Abstract
Over the past 20 years, very few agents have been approved for the treatment of brain tumors. Recent studies have highlighted some of the challenges in assessing activity in novel agents for the treatment of brain tumors. This paper reviews some of the key challenges related to assessment of tumor response to therapy in adult high-grade gliomas and discusses the strengths and limitations of imaging-based endpoints. Although overall survival is considered the “gold standard” endpoint in the field of oncology, progression-free survival and response rate are endpoints that hold great value in neuro-oncology. Particular focus is given to advancements made since the January 2006 Brain Tumor Endpoints Workshop, including the development of Response Assessment in Neuro-Oncology criteria, the value of T2/fluid-attenuated inversion recovery, use of objective response rates and progression-free survival in clinical trials, and the evaluation of pseudoprogression, pseudoresponse, and inflammatory response in radiographic images.
Keywords: clinical trials, endpoints, MRI, RANO, response assessment
Need for Radiographic Measures of Tumor Response to Therapy
Approximately 327 000 new primary brain or CNS tumors are diagnosed each year in the United States, which constitutes ∼21 people per 100 000.1 Of these newly diagnosed tumors, ∼28% are gliomas, which constitute 80% of all malignant tumors.1 Glioblastoma multiforme (GBM), the most common and aggressive type of glioma, is the focal point in this document for 2 reasons. First, it is the most common and aggressive form of malignant glioma, accounting for 54% of all gliomas and 45% of all malignant primary brain and CNS tumors,1 thus it is a high priority area for therapeutic development. Second, GBM is one of the most complex, adaptive, and drug-resistant brain tumors. Therefore, improvements to drug development and measurement of tumor response to therapy in GBM may provide added benefits to other types of brain tumors.
GBM carries a dismal prognosis, with a median survival of around 14 months,2 and fewer than 10% of patients survive beyond 5 years after diagnosis.3 Despite the modest increase in survival observed with the addition of temozolomide to radiotherapy,2 this dismal prognosis has not changed substantially in the past 30 years. Currently, the standard of care for newly diagnosed GBM patients consists of maximum safe surgical resection, followed by radiotherapy plus concomitant and adjuvant temozolomide. At recurrence, however, very few therapeutic options exist. A careful review of the clinical trials from 2006–2012 involving recurrent GBM has shown that only a minority of patients are eligible for a second surgery or reirradiation. At relapse, temozolomide-pretreated patients show progression-free survival (PFS) rates at 6 months of 20%–30% with nitrosoureas, temozolomide rechallenge, or bevacizumab.4 Thus, there is an urgent need for drug development in the setting of recurrent GBM.
Although overall survival (OS) is considered the gold standard for determining whether a cancer treatment is effective, in certain situations OS may not directly reflect the impact of a specific regimen because of potential confounding effects of prognostic factors, additional therapies, and other factors. Therefore, PFS and in particular response rate (RR) are considered valuable endpoints for isolating the relative value of a given treatment.5 Determining response and progression using surrogate measures of tumor burden, however, suffers from issues associated with imaging characteristics (enhancement), measurement variability, false positives, and discordance in radiographic interpretation between observers.6 Therefore, there is a need for refinement of response assessment in neuro-oncology with the objective of minimizing intrinsic errors and enhancing the accuracy of predicting true response to a particular therapy.
Brief History of Glioma Response Assessment
MRI used with the addition of contrast agents that shorten T1 relaxation time constants is the standard for detection, delineation, and response assessment of brain tumors. Using this approach, the tumor region becomes bright on T1-weighted images due to the passing of contrast agent out of abnormal vasculature, through the blood–brain barrier, and into the extracellular space.7 Thus, regions of hyperintensity (brightness) on postcontrast T1-weighted images are thought to reflect the most aggressive portion of the tumor, which has subsequently been confirmed with biopsy observations.8,9
The response criteria proposed by Macdonald and colleagues10 in 1990 attempted to improve upon earlier response assessments, including the Levin criteria11 and the World Health Organization systemic oncology response criteria, which used bidirectional measurements,12 by accounting for corticosteroid use and changes in neurologic status. Similar to its predecessors and the Response Evaluation Criteria in Solid Tumors (RECIST),13 the “Macdonald criteria” used measurements of contrast-enhancing tumor burden to determine tumor response and progression, categorizing response into 4 categories: complete response, partial response, stable disease, and progressive disease. For nearly 20 years the Macdonald criteria, based on contrast enhancement as a surrogate of tumor burden, were used for the evaluation of new therapies. The Macdonald criteria were effectively used for the approval of temozolomide in recurrent anaplastic astrocytoma, where there was a response rate of 35% (8% complete response, 27% partial response) in a single-arm trial. The Macdonald criteria also allowed for the identification of the chemotherapy responsiveness of anaplastic oligodendroglioma, which was subsequently confirmed in OS benefit in phase III trials.14,15
In an effort to acknowledge the need for new agents to treat brain tumors and to address methodological challenges associated with brain tumor clinical trial design, the FDA cosponsored a public workshop on brain tumor clinical trial endpoints with the American Association for Cancer Research and the American Society of Clinical Oncology in January 2006. This workshop focused on clinical trial endpoints intended to support the approval of new drugs for brain cancer and sought debate on the analytic validity of the instrument (eg, imaging or patient-reported outcomes) and on how well individual endpoints reflect clinical benefit. Several issues were discussed and conclusions made:
Objective response: Objective response rates can be reliably assessed in single-arm studies, but the magnitude of response is important given interreviewer variability.
Time to event endpoints (PFS): Need to be evaluated within randomized studies. Future consideration would be given to a landmark-based PFS (ie, 6-mo PFS) if it could be established as a reliable surrogate endpoint, or one that is reasonably likely to predict clinical benefit.
What Is New Since 2006 and What Is the Status of Brain Tumor Imaging Today?
