Review Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Oncol. Sep 24, 2021; 12(9): 767-786
Published online Sep 24, 2021. doi: 10.5306/wjco.v12.i9.767
Re-irradiation for high-grade gliomas: Has anything changed?
Sonia García-Cabezas, Amalia Palacios-Eito, Department of Radiation Oncology, Reina Sofia University Hospital, Cordoba 14004, Spain
Eleonor Rivin del Campo, Department of Radiation Oncology, Tenon University Hospital, Paris 75020, France
Juan Solivera-Vela, Department of Neurosurgery, Reina Sofia University Hospital, Cordoba 14004, Spain
ORCID number: Sonia García-Cabezas (0000-0002-9373-9845); Eleonor Rivin del Campo (0000-0003-3319-7287); Juan Solivera-Vela (0000-0003-2860-6656); Amalia Palacios-Eito (0000-0002-7575-227X).
Author contributions: All authors contributed to the writing and revision of this manuscript.
Conflict-of-interest statement: The authors have no conflict of interest related to the manuscript.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Amalia Palacios-Eito, MD, PhD, Associate Professor, Chief Doctor, Department of Radiation Oncology, Reina Sofia University Hospital, Avda. Menéndez Pidal, s/n, Cordoba 14004, Spain. amalia.palacios.sspa@juntadeandalucia.es
Received: June 2, 2021
Peer-review started: June 2, 2021
First decision: July 16, 2021
Revised: July 21, 2021
Accepted: July 30, 2021
Article in press: July 30, 2021
Published online: September 24, 2021

Abstract

Optimal management after recurrence or progression of high-grade gliomas is still undefined and remains a challenge for neuro-oncology multidisciplinary teams. Improved radiation therapy techniques, new imaging methods, published experience, and a better radiobiological knowledge of brain tissue have positioned re-irradiation (re-RT) as an option for many of these patients. Decisions must be individualized, taking into account the pattern of relapse, previous treatment, and functional status, as well as the patient’s preferences and expected quality of life. Many questions remain unanswered with respect to re-RT: Who is the most appropriate candidate, which dose and fractionation are most effective, how to define the target volume, which imaging technique is best for planning, and what is the optimal timing? This review will focus on describing the most relevant studies that include re-RT as salvage therapy, with the aim of simplifying decision-making and designing the best available therapeutic strategy.

Key Words: Re-irradiation, Recurrent glioma, High-grade gliomas, Glioblastoma, Radiosurgery, Stereotactic radiotherapy

Core Tip: The optimal management after recurrence or progression of high-grade gliomas is still undefined. Improved radiation therapy techniques, new imaging methods, published experience, as well as better radiobiological knowledge of the brain tissue have positioned re-irradiation as a valid alternative for many of these patients. Many questions remain unanswered. This review will focus on describing the most relevant studies that include re-irradiation as salvage treatment, with the aim of simplifying decision-making and designing the best available therapeutic strategy.



INTRODUCTION

High-grade gliomas (HGG) are the most common primary malignant brain neoplasm in adults[1]. The most frequent type, glioblastoma multiforme (GBM), has an incidence of 3 cases/100000 inhabitants[2]. Its treatment is a macroscopically complete tumor resection, whenever possible, followed by external beam radiotherapy (60 Gy in 2 Gy/fr) with concurrent temozolomide (TMZ) and adjuvant TMZ until the completion of six cycles[3]. Nevertheless, approximately 40% of World Health Organization (WHO) grade III gliomas (anaplastic astrocytoma) and 90% of grade IV gliomas (GBM) progress within 2 years. The main site of relapse is in or near the tumor bed[3-5].

With standard treatment, median overall survival (mOS) for GBM is approximately 14.6 mo, and median progression-free survival (mPFS) is 6.9 mo[6]. This tumor has a poor prognosis and is very aggressive and fast-growing. The high rate of local failure suggests secondary therapeutic options for local salvage should be considered.

The first issue during the diagnostic-therapeutic approach is to confirm that we are dealing with true tumor progression. The phenomena of “pseudoprogression”, described in 20%-30% of patients who have received radiochemotherapy and possible radionecrosis (RN), associated or not with tumor, may hinder or delay diagnosis[7]. The Response Assessment in Neuro-Oncology working group criteria[8] for HGG categorization has certain limitations.

Optimal management after recurrence or local progression remains to be defined. It has mostly been established by retrospective studies lacking a quality of life (QoL) evaluation. Established salvage treatment options include a second surgery (re-S), re-irradiation (re-RT), systemic treatment, or some combination thereof[9]. The addition of the “tumor treating field therapy” approach (alternating electrical fields that exert biophysical force on charged and polarizable molecules known as dipoles) has been found to extend survival for patients with newly diagnosed and recurrent GBM (rGBM)[10].

These suboptimal results have motivated multiple lines of research investigating new therapeutic approaches such as the addition of molecular targeted agents, immune checkpoint blockade, vaccines, viral therapy, or other irradiation modalities[11-14].

Current therapeutic approaches, including the radiation therapy techniques and parameters, are very diverse. Thus, a survey of expert radiation oncologists showed high variability, reflecting the scarcity of high-quality prospective data for decision-making[15]. Multiple questions remain unanswered with respect to re-RT: Who is the most appropriate candidate, which dose and fractionation are most effective, how to define the target volume, and which imaging technique is best for planning, as well as the optimal timing? This review will focus on describing the most relevant studies that include re-RT as salvage therapy, with the aim of simplifying decision making and designing the best available therapeutic strategy.

RE-RT IN THE THERAPEUTIC STRATEGY

At present, any ablative treatment option offered to a selected patient with local failure is still palliative and has associated side effects that must be considered. The choice is complex, and the criteria are poorly defined. Decisions must be individualized, taking into account the pattern of relapse, previous treatment, and functional status, as well as the patient’s preferences and expected QoL.

For patients with low functional status, unable to walk and totally dependent for daily activities, the best supportive care should be considered.

Historically, the fear of exceeding the dose tolerance of healthy brain tissue, and therefore the risk of severe side effects, kept radiation oncologists from considering re-RT with ablative doses. Thus, the most offered treatment has been systemic [chemotherapy/bevacizumab (BEV)], with a mOS of 6-9 mo, without a clear advantage of any drug or therapeutic scheme among those used[16,17]. Clearly this is the best strategy for patients with widespread or multifocal disease. However, in the case of a focal relapse, if the patient has favorable clinical criteria, the current trend is to consider a second local treatment such as re-S, re-RT, or both with or without systemic treatment.

The level of evidence supporting this approach is low, probably because the high failure rates (recurrence or progression) of these second treatments make it difficult to compare the different strategies.

Objective parameters are needed to simplify therapeutic decision-making. Scoccianti et al[18], based on a review of the literature, recommend the first algorithm to aid decision-making in daily practice between surgical salvage or re-RT. They consider local treatment for focal relapses in patients with life expectancy > 3 mo. The choice of re-S or re-RT depends on prognostic factors and the expected toxicity of each therapeutic option. The results of combined treatment are encouraging, and the tendency is to recommend it. The therapeutic decision should be interdisciplinary and requires expert neurosurgeons and radiation oncologists. Ultimately, the final decision should be agreed upon with the patient after discussion of the risks and benefits of the available therapeutic options.

RESULTS
Re-resection

A minority of patients (20%-30%) are considered eligible for re-S[19], with a higher morbidity-mortality than before initial resection. After re-S, overall survival from re-RT ranges from 4.9[20] to 13.5 mo[21] and PFS from re-RT from 1.9[22] to 8.3 mo[23]. These results are from retrospective, not comparative series. There is no evidence to suggest that these results are better than can be expected with radiation and/or chemotherapy alone[24,25]. The meta-analysis of Lu et al[26] suggests that re-S of rGBM in select patients confers a significant, prognostic OS advantage independent of other prognostic factors, and a cohort from The Director Trial[27] found that surgery at first recurrence of GBM improved outcome if complete resection of contrast-enhancing tumor was achieved. Preoperative and postoperative Karnofsky performance status (KPS), extent of surgery of first re-S, and chemotherapy after first re-S have been identified as the factors that have the greatest impact on survival[25].

Due to the absence of comparative studies, the role of re-S in rGBM is not yet established. The Randomized Controlled Comparative Phase II Trial on Surgery for Glioblastoma Recurrence trial comparing re-S of recurrence plus second-line treatment, vs second-line treatment without re-S, will quantify the contribution of re-S for rGBM.

Re-RT

Based mostly on retrospective series, selected patients with small recurrent tumors and a good performance status may benefit from re-RT using modern high-precision techniques[28-31]. Prospective studies are very scarce, therefore the exact contribution of re-RT is uncertain.

The tumoricidal dose to be administered is limited by the possibility of generating severe side effects, given that most patients have already received doses in the maximum tolerance range at their first irradiation. Re-RT at the therapeutic doses used at diagnosis (60 Gy) is not recommended.

Potential benefits of re-RT include palliation by reducing corticosteroid use, improving neurologic symptoms, and, in selected patients, increasing PFS and possibly overall survival.

There are three most commonly used external radiation therapy techniques that, depending on the fractionation applied, the treatment volume, and the technology used we refer to as: Stereotactic radiosurgery (SRS), hypofractionated stereotactic radiotherapy (HFSRT), and conventionally fractionated external radiotherapy (CFRT). We also have results with intraoperative techniques[32]. The promising results of particle irradiation are described in the section on new irradiation strategies. The choice of technique, in addition to its geographical availability, depends on the size of the recurrence and consequently of the planning target volume (PTV) generated.

Unfortunately, the lack of comparative trials does not allow their results to be compared. However, even in the absence of randomized data, there is a tendency to use hypofractionated or SRS schemes for small volumes, assuming a slightly higher risk of RN.

Kazmi et al[33] published the first meta-analysis with the results of re-RT in rGBM. They included 50 studies with a total of 2095 patients. Overall survival from re-RT and PFS from re-RT at 6 mo were 73% and 43%, respectively, and at 12 mo were 36% and 17%. They found better PFS at 6 mo with SRS and with short fractionation schedules (≤ 5 fractions), probably due to the lower tumor volume.

SRS as salvage treatment

Table 1 describes a selection of series published since 2005. They are characterized by: Including mostly GBM, a single dose of 12-18 Gy, a median volume of around 10 mL, and a time from the first radiation treatment of between 8.8 mo and 13.8 mo. The Kong series[34] is the largest and the only prospective series. The mOS for GBM is between 7.5 mo and 13 mo, while the range for mPFS, in those series that report it, is between 3.6 mo and 7 mo. Severe toxicity is not reported, except for RN, which in a couple of series is 24%-31% by radiological imaging. These data suggest that patients with small volumes can be safely treated with SRS.

