the disease through the end of the progression-free survival period. RESULTS By the end of RT, tumor volume had significantly decreased from the baseline (P < .0001) and remained decreased through the last available follow-up magnetic resonance imaging study (P = .007632). However, the metabolic profile of the tumor tissue (Cho/Cr, NAA/Cr, and Cho/NAA ratios) did not change significantly over time. CONCLUSION Our data show that longitudinal tumor volume and metabolic profile changes are dissociated in patients with DIPG during progression-free survival. Volume changes, therefore, may not accurately reflect treatment-related changes in tumor burden. This study adds to the existing body of evidence that the value of conventional MRI metrics, including volumetric data, needs to be reevaluated critically and, in infiltrative tumors in particular, may not be useful as study end-points in clinical trials. We submit that advanced quantitative MRI data, including robust, MRS-based metabolic ratios and diffusion and perfusion metrics, may be better surrogate markers of key end-points in clinical trials.
Collapse
|
25
|
Basic Principles and Clinical Applications of Magnetic Resonance Spectroscopy in Neuroradiology. J Comput Assist Tomogr 2016; 40:1-13. [PMID: 26484954 DOI: 10.1097/rct.0000000000000322] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Magnetic resonance spectroscopy is a powerful tool to assist daily clinical diagnostics. This review is intended to give an overview on basic principles of the technology, discuss some of its technical aspects, and present typical applications in daily clinical routine in neuroradiology.
Collapse
|
26
|
Pediatric brainstem gliomas: new understanding leads to potential new treatments for two very different tumors. Curr Oncol Rep 2015; 17:436. [PMID: 25702179 DOI: 10.1007/s11912-014-0436-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Pediatric brainstem gliomas include low-grade focal brainstem gliomas (FBSG) and high-grade diffuse intrinsic pontine gliomas (DIPG). These tumors share a crucial and eloquent area of the brain as their location, which carries common challenges for treatment. Otherwise, though, these two diseases are very different in terms of presentation, biology, treatment, and prognosis. FBSG usually present with greater than 3 months of symptoms, while DIPG are usually diagnosed within 3 months of symptom onset. Surgery remains the preferred initial treatment for FBSG, with chemotherapy used for persistent, recurrent, or inoperable disease; conversely, radiation is the only known effective treatment for DIPG. Recent developments in biological understanding of both tumors have led to new treatment possibilities. In FBSG, two genetic changes related to BRAF characterize the majority of tumors, and key differences in their biological effects are informing strategies for targeted chemotherapy use. In DIPG, widespread histone H3 and ACVR1 mutations have led to new hope for effective targeted treatments. FBSG has an excellent prognosis, while the long-term survival rate of DIPG tragically remains near zero. In this review, we cover the epidemiology, biology, presentation, imaging characteristics, multimodality treatment, and prognosis of FBSG and DIPG, with a focus on recent biological discoveries.
Collapse
|
27
|
Coutinho de Souza P, Mallory S, Smith N, Saunders D, Li XN, McNall-Knapp RY, Fung KM, Towner RA. Inhibition of Pediatric Glioblastoma Tumor Growth by the Anti-Cancer Agent OKN-007 in Orthotopic Mouse Xenografts. PLoS One 2015; 10:e0134276. [PMID: 26248280 PMCID: PMC4527837 DOI: 10.1371/journal.pone.0134276] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 07/08/2015] [Indexed: 12/31/2022] Open
Abstract
Pediatric glioblastomas (pGBM), although rare, are one of the leading causes of cancer-related deaths in children, with tumors essentially refractory to existing treatments. Here, we describe the use of conventional and advanced in vivo magnetic resonance imaging (MRI) techniques to assess a novel orthotopic xenograft pGBM mouse (IC-3752GBM patient-derived culture) model, and to monitor the effects of the anti-cancer agent OKN-007 as an inhibitor of pGBM tumor growth. Immunohistochemistry support data is also presented for cell proliferation and tumor growth signaling. OKN-007 was found to significantly decrease tumor volumes (p<0.05) and increase animal survival (p<0.05) in all OKN-007-treated mice compared to untreated animals. In a responsive cohort of treated animals, OKN-007 was able to significantly decrease tumor volumes (p<0.0001), increase survival (p<0.001), and increase diffusion (p<0.01) and perfusion rates (p<0.05). OKN-007 also significantly reduced lipid tumor metabolism in responsive animals [(Lip1.3 and Lip0.9)-to-creatine ratio (p<0.05)], as well as significantly decrease tumor cell proliferation (p<0.05) and microvessel density (p<0.05). Furthermore, in relationship to the PDGFRα pathway, OKN-007 was able to significantly decrease SULF2 (p<0.05) and PDGFR-α (platelet-derived growth factor receptor-α) (p<0.05) immunoexpression, and significantly increase decorin expression (p<0.05) in responsive mice. This study indicates that OKN-007 may be an effective anti-cancer agent for some patients with pGBMs by inhibiting cell proliferation and angiogenesis, possibly via the PDGFRα pathway, and could be considered as an additional therapy for pediatric brain tumor patients.