Over the last 10 years, the routine implementation of newer imaging techniques, including T2-weighted fluid-attenuated inversion recovery (FLAIR) MRI,16,17 which allows for better visualization of vasogenic edema, surgical and radiation-induced gliosis and infiltrating tumor, and new therapies that drastically change vascular permeability (eg, anti-angiogenic agents and immunotherapies) resulted in the need to evolve the Macdonald response criteria.18,19 The largest differences between these new, evolving response criteria and the standard Macdonald criteria revolve mainly around the identification of nonenhancing, infiltrative tumor.20 Although contrast-enhancing tumor is thought to represent the most aggressive portion of the tumor,8,9 and a large percentage of high-grade gliomas have a significant enhancing component,21 these tumors are known to contain proportions of both neovascularized and infiltrative tumor,22,23 and the relative proportions are thought to reflect different biological phenotypes.21,24,25 In addition to containing nonenhancing tumor at presentation, a substantial proportion of treated tumors can have nonenhancing tumor progression,22,26 and progression of nonenhancing tumor can lead to neurologic decline. These issues, combined with the high incidence of “pseudoresponse” during anti-angiogenic therapy, drove the need for reevaluation of the response criteria in neuro-oncology. The inclusion of evaluating nonenhancing parts of the tumor would be particularly valuable if overall tumor burden (and ultimately patient benefit) would improve prediction of overall outcome better than changes in contrast enhancement. In 2010, the Response Assessment in Neuro-Oncology (RANO) criteria were developed27 to comprehensively reform the Macdonald criteria using the evolving principles and conditions outlined in previous work.18–20 Although one of the main changes was the inclusion of the evaluation of nonenhancing tumor progression, RANO also attempted to correct a number of deficiencies in the Macdonald criteria. These included definitions of measurable and nonmeasurable disease, definitions of progression for patients being considered for enrollment into clinical trials, recommendations to address pseudoprogression (PsP) and pseudoresponse, the requirement of confirmatory scans for response, and recommendations for dealing with patients with equivocal imaging changes. The details of the current RANO criteria and other modifications to RANO are now well documented in various review articles.28–34 Importantly, inherent within RANO is the ability to be fluid and adjust the criteria to new and evolving data.
Anti-angiogenic Therapy, Pseudoresponse, and Infiltrative Tumor Recurrence
Clinical studies examining the efficacy of anti-angiogenic agents in patients with GBM note a dramatic reduction in the amount of contrast enhancement,35–44 translating into very high response rates with bevacizumab (28%–38%,45 63%,46 and 57%47) and cediranib44 (∼50%) compared with response rates of 10% or less using irinotecan at recurrence.48–51 These response rates tend to translate into prolonged PFS but into only a modest change in OS compared with historical series.44,45 It was hypothesized that this was due to the use of the Macdonald response criteria, which are based primarily on change in contrast enhancement and not on change in actual tumor size assessment. Therefore, the phenomenon of pseudoresponse,52 in which contrast agent uptake is reduced due to changes in vascular permeability independent of antitumor effect, potentially represents a significant limitation for early response assessment of anti-angiogenic treatments in recurrent GBM. The awareness of this phenomenon is also the reason for the need to control steroid dose when calling response or progression.
In addition to increased response rates, studies examining tumor relapse/progression while on anti-angiogenic agents note a tendency for growth of nonenhancing, infiltrative tumor prior to emergence or increase of contrast enhancement.38 Approximately 30%–40% of patients are estimated to experience nonenhancing tumor progression prior to changes in contrast enhancement.26 In one study 40% of patients treated with bevacizumab experienced nonenhancing tumor progression, including a subgroup of 21% of patients with circumscribed T2 progression who had an especially poor median survival of 5 months, comparable to patients who never responded to bevacizumab.53
This observation is further supported by preclinical54 and clinical55–57 evidence showing that escape from anti-angiogenic therapy results in tumor invasion into normal brain and upregulation of genes associated with invasion, including those associated with metalloproteinases and insulin-like growth factor binding protein–2. Utilizing a modified Macdonald criteria that integrated qualitative changes in nonenhancing tumor into the response criteria, the BRAIN study (AVF3708g) provided response rate data,45,58 confirmed in NCI06-C-0064E,45 which led to the accelerated approval of bevacizumab for recurrent/progressive glioblastoma. Additionally in the BRAIN study, independent radiology review of objective response utilizing modified Macdonald criteria incorporating change in nonenhancing tumor was similar to the determination by an independent radiologic evaluation by the FDA: 28.2% versus 25.9% (Table 2). These observations helped consolidate incorporation of a definition for nonenhancing tumor progression into the current RANO criteria. This was, however, inserted as a qualitative change, without a quantitative threshold for progressive disease or partial response because of the often complex irregular shape of T2/FLAIR abnormalities making routine measurement an issue.
Table 2.
Summary of recent recurrent GBM studies
| Study Name | Year | No. of Patients | No. Recurr | Multicenter? | Central Review? | Time Post XRT | Meas (M) vs Eval (E) | ORR | Durability | PFS | PFS-6 | OS | Criteria for Response | |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| VEGF Drug Studies | ||||||||||||||
| Friedman et al, J Clin Oncol, 2009 | BRAIN Bev | 2006–07 | 85 | 1–2 | Yes | Yes | 8 wk | M | 28.2 | 5.6 | 4.2 | 42.6 | 9.2 | Modified Macdonald |
| BRAIN Bev Only (FDA) | 2006–07 | 1–2 | Yes | 25.9 | 4.2 | 36 | 9.3 | WHO | ||||||
| Friedman et al, J Clin Oncol, 2009 | BRAIN Bev/CPT-11 | 2006–07 | 82 | 1–2 | Yes | Yes | 8 wk | M | 37.8 | 4.3 | 5.6 | 50.3 | 8.7 | Modified Macdonald |
| Gilbert et al, J Clin Oncol, 2009 | RTOG-0625 Bev/TMZ | 2007 | 60 | Yes | 6 wk | 21 | 39.3 | Macdonald | ||||||
| Gilbert et al, J Clin Oncol, 2009 | RTOG-0625 Bev/CPT11 | 2007 | 57 | Yes | 6 wk | 28 | 38.6 | Macdonald | ||||||
| Boxerman et al, Neuro Oncol, 2013 | RTOG-0625 | 2007 | 107 | 9 | ||||||||||
| Kreisl et al, J Clin Oncol, 2009 | NIH | 2006–07 | 48 | 1+ | No | No | 35 | 3.7 | 24% | 7.2 | Macdonald | |||
| NIH (FDA) | 2006–07 | 56 | 1+ | No | Yes | 19.6 | 3.9 | WHO | ||||||
| Wen et al, J Clin Oncol, 2010 | XL-184 | 151 | 1–2 | Yes | Yes | 15.2 | 3.7–4.7 | 22.3–41.7 | 7.7 to 10.2 | RANO | ||||
