Brain Tumor Res Treat.  2015 Apr;3(1):8-23. 10.14791/btrt.2015.3.1.8.

Modern Brain Tumor Imaging

Affiliations
  • 1Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA. marc.mabray@ucsf.edu

Abstract

The imaging and clinical management of patients with brain tumor continue to evolve over time and now heavily rely on physiologic imaging in addition to high-resolution structural imaging. Imaging remains a powerful noninvasive tool to positively impact the management of patients with brain tumor. This article provides an overview of the current state-of-the art clinical brain tumor imaging. In this review, we discuss general magnetic resonance (MR) imaging methods and their application to the diagnosis of, treatment planning and navigation, and disease monitoring in patients with brain tumor. We review the strengths, limitations, and pitfalls of structural imaging, diffusion-weighted imaging techniques, MR spectroscopy, perfusion imaging, positron emission tomography/MR, and functional imaging. Overall this review provides a basis for understudying the role of modern imaging in the care of brain tumor patients.

Keyword

Magnetic resonance imaging; Brain neoplasms; Glioma; Glioblastoma; Neuroimaging

MeSH Terms

Brain Neoplasms*
Diagnosis
Electrons
Glioblastoma
Glioma
Humans
Magnetic Resonance Imaging
Magnetic Resonance Spectroscopy
Neuroimaging
Perfusion Imaging

Figure

  • Fig. 1 57-year-old male patient who presented with loss of consciousness and seizure and who initially had a non-contrast CT (A) performed that demonstrated a partially calcified mass in the left frontal lobe. Preoperative MR images include axial T2 FLAIR (B), axial T1 post-contrast (C), axial SWI (D), coronal T2 FLAIR (E), axial DWI (F), axial ADC map (G), and DTI tractography of the left corticospinal tract superimposed on axial T2 (H). This T2/FLAIR hyperintense (B, E, and H), non-enhancing mass (C), with internal calcifications visible on CT (A) and as susceptibility on SWI (D) was a grade II oligoastrocytoma (TP53 mutation negative, IDH1 mutation positive, 1p19q co-deletion positive). A gross total resection was performed and the patient remains alive. ADC, apparent diffusion coefficient; CT, computed tomography; DTI, diffusion-tensor imaging; DWI, diffusion weighted imaging; FLAIR, fluid attenuated inversion recovery; MR, magnetic resonance; SWI, susceptibility weighted imaging.

  • Fig. 2 78-year-old female patient with a glioblastoma who presented with gait instability, dizziness, and dysarthria for 6-8 weeks. Preoperative MR images include axial T1 post-contrast (A), coronal T1 post-contrast (B), axial T2 FLAIR (C), axial DWI (D), axial ADC map (E), axial DSC perfusion (F), and corresponding perfusion curve (G). Post-contrast T1 images (A and B) demonstrate irregular rim enhancement and central necrosis of this tumor centered in the right frontal lobe. Surrounding the enhancing component is T2 FLAIR hyperintensity (C) which is likely infiltrative cellular edema. The lesion demonstrates restricted diffusion with low ADC values (D and E) and increased relative cerebral blood volume compared to the contralateral side (F and G) with return to baseline of the perfusion curve consistent with a primary glial tumor. ADC, apparent diffusion coefficient; DSC, dynamic susceptibility contrast-enhanced; DWI, diffusion weighted imaging; FLAIR, fluid attenuated inversion recovery; MRI, magnetic resonance image.

  • Fig. 3 33-year-old female patient with a left insular glioblastoma (19q deletion, 1p intact, IDH1 mutation, PTEN and EGFR mutation negative) who presented with syncope and seizure. Preoperative MR images include axial T1 post-contrast (A), axial T2 FLAIR (B), axial DWI (C), axial ADC map (D), spectroscopy with choline to NAA ratio overlay on axial T1 post-contrast (E), corresponding spectroscopy and choline to NAA ratios (F), axial DSC perfusion (G), corresponding perfusion curve (H), and DTI tractography of the left corticospinal tract (I) and left arcuate fasciculus (J) superimposed on axial T2. Post-contrast T1 image (A) demonstrate irregular patchy partial enhancement within the tumor. T2 FLAIR (B) shows the extent of non-enhancing T2 FLAIR hyperintense tumor. The lesion demonstrates areas of restricted diffusion with low ADC values (C and D) and increased relative cerebral blood volume compared to the contralateral side (G and H) with return to baseline of the perfusion curve consistent with a primary glial tumor. Spectroscopy (E and F) demonstrates elevated choline relative to NAA, consistent with a glioma. ADC, apparent diffusion coefficient; DSC, dynamic susceptibility contrast-enhanced; DTI, diffusion tensor imaging; DWI, diffusion weighted imaging; EGFR, epidermal growth factor receptor; FLAIR, fluid attenuated inversion recovery; IDH, isocitrate dehydrogenase; MRI, magnetic resonance image; NAA, N-acetylaspartate; PTEN, phosphatase and tensin homolog.

  • Fig. 4 49-year-old female patient with two months of headaches and falls and a remote history of right lung lobectomy for reported benign tumor with MR imaging demonstrating a solitary mass in the right cingulate gyrus which upon resection was metastatic adenocarcinoma (ultimately metastatic non-small cell lung cancer). Preoperative MR images include axial T1 post-contrast (A), axial T2 FLAIR (B), axial DSC perfusion (C), and corresponding perfusion curve (D). Post-contrast T1 image (A) demonstrates the solitary enhancing mass. T2 FLAIR (B) shows the large amount of peri-tumoral vasogenic edema. DSC perfusion (C and D) shows elevated cerebral blood volume relative to the contralateral side without return of the curve to baseline, suggesting a solitary metastasis as opposed to a glioma. DSC, dynamic susceptibility contrast-enhanced; FLAIR, fluid attenuated inversion recovery; MRI, magnetic resonance image.