Table 1 Summary of selected publications reporting radiosurgery as salvage treatment in recurrent high-grade gliomas.
Ref.Study typeNo. patientsHistologyRe-irradiation
Median intervalMedian tumor volumeMedian PFS2Median OS2Severe toxicityRadionecrosis
Total dose, median
Dose/fr, median
Combs et al[28], 2005R32All GBM15 Gy63.8 Gy10 mo10 mL7 mo10 mo0%0%
Hsieh et al[104], 2005R26All GBM12 Gy42 GyNR21.6 mLNR10 moNR31.3% by image
Kong et al[34], 2008P11465 GBM, 49 G3G16 Gy72 GyNR10.6 mL4.6 mo (GBM), 8.6 mo (G3G)13 mo (GBM), 26 mo (G3G)0%24.4% by image
Patel et al[68], 2009R26All GBM18 Gy90 Gy12.5 mo10.4 mLNR8.4 moLimited toxicityNR
Skeie et al[30], 2012R51All GBM12.2 Gy43.3 Gy11 mo12.4 mL6 mo12 mo0%0%
Martínez-Carrillo et al[31], 2014R8746 GBM, 41 G3G18 Gy90 Gy13.8 mo8.7 mLNR7.5 mo (GBM); 17 mo (G3G)0%0%
Kim et al[105], 2015R29All GBM15 Gy63.8 Gy8.8 mo11 mL3.6 mo9.2 moNRNR
Fractionated stereotactic radiotherapy as salvage treatment

Hypofractionated schemes have been used mainly in larger recurrent HGG (rHGG). A selection of studies published in 2000 or later, including several prospective series, are presented in Table 2. Some contain anaplastic and low-grade gliomas. The median dose and fractionation used are highly variable, between 25 and 35 Gy (3-7 Gy/fr), with an equivalent dose at 2 Gy (EQD2) range of 37.5-78.7 Gy. The largest series is Fogh et al[29] with 147 patients, of which 42 had anaplastic astrocytomas, with an average dose of 35 Gy (3.5 Gy/fr) and a mOS of 11 mo for rGBM. Severe toxicity is also highly variable, with some series reporting none and others as much as 10.5% and a percentage of radiological RN between 6%-11%.

Table 2 Summary of selected publications reporting hypofractionated stereotactic radiosurgery as salvage treatment in recurrent gliomas.
Ref.Study typeNo. patientsHistologyRe-irradiation
Median intervalMedian tumor volumeMedian PFS2/actuarial PFS2Median OS2Severe toxicityRadionecrosis
Total dose, median
Dose/fr, median
EQD2
Selch et al[106], 2000R2115 GBM, 3 G3G, 2 G2G, 1 no biopsy25 Gy5 Gy43.8 Gy11 mo12 mL5 mo6.7 mo0%0%
Vordermark et al[107], 2005R199 GBM, 10 G2G30 Gy5 Gy52.5 Gy19 mo15 mL4.9 mo, 4.6 mo (GBM)9.3 mo, 7.9 mo (GBM)10.5% other than necrosis0%
Ernst-stecken et al[108], 2007P1510 GBM, 3 G3G. 2 G2G35 Gy7 Gy78.7 Gy10 mo22.4 mL15 mo12 mo20% need to increase steroids dose without evidence of progressive diseaseNR
Fokas et al[78], 2009P53All GBM30 Gy3 Gy37.5 GyNR35 mL22% at 12 mo9 mo0%0%
Fogh et al[29], 2010R147105 GBM, 42 G3G35 Gy3.5 Gy48.1 Gy8 mo22 mLNR11 mo (GBM)0.7% toxicity (severe headaches)0%
Mckenzie et al[69], 2013P3329 GBM, 4 G3G30 Gy5 Gy52.5 GyNR8.54 mL62% at 6 mo8.6 mo9% toxicity other than necrosis9% by image
Ogura et al[80], 2013R3015 GBM, 9 G3G. 6 G2G35 Gy7 Gy78.7 GyNR9 mL3 mo10.2 mo13.3% need to increase steroids dose without evidence of progressive disease6.1% by image
Miwa et al[109], 2014P21All GBM30 Gy5 Gy52.5 Gy12 mo27.4 mLNR11 mo4.8%9.5%
Dincoglan et al[110], 2015R28All GBM25 Gy5 Gy43.8 Gy11.2 mo36.5 mL5.8 mo10.3 mo0%11% G2 by image

A recent study, in a large and heterogeneous series of 198 patients with rHGG, reports a mOS of 7 mo (6 mo for GBM and 14 mo for grade III gliomas) with good tolerance. The most common fractionation schedules were 41.8 Gy-49.4 Gy/3.8 Gy/fr[35].

The main study with CFRT is by Combs et al[36]. They analyzed 59 patients with rGBM treated with 36 Gy/2Gy/fr, achieving an mOS of 8 mo, with only 1.7% of histologically confirmed RN despite a large median tumor volume (49.3 mL). This indicates that it may be an adequate schedule in larger lesions.

Several retrospective papers have compared the different techniques (SRS, HFSRS, CFRT), reporting similar results between them, with mOS of 9.7-11 mo[37,38].

There are very few prospective studies on the efficacy of re-RT vs systemic treatment alone. RTOG 0525[39] has reported mOS of 8.2 mo with re-RT, 10.5 mo with chemotherapy, and 11.3 mo with radiochemotherapy. Patients who only received best supportive care had an mOS of 4.8 mo, probably selected for worse overall status. Available data in rGBM generally suggest that re-RT modestly improves PFS compared with systemic treatment alone, but OS is similar[40].

Re-RT of larger volumes

The main hurdle for re-RT of voluminous relapses has been the risk of RN. Most re-RT studies describe a PTV < 40 mL[41,42]. The available evidence for large volume lesions is sparse and few studies include a median PTV greater than 75 mL. Two authors report the largest series to date. The study by Scholtyssek et al[43], with a median PTV of 110.4 mL and doses of 36 Gy (30 Gy-40.05 Gy) at 2-5 Gy/fr, did not describe severe toxicity or RN. Chan et al[44], with a median PTV of 145.3 mL and dose of 35 Gy/15 fr, in 67 patients, reported 4 cases of radiological RN. The mOS reported in these series were 7.7 and 7.8 mo, in the same range as reported in studies with small treatment volumes. We can conclude that re-RT of large volume disease is feasible, provided that the doses administered are appropriate.

Re-RT with concurrent systemic treatment

Two drugs (TMZ, BEV) are mainly used. Although they have been shown to be safe combinations, their benefit has yet to be demonstrated.

Re-RT with TMZ: Table 3 summarizes the main results. The techniques used have been HFSRS or CFRT. Hematologic ≥ grade 3 toxicity of up to > 40% has been described. RN has been reported, either radiological (7%-8%)[45,46] or histopathological in 4.3%[47]. The mOS for GBM ranges from 9.7-14 mo[45,48] and mPFS between 4-7 mo[46,47], results reported without the combination of TMZ. However, in the Grosu et al[49] and Conti et al[47] series, patients receiving TMZ had higher mOS.

Table 3 Summary of selected publications reporting re-irradiation plus temozolomide as salvage treatment in recurrent high-grade gliomas.
Ref.Study typeNo. patientsHistologyRe-irradiation
Median intervalMedian tumor volume Median PFS2/actuarial PFS2Median OS2/actuarial OS2Severe toxicityRadionecrosis
Total dose, median
Dose/fr, median
EQD2
Grosu et al[49], 2005P44 (TMZ 29)34 GBM, 2 Gliosarcomas, 8 G3G30 Gy5 Gy52.5 Gy16 mo15 mLNR8 mo (11 mo RT + TMZ vs 6 mo without TMZ)0%0%
Combs et al[81], 2008R258 GBM, 10 G3G, 7 G2G36 Gy2 Gy36 Gy36 mo50 mL5 mo; 16% at 12 mo8 mo; 25% at 12 mo0%NR
Minniti et al[45], 2011R36All GBM37.5 Gy2.5 Gy42.2 Gy14 mo13.1 mL5 mo; 8% at 12 mo9.7 mo; 33% at 12 moThrombocytopenia G3: 2.8%8% by image
Conti et al[47], 2012R23 (TMZ 12)All GBM20 Gy10 Gy60 Gy7 mo< 30 mL7 mo (TMZ) vs 4 mo (no TMZ)12 mo (TMZ) vs 7 mo (without TMZ)≥ G3 hematological toxicity > 40%4.3%
Minniti et al[46], 2013R5438 GBM, 16 G3G30 Gy6 Gy60 Gy15.5 mo9.8 mL6 mo (4 mo GBM)12.4 mo (11.4 mo GBM)Thrombocytopenia G3: 3.7%, leukopenia G3: 3.7%7% by image
Greenspoon et al[111], 2014P31All GBM30 Gy5 Gy52.5 GyAt least 6 mo12 mL7 mo9 moNRG3: 9.6%, G4: 3.2%
Aktan et al[48], 2015R21 (17 TMZ)18 GBM, 3 G3G54 Gy2 Gy54 Gy39.4 moRecurrent tumor size was median 5.5 cmNR18 mo (G3G) and 14.1 mo (GBM)0%0%

Overall, concurrent approaches with TMZ do not appear to improve re-RT outcomes and may carry increased risk of toxicity. However, these findings need to be confirmed in prospective series.

Re-RT with BEV: The association of BEV to treatment with first line radiochemotherapy did not demonstrate a benefit in OS in two phase III trials[50,51]. In recurrences, the role of concurrent BEV with re-RT is still not well defined, but several studies have confirmed the safety of this combination with reasonable survival results[52-57]. Table 4 summarizes the main results. The mOS ranges between 9.3-12.2 mo for rGBM, with mPFS between 5.2 and 6.8 mo. This combination has been shown to decrease the risk of RN, especially for re-RT of larger volumes[44,58]. The percentage of symptomatic RN/symptomatic edema, defined as the need for corticosteroids > 6 wk after re-RT, was lower with the BEV combination (21.8% vs 37.8%, P = 0.025), with these differences increasing at 1 year (23.9% vs 54.1%, P = 0.013).