| Batchelor et al, J Clin Oncol, 2013 | Cediranib | 2008–09 | 131 | 1 | Yes | Yes | 12 wk | E | 15.3 | 3.1 | 16.2 | 8 | Macdonald | |
| de Groot et al, J Clin Oncol, 2011 | VEGF Trap | 2007–08 | 42 | 1 | Yes | Yes | M | 18 | 2.8 | 7.7 | 9.1 | Modified Macdonald | ||
| Iwamoto et al, Neuro Oncol, 2010 | Pazopanib | 2007–08 | 35 | 1–3 | Yes | M&E | 5.9 | 2.8 | 3 | 8.2 | Macdonald | |||
| Non-VEGF Drug Studies | ||||||||||||||
| Batchelor et al, J Clin Oncol, 2013 | CCNU | 2008–09 | 65 | 1 | Yes | Yes | 12 wk | E | 8.9 | 2.7 | 25 | 9.8 | Macdonald | |
| Wick et al, J Clin Oncol, 2010 | Enzastaurin | 2006–07 | 174 | 1–2 | Yes | Yes | 12 wk | M | 2.9 | 1.5 | 11 | 6.6 | Levin w/confirm. | |
| Wick et al, J Clin Oncol, 2010 | CCNU | 2006–07 | 92 | 1–2 | Yes | Yes | 12 wk | 4.3 | 1.6 | 19 | 7.1 | Levin w/confirm. | ||
| Yung et al, Br J Cancer, 2000 | TMZ | 112 | 1 | Yes | 5.4 | 21 | 7.5 | Macdonald | ||||||
| Yung et al, Br J Cancer, 2000 | PCB | 113 | 1 | Yes | 5.3 | 8 | 6 | Macdonald | ||||||
| Ballman et al, Neuro Oncol, 2007 | NCCTG | 345 | Yes | NA | 1.8 | 9 | 5 | |||||||
| Lamborn et al, Neuro Oncol, 2008 | NABTC | 437 | 1–3 | Yes | No | 4 wk | M | 7 | 1.9 | 16 | 7 | Macdonald | ||
| Lamborn et al, Neuro Oncol, 2008 | NABTC TMZ | 146 | 1–3 | Yes | 4 wk | M | NA | 3.5 | 28 | 9.3 | Macdonald | |||
| Lamborn, et al, Neuro Oncol, 2008 | NABTC No TMZ | 291 | 1–3 | Yes | 4 wk | M | NA | 1.6 | 9 | 6.1 | Macdonald | |||
| Yung et al, J Clin Oncol, 1999 | TMZ (AG) | 162 | 1 | Yes | Yes | M | 35 | 5.4 | 46 | 13.6 | Macdonald | |||
Abbreviations: XRT, external beam radiotherapy; PFS-6, 6-mo PFS; ORR, overall response rate; Bev, bevacizumab; WHO, World Health Organization; CCNU, lomustine; TMZ, temozolomide; PCB, procarbazine; NCCTG; North Center Cancer Treatment Group; NABTC, North American Brain Tumor Consortium; AG, anaplastic glioma; VEGF, vascular endothelial growth factor.
This summary indicates that RR is never >10% regardless of the drug (non-VEGF agents only) in multicenter, centrally read, first or second recurrence with the inclusion of a 4–12 weeks postradiotherapy window for study entry.
Clinical and Biological Evidence for Incorporating FLAIR Into RANO
Currently, accurate assessment of nonenhancing tumor burden and tumor progression is the most difficult, time-consuming, and expensive portion of RANO evaluation, even though this is to be rated on a subjective scale. While evaluation of nonenhancing tumor burden has been incorporated into a number of registration trials, there remains debate as to whether it provides added value. A bevacizumab-irinotecan study reported by Gallego Perez-Larraya et al26 indicated that RECIST, Macdonald, and RANO had similar estimates of response rate. Also, despite 1/3 of patients experiencing nonenhancing tumor progression during stable or improved contrast enhancement,26 there was no significant difference in PFS (RANO median PFS = 11.7 wk compared with Macdonald PFS = 12.7 wk). Regardless, incorporation of nonenhancing tumor progression did not translate into a significant difference in PFS, and all measures of PFS (including those based on T1 with contrast) were shown to correlate with OS.26 Similar findings were recently reported by Schaub et al59 in a small study of 26 patients showing that recurrence of nonenhancing tumor did not necessarily predict shorter survival. Additionally, a recent study by Radbruch et al60 showed that ∼24% of their 144 patients had T2 recurrence that preceded recurrence via contrast enhancement when T2 progression was defined as >15% increase in bidirectional measurements on T2-weighted images. Of these patients, 62% had contrast enhancement on the subsequent follow-up.60
Consistent with the hypotheses that T2/FLAIR progression is of limited value to predict patient benefit and that early measures of tumor progression via contrast enhancement are still a surrogate of survival even in the presence of anti-angiogenic therapy, recent results from ACRIN-6677/RTOG-0625, a prospective, randomized, phase II multicenter trial by Boxerman et al61 compared bevacizumab with either irinotecan or temozolomide treatment in recurrent GBM. This study demonstrated that response rate at 8 and 16 weeks measured using the Macdonald criteria or 3D enhancing volumes using a central reader paradigm predicted OS in recurrent GBM treated with bevacizumab, whereas T2/FLAIR progression rates alone did not predict OS.62
Preliminary results from Huang et al62 using the BRAIN trial dataset with 160 evaluable patients showed that the RANO criteria reduced median PFS by an average of 1.3 months (5.52 mo with Macdonald; 4.21 mo with RANO; log-rank, P = .0423) but produced no significant difference in overall RR. Tumor progression by Macdonald and RANO at 2, 4, and 6 months predicted OS, but the difference between the 2 criteria was not statistically significant.
It is not clear whether any observed differences in PFS between Macdonald and RANO criteria may account for very different patient reported outcomes and neuropsychological testing data from the 2 phase III trials evaluating the role of bevacizumab in newly diagnosed GBM, AVAglio and RTOG-0825. One potential explanation for the decline in patient reported outcomes and neuropsychological function observed in patients enrolled in RTOG-0825, but not in AVAglio, may be that RTOG-0825 did not include assessment of nonenhancing progression, allowing patients to stay on study longer while they were in fact progressing. Evaluation of the RTOG and other datasets using RANO should help determine the potential utility of nonenhancing tumor progression as a measure of a clinically relevant change in neurologic functioning. If true, this would support the evaluation of T2/FLAIR images as part of outcome assessment.
Biologically, there is some preclinical and clinical evidence to suggest that anti-angiogenic therapy results in a more “infiltrative phenotype”54–57; however, other studies have compelling evidence to suggest that this is not unique to anti-angiogenic therapy63–65 and hypothesize that this is an inherent phenotype to the tumor that exists prior to anti-angiogenic therapy, and/or this phenotype may be relatively common among recurrent GBM regardless of therapy. Additionally, studies examining preclinical response to anti-angiogenic therapy have tangentially noted that prolonged therapy eventually results in reactivation of angiogenesis through upregulation of pro-angiogenic pathways or autocrine vascular endothelial growth factor signaling.54 In summary, the literature suggests that progression may be first via nonenhancing tumor, but tumors will eventually relapse on anti-angiogenic therapy and manifest as an increase in contrast enhancement. Nonenhancing tumor may become apparent prior to changes in contrast enhancement in an individual patient, but this lag is only ∼1 to 2 months on average. There remains significant debate in the field regarding whether this contributes to a substantial difference in PFS, whether it should be evaluated as part of the standard imaging criteria moving forward, and what criteria (improved correlation with OS?) should be used to decide on that issue.