  • Fig. 5 64-year-old male patient who presented with expressive aphasia and dysarthria and ultimately was diagnosed with glioblastoma (IDH1 non-mutated, PTEN deleted, EGFR amplification negative) and underwent a subtotal resection. Preoperative MR images include axial T1 post-contrast (A) demonstrating a peripherally enhancing mass, and task based BOLD-fMRI functional maps superimposed on anatomic images (B-F) (courtesy of Pratik Mukherjee, MD, PhD). A tongue movement paradigm (B and C) demonstrated bilateral supra-sylvian peri-rolandic activation, which on the left abuts the posterior aspect of the tumor anteriorly as demonstrated on axial (B) and sagittal (C) images. A covert visual verb generation paradigm localized expressive language function to the left (D and E), which was anterior to the tumor but approached the anterior inferior aspect of the tumor (not pictured). A passive listening paradigm localized receptive language function to the left (F), separate from the tumor. EGFR, epidermal growth factor receptor; fMRI, functional magnetic resonance image; IDH, isocitrate dehydrogenase; MRI, magnetic resonance image; PTEN, phosphatase and tensin homolog.

  • Fig. 6 Single modality FMISO PET/MR imaging in a 65-year-old man with recurrent left temporal lobe WHO grade III anaplastic astrocytoma. Simultaneously obtained, axial FMISO PET (A), post contrast T1-weighted (B), and fused T1 post contrast FMISO (C) PET/MR demonstrates recurrence of disease evidenced by contrast enhancing focus bordering the posterior margin of an anterior left temporal lobe resection cavity. This region demonstrates increased FMISO uptake. FMISO, 18F-flouromisoidazole; MR, magnetic resonance; PET, positron emission tomography; WHO, World Health Organization.


Cited by  1 articles

White Matter Change Revealed by Diffusion Tensor Imaging in Gliomas
Young Il Won, Chun Kee Chung, Chi Heon Kim, Chul-Kee Park, Bang-Bon Koo, Jong-Min Lee, Hee-Won Jung
Brain Tumor Res Treat. 2016;4(2):100-106.    doi: 10.14791/btrt.2016.4.2.100.