Collapse
Affiliation(s)
- Patricia Coutinho de Souza
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States of America
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, United States of America
| | - Samantha Mallory
- University of Oklahoma Children's Hospital, Oklahoma City, OK, United States of America
| | - Nataliya Smith
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States of America
| | - Debra Saunders
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States of America
| | - Xiao-Nan Li
- Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX, United States of America
| | - Rene Y. McNall-Knapp
- University of Oklahoma Children's Hospital, Oklahoma City, OK, United States of America
| | - Kar-Ming Fung
- Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK, United States of America
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Pathology, Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, United States of America
| | - Rheal A. Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States of America
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, United States of America
- Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK, United States of America
- * E-mail:
| |
Collapse
|
28
|
Warren KE. Measuring the pons: a non-invasive biomarker for pediatric diffuse intrinsic pontine glioma. CNS Oncol 2015; 3:181-3. [PMID: 25055124 DOI: 10.2217/cns.14.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
29
|
Birch R, Peet AC, Arvanitis TN, Wilson M. Sensitivity encoding for fast (1) H MR spectroscopic imaging water reference acquisition. Magn Reson Med 2014; 73:2081-6. [PMID: 25046769 DOI: 10.1002/mrm.25355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 06/10/2014] [Accepted: 06/14/2014] [Indexed: 12/25/2022]
Abstract
PURPOSE Accurate and fast (1) H MR spectroscopic imaging (MRSI) water reference scans are important for absolute quantification of metabolites. However, the additional acquisition time required often precludes the water reference quantitation method for MRSI studies. Sensitivity encoding (SENSE) is a successful MR technique developed to reduce scan time. This study quantitatively assesses the accuracy of SENSE for water reference MRSI data acquisition, compared with the more commonly used reduced resolution technique. METHODS 2D MRSI water reference data were collected from a phantom and three volunteers at 3 Tesla for full acquisition (306 s); 2× reduced resolution (64 s) and SENSE R = 3 (56 s) scans. Water amplitudes were extracted using MRS quantitation software (TARQUIN). Intensity maps and Bland-Altman statistics were generated to assess the accuracy of the fast-MRSI techniques. RESULTS The average mean and standard deviation of differences from the full acquisition were 2.1 ± 3.2% for SENSE and 10.3 ± 10.7% for the reduced resolution technique, demonstrating that SENSE acquisition is approximately three times more accurate than the reduced resolution technique. CONCLUSION SENSE was shown to accurately reconstruct water reference data for the purposes of in vivo absolute metabolite quantification, offering significant improvement over the more commonly used reduced resolution technique.