Treatment-Related Changes, the Inflammatory Response, and Pseudoprogression
During cytotoxic or radiation therapy, damage to epithelial cells66,67 and local tissue inflammation are believed to result in edema and abnormal vessel permeability, which in turn can cause an increase in edema on T2-weighted images and/or new or increased contrast enhancement on MRI or CT.68–70 This process of treatment-related early increases in contrast enhancement mimicking tumor progression (ie, PsP) can be defined as subacute radiographic changes mimicking tumor progression, but in retrospect is likely associated with tissue damage, remodeling, and/or inflammatory response. Although the precise mechanisms of radiation-induced CNS changes are not completely understood to date and are quite complex,71 many consider PsP to represent a part of a continuum of treatment-related changes ranging from early subacute inflammation to frank radionecrosis that typically occurs only months after the end of radiotherapy.68 This hypothesis is supported by a study by Chamberlain et al72 in which 7 of 15 patients (51%) who went to surgery after recurring prior to 6 months following radiotherapy were shown to have histopathological signs of radiation necrosis with no evidence of tumor. Described as early as the early 1990s by Fiegler et al,69 Watne et al,70 and Griebel et al,73 differentiation of PsP from true tumor progression continues to be one of the major diagnostic challenges in the response assessment of malignant gliomas. It also has implications for treatment at relapse, in which the resolution of PsP may suggest activity of inactive agents.74
The true incidence of PsP during standard therapy with radiation/temozolomide is unclear. This limitation is primarily due to the small number of studies on PsP, along with the small number of patients evaluated in these studies. Further, the definition of early progression and PsP are highly variable across studies, and there is an increasing tendency to treat patients at the first signs of radiographic progression, leading to increasing difficulty in verifying whether PsP has indeed occurred. Studies have estimated the occurrence of PsP to range anywhere from 3% to 35% in patients treated with radiochemotherapy72,74–85 (Table 1).76 The rate of PsP incidence is believed to increase with increasing radiation dose and timing86 and the addition of concurrent chemotherapy.76,77 In one of the first well-designed systematic studies involving PsP, de Wit and colleagues74 described a 9% incidence of PsP in patients with malignant gliomas treated with radiation only as part of a phase III clinical trial. This is consistent with a large retrospective evaluation by Ruben et al,87 which noted incidences of radiation necrosis of 2.9%, 5.1%, 9.3%, and 13.3% at 6, 12, 24, and 36 months, respectively, after radiotherapy in 352 glioma patients. However, in a study by Taal et al77 examining radiation therapy with the addition of temozolomide, the incidence of PsP was estimated at around 21% of patients. This higher proportion of PsP in patients treated with radiochemotherapy was also verified by a large study by Brandes et al76 involving 103 patients, ∼31% of whom showed characteristics of PsP.
Table 1.
Summary of current studies focusing on incidence of PsP and actual progression in newly diagnosed malignant gliomas treated with radiation or radiochemotherapy
| Publication | Window for Early Progression | Number of Early Progressors | Number of Confirmed PsP | Total Number of Patients | Early Prog. | PsP in Early Prog. | PsP in All Patients | Criteria for Progression | Treatment |
|---|---|---|---|---|---|---|---|---|---|
| Brandes et al, J Clin Oncol, 2008 | 1 mo | 50 | 32 | 103 | 48.54% | 64.00% | 31.07% | Macdonald | Radiochemotherapy |
| Taal et al, Cancer, 2008 | 1 mo | 36 | 15 | 85 | 42.35% | 41.67% | 17.65% | Macdonald | Radiochemotherapy |
| Mangla et al, Radiology, 2010 | 1 mo | 19 | 7 | 36 | 52.78% | 36.84% | 19.44% | Macdonald | Radiochemotherapy |
| Gerstner et al, J Neurooncol, 2009 | 1 mo | 24 | 13 | 45 | 53.33% | 54.17% | 28.89% | Macdonald | Radiochemotherapy |
| de Wit et al, Neurology, 2004 | 1 mo | 9 | 3 | 32 | 28.13% | 33.33% | 9.38% | Macdonald | Radiation therapy |
| Total | 1 mo | 138 | 70 | 301 | 45.85% | 50.72% | 23.26% | ||
| Sanghera et al, Can J Neurol Sci, 2010 | 2 mo | 27 | 7 | 104 | 25.96% | 25.93% | 6.73% | RECIST | Radiochemotherapy |
| Clarke et al, J Clin Oncol, 2008 (Abstr) | 2 mo | 33 | 8 | 80 | 41.25% | 24.24% | 10.00% | Increased T1 + contrast | Radiochemotherapy |
| Young et al, Neurology, 2011 | 2 mo | 63 | 33 | 93 | 67.74% | 52.38% | 35.48% | Increased T1 + contrast | Radiochemotherapy |
| Chu et al, Radiology, 2013 | 2 mo | 30 | 15 | 52 | 57.69% | 50.00% | 28.85% | Increased T1 + contrast | Radiochemotherapy |
| Total | 2 mo | 153 | 63 | 329 | 46.50% | 41.18% | 19.15% | ||
| Jefferies et al, Clin Oncol, 2007 | 6 mo | 9 | 3 | 15 | 60.00% | 33.33% | 20.00% | Not specified | Radiochemotherapy |
| Chaski et al, Surg Neurol, 2009 | 6 mo | 25 | 3 | 54 | 46.30% | 12.00% | 5.56% | Increased T1 + contrast | Radiochemotherapy |
| Kumar et al, Radiology, 2000 | 6 mo | 14 | 3 | 92 | 15.22% | 21.43% | 3.26% | Increased T1 + contrast | Radiochemotherapy |
| Total | 6 mo | 48 | 9 | 161 | 29.81% | 18.75% | 5.59% |
Advanced imaging techniques have shown some promise in differentiating PsP from true tumor recurrence in recurrent malignant gliomas. For example, studies have shown that relative cerebral blood volume estimated using dynamic susceptibility contrast perfusion MRI is elevated in tumor progression compared with PsP.88–91 However, the particular threshold recommended for best stratification varies widely (from 0.71 to 2.6) and appears highly dependent on acquisition parameters, postprocessing, and how the measurements are performed (eg, spherical regions of interest vs contoured enhancing tumor). Additionally, PET imaging of neutral amino acids including [11C-methyl]-methionine (11C-MET), L-1-[11C]-tyrosine (11C-TYR), O-(2-[18F]-fluoroethyl)-l-tyrosine (18F-FET), and 3,4-dihydroxy-6-[18F]-fluoro-l-phenylalanine (18F-FDOPA) has shown the ability to identify treatment-related changes from tumor growth.92 Although this is promising, large trials with standardized image acquisition are necessary to properly verify the added value of advanced imaging in terms of differentiating PsP from recurrent disease.