Reference

1. Cha S. Update on brain tumor imaging: from anatomy to physiology. AJNR Am J Neuroradiol. 2006; 27:475–487.
2. Belden CJ, Valdes PA, Ran C, et al. Genetics of glioblastoma: a window into its imaging and histopathologic variability. Radiographics. 2011; 31:1717–1740.
Article
3. Bangiyev L, Rossi Espagnet MC, Young R, et al. Adult brain tumor imaging: state of the art. Semin Roentgenol. 2014; 49:39–52.
Article
4. Louis DN, Ohgaki H, Wiestler OD, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007; 114:97–109.
Article
5. Zulch KJ. Histological typing of tumours of the central nervous system. Geneva: World Health Organization;1979.
6. Kleihues P, Sobin LH. World Health Organization classification of tumors. Cancer. 2000; 88:2887.
Article
7. Kleihues P, Burger PC, Scheithauer BW. The new WHO classification of brain tumours. Brain Pathol. 1993; 3:255–268.
Article
8. Gupta K, Salunke P. Molecular markers of glioma: an update on recent progress and perspectives. J Cancer Res Clin Oncol. 2012; 138:1971–1981.
Article
9. Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008; 455:1061–1068.
10. Shiraishi S, Tada K, Nakamura H, et al. Influence of p53 mutations on prognosis of patients with glioblastoma. Cancer. 2002; 95:249–257.
Article
11. Furnari FB, Fenton T, Bachoo RM, et al. Malignant astrocytic glioma: genetics, biology, and paths to treatment. Genes Dev. 2007; 21:2683–2710.
Article
12. Yip S, Iafrate AJ, Louis DN. Molecular diagnostic testing in malignant gliomas: a practical update on predictive markers. J Neuropathol Exp Neurol. 2008; 67:1–15.
Article
13. Shinojima N, Tada K, Shiraishi S, et al. Prognostic value of epidermal growth factor receptor in patients with glioblastoma multiforme. Cancer Res. 2003; 63:6962–6970.
14. Huncharek M, Kupelnick B. Epidermal growth factor receptor gene amplification as a prognostic marker in glioblastoma multiforme: results of a meta-analysis. Oncol Res. 2000; 12:107–112.
Article
15. Mellinghoff IK, Wang MY, Vivanco I, et al. Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N Engl J Med. 2005; 353:2012–2024.
Article
16. Ohgaki H, Kleihues P. Genetic alterations and signaling pathways in the evolution of gliomas. Cancer Sci. 2009; 100:2235–2241.
Article
17. Horbinski C, Miller CR, Perry A. Gone FISHing: clinical lessons learned in brain tumor molecular diagnostics over the last decade. Brain Pathol. 2011; 21:57–73.
Article
18. Diehn M, Nardini C, Wang DS, et al. Identification of noninvasive imaging surrogates for brain tumor gene-expression modules. Proc Natl Acad Sci U S A. 2008; 105:5213–5218.
Article
19. Young RJ, Gupta A, Shah AD, et al. Potential role of preoperative conventional MRI including diffusion measurements in assessing epidermal growth factor receptor gene amplification status in patients with glioblastoma. AJNR Am J Neuroradiol. 2013; 34:2271–2277.
Article
20. Aghi M, Gaviani P, Henson JW, Batchelor TT, Louis DN, Barker FG 2nd. Magnetic resonance imaging characteristics predict epidermal growth factor receptor amplification status in glioblastoma. Clin Cancer Res. 2005; 11(24 Pt 1):8600–8605.
Article
21. Ryoo I, Choi SH, Kim JH, et al. Cerebral blood volume calculated by dynamic susceptibility contrast-enhanced perfusion MR imaging: preliminary correlation study with glioblastoma genetic profiles. PLoS One. 2013; 8:e71704.
Article
22. Hartmann C, Meyer J, Balss J, et al. Type and frequency of IDH1 and IDH2 mutations are related to astrocytic and oligodendroglial differentiation and age: a study of 1,010 diffuse gliomas. Acta Neuropathol. 2009; 118:469–474.
Article
23. Ichimura K, Pearson DM, Kocialkowski S, et al. IDH1 mutations are present in the majority of common adult gliomas but rare in primary glioblastomas. Neuro Oncol. 2009; 11:341–347.
Article
24. Yan H, Parsons DW, Jin G, et al. IDH1 and IDH2 mutations in gliomas. N Engl J Med. 2009; 360:765–773.
25. van den Bent MJ, Dubbink HJ, Marie Y, et al. IDH1 and IDH2 mutations are prognostic but not predictive for outcome in anaplastic oligodendroglial tumors: a report of the European Organization for Research and Treatment of Cancer Brain Tumor Group. Clin Cancer Res. 2010; 16:1597–1604.
Article
26. Wick W, Hartmann C, Engel C, et al. NOA-04 randomized phase III trial of sequential radiochemotherapy of anaplastic glioma with procarbazine, lomustine, and vincristine or temozolomide. J Clin Oncol. 2009; 27:5874–5880.
27. Carrillo JA, Lai A, Nghiemphu PL, et al. Relationship between tumor enhancement, edema, IDH1 mutational status, MGMT promoter methylation, and survival in glioblastoma. AJNR Am J Neuroradiol. 2012; 33:1349–1355.
Article
28. Seiz M, Tuettenberg J, Meyer J, et al. Detection of IDH1 mutations in gliomatosis cerebri, but only in tumors with additional solid component: evidence for molecular subtypes. Acta Neuropathol. 2010; 120:261–267.
Article
29. Choi C, Ganji SK, DeBerardinis RJ, et al. 2-hydroxyglutarate detection by magnetic resonance spectroscopy in IDH-mutated patients with gliomas. Nat Med. 2012; 18:624–629.
Article
30. Riemenschneider MJ, Jeuken JW, Wesseling P, Reifenberger G. Molecular diagnostics of gliomas: state of the art. Acta Neuropathol. 2010; 120:567–584.
Article
31. Labussière M, Idbaih A, Wang XW, et al. All the 1p19q codeleted gliomas are mutated on IDH1 or IDH2. Neurology. 2010; 74:1886–1890.
Article
32. Cairncross JG, Ueki K, Zlatescu MC, et al. Specific genetic predictors of chemotherapeutic response and survival in patients with anaplastic oligodendrogliomas. J Natl Cancer Inst. 1998; 90:1473–1479.
Article
33. van den Bent MJ, Carpentier AF, Brandes AA, et al. Adjuvant procarbazine, lomustine, and vincristine improves progression-free survival but not overall survival in newly diagnosed anaplastic oligodendrogliomas and oligoastrocytomas: a randomized European Organisation for Research and Treatment of Cancer phase III trial. J Clin Oncol. 2006; 24:2715–2722.
Article
34. Trost D, Ehrler M, Fimmers R, et al. Identification of genomic aberrations associated with shorter overall survival in patients with oligodendroglial tumors. Int J Cancer. 2007; 120:2368–2376.