Table 4 Summary of selected publications reporting re-irradiation plus bevacizumab as salvage treatment in recurrent high-grade gliomas.
Ref.Study typeNo. patientsHistologyRe-irradiation
Median intervalMedian tumor volumeMedian PFS2Median OS2Severe toxicityRadionecrosis
Total dose, median
Dose/fr, median
EQD2
Gutin et al[56], 2009P2520 GBM, 5 G3G30 Gy6 Gy60 Gy14.5 mo34 mL7.3 mo12.5 moG3: 1 hemorrhage; G4: 3 (1 bowel perforation, 1 wound dehiscence and 1 GI bleed)0%
Cuneo et al[54], 2012R63 (41 BEV)49 GBM, 8 G3G, 6 prior G2G15 Gy15 Gy63.8 Gy21 mo4.8 mLGBM: 5.2 mo (BEV) vs 2.1 mo (without BEV). 6 mo whole seriesGBM: 11.2 mo (BEV) vs 3.9 mo (without BEV). 10 mo whole series11%10%
Niyazi et al[52], 2012R30 (20 BEV)22 GBM, 8 G3G36 Gy2 Gy36 GyNRNR8 moMean 12 moG3:1; G4: 1 wound dehiscence0%
Shapiro et al[112], 2013R2420 GBM, 1 G3G, 3 G2G30 Gy6 Gy60 Gy12.6 mo35.3 mL7.5 mo (6.8 mo GBM)12.2 mo (whole series and GBM)Toxicity BEV: G4: 12.5%0%
Cabrera et al[113], 2013P158 GBM, 7 G3G18 Gy. 25 Gy18 Gy. 5 Gy90 Gy. 43.8 Gy20 moNR (< 5 cm)3.9 mo14.4 moG3:10%
Flieger et al[57], 2014P71 (57 BEV)52 GBM, 19 G3G and G2G36 Gy2 Gy36 GyNRNR5.6 mo (BEV) vs 2.5 mo (without BEV)GBM: 9.3 mo (BEV) vs 6.1 mo (without BEV)Toxicity BEV: G4: 5.3%4.2% (BEV) by image or histologically

The highly anticipated results from the NRG Oncology/RTOG 1205 phase II clinical trial (NCT01730950) are expected in 2023. It randomizes patients with recurrence to BEV alone or BEV with concurrent re-RT (35 Gy in 10 fractions for tumors smaller than 5 cm). Preliminary results of this study have confirmed the safety of the BEV-Re-HFSRT combination and that it provides a benefit in PFS at 6 mo, even without a benefit in mOS, as observed in first line.

Re-RT after progression to BEV

Recently, a new scenario of re-RT has been explored, after progression to BEV. Several groups have published data on this approach, showing an mOS of 5.4 mo[59] and 4.8 mo[39]. The combination of minocycline, BEV, and fractionated re-RT after progression to BEV has been investigated in a phase I trial[60]. PFS3 was 64.6%, and mOS was 6.4 mo. This study adds a prospective trial to the literature showing that re-RT of HGG after BEV failure can be performed with acceptable tolerability. Another recently published phase I trial included 32 patients with rHGG and the combination of pembrolizumab, an anti-programmed cell death protein 1 (PD-1) monoclonal antibody, HFSRT, and BEV, with an mOS and mPFS of 13.4 mo and 7.9 mo, respectively. The authors concluded that this combination is safe and well tolerated, meriting further investigation[61].

Re-resection and re-radiation therapy

Straube et al[62] was the first author to suggest that this strategy could be beneficial, after concluding that the pattern of relapse in 26 patients with complete re-S was solely local in 70%. Based on this, and taking into account the maximal safe resection, several groups have demonstrated the value of additional re-RT with different techniques[38,63-65].

Combs et al[63] published the first study after rHGG re-S followed by early re-RT. It included 108 patients, most of whom received 36 Gy at 2-3 Gy per fraction. The mOS was 12 mo, with no serious toxicity. In multivariate analysis, the extent of surgery, methylguanine-DNA methyltransferase (MGMT) methylation, interval time between first and second irradiation, and KPS were independent prognostic factors for OS. A subsequent study[64], with 25 interventional rGBM cases treated with HFSRS and simultaneous integrated boost (37.5 Gy and 45 Gy in 15 fractions), reported an mPFS of 13 mo and mOS of 16 mo, with better outcomes in smaller recurrences, without eloquent area involvement and in patients with a good general condition.

On multivariate analysis, the macroscopic tumor volume (GTV) ≥ 100 mL vs < 100 mL was confirmed as an independent prognostic factor affecting OS. Radiologically suspected RN was observed in 16 patients (64%) at a median of 9 mo after re-RT, and 8 patients developed grade 3 RN requiring hospitalization.

In the series by Chun et al[65] with 84 patients, the addition of radiation therapy (median dose of 45 Gy at 1.8 Gy/fr) to re-S was associated with a significant benefit in PFS, with mPFS for re-S being 3.5 mo and 9 mo for re-S plus re-RT. The benefit in OS was marginal, with an mOS of 12.7 mo with re-S vs 28.1 mo with re-S plus re-RT (P = 0.066). Three risk factors (age ≥ 50, WHO grade IV, and unmethylated promoter of MGMT) were significantly associated with poor OS in multivariate analysis. The authors established three categories of risk groups based on these factors. The benefit of re-RT in both OS and PFS was established in patients with two or more risk factors (intermediate and high risk groups). There was no radiological or pathological evidence of RN during or after re-RT.

Results of the GlioCAVE/NOA 17 trial (NCT02715297) should better determine the contribution of early adjuvant radiotherapy after re-S in rGBM. It is a prospective phase II study with a schedule of 46 Gy at 2 Gy/fr or 36 Gy at 3 Gy/fr, with PFS as the primary endpoint.

Prognostic scales for re-RT

The first scale to predict OS after re-RT published by Combs[66] derived from 233 patients with recurrent low- and HGG and included: WHO grade, age at the time of re-RT, and the time interval to re-RT. The same group published a modified version, the New Combs Score, which added other factors such as KPS, tumor volume, and re-S prior to re-RT[67]. This new revalidated scale[38,63] with a simple approach, is practical, useful, and widely used for decision making (Table 5).

Table 5 Scoring scheme and new prognostic groups of the “New Combs Score”.
Prognostic factors
Prognostic value
Primary histology
Glioblastoma, WHO IV2
Anaplastic glioma, WHO III1
Low-grade glioma, WHO I/II0
Age
≥ 50 yr1
< 50 yr0
Time between primary RT and re-RT
≤ 12 meses1
> 12 meses0
Re-resection performed
No1
Yes0
KPS
< 80%1
≥ 80%0
Tumor volume (PTV)
> 47 mL1
≤ 47 mL0
Scoring groupScoring value/mOS
a0–1/19.5 mo
b2–3/11.3 mo
c4–5/8.1 mo
d6–7/5.5 mo

Other reported prognostic features include the re-RT dose[31,57], use of salvage chemotherapy[43,57], extent of resection[28,43], MGMT promoter methylation status[46], and radiographic response[68-70]. However, how they should be quantified remains to be described.

Interestingly, Chapman et al[38], without finding an association of irradiation technique (SRS vs non SRS) or fractionation with survival, identified a threshold dose as a function of PTV size that should not be exceeded to minimize toxicity: 40 Gy Biological Equivalent Dose 10 for SRS (16 Gy in 1 fraction) and 45 Gy Biological Equivalent Dose 10 for non-SRS treatments (approximately 30 Gy in 5 fractions, 35 Gy in 10 fractions or 40 Gy in 20 fractions), from the same range as those identified in other series[29,57,71]. And, globally, it identifies a group of patients who can achieve an advantage in OS and PFS with re-RT, in particular young patients with good KPS, longer time interval from initial radiation to first progression, small recurrence volume, and an adequate re-RT dose.

RADIOTHERAPY SPECIFICATIONS
Treatment volumes

Definition of target volumes: The definition of re-RT target volumes should be conservative, minimizing the irradiation of healthy tissues to avoid severe toxicities (RN). It requires not only extreme precision and conformality during treatment but also precise images that identify the exact location of tumor tissues. Inaccuracies in tumor delineation may diminish any gain in local control achieved by dose escalation. One aspect to consider would be whether the relapse is located in the area of previous maximum dose or is marginal or remote from the first irradiation. In this case, and depending on the volume of the relapse, the dose prescription can be less conservative.

Several studies have shown that standard anatomic imaging modalities [computed tomography, magnetic resonance imaging (MRI)], while very accurate in visualizing normal anatomic structures, are limited in defining the exact extent of the tumor. Classically, volume delineation for irradiation is based on T1-weighted MRI with gadolinium. Contrast uptake is a consequence of blood-brain barrier disruption and does not necessarily reflect the actual tumor extent in gliomas. Macroscopic tumor masses far from the margins of contrast enhancement have been detected in surrounding edema and even in adjacent normal-appearing brain tissue[72-74]. Antiangiogenic drugs may also condition contrast uptake, as they may initially have a stabilizing effect on the blood-brain barrier[75].

Multiple studies correlating imaging findings with histopathologic evaluation in surgically treated patients with HGG have indicated that molecular imaging with amino acid positron emission tomography (PET) is more specific and equally sensitive for tumor detection than MRI (T1 with gadolinium). Grosu et al[49] have postulated that target volumes for re-RT should be based on amino acid PET imaging in addition to MRI, to include the actual tumor dimension. Other imaging modalities have been used to delineate GTV, including spectroscopy MRI, perfusion-weighted imaging and diffusion-weighted imaging[76], 11C-methionine PET[49], and 18 F-dihydroxyphenylalanine PET[46]. However, there are no randomized trials that have evaluated the impact of molecular or functional imaging-based radiotherapy on the outcomes achieved.

The ongoing phase II GLIAA (NOA 10/ARO 2013-1) trial[77] is the first randomized study evaluating the impact of differences in planning volumes designed with molecular vs MRI imaging on PFS after re-RT in patients with rGBM. The limited availability of molecular and/or functional imaging equipment together with the lack of evidence of its superiority in the design of planning volumes conditions the continued use of MRI images for re-RT volume definition.

Volumes-exclusive radiotherapy: The definition of the target volume generally includes the GTV, defined as any contrast-enhancing lesion on T1-weighted MRI. In most studies, the clinical target volume (CTV) equals GTV[28,29,78]. Some papers add a CTV to include the peritumoral edema visualized in the fluid-attenuated inversion recovery sequence of the MRI, since it is known that tumor cells can be found in this location[79,80]. Subsequently, a margin usually ≤ 5 mm is added for PTV expansion[45,49,78], although some authors include up to 1 cm[57,81].

Volumes-adjuvant radiotherapy: For re-S patients, Straube et al[62] proposed a GTV including the resection cavity and contrast enhancement areas, with a margin of 5-10 mm to generate the CTV and 1-3 mm to create the PTV. The GLIOCAVE-NOA 17 study[82] meets these criteria. The CTV encompasses the margins of the resection cavity, including all areas of contrast enhancement plus 5 mm.

DOSAGE AND FRACTIONATION

The optimal dose and fractionation schedule in these patients is unknown. Re-RT is a well-known factor contributing to the risk of RN, which is directly associated with dose and irradiated volume.