Conclusions
In general, changes in contrast enhancement follow change in tumor burden in recurrent glioblastoma, with a few exceptions. First, contrast enhancement is altered by changes in corticosteroid dose. This is mitigated by the RANO requirements for stable steroid dosage at baseline and limiting objective response determination if steroid dose is increased. Second, increased contrast uptake on scans obtained during the first 12 weeks postradiotherapy may reflect only treatment-related changes, reducing the ability for contrast enhancement to serve as an accurate surrogate for tumor burden in this situation. This is mitigated by the RANO requirement that limits enrollment of patients into recurrent studies who have progressive lesions 12 weeks or later following external beam radiotherapy. (Important to note, however, is that several studies have suggested that PsP can occur several months after the end of radiotherapy. Hence, this criteria does not fully mitigate PsP as a potential confounding factor.) Third, the RANO criteria require that durable response be demonstrated on subsequent MRI scans in order to identify true response from a transient permeability effect. Lastly, the use of agents that directly impact vascular permeability (eg, anti–vascular endothelial growth factor therapies) may also reduce the accuracy of contrast enhancement as a surrogate for tumor burden in recurrent glioblastoma. This is currently mitigated by the RANO requirement of nonenhancing tumor evaluation; however, it is conceivable that the overall RR threshold for determining success may need to be adjusted for these agents.
Additionally, there is significant uncertainty regarding the value of nonenhancing tumor assessment via T2/FLAIR images in recurrent glioblastoma. While 30%–40% of tumors initially develop nonenhancing tumor after anti–vascular endothelial growth factor therapies, most tumors subsequently develop enhancing disease that may be more easily measured. Currently, quantitative evaluation of nonenhancing tumor is not performed, leading to concerns regarding the reproducibility of determining nonenhancing tumor progression with no specific guidelines. Evaluation of nonenhancing tumor remains intuitively meaningful; however, there is little evidence in the current literature to support the added value of nonenhancing tumor assessment.
Funding
This work was supported by research grants from Genentech/Roche and Siemens to B.M.E.; research grants from Roche and Abbvie to M.J.v.d.B.; and grants to T.F.C. from the Henry Singleton Brain Cancer Research Program, Art of the Brain, Ziering Family Foundation, Ben and Catherine Ivy Foundation, and Defeat GBM (National Brain Tumor Society).
Acknowledgments
Conflict of interest statement. B.M.E. is a consultant for Genentech/Roche and Siemens Healthcare. P.Y.W. is a consultant and on the advisory boards of Genentech/Roche, Sigma Tau, Celldex, Momenta, Vascular Biogenics, and Meditech, is a speaker for Merck, editor-in-chief of Neuro-Oncology, and the editor of the Neuro-Oncology section of Up-to-Date. M.J.v.d.B is a consultant and a research supporter at Roche and Abbvie, a consultant at Celldex, Amgen, and Actelion, a consultant and on the speakers bureau of Merck Ag, and has received honoraria from Up-to-Date. T.F.C. is a consultant and on the advisory board of Genentech/Roche, Tocagen, Lpath, Newgen, Celegene, and Amgen.
References
- 1.Ostrom QT, Gittleman H, Farah P, et al. CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2006–2010. Neuro Oncol. 2013;15(Suppl 2):ii1–i56. doi: 10.1093/neuonc/not151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–996. doi: 10.1056/NEJMoa043330. [DOI] [PubMed] [Google Scholar]
- 3.Stupp R, Hegi ME, Mason WP, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009;10(5):459–466. doi: 10.1016/S1470-2045(09)70025-7. [DOI] [PubMed] [Google Scholar]
- 4.Weller M, Cloughesy T, Perry JR, et al. Standards of care for treatment of recurrent glioblastoma—are we there yet? Neuro Oncol. 2013;15(1):4–27. doi: 10.1093/neuonc/nos273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Lamborn KR, Yung WK, Chang SM, et al. Progression-free survival: an important end point in evaluating therapy for recurrent high-grade gliomas. Neuro Oncol. 2008;10(2):162–170. doi: 10.1215/15228517-2007-062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Provenzale JM, Ison C, Delong D. Bidimensional measurements in brain tumors: assessment of interobserver variability. Am J Roentgenol. 2009;193(6):W515–W522. doi: 10.2214/AJR.09.2615. [DOI] [PubMed] [Google Scholar]
- 7.Niendorf HP, Laniado M, Semmler W, et al. Dose administration of gadolinium-DTPA in MR imaging of intracranial tumors. Am J Neuroradiol. 1987;8(5):803–815. [PMC free article] [PubMed] [Google Scholar]
- 8.Kelly PJ, Daumas-Duport C, Scheithauer BW, et al. Stereotactic histologic correlations of computed tomography- and magnetic resonance imaging-defined abnormalities in patients with glial neoplasms. Mayo Clin Proc. 1987;62(6):450–459. doi: 10.1016/s0025-6196(12)65470-6. [DOI] [PubMed] [Google Scholar]
- 9.Kelly PJ, Daumas-Duport C, Kispert DB, et al. Imaging-based stereotaxic serial biopsies in untreated intracranial glial neoplasms. J Neurosurg. 1987;66(6):865–874. doi: 10.3171/jns.1987.66.6.0865. [DOI] [PubMed] [Google Scholar]
- 10.Macdonald DR, Cascino TL, Schold SC, Jr, et al. Response criteria for phase II studies of supratentorial malignant glioma. J Clin Oncol. 1990;8(7):1277–1280. doi: 10.1200/JCO.1990.8.7.1277. [DOI] [PubMed] [Google Scholar]
- 11.Levin VA, Crafts DC, Norman DM, et al. Criteria for evaluating patients undergoing chemotherapy for malignant brain tumors. J Neurosurg. 1977;47(3):329–335. doi: 10.3171/jns.1977.47.3.0329. [DOI] [PubMed] [Google Scholar]
- 12.Miller AB, Hoogstraten B, Staquet M, et al. Reporting results of cancer treatment. Cancer. 1981;47(1):207–214. doi: 10.1002/1097-0142(19810101)47:1<207::aid-cncr2820470134>3.0.co;2-6. [DOI] [PubMed] [Google Scholar]
- 13.Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst. 2000;92(3):205–216. doi: 10.1093/jnci/92.3.205. [DOI] [PubMed] [Google Scholar]