Article
35. Cairncross G, Wang M, Shaw E, et al. Phase III trial of chemoradiotherapy for anaplastic oligodendroglioma: long-term results of RTOG 9402. J Clin Oncol. 2013; 31:337–343.
Article
36. Weller M, Pfister SM, Wick W, Hegi ME, Reifenberger G, Stupp R. Molecular neuro-oncology in clinical practice: a new horizon. Lancet Oncol. 2013; 14:e370–e379.
Article
37. Idbaih A, Marie Y, Pierron G, et al. Two types of chromosome 1p losses with opposite significance in gliomas. Ann Neurol. 2005; 58:483–487.
Article
38. Jenkinson MD, du Plessis DG, Smith TS, Joyce KA, Warnke PC, Walker C. Histological growth patterns and genotype in oligodendroglial tumours: correlation with MRI features. Brain. 2006; 129(Pt 7):1884–1891.
Article
39. Whitmore RG, Krejza J, Kapoor GS, et al. Prediction of oligodendroglial tumor subtype and grade using perfusion weighted magnetic resonance imaging. J Neurosurg. 2007; 107:600–609.
Article
40. Kapoor GS, Gocke TA, Chawla S, et al. Magnetic resonance perfusion-weighted imaging defines angiogenic subtypes of oligodendroglioma according to 1p19q and EGFR status. J Neurooncol. 2009; 92:373–386.
Article
41. Nakagawachi T, Soejima H, Urano T, et al. Silencing effect of CpG island hypermethylation and histone modifications on O6-methylguanine-DNA methyltransferase (MGMT) gene expression in human cancer. Oncogene. 2003; 22:8835–8844.
Article
42. Esteller M, Garcia-Foncillas J, Andion E, et al. Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl J Med. 2000; 343:1350–1354.
Article
43. Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 2005; 352:997–1003.
Article
44. Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005; 352:987–996.
Article
45. Gorlia T, van den Bent MJ, Hegi ME, et al. Nomograms for predicting survival of patients with newly diagnosed glioblastoma: prognostic factor analysis of EORTC and NCIC trial 26981-22981/CE.3. Lancet Oncol. 2008; 9:29–38.
Article
46. Brandes AA, Franceschi E, Tosoni A, et al. MGMT promoter methylation status can predict the incidence and outcome of pseudoprogression after concomitant radiochemotherapy in newly diagnosed glioblastoma patients. J Clin Oncol. 2008; 26:2192–2197.
Article
47. Kong DS, Kim ST, Kim EH, et al. Diagnostic dilemma of pseudoprogression in the treatment of newly diagnosed glioblastomas: the role of assessing relative cerebral blood flow volume and oxygen-6-methylguanine-DNA methyltransferase promoter methylation status. AJNR Am J Neuroradiol. 2011; 32:382–387.
Article
48. von Deimling A, Korshunov A, Hartmann C. The next generation of glioma biomarkers: MGMT methylation, BRAF fusions and IDH1 mutations. Brain Pathol. 2011; 21:74–87.
Article
49. Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene in human cancer. Nature. 2002; 417:949–954.
Article
50. Jones DT, Kocialkowski S, Liu L, Pearson DM, Ichimura K, Collins VP. Oncogenic RAF1 rearrangement and a novel BRAF mutation as alternatives to KIAA1549:BRAF fusion in activating the MAPK pathway in pilocytic astrocytoma. Oncogene. 2009; 28:2119–2123.
Article
51. Jones DT, Kocialkowski S, Liu L, et al. Tandem duplication producing a novel oncogenic BRAF fusion gene defines the majority of pilocytic astrocytomas. Cancer Res. 2008; 68:8673–8677.
Article
52. Horbinski C, Nikiforova MN, Hagenkord JM, Hamilton RL, Pollack IF. Interplay among BRAF, p16, p53, and MIB1 in pediatric low-grade gliomas. Neuro Oncol. 2012; 14:777–789.
Article
53. Heaphy CM, de Wilde RF, Jiao Y, et al. Altered telomeres in tumors with ATRX and DAXX mutations. Science. 2011; 333:425.
Article
54. Lovejoy CA, Li W, Reisenweber S, et al. Loss of ATRX, genome instability, and an altered DNA damage response are hallmarks of the alternative lengthening of telomeres pathway. PLoS Genet. 2012; 8:e1002772.
Article
55. Jiao Y, Killela PJ, Reitman ZJ, et al. Frequent ATRX, CIC, FUBP1 and IDH1 mutations refine the classification of malignant gliomas. Oncotarget. 2012; 3:709–722.
Article
56. Liu XY, Gerges N, Korshunov A, et al. Frequent ATRX mutations and loss of expression in adult diffuse astrocytic tumors carrying IDH1/IDH2 and TP53 mutations. Acta Neuropathol. 2012; 124:615–625.
Article
57. Khuong-Quang DA, Buczkowicz P, Rakopoulos P, et al. K27M mutation in histone H3.3 defines clinically and biologically distinct subgroups of pediatric diffuse intrinsic pontine gliomas. Acta Neuropathol. 2012; 124:439–447.
Article
58. Schwartzentruber J, Korshunov A, Liu XY, et al. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature. 2012; 482:226–231.
59. Wiestler B, Capper D, Holland-Letz T, et al. ATRX loss refines the classification of anaplastic gliomas and identifies a subgroup of IDH mutant astrocytic tumors with better prognosis. Acta Neuropathol. 2013; 126:443–451.
Article
60. Wu G, Broniscer A, McEachron TA, et al. Somatic histone H3 alterations in pediatric diffuse intrinsic pontine gliomas and non-brainstem glioblastomas. Nat Genet. 2012; 44:251–253.
Article
61. Sturm D, Witt H, Hovestadt V, et al. Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of glioblastoma. Cancer Cell. 2012; 22:425–437.
62. Lee YY, Van Tassel P. Intracranial oligodendrogliomas: imaging findings in 35 untreated cases. AJR Am J Roentgenol. 1989; 152:361–369.
Article
63. Eskandary H, Sabba M, Khajehpour F, Eskandari M. Incidental findings in brain computed tomography scans of 3000 head trauma patients. Surg Neurol. 2005; 63:550–553. discussion 553.
Article
64. Gumprecht HK, Widenka DC, Lumenta CB. BrainLab VectorVision Neuronavigation System: technology and clinical experiences in 131 cases. Neurosurgery. 1999; 44:97–104. discussion 104-5.
Article
65. Elhawary H, Liu H, Patel P, et al. Intraoperative real-time querying of white matter tracts during frameless stereotactic neuronavigation. Neurosurgery. 2011; 68:506–516. discussion 516.
Article
66. Zhang B, MacFadden D, Damyanovich AZ, et al. Development of a geometrically accurate imaging protocol at 3 Tesla MRI for stereotactic radiosurgery treatment planning. Phys Med Biol. 2010; 55:6601–6615.
Article