Collapse
Affiliation(s)
- Rebecca Birch
- PSIBS Doctoral Training Centre, University of Birmingham, United Kingdom.,Department of Oncology, Birmingham Children's Hospital NHS Foundation Trust, Birmingham, United Kingdom
| | - Andrew C Peet
- Department of Oncology, Birmingham Children's Hospital NHS Foundation Trust, Birmingham, United Kingdom.,School of Cancer Sciences, University of Birmingham, United Kingdom
| | - Theodoros N Arvanitis
- Department of Oncology, Birmingham Children's Hospital NHS Foundation Trust, Birmingham, United Kingdom.,Institute of Digital Healthcare, WMG, University of Warwick, Coventry, United Kingdom
| | - Martin Wilson
- Department of Oncology, Birmingham Children's Hospital NHS Foundation Trust, Birmingham, United Kingdom.,School of Cancer Sciences, University of Birmingham, United Kingdom
| |
Collapse
|
30
|
Öz G, Alger JR, Barker PB, Bartha R, Bizzi A, Boesch C, Bolan PJ, Brindle KM, Cudalbu C, Dinçer A, Dydak U, Emir UE, Frahm J, González RG, Gruber S, Gruetter R, Gupta RK, Heerschap A, Henning A, Hetherington HP, Howe FA, Hüppi PS, Hurd RE, Kantarci K, Klomp DWJ, Kreis R, Kruiskamp MJ, Leach MO, Lin AP, Luijten PR, Marjańska M, Maudsley AA, Meyerhoff DJ, Mountford CE, Nelson SJ, Pamir MN, Pan JW, Peet AC, Poptani H, Posse S, Pouwels PJW, Ratai EM, Ross BD, Scheenen TWJ, Schuster C, Smith ICP, Soher BJ, Tkáč I, Vigneron DB, Kauppinen RA. Clinical proton MR spectroscopy in central nervous system disorders. Radiology 2014; 270:658-79. [PMID: 24568703 PMCID: PMC4263653 DOI: 10.1148/radiol.13130531] [Citation(s) in RCA: 459] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A large body of published work shows that proton (hydrogen 1 [(1)H]) magnetic resonance (MR) spectroscopy has evolved from a research tool into a clinical neuroimaging modality. Herein, the authors present a summary of brain disorders in which MR spectroscopy has an impact on patient management, together with a critical consideration of common data acquisition and processing procedures. The article documents the impact of (1)H MR spectroscopy in the clinical evaluation of disorders of the central nervous system. The clinical usefulness of (1)H MR spectroscopy has been established for brain neoplasms, neonatal and pediatric disorders (hypoxia-ischemia, inherited metabolic diseases, and traumatic brain injury), demyelinating disorders, and infectious brain lesions. The growing list of disorders for which (1)H MR spectroscopy may contribute to patient management extends to neurodegenerative diseases, epilepsy, and stroke. To facilitate expanded clinical acceptance and standardization of MR spectroscopy methodology, guidelines are provided for data acquisition and analysis, quality assessment, and interpretation. Finally, the authors offer recommendations to expedite the use of robust MR spectroscopy methodology in the clinical setting, including incorporation of technical advances on clinical units.
Collapse
Affiliation(s)
- Gülin Öz
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Jeffry R. Alger
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Peter B. Barker
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Robert Bartha
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Alberto Bizzi
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Chris Boesch
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Patrick J. Bolan
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Kevin M. Brindle
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Cristina Cudalbu
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Alp Dinçer
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Ulrike Dydak
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Uzay E. Emir
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Jens Frahm
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Ramón Gilberto González
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Stephan Gruber
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Rolf Gruetter
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Rakesh K. Gupta
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Arend Heerschap
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Anke Henning
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Hoby P. Hetherington
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Franklyn A. Howe
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Petra S. Hüppi
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Ralph E. Hurd
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Kejal Kantarci
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Dennis W. J. Klomp
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Roland Kreis
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Marijn J. Kruiskamp
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Martin O. Leach
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Alexander P. Lin
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Peter R. Luijten
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Małgorzata Marjańska
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Andrew A. Maudsley
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Dieter J. Meyerhoff
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Carolyn E. Mountford
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Sarah J. Nelson
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - M. Necmettin Pamir
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Jullie W. Pan
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Andrew C. Peet
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Harish Poptani
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Stefan Posse
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Petra J. W. Pouwels
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Eva-Maria Ratai
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Brian D. Ross
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Tom W. J. Scheenen
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Christian Schuster
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Ian C. P. Smith
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Brian J. Soher
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Ivan Tkáč
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | - Daniel B. Vigneron
- From the Center for Magnetic Resonance Research, University of Minnesota,
2021 6th St SE, Minneapolis, MN 55455 (G.O.)