- 14.van den Bent MJ, Brandes AA, Taphoorn MJ, et al. Adjuvant procarbazine, lomustine, and vincristine chemotherapy in newly diagnosed anaplastic oligodendroglioma: long-term follow-up of EORTC brain tumor group study 26951. J Clin Oncol. 2013;31(3):344–350. doi: 10.1200/JCO.2012.43.2229. [DOI] [PubMed] [Google Scholar]
- 15.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(3):337–343. doi: 10.1200/JCO.2012.43.2674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Hajnal JV, De Coene B, Lewis PD, et al. High signal regions in normal white matter shown by heavily T2-weighted CSF nulled IR sequences. J Comput Assist Tomogr. 1992;16(4):506–513. doi: 10.1097/00004728-199207000-00002. [DOI] [PubMed] [Google Scholar]
- 17.De Coene B, Hajnal JV, Gatehouse P, et al. MR of the brain using fluid-attenuated inversion recovery (FLAIR) pulse sequences. Am J Neuroradiol. 1992;13(6):1555–1564. [PMC free article] [PubMed] [Google Scholar]
- 18.Henson JW, Ulmer S, Harris GJ. Brain tumor imaging in clinical trials. Am J Neuroradiol. 2008;29(3):419–424. doi: 10.3174/ajnr.A0963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Sorensen AG, Batchelor TT, Wen PY, et al. Response criteria for glioma. Nat Clin Pract Oncol. 2008;5(11):634–644. doi: 10.1038/ncponc1204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.van den Bent MJ, Vogelbaum MA, Wen PY, et al. End point assessment in gliomas: novel treatments limit usefulness of classical Macdonald's criteria. J Clin Oncol. 2009;27(18):2905–2908. doi: 10.1200/JCO.2009.22.4998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ellingson BM, Lai A, Harris RJ, et al. Probabilistic radiographic atlas of glioblastoma phenotypes. Am J Neuroradiol. 2013;34(3):533–540. doi: 10.3174/ajnr.A3253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chamberlain MC. Radiographic patterns of relapse in glioblastoma. J Neurooncol. 2011;101(2):319–323. doi: 10.1007/s11060-010-0251-4. [DOI] [PubMed] [Google Scholar]
- 23.Jain RK, di Tomaso E, Duda DG, et al. Angiogenesis in brain tumours. Nat Rev Neurosci. 2007;8(8):610–622. doi: 10.1038/nrn2175. [DOI] [PubMed] [Google Scholar]
- 24.Zinn PO, Mahajan B, Sathyan P, et al. Radiogenomic mapping of edema/cellular invasion MRI-phenotypes in glioblastoma multiforme. PLoS One. 2011;6(10):e25451. doi: 10.1371/journal.pone.0025451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Pope WB, Chen JH, Dong J, et al. Relationship between gene expression and enhancement in glioblastoma multiforme: exploratory DNA microarray analysis. Radiology. 2008;249(1):268–277. doi: 10.1148/radiol.2491072000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Gallego Perez-Larraya J, Lahutte M, Petrirena G, et al. Response assessment in recurrent glioblastoma treated with irinotecan-bevacizumab: comparative analysis of the Macdonald, RECIST, RANO, and RECIST + F criteria. Neuro Oncol. 2012;14(5):667–673. doi: 10.1093/neuonc/nos070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.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(11):1963–1972. doi: 10.1200/JCO.2009.26.3541. [DOI] [PubMed] [Google Scholar]
- 28.Mehta AI, Kanaly CW, Friedman AH, et al. Monitoring radiographic brain tumor progression. Toxins (Basel) 2011;3(3):191–200. doi: 10.3390/toxins3030191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Chinot OL, Macdonald DR, Abrey LE, et al. Response assessment criteria for glioblastoma: practical adaptation and implementation in clinical trials of antiangiogenic therapy. Curr Neurol Neurosci Rep. 2013;13(5):347. doi: 10.1007/s11910-013-0347-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Pope WB, Young JR, Ellingson BM. Advances in MRI assessment of gliomas and response to anti-VEGF therapy. Curr Neurol Neurosci Rep. 2011;11(3):336–344. doi: 10.1007/s11910-011-0179-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Quant EC, Wen PY. Response assessment in neuro-oncology. Curr Oncol Rep. 2011;13(1):50–56. doi: 10.1007/s11912-010-0143-y. [DOI] [PubMed] [Google Scholar]
- 32.Pope WB, Hessel C. Response assessment in neuro-oncology criteria: implementation challenges in multicenter neuro-oncology trials. Am J Neuroradiol. 2011;32(5):794–797. doi: 10.3174/ajnr.A2582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Reardon DA, Galanis E, DeGroot JF, et al. Clinical trial end points for high-grade glioma: the evolving landscape. Neuro Oncol. 2011;13(3):353–361. doi: 10.1093/neuonc/noq203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Khan SN, Linetsky M, Ellingson BM, et al. Magnetic resonance imaging of glioma in the era of antiangiogenic therapy. PET Clinics. 2013;8(2):163–182. doi: 10.1016/j.cpet.2012.09.004. [DOI] [PubMed] [Google Scholar]
- 35.Pope WB, Lai A, Nghiemphu P, et al. MRI in patients with high-grade gliomas treated with bevacizumab and chemotherapy. Neurology. 2006;66(8):1258–1260. doi: 10.1212/01.wnl.0000208958.29600.87. [DOI] [PubMed] [Google Scholar]
- 36.Chamberlain MC. MRI in patients with high-grade gliomas treated with bevacizumab and chemotherapy. Neurology. 2006;67(11):2089. doi: 10.1212/01.wnl.0000250628.10420.d8. author reply 2089. [DOI] [PubMed] [Google Scholar]
- 37.Sathornsumetee S, Cao Y, Marcello JE, et al. Tumor angiogenic and hypoxic profiles predict radiographic response and survival in malignant astrocytoma patients treated with bevacizumab and irinotecan. J Clin Oncol. 2008;26(2):271–278. doi: 10.1200/JCO.2007.13.3652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Norden AD, Young GS, Setayesh K, et al. Bevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrence. Neurology. 2008;70(10):779–787. doi: 10.1212/01.wnl.0000304121.57857.38. [DOI] [PubMed] [Google Scholar]
- 39.Bokstein F, Shpigel S, Blumenthal DT. Treatment with bevacizumab and irinotecan for recurrent high-grade glial tumors. Cancer. 2008;112(10):2267–2273. doi: 10.1002/cncr.23401. [DOI] [PubMed] [Google Scholar]
- 40.Ananthnarayan S, Bahng J, Roring J, et al. Time course of imaging changes of GBM during extended bevacizumab treatment. J Neurooncol. 2008;88(3):339–347. doi: 10.1007/s11060-008-9573-x. [DOI] [PubMed] [Google Scholar]
- 41.Kang TY, Jin T, Elinzano H, et al. Irinotecan and bevacizumab in progressive primary brain tumors, an evaluation of efficacy and safety. J Neurooncol. 2008;89(1):113–118. doi: 10.1007/s11060-008-9599-0. [DOI] [PubMed] [Google Scholar]