67. Saconn PA, Shaw EG, Chan MD, et al. Use of 3.0-T MRI for stereotactic radiosurgery planning for treatment of brain metastases: a single-institution retrospective review. Int J Radiat Oncol Biol Phys. 2010; 78:1142–1146.
Article
68. Haacke EM. Susceptibility weighted imaging (SWI). Z Med Phys. 2006; 16:237.
Article
69. Haacke EM, Mittal S, Wu Z, Neelavalli J, Cheng YC. Susceptibility-weighted imaging: technical aspects and clinical applications, part 1. AJNR Am J Neuroradiol. 2009; 30:19–30.
Article
70. Bian W, Hess CP, Chang SM, Nelson SJ, Lupo JM. Susceptibility-weighted MR imaging of radiation therapy-induced cerebral microbleeds in patients with glioma: a comparison between 3T and 7T. Neuroradiology. 2014; 56:91–96.
Article
71. Borja MJ, Plaza MJ, Altman N, Saigal G. Conventional and advanced MRI features of pediatric intracranial tumors: supratentorial tumors. AJR Am J Roentgenol. 2013; 200:W483–W503.
Article
72. Sze G, Milano E, Johnson C, Heier L. Detection of brain metastases: comparison of contrast-enhanced MR with unenhanced MR and enhanced CT. AJNR Am J Neuroradiol. 1990; 11:785–791.
73. White ML, Zhang Y, Kirby P, Ryken TC. Can tumor contrast enhancement be used as a criterion for differentiating tumor grades of oligodendrogliomas? AJNR Am J Neuroradiol. 2005; 26:784–790.
74. Ginsberg LE, Fuller GN, Hashmi M, Leeds NE, Schomer DF. The significance of lack of MR contrast enhancement of supratentorial brain tumors in adults: histopathological evaluation of a series. Surg Neurol. 1998; 49:436–440.
Article
75. Garzón B, Emblem KE, Mouridsen K, et al. Multiparametric analysis of magnetic resonance images for glioma grading and patient survival time prediction. Acta Radiol. 2011; 52:1052–1060.
Article
76. Koeller KK, Rushing EJ. From the archives of the AFIP: pilocytic astrocytoma: radiologic-pathologic correlation. Radiographics. 2004; 24:1693–1708.
77. Lee YY, Van Tassel P, Bruner JM, Moser RP, Share JC. Juvenile pilocytic astrocytomas: CT and MR characteristics. AJR Am J Roentgenol. 1989; 152:1263–1270.
Article
78. Klatzo I. Presidental address. Neuropathological aspects of brain edema. J Neuropathol Exp Neurol. 1967; 26:1–14.
79. Lu S, Ahn D, Johnson G, Cha S. Peritumoral diffusion tensor imaging of high-grade gliomas and metastatic brain tumors. AJNR Am J Neuroradiol. 2003; 24:937–941.
80. Law M, Cha S, Knopp EA, Johnson G, Arnett J, Litt AW. High-grade gliomas and solitary metastases: differentiation by using perfusion and proton spectroscopic MR imaging. Radiology. 2002; 222:715–721.
Article
81. Barajas RF Jr, Hess CP, Phillips JJ, et al. Super-resolution track density imaging of glioblastoma: histopathologic correlation. AJNR Am J Neuroradiol. 2013; 34:1319–1325.
Article
82. Cha S, Lupo JM, Chen MH, et al. Differentiation of glioblastoma multiforme and single brain metastasis by peak height and percentage of signal intensity recovery derived from dynamic susceptibility-weighted contrast-enhanced perfusion MR imaging. AJNR Am J Neuroradiol. 2007; 28:1078–1084.
Article
83. Cha S, Pierce S, Knopp EA, et al. Dynamic contrast-enhanced T2*-weighted MR imaging of tumefactive demyelinating lesions. AJNR Am J Neuroradiol. 2001; 22:1109–1116.
84. Stadnik TW, Chaskis C, Michotte A, et al. Diffusion-weighted MR imaging of intracerebral masses: comparison with conventional MR imaging and histologic findings. AJNR Am J Neuroradiol. 2001; 22:969–976.
85. Tung GA, Evangelista P, Rogg JM, Duncan JA 3rd. Diffusion-weighted MR imaging of rim-enhancing brain masses: is markedly decreased water diffusion specific for brain abscess? AJR Am J Roentgenol. 2001; 177:709–712.
86. Ebisu T, Tanaka C, Umeda M, et al. Discrimination of brain abscess from necrotic or cystic tumors by diffusion-weighted echo planar imaging. Magn Reson Imaging. 1996; 14:1113–1116.
Article
87. Desprechins B, Stadnik T, Koerts G, Shabana W, Breucq C, Osteaux M. Use of diffusion-weighted MR imaging in differential diagnosis between intracerebral necrotic tumors and cerebral abscesses. AJNR Am J Neuroradiol. 1999; 20:1252–1257.
88. Raz E, Zagzag D, Saba L, et al. Cyst with a mural nodule tumor of the brain. Cancer Imaging. 2012; 12:237–244.
Article
89. Kondziolka D, Bernstein M, Resch L, et al. Significance of hemorrhage into brain tumors: clinicopathological study. J Neurosurg. 1987; 67:852–857.
Article
90. Rees JH, Smirniotopoulos JG, Jones RV, Wong K. Glioblastoma multiforme: radiologic-pathologic correlation. Radiographics. 1996; 16:1413–1438. quiz 1462-3.
Article
91. Wen PY, Macdonald DR, Reardon DA, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol. 2010; 28:1963–1972.
Article
92. Batchelor TT, Sorensen AG, di Tomaso E, et al. AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell. 2007; 11:83–95.
Article
93. Pope WB, Lai A, Nghiemphu P, Mischel P, Cloughesy TF. MRI in patients with high-grade gliomas treated with bevacizumab and chemotherapy. Neurology. 2006; 66:1258–1260.
Article
94. Thompson EM, Frenkel EP, Neuwelt EA. The paradoxical effect of bevacizumab in the therapy of malignant gliomas. Neurology. 2011; 76:87–93.
Article
95. Taal W, Brandsma D, de Bruin HG, et al. Incidence of early pseudo-pro-gression in a cohort of malignant glioma patients treated with chemoirradiation with temozolomide. Cancer. 2008; 113:405–410.
Article
96. Brandsma D, Stalpers L, Taal W, Sminia P, van den Bent MJ. Clinical features, mechanisms, and management of pseudoprogression in malignant gliomas. Lancet Oncol. 2008; 9:453–461.
Article
97. Gaensler EH, Dillon WP, Edwards MS, Larson DA, Rosenau W, Wilson CB. Radiation-induced telangiectasia in the brain simulates cryptic vascular malformations at MR imaging. Radiology. 1994; 193:629–636.
Article
98. Lupo JM, Chuang CF, Chang SM, et al. 7-Tesla susceptibility-weighted imaging to assess the effects of radiotherapy on normal-appearing brain in patients with glioma. Int J Radiat Oncol Biol Phys. 2012; 82:e493–e500.
Article
99. Barajas RF Jr, Rubenstein JL, Chang JS, Hwang J, Cha S. Diffusion-weighted MR imaging derived apparent diffusion coefficient is predictive of clinical outcome in primary central nervous system lymphoma. AJNR Am J Neuroradiol. 2010; 31:60–66.