| | | |
Collapse
|
31
|
Diffusion-weighted MRI derived apparent diffusion coefficient identifies prognostically distinct subgroups of pediatric diffuse intrinsic pontine glioma. J Neurooncol 2014; 117:175-82. [DOI: 10.1007/s11060-014-1375-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 01/19/2014] [Indexed: 02/06/2023]
|
32
|
Measurements of the pons as a biomarker of progression for pediatric DIPG. J Neurooncol 2013; 116:127-33. [PMID: 24113877 DOI: 10.1007/s11060-013-1266-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 09/22/2013] [Indexed: 10/26/2022]
Abstract
Treatment of pediatric diffuse intrinsic pontine glioma (DIPG) remains challenging, and reliable biomarkers of response are lacking. Radiographic response is a primary endpoint in many investigational studies of brain tumors, but there is no standard method of tumor measurement for DIPG, significant inter-observer variability exists given the invasive nature of these tumors, and tumor measurements are not predictive of outcome. Because DIPGs involve a significant portion of the pons, we evaluated the reliability and prognostic value of one-dimensional (1D) and two-dimensional (2D) pons measurements using anatomical landmarks rather than tumor boundaries. Patients with DIPG (n = 75) were evaluated longitudinally at our institution using MRI. Four readers independently performed 1D and 2D measurements of the pons using FLAIR images. Agreement and inter-reader variability were evaluated using differences among the six reader pairs and the coefficient of variation (CV). Prognostic value of pons measurements was calculated using Cox proportional hazards models, where relative hazard (RH) represents risk of death. Readers evaluated 384 exams. Agreement of readers' 1D and 2D measurements was strong (median difference between reader pairs 3.1 and 5.4%, respectively), with low inter-reader variability (median CV = 3.1% and median CV = 4.8%, respectively). Increases in 1D and 2D pons measurements over time indicated poorer prognosis (RH = 2.29, p = 0.0025 and RH = 1.13, p = 0.0016, respectively), with shorter overall survival. Pons measurements had low inter-reader variability compared to previously reported tumor measurement techniques and correlated with outcome in children with DIPG. Measurements of the pons (as opposed to direct measurements of tumor) are a viable in vivo biomarker for DIPG.
Collapse
|
33
|
Warren KE. Diffuse intrinsic pontine glioma: poised for progress. Front Oncol 2012; 2:205. [PMID: 23293772 PMCID: PMC3531714 DOI: 10.3389/fonc.2012.00205] [Citation(s) in RCA: 207] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 12/11/2012] [Indexed: 12/21/2022] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are amongst the most challenging tumors to treat. Surgery is not an option, the effects of radiation therapy are temporary, and no chemotherapeutic agent has demonstrated significant efficacy. Numerous clinical trials of new agents and novel therapeutic approaches have been performed over the course of several decades in efforts to improve the outcome of children with DIPG, yet without success. The diagnosis of DIPG is based on radiographic findings in the setting of a typical clinical presentation, and tissue is not routinely obtained as the standard of care. The paradigm for treating children with these tumors has been based on that for supratentorial high-grade gliomas in adults as the biology of these lesions were presumed to be similar. However, recent pivotal studies demonstrate that DIPGs appear to be their own entity. Simply identifying this fact releases a number of constraints and opens opportunities for biologic investigation of these lesions, setting the stage to move forward in identifying DIPG-specific treatments. This review will summarize the current state of knowledge of DIPG, discuss obstacles to therapy, and summarize results of recent biologic studies.
Collapse
Affiliation(s)
- Katherine E Warren
- Pediatric Neuro-Oncology Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health Bethesda, MD, USA
| |
Collapse
|
34
|
Steffen-Smith EA, Venzon DJ, Bent RS, Hipp SJ, Warren KE. Single- and multivoxel proton spectroscopy in pediatric patients with diffuse intrinsic pontine glioma. Int J Radiat Oncol Biol Phys 2012; 84:774-9. [PMID: 22445531 PMCID: PMC3386374 DOI: 10.1016/j.ijrobp.2012.01.032] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Revised: 01/03/2012] [Accepted: 01/08/2012] [Indexed: 11/15/2022]
Abstract
PURPOSE To determine the feasibility of two magnetic resonance spectroscopy (MRS) techniques for treating pediatric patients with diffuse intrinsic pontine gliomas (DIPGs) and to evaluate the relationship of metabolic profiles determined by each technique. Utility of each technique for improving patient management is also discussed. METHODS AND MATERIALS Children with DIPG (n = 36) were evaluated using single-voxel spectroscopy (SVS) and magnetic resonance spectroscopic imaging (MRSI) during the same imaging session. Patients were followed longitudinally (n = 150 total studies). Technical feasibility was defined by sufficient water and lipid suppression for detection of metabolites. Correlation of metabolic data obtained by SVS and MRSI was determined using the Spearman rank method. Metabolite ratios, including choline:N-acetyl-aspartate (Cho:NAA) and Cho:creatine (Cho:Cr), were obtained from SVS and MRSI. RESULTS SVS and MRSI acquisitions were feasible in >90% of studies. Maximum Cho:NAA and Cho:Cr from MRSI analysis were strongly associated with Cho:NAA and Cho:Cr obtained by SVS (r = 0.67 and 0.76, respectively). MRSI Cho:NAA values were more heterogeneous than Cho:Cr values within the same lesion, and a strong linear relationship between the range and maximum Cho:NAA values was observed. CONCLUSIONS SVS and MRSI acquisitions were feasible, with a strong correlation in metabolic data. Both techniques may improve diagnostic evaluation and management of DIPG. SVS is recommended for global assessment of tumor metabolism before and after therapy. MRSI showed heterogeneous patterns of metabolic activity within these tumors and is recommended for planning and monitoring targeted therapies and evaluating nearby tissue for tumor invasion.