Sminia and Mayer[83] examined > 25 glioma re-RT studies to assess tolerance dose and treatment volume of normal brain tissue. RN occurred with a cumulative EQD2 dose (α/β = 3) > 100 Gy for CFRT, > 105 Gy for fractionated stereotactic radiotherapy (FSRT), and 135 Gy for SRS.

Given that these patients have already received 60 Gy after initial diagnosis, there is a margin of at least 40 Gy for re-RT. Hence, the prescribed doses for re-RT in most published studies ranged from 30-45 Gy, thus maintaining a cumulative EQD2 of approximately 100 Gy[64]. However, given that brain tissue recovers over time, it seems safe to administer higher doses to smaller volumes, using FSRT or SRS, without increasing the likelihood of RN[83].

Scoccianti et al[42], after an extensive review of published series and always proposing schemes with reported severe toxicity ≤ 3.5%, described a treatment strategy depending on the volume to be irradiated. Thus, for small volumes (≤ 12.5 mL) SRS schemes are safe (e.g., 12-15 Gy) provided that the EQD2 value does not exceed 65 Gy; HFSRT (e.g., 5 × 5 Gy) for medium-sized lesions (> 12.5-35 mL), provided that the EQD2 value does not exceed 50 Gy and CFRT (e.g., 36 Gy in 20 fr) for larger lesions (> 35-50 mL). These authors pointed out that this recommended strategy should be confirmed in prospective studies.

Whenever possible, hypofractionated schemes are preferred, avoiding unnecessary transfers in these patients with limited life expectancy.

Organ-at-risk tolerance dose

In primary treatment, the maximum doses to the brainstem, chiasm, and optic nerves to avoid the risk of myelopathy are well defined[84,85]. In the context of re-RT in HGG, current evidence is limited[15,44]. Preclinical data suggest a 61% recovery in the spinal cord after 1 year since the first irradiation, and it is believed that this is likely to be applicable to other central nervous system tissues[86]. These models indicate that, in the context of re-RT, maximum summed doses of up to 86 Gy could be tolerated for the optic chiasm and brainstem.

Two series with low recorded toxicity analyzed cumulative dose in organ-at-risk with different doses and fractionations. Shen et al[71] reported a median maximum dose in the brainstem of 76.9 Gy and 56 Gy in the optic pathway, with a CFRT schedule and a mean dose of 41.4 Gy. In the series of Chan et al[44], with a dose mostly of 35-40 Gy/15 fr, the median maximum dose was 64 Gy for the brainstem and 54.9 Gy for the optic chiasm, although it is noted that concomitant BEV was administered, which may reduce the risk of RN.

It is essential to record and communicate doses to organ-at-risk before re-RTs in order to be able to design a toxicity risk model.

TOXICITY AND QOL
Toxicity

Data on re-RT toxicity are scarce in the literature (Tables 1-4), and its analysis and quantification are difficult. Late toxicity assessment is limited by poor prognosis, difficult differentiation between tumor recurrence, and RN, which is associated with the variety of techniques and fractionations used.

The only existing meta-analysis[33] reported a grade ≥ 3 toxicity rate of 7%, and the morbidity and mortality rate for re-RT ranged from 0%-31% and 0%-1%, respectively.

QoL

Disease progression is associated with deterioration of neurocognitive function. The evidence supporting treatment in this population is evolving, but little is known about its impact on QoL. The survival benefit is desirable but must be carefully weighed against expected morbidities.

Analysis of pooled data from over 300 GBM patients from 13 published articles showed that overall, re-RT resulted in clinical improvement in 24%-45% of patients and a reduction in corticosteroid dependence in 20%-60% of patients. However, the subgroup with KPS < 70 appeared to have a higher risk of early progression and apparently had less benefit from re-RT[87].

Very few studies prospectively evaluate the impact on QoL and activities of daily living in the setting of salvage re-RT. Wick et al[88] analyzed QoL in 84 patients with rGBM from a phase II trial with Asunercept/APG 101 and re-RT vs re-RT alone, with a dose of 36 Gy at 2 Gy/fr. The EORTC QLQ-C15-PAL, EORTC QLQ-BN20, and Medical Research Council scale questionnaires were used, concluding that Asunercept plus re-RT significantly prolonged time to deterioration of QoL vs re-RT alone. More recently, Maitre et al[89] reported prospective data on QoL and activities of daily living in patients with recurrent/progressive glioma treated with re-RT (median dose EQD2 51.4 Gy). They used the QLQ-C30 and QLQ-BN20 questionnaires and the modified Barthel index. They performed 225 evaluations in 60 patients, concluding that high-dose re-RT in selected patients is associated with stabilization of QoL and greater functional independence.

NEW STRATEGIES FOR RE-RT

New re-RT strategies for the treatment of HGG recurrences include particle radiotherapy, as well as intraoperative radiation therapy (IORT) and brachytherapy. Although they are not novel techniques, they are re-emerging in recent years with technological advances.

Particle irradiation

Proton therapy is emerging for the treatment of these patients. Due to its physical and radiobiological properties, this radiation modality offers dosimetric advantages over photons, achieving a better dose distribution and decreasing the irradiation of healthy tissue. The Proton Collaborative Group has published the largest series to date[90]. They analyzed 45 patients with a median of 20.2 mo between initial diagnosis and recurrence. The median dose was 46.2 Gy (range, 25-60 Gy), with a mean of 2.2 Gy/fr, achieving mPFS and mOS of 13.9 and 14.2 mo, respectively. The treatment was well tolerated, and the appreciated toxicity was related to a dose higher than 41 Gy (EQD2). Only prior surgery was positively associated with PFS and OS.

The first study of re-RT with carbon ion beams in rHGG analyzed 30 patients with a median interval between initial radiotherapy and re-RT of 10 mo[91]. The dose administered was 45 Gy in 15 fractions, with a mOS of 13 mo. Eight patients had grade 3 toxicity. Only initial histology with a Ki67 < 20% was a prognostic factor. Resection or chemotherapy did not significantly improve OS. A phase I/II trial to compare re-RT of recurrent gliomas with carbon ions vs re-RT with photons is ongoing (NCT 01166308).

IORT

IORT data come from older series, mainly from HGG at diagnosis, and only a few papers included rGBM[32]. The results were promising, but the complexity of the procedure led to abandoning its use. The development of portable systems capable of being moved to the operating room has sparked interest in this technique. This approach is conceptually attractive because it allows the delivery of a large dose of radiation to the tumor bed and tumor debris close to the surgical cavity immediately after resection, while respecting the surrounding brain tissue, decreasing the likelihood of RN. In addition, local and systemic immune responses may be promoted, which could benefit oncological outcomes[92,93].

Recently, although in newly diagnosed GBM, Giordano et al[94] reported the results of a phase I/II dose-escalation trial, evaluating the safety and efficacy of the Zeiss INTRABEAM system, a miniaturized 50 keV LINAC with spherical applicators. Fifteen patients, mainly with subtotal resection, were included, receiving a dose of 30 and 40 Gy, with no evidence of limiting toxicity, achieving a PFS of 17.7 mo.

Brachytherapy

Like IORT, it has the advantage of allowing immediate irradiation of the surgical cavity[95], without having to wait the usual 4 wk until the surgical wound is completely healed to start external radiotherapy. This delay is not desirable in HGGs, where in as little as 3 wk there is already a high rate of tumor repopulation. The most commonly used technique is permanent seed implantation. Initially the isotope used was I-125, but high complication rates were reported[96]. Suture-stranded Cs-131 seeds, with a shorter half-life, are now the most commonly used isotope. A study combining re-S with insertion of suture-stranded Cs-131 seeds and BEV (before or after the procedure) has recently been published[95]. Twenty patients were analyzed, with a dose of 80 Gy administered at 0.5 cm from the surface of the resection cavity. Seven patients had been previously salvaged with external radiotherapy. Local control was 85% and mOS was 9 mo. There were two wound infections and three seizures, with no case of RN.

These radiation techniques are safe and effective, but further prospective and comparative research is needed to draw solid conclusions.

SPECIAL PATIENT GROUPS
Elderly patients

As in younger patients, radiotherapy is the cornerstone of first-line treatment of older patients with GBM. However, they receive poor care after recurrence[97]. The evidence for re-RT in older patients is very scarce, as the median age in published papers is around 53 years[33]. However, the aging population is growing and treatment decisions in patients with rGBM and good general condition are increasing. To our knowledge, only one study on re-RT in older patients has been published. Straube et al[98] reported the results of 25 patients with a median age of 69.6 years (range 65-79) who received re-RT, most after reintervention. The mOS was 6.9 mo and mPFS at 4.3 mo, with no case of severe toxicity attributable to re-RT. This survival is within the range of series reported in younger patients[28]. Therefore, although prospective trials are needed, these results suggest that second-line salvage therapy should not be dismissed on the basis of age alone.

Pediatric patients

As with adults, children with rHGG have limited treatment options. Re-RT has an emerging role as a palliative treatment for children with recurrent brainstem glioma (diffuse intrinsic pontine glioma or DIPG)[99-101], being associated with symptomatic improvement and longer survival compared to non-re-irradiated patients[99]. Indeed, re-RT in DIPGs is the subject of several ongoing or completed prospective studies (NCT01777633 and NCT03126266). Given that the irradiation dose tolerance of the supratentorial brain is higher than that of the brainstem, it stands to reason that re-RT in supratentorial rHGG should be equally safe and effective[102]. However, the role of re-RT has been little studied in non-pontine gliomas.

Recently, Tsang et al[103] have published the results of the largest known cohort of children with recurrent supratentorial HGG treated with re-RT compared to a group of non-re-irradiated children. They retrospectively analyzed 40 patients ≤ 18 years. Fourteen patients, with an interval of at least 6 mo after the first radiotherapy, were re-irradiated. Doses administered ranged from 30-54 Gy at 1.8 Gy/fr. Median survival was 9.4 mo for re-RT patients compared to 3.8 mo for the 26 who did not receive re-RT. The time elapsed between the first and second irradiation determined significant differences, being higher in children with an interval ≥ 12 mo. One patient presented grade 3 RN 4 mo after re-RT. There were no significant differences between patients with initial vs distant field re-RT, between those who received concurrent chemotherapy vs exclusive re-RT, or between those who were previously operated vs those who received radiotherapy alone. Thus, offering re-RT to these patients is associated with reasonable short-term control and survival without significant toxicity.

CONCLUSION

The rHGG scenario remains devastating. Nevertheless, the available evidence, albeit low level, suggests that re-RT, at recommended doses and in selected patients, is safe and provides encouraging local control and survival rates.

The combination of re-S with early re-RT appears to be the most promising option.