- 42.de Groot JF, Yung WK. Bevacizumab and irinotecan in the treatment of recurrent malignant gliomas. Cancer J. 2008;14(5):279–285. doi: 10.1097/PPO.0b013e3181867bd6. [DOI] [PubMed] [Google Scholar]
- 43.Zuniga RM, Torcuator R, Jain R, et al. Efficacy, safety and patterns of response and recurrence in patients with recurrent high-grade gliomas treated with bevacizumab plus irinotecan. J Neurooncol. 2009;91(3):329–336. doi: 10.1007/s11060-008-9718-y. [DOI] [PubMed] [Google Scholar]
- 44.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(1):83–95. doi: 10.1016/j.ccr.2006.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Friedman HS, Prados MD, Wen PY, et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol. 2009;27(28):4733–4740. doi: 10.1200/JCO.2008.19.8721. [DOI] [PubMed] [Google Scholar]
- 46.Vredenburgh JJ, Desjardins A, Herndon JE, 2nd, et al. Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin Cancer Res. 2007;13(4):1253–1259. doi: 10.1158/1078-0432.CCR-06-2309. [DOI] [PubMed] [Google Scholar]
- 47.Vredenburgh JJ, Desjardins A, Herndon JE, 2nd, et al. Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol. 2007;25(30):4722–4729. doi: 10.1200/JCO.2007.12.2440. [DOI] [PubMed] [Google Scholar]
- 48.Friedman HS, Petros WP, Friedman AH, et al. Irinotecan therapy in adults with recurrent or progressive malignant glioma. J Clin Oncol. 1999;17(5):1516–1525. doi: 10.1200/JCO.1999.17.5.1516. [DOI] [PubMed] [Google Scholar]
- 49.Cloughesy TF, Filka E, Kuhn J, et al. Two studies evaluating irinotecan treatment for recurrent malignant glioma using an every-3-week regimen. Cancer. 2003;97(9 Suppl):2381–2386. doi: 10.1002/cncr.11306. [DOI] [PubMed] [Google Scholar]
- 50.Raymond E, Fabbro M, Boige V, et al. Multicentre phase II study and pharmacokinetic analysis of irinotecan in chemotherapy-naive patients with glioblastoma. Ann Oncol. 2003;14(4):603–614. doi: 10.1093/annonc/mdg159. [DOI] [PubMed] [Google Scholar]
- 51.Prados MD, Lamborn K, Yung WK, et al. A phase 2 trial of irinotecan (CPT-11) in patients with recurrent malignant glioma: a North American Brain Tumor Consortium study. Neuro Oncol. 2006;8(2):189–193. doi: 10.1215/15228517-2005-010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Hygino da Cruz LC, Jr, Rodriguez I, Domingues RC, et al. Pseudoprogression and pseudoresponse: imaging challenges in the assessment of posttreatment glioma. Am J Neuroradiol. 2011;32(11):1978–1985. doi: 10.3174/ajnr.A2397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Nowosielski M, Wiestler B, Goebel G, et al. Progression types after anti-angiogenic therapy are related to outcome in recurrent glioblastoma. Neurology. 2014;82(19):1684–1692. doi: 10.1212/WNL.0000000000000402. [DOI] [PubMed] [Google Scholar]
- 54.Lucio-Eterovic AK, Piao Y, de Groot JF. Mediators of glioblastoma resistance and invasion during antivascular endothelial growth factor therapy. Clin Cancer Res. 2009;15(14):4589–4599. doi: 10.1158/1078-0432.CCR-09-0575. [DOI] [PubMed] [Google Scholar]
- 55.de Groot JF, Fuller G, Kumar AJ, et al. Tumor invasion after treatment of glioblastoma with bevacizumab: radiographic and pathologic correlation in humans and mice. Neuro Oncol. 2010;12(3):233–242. doi: 10.1093/neuonc/nop027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Narayana A, Kelly P, Golfinos J, et al. Antiangiogenic therapy using bevacizumab in recurrent high-grade glioma: impact on local control and patient survival. J Neurosurg. 2009;110(1):173–180. doi: 10.3171/2008.4.17492. [DOI] [PubMed] [Google Scholar]
- 57.Iwamoto FM, Abrey LE, Beal K, et al. Patterns of relapse and prognosis after bevacizumab failure in recurrent glioblastoma. Neurology. 2009;73(15):1200–1206. doi: 10.1212/WNL.0b013e3181bc0184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Prados M, Cloughesy T, Samant M, et al. Response as a predictor of survival in patients with recurrent glioblastoma treated with bevacizumab. Neuro Oncol. 2011;13(1):143–151. doi: 10.1093/neuonc/noq151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Schaub C, Greschus S, Seifert M, et al. FLAIR-only progression in bevacizumab-treated relapsing glioblastoma does not predict short survival. Oncology. 2013;85(3):191–195. doi: 10.1159/000354692. [DOI] [PubMed] [Google Scholar]
- 60.Radbruch A, Lutz K, Wiestler B, et al. Relevance of T2 signal changes in the assessment of progression of glioblastoma according to the Response Assessment in Neurooncology criteria. Neuro Oncol. 2012;14(2):222–229. doi: 10.1093/neuonc/nor200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Boxerman JL, Zhang Z, Safriel Y, et al. Early post-bevacizumab progression on contrast-enhanced MRI as a prognostic marker for overall survival in recurrent glioblastoma: results from the ACRIN 6677/RTOG 0625 Central Reader Study. Neuro Oncol. 2013;15(7):945–954. doi: 10.1093/neuonc/not049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Huang RY, Rahman R, Pope WB, et al. Validation of RANO criteria: contribution of T2/FLAIR assessment in patients with recurrent glioblastoma treated with bevacizumab. Paper presented at: ASCO Annual Meeting; Chicago, IL. 2014. [Google Scholar]
- 63.Wick A, Dorner N, Schafer N, et al. Bevacizumab does not increase the risk of remote relapse in malignant glioma. Ann Neurol. 2011;69(3):586–592. doi: 10.1002/ana.22336. [DOI] [PubMed] [Google Scholar]
- 64.Pope WB, Xia Q, Paton VE, et al. Patterns of progression in patients with recurrent glioblastoma treated with bevacizumab. Neurology. 2011;76(5):432–437. doi: 10.1212/WNL.0b013e31820a0a8a. [DOI] [PubMed] [Google Scholar]
- 65.Wick W, Wick A, Weiler M, et al. Patterns of progression in malignant glioma following anti-VEGF therapy: perceptions and evidence. Curr Neurol Neurosci Rep. 2011;11(3):305–312. doi: 10.1007/s11910-011-0184-0. [DOI] [PubMed] [Google Scholar]
- 66.Li YQ, Chen P, Haimovitz-Friedman A, et al. Endothelial apoptosis initiates acute blood-brain barrier disruption after ionizing radiation. Cancer Res. 2003;63(18):5950–5956. [PubMed] [Google Scholar]
- 67.Fajardo LF, Bathrong M, Anderson RE. Radiation Pathology. New York: Oxford University Press; 2001. [Google Scholar]