Article
100. Guo AC, Cummings TJ, Dash RC, Provenzale JM. Lymphomas and high-grade astrocytomas: comparison of water diffusibility and histologic characteristics. Radiology. 2002; 224:177–183.
Article
101. Herneth AM, Guccione S, Bednarski M. Apparent diffusion coefficient: a quantitative parameter for in vivo tumor characterization. Eur J Radiol. 2003; 45:208–213.
Article
102. Rumboldt Z, Camacho DL, Lake D, Welsh CT, Castillo M. Apparent diffusion coefficients for differentiation of cerebellar tumors in children. AJNR Am J Neuroradiol. 2006; 27:1362–1369.
103. Hilario A, Sepulveda JM, Perez-Nuñez A, et al. A prognostic model based on preoperative MRI predicts overall survival in patients with diffuse gliomas. AJNR Am J Neuroradiol. 2014; 35:1096–1102.
Article
104. Bulakbasi N, Guvenc I, Onguru O, Erdogan E, Tayfun C, Ucoz T. The added value of the apparent diffusion coefficient calculation to magnetic resonance imaging in the differentiation and grading of malignant brain tumors. J Comput Assist Tomogr. 2004; 28:735–746.
Article
105. Kono K, Inoue Y, Nakayama K, et al. The role of diffusion-weighted imaging in patients with brain tumors. AJNR Am J Neuroradiol. 2001; 22:1081–1088.
106. Valles FE, Perez-Valles CL, Regalado S, Barajas RF, Rubenstein JL, Cha S. Combined diffusion and perfusion MR imaging as biomarkers of prognosis in immunocompetent patients with primary central nervous system lymphoma. AJNR Am J Neuroradiol. 2013; 34:35–40.
Article
107. Lee EJ, terBrugge K, Mikulis D, et al. Diagnostic value of peritumoral minimum apparent diffusion coefficient for differentiation of glioblastoma multiforme from solitary metastatic lesions. AJR Am J Roentgenol. 2011; 196:71–76.
108. Oh J, Cha S, Aiken AH, et al. Quantitative apparent diffusion coefficients and T2 relaxation times in characterizing contrast enhancing brain tumors and regions of peritumoral edema. J Magn Reson Imaging. 2005; 21:701–708.
Article
109. Server A, Kulle B, Maehlen J, et al. Quantitative apparent diffusion coefficients in the characterization of brain tumors and associated peritumoral edema. Acta Radiol. 2009; 50:682–689.
Article
110. Smith JS, Cha S, Mayo MC, et al. Serial diffusion-weighted magnetic resonance imaging in cases of glioma: distinguishing tumor recurrence from postresection injury. J Neurosurg. 2005; 103:428–438.
Article
111. Jain R, Scarpace LM, Ellika S, et al. Imaging response criteria for recurrent gliomas treated with bevacizumab: role of diffusion weighted imaging as an imaging biomarker. J Neurooncol. 2010; 96:423–431.
Article
112. Pope WB, Qiao XJ, Kim HJ, et al. Apparent diffusion coefficient histogram analysis stratifies progression-free and overall survival in patients with recurrent GBM treated with bevacizumab: a multi-center study. J Neurooncol. 2012; 108:491–498.
Article
113. Lee WJ, Choi SH, Park CK, et al. Diffusion-weighted MR imaging for the differentiation of true progression from pseudoprogression following concomitant radiotherapy with temozolomide in patients with newly diagnosed high-grade gliomas. Acad Radiol. 2012; 19:1353–1361.
Article
114. Matsusue E, Fink JR, Rockhill JK, Ogawa T, Maravilla KR. Distinction between glioma progression and post-radiation change by combined physiologic MR imaging. Neuroradiology. 2010; 52:297–306.
Article
115. Okada T, Mikuni N, Miki Y, et al. Corticospinal tract localization: integration of diffusion-tensor tractography at 3-T MR imaging with intraoperative white matter stimulation mapping--preliminary results. Radiology. 2006; 240:849–857.
Article
116. Haris M, Kumar S, Raj MK, et al. Serial diffusion tensor imaging to characterize radiation-induced changes in normal-appearing white matter following radiotherapy in patients with adult low-grade gliomas. Radiat Med. 2008; 26:140–150.
Article
117. Kitahara S, Nakasu S, Murata K, Sho K, Ito R. Evaluation of treatment-induced cerebral white matter injury by using diffusion-tensor MR imaging: initial experience. AJNR Am J Neuroradiol. 2005; 26:2200–2206.
118. Aronen HJ, Gazit IE, Louis DN, et al. Cerebral blood volume maps of gliomas: comparison with tumor grade and histologic findings. Radiology. 1994; 191:41–51.
Article
119. Knopp EA, Cha S, Johnson G, et al. Glial neoplasms: dynamic contrast-enhanced T2*-weighted MR imaging. Radiology. 1999; 211:791–798.
Article
120. Lev MH, Ozsunar Y, Henson JW, et al. Glial tumor grading and outcome prediction using dynamic spin-echo MR susceptibility mapping compared with conventional contrast-enhanced MR: confounding effect of elevated rCBV of oligodendrogliomas [corrected]. AJNR Am J Neuroradiol. 2004; 25:214–221.
121. Cha S, Tihan T, Crawford F, et al. Differentiation of low-grade oligodendrogliomas from low-grade astrocytomas by using quantitative blood-volume measurements derived from dynamic susceptibility contrast-enhanced MR imaging. AJNR Am J Neuroradiol. 2005; 26:266–273.
122. Law M, Yang S, Babb JS, et al. Comparison of cerebral blood volume and vascular permeability from dynamic susceptibility contrast-enhanced perfusion MR imaging with glioma grade. AJNR Am J Neuroradiol. 2004; 25:746–755.
123. Cha S, Knopp EA, Johnson G, Wetzel SG, Litt AW, Zagzag D. Intracranial mass lesions: dynamic contrast-enhanced susceptibility-weighted echo-planar perfusion MR imaging. Radiology. 2002; 223:11–29.
Article
124. Barajas RF, Chang JS, Sneed PK, Segal MR, McDermott MW, Cha S. Distinguishing recurrent intra-axial metastatic tumor from radiation necrosis following gamma knife radiosurgery using dynamic susceptibility-weighted contrast-enhanced perfusion MR imaging. AJNR Am J Neuroradiol. 2009; 30:367–372.
Article
125. Barajas RF Jr, Chang JS, Segal MR, et al. Differentiation of recurrent glioblastoma multiforme from radiation necrosis after external beam radiation therapy with dynamic susceptibility-weighted contrast-enhanced perfusion MR imaging. Radiology. 2009; 253:486–496.
Article
126. Roberts HC, Roberts TP, Ley S, Dillon WP, Brasch RC. Quantitative estimation of microvascular permeability in human brain tumors: correlation of dynamic Gd-DTPA-enhanced MR imaging with histopathologic grading. Acad Radiol. 2002; 9:Suppl 1. S151–S155.