Randomized clinical trials are needed to establish the optimal treatment strategy for these patients.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Oncology

Country/Territory of origin: Spain

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Gamarra LF S-Editor: Fan JR L-Editor: Filipodia P-Editor: Yuan YY

References
1.  Ostrom QT, Patil N, Cioffi G, Waite K, Kruchko C, Barnholtz-Sloan JS. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2013-2017. Neuro Oncol. 2020;22:iv1-iv96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 637]  [Cited by in F6Publishing: 1023]  [Article Influence: 341.0]  [Reference Citation Analysis (0)]
2.  Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P, Ellison DW. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol. 2016;131:803-820.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10993]  [Cited by in F6Publishing: 9857]  [Article Influence: 1232.1]  [Reference Citation Analysis (0)]
3.  Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, Curschmann J, Janzer RC, Ludwin SK, Gorlia T, Allgeier A, Lacombe D, Cairncross JG, Eisenhauer E, Mirimanoff RO; European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups;  National Cancer Institute of Canada Clinical Trials Group. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987-996.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14033]  [Cited by in F6Publishing: 14470]  [Article Influence: 761.6]  [Reference Citation Analysis (0)]
4.  Cairncross G, Wang M, Shaw E, Jenkins R, Brachman D, Buckner J, Fink K, Souhami L, Laperriere N, Curran W, Mehta M. Phase III trial of chemoradiotherapy for anaplastic oligodendroglioma: long-term results of RTOG 9402. J Clin Oncol. 2013;31:337-343.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 779]  [Cited by in F6Publishing: 733]  [Article Influence: 61.1]  [Reference Citation Analysis (0)]
5.  van den Bent MJ, Baumert B, Erridge SC, Vogelbaum MA, Nowak AK, Sanson M, Brandes AA, Clement PM, Baurain JF, Mason WP, Wheeler H, Chinot OL, Gill S, Griffin M, Brachman DG, Taal W, Rudà R, Weller M, McBain C, Reijneveld J, Enting RH, Weber DC, Lesimple T, Clenton S, Gijtenbeek A, Pascoe S, Herrlinger U, Hau P, Dhermain F, van Heuvel I, Stupp R, Aldape K, Jenkins RB, Dubbink HJ, Dinjens WNM, Wesseling P, Nuyens S, Golfinopoulos V, Gorlia T, Wick W, Kros JM. Interim results from the CATNON trial (EORTC study 26053-22054) of treatment with concurrent and adjuvant temozolomide for 1p/19q non-co-deleted anaplastic glioma: a phase 3, randomised, open-label intergroup study. Lancet. 2017;390:1645-1653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 262]  [Cited by in F6Publishing: 232]  [Article Influence: 33.1]  [Reference Citation Analysis (0)]
6.  Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, Hau P, Brandes AA, Gijtenbeek J, Marosi C, Vecht CJ, Mokhtari K, Wesseling P, Villa S, Eisenhauer E, Gorlia T, Weller M, Lacombe D, Cairncross JG, Mirimanoff RO. Effects of radiotherapy with concomitant and adjuvant temozolomide vs radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4968]  [Cited by in F6Publishing: 5461]  [Article Influence: 364.1]  [Reference Citation Analysis (1)]
7.  Wen PY, Macdonald DR, Reardon DA, Cloughesy TF, Sorensen AG, Galanis E, Degroot J, Wick W, Gilbert MR, Lassman AB, Tsien C, Mikkelsen T, Wong ET, Chamberlain MC, Stupp R, Lamborn KR, Vogelbaum MA, van den Bent MJ, Chang SM. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol. 2010;28:1963-1972.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2528]  [Cited by in F6Publishing: 2695]  [Article Influence: 192.5]  [Reference Citation Analysis (0)]
8.  Lin NU, Lee EQ, Aoyama H, Barani IJ, Barboriak DP, Baumert BG, Bendszus M, Brown PD, Camidge DR, Chang SM, Dancey J, de Vries EG, Gaspar LE, Harris GJ, Hodi FS, Kalkanis SN, Linskey ME, Macdonald DR, Margolin K, Mehta MP, Schiff D, Soffietti R, Suh JH, van den Bent MJ, Vogelbaum MA, Wen PY; Response Assessment in Neuro-Oncology (RANO) group. Response assessment criteria for brain metastases: proposal from the RANO group. Lancet Oncol. 2015;16:e270-e278.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 501]  [Cited by in F6Publishing: 599]  [Article Influence: 66.6]  [Reference Citation Analysis (0)]
9.  Niyazi M, Siefert A, Schwarz SB, Ganswindt U, Kreth FW, Tonn JC, Belka C. Therapeutic options for recurrent malignant glioma. Radiother Oncol. 2011;98:1-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 99]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
10.  Stupp R, Wong ET, Kanner AA, Steinberg D, Engelhard H, Heidecke V, Kirson ED, Taillibert S, Liebermann F, Dbalý V, Ram Z, Villano JL, Rainov N, Weinberg U, Schiff D, Kunschner L, Raizer J, Honnorat J, Sloan A, Malkin M, Landolfi JC, Payer F, Mehdorn M, Weil RJ, Pannullo SC, Westphal M, Smrcka M, Chin L, Kostron H, Hofer S, Bruce J, Cosgrove R, Paleologous N, Palti Y, Gutin PH. NovoTTF-100A versus physician's choice chemotherapy in recurrent glioblastoma: a randomised phase III trial of a novel treatment modality. Eur J Cancer. 2012;48:2192-2202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 528]  [Cited by in F6Publishing: 517]  [Article Influence: 43.1]  [Reference Citation Analysis (0)]
11.  van den Bent MJ, Klein M, Smits M, Reijneveld JC, French PJ, Clement P, de Vos FYF, Wick A, Mulholland PJ, Taphoorn MJB, Lewis J, Weller M, Chinot OL, Kros JM, de Heer I, Verschuere T, Coens C, Golfinopoulos V, Gorlia T, Idbaih A. Bevacizumab and temozolomide in patients with first recurrence of WHO grade II and III glioma, without 1p/19q co-deletion (TAVAREC): a randomised controlled phase 2 EORTC trial. Lancet Oncol. 2018;19:1170-1179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 49]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
12.  Reardon DA, Brandes AA, Omuro A, Mulholland P, Lim M, Wick A, Baehring J, Ahluwalia MS, Roth P, Bähr O, Phuphanich S, Sepulveda JM, De Souza P, Sahebjam S, Carleton M, Tatsuoka K, Taitt C, Zwirtes R, Sampson J, Weller M. Effect of Nivolumab vs Bevacizumab in Patients With Recurrent Glioblastoma: The CheckMate 143 Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020;6:1003-1010.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 411]  [Cited by in F6Publishing: 727]  [Article Influence: 242.3]  [Reference Citation Analysis (0)]
13.  Lang FF, Conrad C, Gomez-Manzano C, Yung WKA, Sawaya R, Weinberg JS, Prabhu SS, Rao G, Fuller GN, Aldape KD, Gumin J, Vence LM, Wistuba I, Rodriguez-Canales J, Villalobos PA, Dirven CMF, Tejada S, Valle RD, Alonso MM, Ewald B, Peterkin JJ, Tufaro F, Fueyo J. Phase I Study of DNX-2401 (Delta-24-RGD) Oncolytic Adenovirus: Replication and Immunotherapeutic Effects in Recurrent Malignant Glioma. J Clin Oncol. 2018;36:1419-1427.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 442]  [Cited by in F6Publishing: 437]  [Article Influence: 72.8]  [Reference Citation Analysis (0)]
14.  Liau LM, Ashkan K, Tran DD, Campian JL, Trusheim JE, Cobbs CS, Heth JA, Salacz M, Taylor S, D'Andre SD, Iwamoto FM, Dropcho EJ, Moshel YA, Walter KA, Pillainayagam CP, Aiken R, Chaudhary R, Goldlust SA, Bota DA, Duic P, Grewal J, Elinzano H, Toms SA, Lillehei KO, Mikkelsen T, Walbert T, Abram SR, Brenner AJ, Brem S, Ewend MG, Khagi S, Portnow J, Kim LJ, Loudon WG, Thompson RC, Avigan DE, Fink KL, Geoffroy FJ, Lindhorst S, Lutzky J, Sloan AE, Schackert G, Krex D, Meisel HJ, Wu J, Davis RP, Duma C, Etame AB, Mathieu D, Kesari S, Piccioni D, Westphal M, Baskin DS, New PZ, Lacroix M, May SA, Pluard TJ, Tse V, Green RM, Villano JL, Pearlman M, Petrecca K, Schulder M, Taylor LP, Maida AE, Prins RM, Cloughesy TF, Mulholland P, Bosch ML. First results on survival from a large Phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma. J Transl Med. 2018;16:142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 325]  [Article Influence: 54.2]  [Reference Citation Analysis (0)]
15.  Krauze AV, Attia A, Braunstein S, Chan M, Combs SE, Fietkau R, Fiveash J, Flickinger J, Grosu A, Howard S, Nieder C, Niyazi M, Rowe L, Smart DD, Tsien C, Camphausen K. Expert consensus on re-irradiation for recurrent glioma. Radiat Oncol. 2017;12:194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
16.  Birk HS, Han SJ, Butowski NA. Treatment options for recurrent high-grade gliomas. CNS Oncol. 2017;6:61-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 43]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
17.  Weller M, Cloughesy T, Perry JR, Wick W. Standards of care for treatment of recurrent glioblastoma--are we there yet? Neuro Oncol. 2013;15:4-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 530]  [Cited by in F6Publishing: 509]  [Article Influence: 46.3]  [Reference Citation Analysis (0)]
18.  Scoccianti S, Perna M, Olmetto E, Delli Paoli C, Terziani F, Ciccone LP, Detti B, Greto D, Simontacchi G, Grassi R, Scoccimarro E, Bonomo P, Mangoni M, Desideri I, Di Cataldo V, Vernaleone M, Casati M, Pallotta S, Livi L. Local treatment for relapsing glioblastoma: A decision-making tree for choosing between reirradiation and second surgery. Crit Rev Oncol Hematol. 2021;157:103184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
19.  Hou LC, Veeravagu A, Hsu AR, Tse VC. Recurrent glioblastoma multiforme: a review of natural history and management options. Neurosurg Focus. 2006;20:E5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 190]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
20.  Rusthoven KE, Olsen C, Franklin W, Kleinschmidt-DeMasters BK, Kavanagh BD, Gaspar LE, Lillehei K, Waziri A, Damek DM, Chen C. Favorable prognosis in patients with high-grade glioma with radiation necrosis: the University of Colorado reoperation series. Int J Radiat Oncol Biol Phys. 2011;81:211-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 33]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
21.  Chen MW, Morsy AA, Liang S, Ng WH. Re-do Craniotomy for Recurrent Grade IV Glioblastomas: Impact and Outcomes from the National Neuroscience Institute Singapore. World Neurosurg. 2016;87:439-445.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
22.  Clarke JL, Ennis MM, Yung WK, Chang SM, Wen PY, Cloughesy TF, Deangelis LM, Robins HI, Lieberman FS, Fine HA, Abrey L, Gilbert MR, Mehta M, Kuhn JG, Aldape KD, Lamborn KR, Prados MD; North American Brain Tumor Consortium. Is surgery at progression a prognostic marker for improved 6-month progression-free survival or overall survival for patients with recurrent glioblastoma? Neuro Oncol. 2011;13:1118-1124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 86]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
23.  Tully PA, Gogos AJ, Love C, Liew D, Drummond KJ, Morokoff AP. Reoperation for Recurrent Glioblastoma and Its Association With Survival Benefit. Neurosurgery. 2016;79:678-689.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 38]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