- 68.Brandsma D, Stalpers L, Taal W, et al. Clinical features, mechanisms, and management of pseudoprogression in malignant gliomas. Lancet Oncol. 2008;9(5):453–461. doi: 10.1016/S1470-2045(08)70125-6. [DOI] [PubMed] [Google Scholar]
- 69.Fiegler W, Langer M, Scheer M, et al. [Reversible computed tomographic changes following brain tumor irradiation induced by the ‘early-delayed reaction’ after radiation] Radiologe. 1986;26(4):206–209. [PubMed] [Google Scholar]
- 70.Watne K, Hager B, Heier M, et al. Reversible oedema and necrosis after irradiation of the brain. Diagnostic procedures and clinical manifestations. Acta Oncol. 1990;29(7):891–895. doi: 10.3109/02841869009096385. [DOI] [PubMed] [Google Scholar]
- 71.Soussain C, Ricard D, Fike JR, et al. CNS complications of radiotherapy and chemotherapy. Lancet. 2009;374(9701):1639–1651. doi: 10.1016/S0140-6736(09)61299-X. [DOI] [PubMed] [Google Scholar]
- 72.Chamberlain MC, Glantz MJ, Chalmers L, et al. Early necrosis following concurrent Temodar and radiotherapy in patients with glioblastoma. J Neurooncol. 2007;82(1):81–83. doi: 10.1007/s11060-006-9241-y. [DOI] [PubMed] [Google Scholar]
- 73.Griebel M, Friedman HS, Halperin EC, et al. Reversible neurotoxicity following hyperfractionated radiation therapy of brain stem glioma. Med Pediatr Oncol. 1991;19(3):182–186. doi: 10.1002/mpo.2950190307. [DOI] [PubMed] [Google Scholar]
- 74.de Wit MC, de Bruin HG, Eijkenboom W, et al. Immediate post-radiotherapy changes in malignant glioma can mimic tumor progression. Neurology. 2004;63(3):535–537. doi: 10.1212/01.wnl.0000133398.11870.9a. [DOI] [PubMed] [Google Scholar]
- 75.Young RJ, Gupta A, Shah AD, et al. Potential utility of conventional MRI signs in diagnosing pseudoprogression in glioblastoma. Neurology. 2011;76(22):1918–1924. doi: 10.1212/WNL.0b013e31821d74e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.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(13):2192–2197. doi: 10.1200/JCO.2007.14.8163. [DOI] [PubMed] [Google Scholar]
- 77.Taal W, Brandsma D, de Bruin HG, et al. Incidence of early pseudo-progression in a cohort of malignant glioma patients treated with chemoirradiation with temozolomide. Cancer. 2008;113(2):405–410. doi: 10.1002/cncr.23562. [DOI] [PubMed] [Google Scholar]
- 78.Mangla R, Singh G, Ziegelitz D, et al. Changes in relative cerebral blood volume 1 month after radiation-temozolomide therapy can help predict overall survival in patients with glioblastoma. Radiology. 2010;256(2):575–584. doi: 10.1148/radiol.10091440. [DOI] [PubMed] [Google Scholar]
- 79.Gerstner ER, McNamara MB, Norden AD, et al. Effect of adding temozolomide to radiation therapy on the incidence of pseudo-progression. J Neurooncol. 2009;94(1):97–101. doi: 10.1007/s11060-009-9809-4. [DOI] [PubMed] [Google Scholar]
- 80.Sanghera P, Perry J, Sahgal A, et al. Pseudoprogression following chemoradiotherapy for glioblastoma multiforme. Can J Neurol Sci. 2010;37(1):36–42. doi: 10.1017/s0317167100009628. [DOI] [PubMed] [Google Scholar]
- 81.Clarke JL, Abrey LE, Karimi S, et al. Pseudoprogression (PsPr) after concurrent radiotherapy (RT) and temozolomide (TMZ) for newly diagnosed glioblastoma multiforme (GBM) [abstract] J Clin Oncol. 2008;26(15S May 20 Suppl):2025. [Google Scholar]
- 82.Chu HH, Choi SH, Ryoo I, et al. Differentiation of true progression from pseudoprogression in glioblastoma treated with radiation therapy and concomitant temozolomide: comparison study of standard and high-b-value diffusion-weighted imaging. Radiology. 2013;269(3):831–840. doi: 10.1148/radiol.13122024. [DOI] [PubMed] [Google Scholar]
- 83.Jefferies S, Burton K, Jones P, Burnet N, et al. Interpretation of early imaging after concurrent radiotherapy and temozolomide in glioblastoma. Clin Oncol (R Coll Radiol) 2007;19(3 Suppl):S33. [Google Scholar]
- 84.Chaskis C, Neyns B, Michotte A, et al. Pseudoprogression after radiotherapy with concurrent temozolomide for high-grade glioma: clinical observations and working recommendations. Surg Neurol. 2009;72(4):423–428. doi: 10.1016/j.surneu.2008.09.023. [DOI] [PubMed] [Google Scholar]
- 85.Kumar AJ, Leeds NE, Fuller GN, et al. Malignant gliomas: MR imaging spectrum of radiation therapy- and chemotherapy-induced necrosis of the brain after treatment. Radiology. 2000;217(2):377–384. doi: 10.1148/radiology.217.2.r00nv36377. [DOI] [PubMed] [Google Scholar]
- 86.Yoneoka Y, Satoh M, Akiyama K, et al. An experimental study of radiation-induced cognitive dysfunction in an adult rat model. Br J Radiol. 1999;72(864):1196–1201. doi: 10.1259/bjr.72.864.10703477. [DOI] [PubMed] [Google Scholar]
- 87.Ruben JD, Dally M, Bailey M, et al. Cerebral radiation necrosis: incidence, outcomes, and risk factors with emphasis on radiation parameters and chemotherapy. Int J Radiat Oncol Biol Phys. 2006;65(2):499–508. doi: 10.1016/j.ijrobp.2005.12.002. [DOI] [PubMed] [Google Scholar]
- 88.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(2):486–496. doi: 10.1148/radiol.2532090007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Hu LS, Baxter LC, Smith KA, et al. Relative cerebral blood volume values to differentiate high-grade glioma recurrence from posttreatment radiation effect: direct correlation between image-guided tissue histopathology and localized dynamic susceptibility-weighted contrast-enhanced perfusion MR imaging measurements. Am J Neuroradiol. 2009;30(3):552–558. doi: 10.3174/ajnr.A1377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Sugahara T, Korogi Y, Tomiguchi S, et al. Posttherapeutic intraaxial brain tumor: the value of perfusion-sensitive contrast-enhanced MR imaging for differentiating tumor recurrence from nonneoplastic contrast-enhancing tissue. Am J Neuroradiol. 2000;21(5):901–909. [PMC free article] [PubMed] [Google Scholar]
- 91.Baek HJ, Kim HS, Kim N, et al. Percent change of perfusion skewness and kurtosis: a potential imaging biomarker for early treatment response in patients with newly diagnosed glioblastomas. Radiology. 2012;264(3):834–843. doi: 10.1148/radiol.12112120. [DOI] [PubMed] [Google Scholar]
- 92.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. doi: 10.1093/brain/awh399. [DOI] [PubMed] [Google Scholar]