127. Roberts HC, Roberts TP, Bollen AW, Ley S, Brasch RC, Dillon WP. Correlation of microvascular permeability derived from dynamic contrast-enhanced MR imaging with histologic grade and tumor labeling index: a study in human brain tumors. Acad Radiol. 2001; 8:384–391.
Article
128. Roberts HC, Roberts TP, Brasch RC, Dillon WP. Quantitative measurement of microvascular permeability in human brain tumors achieved using dynamic contrast-enhanced MR imaging: correlation with histologic grade. AJNR Am J Neuroradiol. 2000; 21:891–899.
129. Patankar TF, Haroon HA, Mills SJ, et al. Is volume transfer coefficient (K(trans)) related to histologic grade in human gliomas? AJNR Am J Neuroradiol. 2005; 26:2455–2465.
130. Narang J, Jain R, Arbab AS, et al. Differentiating treatment-induced necrosis from recurrent/progressive brain tumor using nonmodel-based semiquantitative indices derived from dynamic contrast-enhanced T1-weighted MR perfusion. Neuro Oncol. 2011; 13:1037–1046.
Article
131. Horská A, Barker PB. Imaging of brain tumors: MR spectroscopy and metabolic imaging. Neuroimaging Clin N Am. 2010; 20:293–310.
Article
132. Castillo M, Smith JK, Kwock L. Correlation of myo-inositol levels and grading of cerebral astrocytomas. AJNR Am J Neuroradiol. 2000; 21:1645–1649.
133. Howe FA, Barton SJ, Cudlip SA, et al. Metabolic profiles of human brain tumors using quantitative in vivo 1H magnetic resonance spectroscopy. Magn Reson Med. 2003; 49:223–232.
Article
134. Haris M, Cai K, Singh A, Hariharan H, Reddy R. In vivo mapping of brain myo-inositol. Neuroimage. 2011; 54:2079–2085.
Article
135. Tien RD, Lai PH, Smith JS, Lazeyras F. Single-voxel proton brain spectroscopy exam (PROBE/SV) in patients with primary brain tumors. AJR Am J Roentgenol. 1996; 167:201–209.
Article
136. McKnight TR, von dem Bussche MH, Vigneron DB, et al. Histopathological validation of a three-dimensional magnetic resonance spectroscopy index as a predictor of tumor presence. J Neurosurg. 2002; 97:794–802.
Article
137. Fountas KN, Kapsalaki EZ, Vogel RL, Fezoulidis I, Robinson JS, Gotsis ED. Noninvasive histologic grading of solid astrocytomas using proton magnetic resonance spectroscopy. Stereotact Funct Neurosurg. 2004; 82:90–97.
Article
138. Huang Y, Lisboa PJ, El-Deredy W. Tumour grading from magnetic resonance spectroscopy: a comparison of feature extraction with variable selection. Stat Med. 2003; 22:147–164.
Article
139. Server A, Josefsen R, Kulle B, et al. Proton magnetic resonance spectroscopy in the distinction of high-grade cerebral gliomas from single metastatic brain tumors. Acta Radiol. 2010; 51:316–325.
Article
140. Plotkin M, Eisenacher J, Bruhn H, et al. 123I-IMT SPECT and 1H MR-spectroscopy at 3.0 T in the differential diagnosis of recurrent or residual gliomas: a comparative study. J Neurooncol. 2004; 70:49–58.
Article
141. Träber F, Block W, Flacke S, et al. [1H-MR Spectroscopy of brain tumors in the course of radiation therapy: Use of fast spectroscopic imaging and single-voxel spectroscopy for diagnosing recurrence]. Rofo. 2002; 174:33–42.
142. Ando K, Ishikura R, Nagami Y, et al. [Usefulness of Cho/Cr ratio in proton MR spectroscopy for differentiating residual/recurrent glioma from non-neoplastic lesions]. Nihon Igaku Hoshasen Gakkai Zasshi. 2004; 64:121–126.
143. Hygino da Cruz LC Jr, Rodriguez I, Domingues RC, Gasparetto EL, Sorensen AG. Pseudoprogression and pseudoresponse: imaging challenges in the assessment of posttreatment glioma. AJNR Am J Neuroradiol. 2011; 32:1978–1985.
Article
144. Henry RG, Vigneron DB, Fischbein NJ, et al. Comparison of relative cerebral blood volume and proton spectroscopy in patients with treated gliomas. AJNR Am J Neuroradiol. 2000; 21:357–366.
145. Park I, Chen AP, Zierhut ML, Ozturk-Isik E, Vigneron DB, Nelson SJ. Implementation of 3 T lactate-edited 3D 1H MR spectroscopic imaging with flyback echo-planar readout for gliomas patients. Ann Biomed Eng. 2011; 39:193–204.
Article
146. Li X, Vigneron DB, Cha S, et al. Relationship of MR-derived lactate, mobile lipids, and relative blood volume for gliomas in vivo. AJNR Am J Neuroradiol. 2005; 26:760–769.
147. Crawford FW, Khayal IS, McGue C, et al. Relationship of pre-surgery metabolic and physiological MR imaging parameters to survival for patients with untreated GBM. J Neurooncol. 2009; 91:337–351.
Article
148. Nelson SJ, Vigneron D, Kurhanewicz J, Chen A, Bok R, Hurd R. DNP-Hyperpolarized C Magnetic Resonance Metabolic Imaging for Cancer Applications. Appl Magn Reson. 2008; 34:533–544.
Article
149. Park I, Larson PE, Zierhut ML, et al. Hyperpolarized 13C magnetic resonance metabolic imaging: application to brain tumors. Neuro Oncol. 2010; 12:133–144.
Article
150. Park I, Bok R, Ozawa T, et al. Detection of early response to temozolomide treatment in brain tumors using hyperpolarized 13C MR metabolic imaging. J Magn Reson Imaging. 2011; 33:1284–1290.
Article
151. Chaumeil MM, Larson PE, Yoshihara HA, et al. Non-invasive in vivo assessment of IDH1 mutational status in glioma. Nat Commun. 2013; 4:2429.
Article
152. Lee MH, Smyser CD, Shimony JS. Resting-state fMRI: a review of methods and clinical applications. AJNR Am J Neuroradiol. 2013; 34:1866–1872.
Article
153. Fox MD, Snyder AZ, Vincent JL, Corbetta M, Van Essen DC, Raichle ME. The human brain is intrinsically organized into dynamic, anticorrelated functional networks. Proc Natl Acad Sci U S A. 2005; 102:9673–9678.
Article
154. De Luca M, Beckmann CF, De Stefano N, Matthews PM, Smith SM. fMRI resting state networks define distinct modes of long-distance interactions in the human brain. Neuroimage. 2006; 29:1359–1367.
Article
155. Biswal B, Yetkin FZ, Haughton VM, Hyde JS. Functional connectivity in the motor cortex of resting human brain using echo-planar MRI. Magn Reson Med. 1995; 34:537–541.
Article
156. Binder JR, Swanson SJ, Hammeke TA, et al. Determination of language dominance using functional MRI: a comparison with the Wada test. Neurology. 1996; 46:978–984.
Article