24.  Sastry RA, Shankar GM, Gerstner ER, Curry WT. The impact of surgery on survival after progression of glioblastoma: A retrospective cohort analysis of a contemporary patient population. J Clin Neurosci. 2018;53:41-47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 17]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
25.  Ringel F, Pape H, Sabel M, Krex D, Bock HC, Misch M, Weyerbrock A, Westermaier T, Senft C, Schucht P, Meyer B, Simon M; SN1 study group. Clinical benefit from resection of recurrent glioblastomas: results of a multicenter study including 503 patients with recurrent glioblastomas undergoing surgical resection. Neuro Oncol. 2016;18:96-104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 157]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
26.  Lu VM, Jue TR, McDonald KL, Rovin RA. The Survival Effect of Repeat Surgery at Glioblastoma Recurrence and its Trend: A Systematic Review and Meta-Analysis. World Neurosurg. 2018;115:453-459.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 47]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
27.  Suchorska B, Weller M, Tabatabai G, Senft C, Hau P, Sabel MC, Herrlinger U, Ketter R, Schlegel U, Marosi C, Reifenberger G, Wick W, Tonn JC, Wirsching HG. Complete resection of contrast-enhancing tumor volume is associated with improved survival in recurrent glioblastoma-results from the DIRECTOR trial. Neuro Oncol. 2016;18:549-556.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 160]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
28.  Combs SE, Widmer V, Thilmann C, Hof H, Debus J, Schulz-Ertner D. Stereotactic radiosurgery (SRS): treatment option for recurrent glioblastoma multiforme (GBM). Cancer. 2005;104:2168-2173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 161]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
29.  Fogh SE, Andrews DW, Glass J, Curran W, Glass C, Champ C, Evans JJ, Hyslop T, Pequignot E, Downes B, Comber E, Maltenfort M, Dicker AP, Werner-Wasik M. Hypofractionated stereotactic radiation therapy: an effective therapy for recurrent high-grade gliomas. J Clin Oncol. 2010;28:3048-3053.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 226]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
30.  Skeie BS, Enger PO, Brogger J, Ganz JC, Thorsen F, Heggdal JI, Pedersen PH. Gamma knife surgery vs reoperation for recurrent glioblastoma multiforme. World Neurosurg. 2012;78:658-669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 80]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
31.  Martínez-Carrillo M, Tovar-Martín I, Zurita-Herrera M, Del Moral-Ávila R, Guerrero-Tejada R, Saura-Rojas E, Osorio-Ceballos JL, Arrebola-Moreno JP, Expósito-Hernández J. Salvage radiosurgery for selected patients with recurrent malignant gliomas. Biomed Res Int. 2014;2014:657953.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 36]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
32.  Krivoshapkin A, Gaytan A, Salim N, Abdullaev O, Sergeev G, Marmazeev I, Cesnulis E, Killeen T. Repeat Resection and Intraoperative Radiotherapy for Malignant Gliomas of the Brain: A History and Review of Current Techniques. World Neurosurg. 2019;132:356-362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 3]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
33.  Kazmi F, Soon YY, Leong YH, Koh WY, Vellayappan B. Re-irradiation for recurrent glioblastoma (GBM): a systematic review and meta-analysis. J Neurooncol. 2019;142:79-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 91]  [Article Influence: 15.2]  [Reference Citation Analysis (0)]
34.  Kong DS, Lee JI, Park K, Kim JH, Lim DH, Nam DH. Efficacy of stereotactic radiosurgery as a salvage treatment for recurrent malignant gliomas. Cancer. 2008;112:2046-2051.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 120]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
35.  Kaul D, Pudlitz V, Böhmer D, Wust P, Budach V, Grün A. Reirradiation of High-Grade Gliomas: A Retrospective Analysis of 198 Patients Based on the Charité Data Set. Adv Radiat Oncol. 2020;5:959-964.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
36.  Combs SE, Thilmann C, Edler L, Debus J, Schulz-Ertner D. Efficacy of fractionated stereotactic reirradiation in recurrent gliomas: long-term results in 172 patients treated in a single institution. J Clin Oncol. 2005;23:8863-8869.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 245]  [Cited by in F6Publishing: 223]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
37.  Post CCB, Kramer MCA, Smid EJ, van der Weide HL, Kleynen CE, Heesters MAAM, Verhoeff JJC. Patterns of re-irradiation for recurrent gliomas and validation of a prognostic score. Radiother Oncol. 2019;130:156-163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
38.  Chapman CH, Hara JH, Molinaro AM, Clarke JL, Oberheim Bush NA, Taylor JW, Butowski NA, Chang SM, Fogh SE, Sneed PK, Nakamura JL, Raleigh DR, Braunstein SE. Reirradiation of recurrent high-grade glioma and development of prognostic scores for progression and survival. Neurooncol Pract. 2019;6:364-374.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
39.  Shi W, Scannell Bryan M, Gilbert MR, Mehta MP, Blumenthal DT, Brown PD, Valeinis E, Hopkins K, Souhami L, Andrews DW, Tzuk-Shina T, Howard SP, Youssef EF, Lessard N, Dignam JJ, Werner-Wasik M. Investigating the Effect of Reirradiation or Systemic Therapy in Patients With Glioblastoma After Tumor Progression: A Secondary Analysis of NRG Oncology/Radiation Therapy Oncology Group Trial 0525. Int J Radiat Oncol Biol Phys. 2018;100:38-44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 44]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
40.  Wen PY, Weller M, Lee EQ, Alexander BM, Barnholtz-Sloan JS, Barthel FP, Batchelor TT, Bindra RS, Chang SM, Chiocca EA, Cloughesy TF, DeGroot JF, Galanis E, Gilbert MR, Hegi ME, Horbinski C, Huang RY, Lassman AB, Le Rhun E, Lim M, Mehta MP, Mellinghoff IK, Minniti G, Nathanson D, Platten M, Preusser M, Roth P, Sanson M, Schiff D, Short SC, Taphoorn MJB, Tonn JC, Tsang J, Verhaak RGW, von Deimling A, Wick W, Zadeh G, Reardon DA, Aldape KD, van den Bent MJ. Glioblastoma in adults: a Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions. Neuro Oncol. 2020;22:1073-1113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 512]  [Cited by in F6Publishing: 492]  [Article Influence: 123.0]  [Reference Citation Analysis (0)]
41.  Ho ALK, Jena R. Glioma Re-irradiation and Neurocognitive Dysfunction. Clin Oncol (R Coll Radiol). 2018;30:457.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
42.  Scoccianti S, Francolini G, Carta GA, Greto D, Detti B, Simontacchi G, Visani L, Baki M, Poggesi L, Bonomo P, Mangoni M, Desideri I, Pallotta S, Livi L. Re-irradiation as salvage treatment in recurrent glioblastoma: A comprehensive literature review to provide practical answers to frequently asked questions. Crit Rev Oncol Hematol. 2018;126:80-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 37]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
43.  Scholtyssek F, Zwiener I, Schlamann A, Seidel C, Meixensberger J, Bauer M, Hoffmann KT, Combs SE, von Bueren AO, Kortmann RD, Müller K. Reirradiation in progressive high-grade gliomas: outcome, role of concurrent chemotherapy, prognostic factors and validation of a new prognostic score with an independent patient cohort. Radiat Oncol. 2013;8:161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 43]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
44.  Chan J, Jayamanne D, Wheeler H, Khasraw M, Wong M, Kastelan M, Guo L, Back M. The role of large volume re-irradiation with Bevacizumab in chemorefractory high grade glioma. Clin Transl Radiat Oncol. 2020;22:33-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
45.  Minniti G, Armosini V, Salvati M, Lanzetta G, Caporello P, Mei M, Osti MF, Maurizi RE. Fractionated stereotactic reirradiation and concurrent temozolomide in patients with recurrent glioblastoma. J Neurooncol. 2011;103:683-691.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 60]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
46.  Minniti G, Scaringi C, De Sanctis V, Lanzetta G, Falco T, Di Stefano D, Esposito V, Enrici RM. Hypofractionated stereotactic radiotherapy and continuous low-dose temozolomide in patients with recurrent or progressive malignant gliomas. J Neurooncol. 2013;111:187-194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 39]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
47.  Conti A, Pontoriero A, Arpa D, Siragusa C, Tomasello C, Romanelli P, Cardali S, Granata F, De Renzis C, Tomasello F. Efficacy and toxicity of CyberKnife re-irradiation and "dose dense" temozolomide for recurrent gliomas. Acta Neurochir (Wien). 2012;154:203-209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 48]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
48.  Aktan M, Koc M, Kanyilmaz G. Survival following reirradiation using intensity-modulated radiation therapy with temozolomide in selected patients with recurrent high grade gliomas. Ann Transl Med. 2015;3:304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
49.  Grosu AL, Weber WA, Franz M, Stärk S, Piert M, Thamm R, Gumprecht H, Schwaiger M, Molls M, Nieder C. Reirradiation of recurrent high-grade gliomas using amino acid PET (SPECT)/CT/MRI image fusion to determine gross tumor volume for stereotactic fractionated radiotherapy. Int J Radiat Oncol Biol Phys. 2005;63:511-519.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 277]  [Cited by in F6Publishing: 224]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
50.  Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishikawa R, Carpentier AF, Hoang-Xuan K, Kavan P, Cernea D, Brandes AA, Hilton M, Abrey L, Cloughesy T. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med. 2014;370:709-722.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1748]  [Cited by in F6Publishing: 1731]  [Article Influence: 173.1]  [Reference Citation Analysis (0)]
51.  Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT, Vogelbaum MA, Colman H, Chakravarti A, Pugh S, Won M, Jeraj R, Brown PD, Jaeckle KA, Schiff D, Stieber VW, Brachman DG, Werner-Wasik M, Tremont-Lukats IW, Sulman EP, Aldape KD, Curran WJ Jr, Mehta MP. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med. 2014;370:699-708.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1903]  [Cited by in F6Publishing: 1905]  [Article Influence: 190.5]  [Reference Citation Analysis (0)]
52.  Niyazi M, Ganswindt U, Schwarz SB, Kreth FW, Tonn JC, Geisler J, la Fougère C, Ertl L, Linn J, Siefert A, Belka C. Irradiation and bevacizumab in high-grade glioma retreatment settings. Int J Radiat Oncol Biol Phys. 2012;82:67-76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 73]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
53.  Palmer JD, Bhamidipati D, Song A, Eldredge-Hindy HB, Siglin J, Dan TD, Champ CE, Zhang I, Bar-Ad V, Kim L, Glass J, Evans JJ, Andrews DW, Werner-Wasik M, Shi W. Bevacizumab and re-irradiation for recurrent high grade gliomas: does sequence matter? J Neurooncol. 2018;140:623-628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 16]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
54.  Cuneo KC, Vredenburgh JJ, Sampson JH, Reardon DA, Desjardins A, Peters KB, Friedman HS, Willett CG, Kirkpatrick JP. Safety and efficacy of stereotactic radiosurgery and adjuvant bevacizumab in patients with recurrent malignant gliomas. Int J Radiat Oncol Biol Phys. 2012;82:2018-2024.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 129]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
55.  Park KJ, Kano H, Iyer A, Liu X, Niranjan A, Flickinger JC, Lieberman FS, Lunsford LD, Kondziolka D. Salvage gamma knife stereotactic radiosurgery followed by bevacizumab for recurrent glioblastoma multiforme: a case-control study. J Neurooncol. 2012;107:323-333.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 52]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
56.  Gutin PH, Iwamoto FM, Beal K, Mohile NA, Karimi S, Hou BL, Lymberis S, Yamada Y, Chang J, Abrey LE. Safety and efficacy of bevacizumab with hypofractionated stereotactic irradiation for recurrent malignant gliomas. Int J Radiat Oncol Biol Phys. 2009;75:156-163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 294]  [Cited by in F6Publishing: 278]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
57.  Flieger M, Ganswindt U, Schwarz SB, Kreth FW, Tonn JC, la Fougère C, Ertl L, Linn J, Herrlinger U, Belka C, Niyazi M. Re-irradiation and bevacizumab in recurrent high-grade glioma: an effective treatment option. J Neurooncol. 2014;117:337-345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 57]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
58.  Fleischmann DF, Jenn J, Corradini S, Ruf V, Herms J, Forbrig R, Unterrainer M, Thon N, Kreth FW, Belka C, Niyazi M. Bevacizumab reduces toxicity of reirradiation in recurrent high-grade glioma. Radiother Oncol. 2019;138:99-105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 27]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
59.  Schernberg A, Dhermain F, Ammari S, Dumont SN, Domont J, Patrikidou A, Pallud J, Dezamis É, Deutsch É, Louvel G. Reirradiation with concurrent bevacizumab for recurrent high-grade gliomas in adult patients. Cancer Radiother. 2018;22:9-16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
60.  Cohen AL, Anker CJ, Johnson B, Burt LM, Shrieve DC, Salzman K, Jensen R, Boucher K, Colman H. Repeat radiation with bevacizumab and minocycline in bevacizumab-refractory high grade gliomas: a prospective phase 1 trial. J Neurooncol. 2020;148:577-585.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
61.  Sahebjam S, Forsyth PA, Tran ND, Arrington JA, Macaulay R, Etame AB, Walko CM, Boyle T, Peguero EN, Jaglal M, Mokhtari S, Enderling H, Raghunand N, Gatewood T, Long W, Dzierzeski JL, Evernden B, Robinson T, Wicklund MC, Kim S, Thompson ZJ, Chen DT, Chinnaiyan P, Yu HM. Hypofractionated stereotactic re-irradiation with pembrolizumab and bevacizumab in patients with recurrent high-grade gliomas: results from a phase I study. Neuro Oncol. 2021;23:677-686.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 53]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
62.  Straube C, Elpula G, Gempt J, Gerhardt J, Bette S, Zimmer C, Schmidt-Graf F, Meyer B, Combs SE. Re-irradiation after gross total resection of recurrent glioblastoma : Spatial pattern of recurrence and a review of the literature as a basis for target volume definition. Strahlenther Onkol. 2017;193:897-909.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 22]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
63.  Combs SE, Kessel KA, Hesse J, Straube C, Zimmer C, Schmidt-Graf F, Schlegel J, Gempt J, Meyer B. Moving Second Courses of Radiotherapy Forward: Early Re-Irradiation After Surgical Resection for Recurrent Gliomas Improves Efficacy With Excellent Tolerability. Neurosurgery. 2018;83:1241-1248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
64.  Lee J, Ahn SS, Chang JH, Suh CO. Hypofractionated Re-irradiation after Maximal Surgical Resection for Recurrent Glioblastoma: Therapeutic Adequacy and Its Prognosticators of Survival. Yonsei Med J. 2018;59:194-201.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
65.  Chun SJ, Park SH, Park CK, Kim JW, Kim TM, Choi SH, Lee ST, Kim IH. Survival gain with re-Op/RT for recurred high-grade gliomas depends upon risk groups. Radiother Oncol. 2018;128:254-259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
66.  Combs SE, Edler L, Rausch R, Welzel T, Wick W, Debus J. Generation and validation of a prognostic score to predict outcome after re-irradiation of recurrent glioma. Acta Oncol. 2013;52:147-152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 88]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
67.  Kessel KA, Hesse J, Straube C, Zimmer C, Schmidt-Graf F, Schlegel J, Meyer B, Combs SE. Modification and optimization of an established prognostic score after re-irradiation of recurrent glioma. PLoS One. 2017;12:e0180457.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
68.  Patel M, Siddiqui F, Jin JY, Mikkelsen T, Rosenblum M, Movsas B, Ryu S. Salvage reirradiation for recurrent glioblastoma with radiosurgery: radiographic response and improved survival. J Neurooncol. 2009;92:185-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 120]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
69.  McKenzie JT, Guarnaschelli JN, Vagal AS, Warnick RE, Breneman JC. Hypofractionated stereotactic radiotherapy for unifocal and multifocal recurrence of malignant gliomas. J Neurooncol. 2013;113:403-409.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 28]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
70.  Yazici G, Cengiz M, Ozyigit G, Eren G, Yildiz F, Akyol F, Gurkaynak M, Zorlu F. Hypofractionated stereotactic reirradiation for recurrent glioblastoma. J Neurooncol. 2014;120:117-123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 38]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
71.  Shen CJ, Kummerlowe MN, Redmond KJ, Martinez-Gutierrez JC, Usama SM, Holdhoff M, Grossman SA, Laterra JJ, Strowd RE, Kleinberg LR. Re-irradiation for malignant glioma: Toward patient selection and defining treatment parameters for salvage. Adv Radiat Oncol. 2018;3:582-590.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
72.  Watanabe M, Tanaka R, Takeda N. Magnetic resonance imaging and histopathology of cerebral gliomas. Neuroradiology. 1992;34:463-469.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 258]  [Cited by in F6Publishing: 268]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
73.  Li Y, Rey-Dios R, Roberts DW, Valdés PA, Cohen-Gadol AA. Intraoperative fluorescence-guided resection of high-grade gliomas: a comparison of the present techniques and evolution of future strategies. World Neurosurg. 2014;82:175-185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 105]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
74.  Chang PD, Chow DS, Yang PH, Filippi CG, Lignelli A. Predicting Glioblastoma Recurrence by Early Changes in the Apparent Diffusion Coefficient Value and Signal Intensity on FLAIR Images. AJR Am J Roentgenol. 2017;208:57-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 43]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
75.  Batchelor TT, Sorensen AG, di Tomaso E, Zhang WT, Duda DG, Cohen KS, Kozak KR, Cahill DP, Chen PJ, Zhu M, Ancukiewicz M, Mrugala MM, Plotkin S, Drappatz J, Louis DN, Ivy P, Scadden DT, Benner T, Loeffler JS, Wen PY, Jain RK. AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell. 2007;11:83-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1442]  [Cited by in F6Publishing: 1325]  [Article Influence: 77.9]  [Reference Citation Analysis (0)]
76.  Popp I, Bott S, Mix M, Oehlke O, Schimek-Jasch T, Nieder C, Nestle U, Bock M, Yuh WTC, Meyer PT, Weber WA, Urbach H, Mader I, Grosu AL. Diffusion-weighted MRI and ADC versus FET-PET and GdT1w-MRI for gross tumor volume (GTV) delineation in re-irradiation of recurrent glioblastoma. Radiother Oncol. 2019;130:121-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
77.  Oehlke O, Mix M, Graf E, Schimek-Jasch T, Nestle U, Götz I, Schneider-Fuchs S, Weyerbrock A, Mader I, Baumert BG, Short SC, Meyer PT, Weber WA, Grosu AL. Amino-acid PET versus MRI guided re-irradiation in patients with recurrent glioblastoma multiforme (GLIAA) - protocol of a randomized phase II trial (NOA 10/ARO 2013-1). BMC Cancer. 2016;16:769.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 51]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
78.  Fokas E, Wacker U, Gross MW, Henzel M, Encheva E, Engenhart-Cabillic R. Hypofractionated stereotactic reirradiation of recurrent glioblastomas : a beneficial treatment option after high-dose radiotherapy? Strahlenther Onkol. 2009;185:235-240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 102]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
79.  Kim B, Soisson E, Duma C, Chen P, Hafer R, Cox C, Cubellis J, Minion A, Plunkett M, Mackintosh R. Treatment of recurrent high grade gliomas with hypofractionated stereotactic image-guided helical tomotherapy. Clin Neurol Neurosurg. 2011;113:509-512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 15]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
80.  Ogura K, Mizowaki T, Arakawa Y, Ogura M, Sakanaka K, Miyamoto S, Hiraoka M. Initial and cumulative recurrence patterns of glioblastoma after temozolomide-based chemoradiotherapy and salvage treatment: a retrospective cohort study in a single institution. Radiat Oncol. 2013;8:97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 37]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
81.  Combs SE, Bischof M, Welzel T, Hof H, Oertel S, Debus J, Schulz-Ertner D. Radiochemotherapy with temozolomide as re-irradiation using high precision fractionated stereotactic radiotherapy (FSRT) in patients with recurrent gliomas. J Neurooncol. 2008;89:205-210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 61]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
82.  Straube C, Scherb H, Gempt J, Kirschke J, Zimmer C, Schmidt-Graf F, Meyer B, Combs SE. Adjuvant stereotactic fractionated radiotherapy to the resection cavity in recurrent glioblastoma - the GlioCave study (NOA 17 - ARO 2016/3 - DKTK ROG trial). BMC Cancer. 2018;18:15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 21]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
83.  Sminia P, Mayer R. External beam radiotherapy of recurrent glioma: radiation tolerance of the human brain. Cancers (Basel). 2012;4:379-399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 45]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
84.  Mayo C, Yorke E, Merchant TE. Radiation associated brainstem injury. Int J Radiat Oncol Biol Phys. 2010;76:S36-S41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 220]  [Cited by in F6Publishing: 210]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
85.  Mayo C, Martel MK, Marks LB, Flickinger J, Nam J, Kirkpatrick J. Radiation dose-volume effects of optic nerves and chiasm. Int J Radiat Oncol Biol Phys. 2010;76:S28-S35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 358]  [Cited by in F6Publishing: 305]  [Article Influence: 21.8]  [Reference Citation Analysis (0)]
86.  Kirkpatrick JP, van der Kogel AJ, Schultheiss TE. Radiation dose-volume effects in the spinal cord. Int J Radiat Oncol Biol Phys. 2010;76:S42-S49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 354]  [Cited by in F6Publishing: 319]  [Article Influence: 22.8]  [Reference Citation Analysis (0)]
87.  Nieder C, Astner ST, Mehta MP, Grosu AL, Molls M. Improvement, clinical course, and quality of life after palliative radiotherapy for recurrent glioblastoma. Am J Clin Oncol. 2008;31:300-305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 61]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
88.  Wick W, Fricke H, Junge K, Kobyakov G, Martens T, Heese O, Wiestler B, Schliesser MG, Von Deimling A, Pichler J, Vetlova E, Harting I, Debus J, Hartmann C, Kunz C, Platten M, Bendszus M, Combs SE. A phase II, randomized, study of weekly APG101+reirradiation vs reirradiation in progressive glioblastoma. Clin Cancer Res. 2014;20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 97]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
89.  Maitre P, Gupta T, Maitre M, Goda J, Krishnatry R, Chatterjee A, Sridhar E, Sahay A, Mokal S, Moiyadi A, Shetty P, Patil V, Jalali R. Prospective Longitudinal Assessment of Quality of Life and Activities of Daily Living as Patient-Reported Outcome Measures in Recurrent/Progressive Glioma Treated with High-dose Salvage Re-irradiation. Clin Oncol (R Coll Radiol). 2021;33:e155-e165.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
90.  Saeed AM, Khairnar R, Sharma AM, Larson GL, Tsai HK, Wang CJ, Halasz LM, Chinnaiyan P, Vargas CE, Mishra MV. Clinical Outcomes in Patients with Recurrent Glioblastoma Treated with Proton Beam Therapy Reirradiation: Analysis of the Multi-Institutional Proton Collaborative Group Registry. Adv Radiat Oncol. 2020;5:978-983.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
91.  Eberle F, Lautenschläger S, Engenhart-Cabillic R, Jensen AD, Carl B, Stein M, Debus J, Hauswald H. Carbon Ion Beam Reirradiation in Recurrent High-Grade Glioma. Cancer Manag Res. 2020;12:633-639.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
92.  Veldwijk MR, Zhang B, Wenz F, Herskind C. The biological effect of large single doses: a possible role for non-targeted effects in cell inactivation. PLoS One. 2014;9:e84991.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
93.  Herskind C, Wenz F. Radiobiological aspects of intraoperative tumour-bed irradiation with low-energy X-rays (LEX-IORT). Transl Cancer Res. 2014;3:3-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 79]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
94.  Giordano FA, Brehmer S, Mürle B, Welzel G, Sperk E, Keller A, Abo-Madyan Y, Scherzinger E, Clausen S, Schneider F, Herskind C, Glas M, Seiz-Rosenhagen M, Groden C, Hänggi D, Schmiedek P, Emami B, Souhami L, Petrecca K, Wenz F. Intraoperative Radiotherapy in Newly Diagnosed Glioblastoma (INTRAGO): An Open-Label, Dose-Escalation Phase I/II Trial. Neurosurgery. 2019;84:41-49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
95.  Wernicke AG, Taube S, Smith AW, Herskovic A, Parashar B, Schwartz TH. Cs-131 brachytherapy for patients with recurrent glioblastoma combined with bevacizumab avoids radiation necrosis while maintaining local control. Brachytherapy. 2020;19:705-712.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 14]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
96.  Davis ME. Glioblastoma: Overview of Disease and Treatment. Clin J Oncol Nurs. 2016;20:S2-S8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 729]  [Cited by in F6Publishing: 661]  [Article Influence: 82.6]  [Reference Citation Analysis (0)]
97.  Zanello M, Roux A, Ursu R, Peeters S, Bauchet L, Noel G, Guyotat J, Le Reste PJ, Faillot T, Litre F, Desse N, Emery E, Petit A, Peltier J, Voirin J, Caire F, Barat JL, Vignes JR, Menei P, Langlois O, Dezamis E, Carpentier A, Dam Hieu P, Metellus P, Pallud J; On the Behalf of the Club de Neuro-Oncologie of the Société Française de Neurochirurgie. Recurrent glioblastomas in the elderly after maximal first-line treatment: does preserved overall condition warrant a maximal second-line treatment? J Neurooncol. 2017;135:285-297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 28]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
98.  Straube C, Antoni S, Gempt J, Zimmer C, Meyer B, Schlegel J, Schmidt-Graf F, Combs SE. Re-irradiation in elderly patients with glioblastoma: a single institution experience. J Neurooncol. 2019;142:327-335.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 11]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
99.  Lassaletta A, Strother D, Laperriere N, Hukin J, Vanan MI, Goddard K, Lafay-Cousin L, Johnston DL, Zelcer S, Zapotocky M, Rajagopal R, Ramaswamy V, Hawkins C, Tabori U, Huang A, Bartels U, Bouffet E. Reirradiation in patients with diffuse intrinsic pontine gliomas: The Canadian experience. Pediatr Blood Cancer. 2018;65:e26988.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 38]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
100.  Lobon-Iglesias MJ, Giraud G, Castel D, Philippe C, Debily MA, Briandet C, Fouyssac F, de Carli E, Dufour C, Valteau-Couanet D, Sainte-Rose C, Blauwblomme T, Beccaria K, Zerah M, Puget S, Calmon R, Boddaert N, Bolle S, Varlet P, Grill J. Diffuse intrinsic pontine gliomas (DIPG) at recurrence: is there a window to test new therapies in some patients? J Neurooncol. 2018;137:111-118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 10]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
101.  Amsbaugh MJ, Mahajan A, Thall PF, McAleer MF, Paulino AC, Grosshans D, Khatua S, Ketonen L, Fontanilla H, McGovern SL. A Phase 1/2 Trial of Reirradiation for Diffuse Intrinsic Pontine Glioma. Int J Radiat Oncol Biol Phys. 2019;104:144-148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 34]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
102.  Marks LB, Yorke ED, Jackson A, Ten Haken RK, Constine LS, Eisbruch A, Bentzen SM, Nam J, Deasy JO. Use of normal tissue complication probability models in the clinic. Int J Radiat Oncol Biol Phys. 2010;76:S10-S19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1214]  [Cited by in F6Publishing: 1071]  [Article Influence: 76.5]  [Reference Citation Analysis (1)]
103.  Tsang DS, Oliveira C, Bouffet E, Hawkins C, Ramaswamy V, Yee R, Tabori U, Bartels U, Huang A, Millar BA, Crooks B, Bowes L, Zelcer S, Laperriere N. Repeat irradiation for children with supratentorial high-grade glioma. Pediatr Blood Cancer. 2019;66:e27881.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 12]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
104.  Hsieh PC, Chandler JP, Bhangoo S, Panagiotopoulos K, Kalapurakal JA, Marymont MH, Cozzens JW, Levy RM, Salehi S. Adjuvant gamma knife stereotactic radiosurgery at the time of tumor progression potentially improves survival for patients with glioblastoma multiforme. Neurosurgery. 2005;57:684-692.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
105.  Kim HR, Kim KH, Kong DS, Seol HJ, Nam DH, Lim DH, Lee JI. Outcome of salvage treatment for recurrent glioblastoma. J Clin Neurosci. 2015;22:468-473.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
106.  Selch MT, DeSalles AAF, Solberg TD, Wallace RE, Do TM, Ford J, Cabatan-Awang C, Withers HR. Hypofractionated Stereotactic Radiotherapy for Recurrent Malignant Gliomas. J Radiosurgery. 2000;3:3-12.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Vordermark D, Kölbl O, Ruprecht K, Vince GH, Bratengeier K, Flentje M. Hypofractionated stereotactic re-irradiation: treatment option in recurrent malignant glioma. BMC Cancer. 2005;5:55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 131]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
108.  Ernst-Stecken A, Ganslandt O, Lambrecht U, Sauer R, Grabenbauer G. Survival and quality of life after hypofractionated stereotactic radiotherapy for recurrent malignant glioma. J Neurooncol. 2007;81:287-294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 72]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
109.  Miwa K, Matsuo M, Ogawa S, Shinoda J, Yokoyama K, Yamada J, Yano H, Iwama T. Re-irradiation of recurrent glioblastoma multiforme using 11C-methionine PET/CT/MRI image fusion for hypofractionated stereotactic radiotherapy by intensity modulated radiation therapy. Radiat Oncol. 2014;9:181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 51]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
110.  Dincoglan F, Beyzadeoglu M, Sager O, Demiral S, Gamsiz H, Uysal B, Ebruli C, Akin M, Oysul K, Sirin S, Dirican B. Management of patients with recurrent glioblastoma using hypofractionated stereotactic radiotherapy. Tumori. 2015;101:179-184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 43]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
111.  Greenspoon JN, Sharieff W, Hirte H, Overholt A, Devillers R, Gunnarsson T, Whitton A. Fractionated stereotactic radiosurgery with concurrent temozolomide chemotherapy for locally recurrent glioblastoma multiforme: a prospective cohort study. Onco Targets Ther. 2014;7:485-490.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 42]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
112.  Shapiro LQ, Beal K, Goenka A, Karimi S, Iwamoto FM, Yamada Y, Zhang Z, Lassman AB, Abrey LE, Gutin PH. Patterns of failure after concurrent bevacizumab and hypofractionated stereotactic radiation therapy for recurrent high-grade glioma. Int J Radiat Oncol Biol Phys. 2013;85:636-642.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 50]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
113.  Cabrera AR, Cuneo KC, Desjardins A, Sampson JH, McSherry F, Herndon JE 2nd, Peters KB, Allen K, Hoang JK, Chang Z, Craciunescu O, Vredenburgh JJ, Friedman HS, Kirkpatrick JP. Concurrent stereotactic radiosurgery and bevacizumab in recurrent malignant gliomas: a prospective trial. Int J Radiat Oncol Biol Phys. 2013;86:873-879.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 63]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]