Collapse
Affiliation(s)
- Emilie A. Steffen-Smith
- Pediatric Oncology Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD
| | - David J. Venzon
- Biostatistics and Data Management Section, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Robyn S. Bent
- Pediatric Oncology Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD
| | - Sean J. Hipp
- Pediatric Oncology Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD
- Walter Reed National Military Medical Center, Department of Pediatrics, Bethesda, MD
- Uniformed Services University of the Health Sciences, Department of Pediatrics, Bethesda, MD
| | - Katherine E. Warren
- Pediatric Oncology Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD
| |
Collapse
|
35
|
Hipp SJ, Steffen-Smith EA, Patronas N, Herscovitch P, Solomon JM, Bent RS, Steinberg SM, Warren KE. Molecular imaging of pediatric brain tumors: comparison of tumor metabolism using ¹⁸F-FDG-PET and MRSI. J Neurooncol 2012; 109:521-7. [PMID: 22760419 DOI: 10.1007/s11060-012-0918-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 06/19/2012] [Indexed: 01/21/2023]
Abstract
Magnetic resonance spectroscopic imaging (MRSI) and (18)F-fluorodeoxyglucose positron emission tomography (FDG-PET) are non-invasive imaging techniques routinely used to evaluate tumor malignancy in adults with brain tumors. We compared the metabolic activity of pediatric brain tumors using FDG-PET and MRSI. Children (n = 37) diagnosed with a primary brain tumor underwent FDG-PET and MRSI within two weeks of each other. Tumor metabolism was classified as inactive, active or highly active using the maximum choline:N-acetyl-asparate (Cho:NAA) on MRSI and the highest tumor uptake on FDG-PET. A voxel-wise comparison was used to evaluate the area with the greatest abnormal metabolism. Agreement between methods was assessed using the percent agreement and the kappa statistic (κ). Pediatric brain tumors were metabolically heterogeneous on FDG-PET and MRSI studies. Active tumor metabolism was observed more frequently using MRSI compared to FDG-PET, and agreement in tumor classification was weak (κ = 0.16, p = 0.12), with 42 % agreement (95 % CI = 25-61 %). Voxel-wise comparison for identifying the area of greatest metabolic activity showed overlap in the majority (62 %) of studies, though exact agreement between techniques was low (29.4 %, 95 % CI = 15.1-47.5 %). These results indicate that FDG-PET and MRSI detect similar but not always identical regions of tumor activity, and there is little agreement in the degree of tumor metabolic activity between the two techniques.
Collapse
Affiliation(s)
- Sean J Hipp
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
High-grade gliomas (HGGs) are malignant tumors and typically include glioblastoma multiforme and anaplastic astrocytoma subtypes. Brainstem gliomas and ependymomas are separate entities with respect to clinical presentation, treatment, prognosis, and outcome in comparison with supratentorial HGGs. In children, these tumors account for 3% to 7% of newly diagnosed brain tumors and 20% of all diagnoses of pediatric supratentorial brain tumors. These neoplasms are highly proliferative and mitotically active and of glial origin. This article reviews clinical, diagnostic, and pathologic features of HGG and current treatments and potential future therapies specific to pediatric patients with HGGs.
Collapse
Affiliation(s)
- Tene A Cage
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143-0112, USA.
| | | | | | | |
Collapse
|