157. Adcock JE, Wise RG, Oxbury JM, Oxbury SM, Matthews PM. Quantitative fMRI assessment of the differences in lateralization of language-related brain activation in patients with temporal lobe epilepsy. Neuroimage. 2003; 18:423–438.
Article
158. Vlieger EJ, Majoie CB, Leenstra S, Den Heeten GJ. Functional magnetic resonance imaging for neurosurgical planning in neurooncology. Eur Radiol. 2004; 14:1143–1153.
Article
159. Håberg A, Kvistad KA, Unsgård G, Haraldseth O. Preoperative blood oxygen level-dependent functional magnetic resonance imaging in patients with primary brain tumors: clinical application and outcome. Neurosurgery. 2004; 54:902–914. discussion 914-5.
Article
160. Shimony JS, Zhang D, Johnston JM, Fox MD, Roy A, Leuthardt EC. Resting-state spontaneous fluctuations in brain activity: a new paradigm for presurgical planning using fMRI. Acad Radiol. 2009; 16:578–583.
161. Zhang D, Johnston JM, Fox MD, et al. Preoperative sensorimotor mapping in brain tumor patients using spontaneous fluctuations in neuronal activity imaged with functional magnetic resonance imaging: initial experience. Neurosurgery. 2009; 65:6 Suppl. 226–236.
Article
162. Kokkonen SM, Nikkinen J, Remes J, et al. Preoperative localization of the sensorimotor area using independent component analysis of resting-state fMRI. Magn Reson Imaging. 2009; 27:733–740.
Article
163. Mickevicius N, Sabsevitz D, Bovi J, LaViolette PS. Effects of whole-brain radiation therapy on resting state connectivity: a case study. Int J Radiat Oncol Biol Phys. 2014; 90:S325.
Article
164. Sours C, Mistry N, Zhang H, et al. Feasibility study testing the incorporation of resting state fMRI data in radiation therapy planning to limit dose to cognitive function networks in patients with primary brain tumors. Int J Radiat Oncol Biol Phys. 2013; 87:S254–S255.
Article
165. Harris RJ, Bookheimer SY, Cloughesy TF, et al. Altered functional connectivity of the default mode network in diffuse gliomas measured with pseudo-resting state fMRI. J Neurooncol. 2014; 116:373–379.
Article
166. Werner P, Barthel H, Drzezga A, Sabri O. Current status and future role of brain PET/MRI in clinical and research settings. Eur J Nucl Med Mol Imaging. 2015; 42:512–526.
Article
167. Puttick S, Bell C, Dowson N, Rose S, Fay M. PET, MRI, and simultaneous PET/MRI in the development of diagnostic and therapeutic strategies for glioma. Drug Discov Today. 2014; 11. 06. [Epub]. http://dx.doi.org/10.1016/j.drudis.2014.10.016.
Article
168. Ricci PE, Karis JP, Heiserman JE, Fram EK, Bice AN, Drayer BP. Differentiating recurrent tumor from radiation necrosis: time for re-evaluation of positron emission tomography? AJNR Am J Neuroradiol. 1998; 19:407–413.
169. Weber W, Bartenstein P, Gross MW, et al. Fluorine-18-FDG PET and iodine-123-IMT SPECT in the evaluation of brain tumors. J Nucl Med. 1997; 38:802–808.
170. Singhal T, Narayanan TK, Jacobs MP, Bal C, Mantil JC. 11C-methionine PET for grading and prognostication in gliomas: a comparison study with 18F-FDG PET and contrast enhancement on MRI. J Nucl Med. 2012; 53:1709–1715.
Article
171. Floeth FW, Pauleit D, Wittsack HJ, et al. Multimodal metabolic imaging of cerebral gliomas: positron emission tomography with [18F]fluoroethyl-L-tyrosine and magnetic resonance spectroscopy. J Neurosurg. 2005; 102:318–327.
Article
172. Pauleit D, Floeth F, Hamacher K, et al. O-(2-[18F]fluoroethyl)-L-tyrosine PET combined with MRI improves the diagnostic assessment of cerebral gliomas. Brain. 2005; 128(Pt 3):678–687.
Article
173. Pöpperl G, Kreth FW, Mehrkens JH, et al. FET PET for the evaluation of untreated gliomas: correlation of FET uptake and uptake kinetics with tumour grading. Eur J Nucl Med Mol Imaging. 2007; 34:1933–1942.
Article
174. Terakawa Y, Tsuyuguchi N, Iwai Y, et al. Diagnostic accuracy of 11C-methionine PET for differentiation of recurrent brain tumors from radiation necrosis after radiotherapy. J Nucl Med. 2008; 49:694–699.
Article
175. Shishido H, Kawai N, Miyake K, Yamamoto Y, Nishiyama Y, Tamiya T. Diagnostic Value of 11C-Methionine (MET) and 18F-Fluorothymidine (FLT) Positron Emission Tomography in Recurrent High-Grade Gliomas; Differentiation from Treatment-Induced Tissue Necrosis. Cancers (Basel). 2012; 4:244–256.
Article
176. Chen W, Silverman DH, Delaloye S, et al. 18F-FDOPA PET imaging of brain tumors: comparison study with 18F-FDG PET and evaluation of diagnostic accuracy. J Nucl Med. 2006; 47:904–911.
177. Ledezma CJ, Chen W, Sai V, et al. 18F-FDOPA PET/MRI fusion in patients with primary/recurrent gliomas: initial experience. Eur J Radiol. 2009; 71:242–248.
Article
178. Lizarraga KJ, Allen-Auerbach M, Czernin J, et al. (18)F-FDOPA PET for differentiating recurrent or progressive brain metastatic tumors from late or delayed radiation injury after radiation treatment. J Nucl Med. 2014; 55:30–36.
Article
179. Bruehlmeier M, Roelcke U, Schubiger PA, Ametamey SM. Assessment of hypoxia and perfusion in human brain tumors using PET with 18F-fluoromisonidazole and 15O-H2O. J Nucl Med. 2004; 45:1851–1859.
180. Cher LM, Murone C, Lawrentschuk N, et al. Correlation of hypoxic cell fraction and angiogenesis with glucose metabolic rate in gliomas using 18F-fluoromisonidazole, 18F-FDG PET, and immunohistochemical studies. J Nucl Med. 2006; 47:410–418.
181. Szeto MD, Chakraborty G, Hadley J, et al. Quantitative metrics of net proliferation and invasion link biological aggressiveness assessed by MRI with hypoxia assessed by FMISO-PET in newly diagnosed glioblastomas. Cancer Res. 2009; 69:4502–4509.
Article
182. Hirata K, Terasaka S, Shiga T, et al. 18F-Fluoromisonidazole positron emission tomography may differentiate glioblastoma multiforme from less malignant gliomas. Eur J Nucl Med Mol Imaging. 2012; 39:760–770.
Article
183. Spence AM, Muzi M, Swanson KR, et al. Regional hypoxia in glioblastoma multiforme quantified with [18F]fluoromisonidazole positron emission tomography before radiotherapy: correlation with time to progression and survival. Clin Cancer Res. 2008; 14:2623–2630.
Article
Full Text Links
  • BTRT
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr