Open access peer-reviewed chapter

Stereotactic Radiosurgery for Recurrent Glioblastoma Multiforme

Written By

Cheng-Ta Hsieh and Da-Tong Ju

Submitted: 21 September 2019 Reviewed: 13 January 2020 Published: 20 February 2020

DOI: 10.5772/intechopen.91163

From the Edited Volume

Tumor Progression and Metastasis

Edited by Ahmed Lasfar and Karine Cohen-Solal

Chapter metrics overview

997 Chapter Downloads

View Full Metrics

Abstract

Glioblastoma multiforme (GBM) is the most aggressive intracranial tumor that primarily affects adults. Since the introduction of temozolomide in 2005, maximal resection surgery with concurrent chemoradiation has become the standard treatment method for patients with newly diagnosed GBM. Although newly discovered chemoagents have been demonstrated to improve the median survival time, GBM still recurs in most patients. Recurrent GBM is still a therapeutic challenge for clinical physicians. Surgical intervention and other conventional chemoagents have been applied to manage recurrent GBM. Stereotactic radiosurgery (SRS) provides a highly precise radiation dose to the tumor lesion and reduces the dose to the adjacent normal brain tissue. After standard treatment for newly diagnosed GBM is completed, conventional re-irradiation therapy is not suitable for patients with recurrent GBMs. Therefore, SRS may become an alternative option in the treatment of recurrent GBMs. In this review, we discuss the relevant literature regarding SRS for recurrent GBMs and provide treatment advice for clinical physicians.

Keywords

  • stereotactic radiosurgery
  • recurrent glioblastoma multiforme
  • re-irradiation
  • survival time
  • prognosis

1. Introduction

Glioblastoma multiforme (GBM) is the most common primary brain neoplasm in adults [1, 2]. There are 1.6 times more males than females who develop a GBM [1]. According to the latest statistical report of the Central Brain Tumor Registry of the USA, the annual incidence of GBM has been estimated at approximately 3.22 cases per 100,000 people, and the median age is 65 years [3]. The current standard treatment of patients with a newly diagnosed GBM was established in 2005, and it consists of maximal surgical resection of the tumor followed by chemotherapy and conventional radiotherapy [4, 5]. Despite this therapy, the median overall survival time is approximately 15–17 months [2].

GBM is a refractory malignant and infiltrating tumor that may recur any time after initial multimodal treatments are completed [6]. Managing recurrent GBM has always been challenging, and a balance has to be achieved between significant treatment toxicity and associated morbidities and mortalities [6, 7]. Reoperation with maximal resection at recurrence remains as an independent predictor to improve overall survival [8, 9]. However, repeat gross-total resection may not be easily achieved when recurrent GBM involves important eloquent brain structures, such as the brainstem or motor area. Extensive bevacizumab and temozolomide are the two main FDA-approved chemoagents used to treat patients with recurrent GBMs, but the prognosis remains poor [10].

Re-irradiation is an alternative option for managing recurrent GBMs [11]. The majority of recurrent tumors occur at the initial or adjacent regional sites [12]. Because a total dose of 60 Gy in 30 fractions has been prescribed for initial radiation therapy, re-irradiation with further dose escalation appears to produce more significant toxicity [10, 11]. Stereotactic radiosurgery (SRS) is a noninvasive treatment that provides a highly precise, targeted additional radiation boost to the tumor lesion, and it maintains an acceptable rate of adverse radiation effects while reducing the dose to adjacent normal brain tissues [13]. In this review, we searched the relevant literature and investigated the role of SRS in the management of recurrent GBMs. Our results may provide treatment information for clinical treatment.

Advertisement

2. Search methodology

In this study, different combinations of the keywords “recurrent glioblastoma multiple,” “high-grade glioma,” “stereotactic radiosurgery,” and “re-irradiation” were used to search the published literature in the PubMed database until October 31, 2019. The inclusion criteria of the study were (1) patients with recurrent GBMs, (2) treatment with stereotactic radiosurgery (SRS) or a fractionated radiosurgery (less than 5 fractions), and (3) outcomes with overall survival time. Tumor progression was also accepted as a recurrent disease. Potentially relevant studies were identified from the reference lists of the studies obtained from the database search. Articles excluded from the review were those written in languages other than English and those that lacked survival response data. Finally, a total of 49 studies were included in this review.

Advertisement

3. The summary of patients with recurrent GBMs treated with SRS

A total of 49 studies published from 1994 to 2019 were enrolled in this review, as summarized in Table 1 [13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61]. There were 6 prospective studies and 43 retrospective studies. About 2066 patients with recurrent glioblastomas treated with SRS, including linear accelerator (LINAC) radiosurgery, Gamma Knife radiosurgery, and Cyberknife radiosurgery, are reported. In all studies, the median age of the patients who received SRS treatment for recurrent GBM ranged from 34 to 62 years. The majority of patients were males. The median prescribed dose of SRS ranged from 6 to 30 Gy. The median targeted volume for treatment ranged from 1.35 to 21.3 cc. The overall survival time from treatment for SRS ranged from 3.9 to 17.9 months, where the progression-free survival time from the treatment of SRS ranged from 2.1 to 14.9 months. In the prognostic analysis of survival time in patients with recurrent GBMs treated with SRS, a small tumor volume, younger age, higher Karnofsky performance scale (KPS) score, lower recursive partitioning analysis (RPA) class, adjuvant bevacizumab, methylated O6-methylguanine-DNA-methyltransferase (MGMT) promoter, and longer interval between the original surgery and SRS were significantly associated with patients’ survival outcomes.

Table 1.

Summary of the published literature on stereotactic radiosurgery in patients with recurrent glioblastoma multiforme.

3.1 The effect of LINAC radiosurgery in patients with recurrent GBMs

From 1994 to 2018, a total of 501 patients with recurrent GBMs treated with LINAC SRS were enrolled in 22 studies, including 3 prospective trials and 17 retrospective studies [14, 15, 16, 19, 21, 23, 24, 25, 26, 28, 32, 33, 35, 39, 40, 43, 49, 51, 54, 56, 60, 61]. The median age ranged from 34 to 54 years. The median prescribed dose ranged from 13 to 30 Gy. The median targeted tumor volume was 4.5 to 41.3 cc. The median overall survival time from the treatment of SRS ranged from 3.9 to 14.4 months, whereas the median progression-free survival time was 2.1 to 11 months.

The first study about LINAC radiosurgery for recurrent GBMs was described by Chamerian et al. [14]. The median prescribed dose was 13.4 Gy, and the median treated tumor volume was 17 cc. The median overall survival time was only 8 months, whereas the median progression-free survival time was 4 months. After that, only one retrospective study of more than 100 patients with recurrent high-grade gliomas treated with LINAC SRS has been reported [16]. Shrieve et al. showed that the median survival time of 72 recurrent GBM patients was 10.2 months [16]. Younger age (less than 46 years) and small tumor volume (less than 10.1 cc) were the significant prognostic factors associated with survival time. There were two studies that enrolled patients with only recurrent GBMs [21, 33]. In 2005, Combs et al. reported 32 patients, including 19 males and 13 females with recurrent GBMs treated with LINAC SRS [21]. The median age was 56 years, ranging from 33 to 76 years. The median prescribed radiation dose was 15 Gy, ranging from 10 to 20 Gy. The median targeted tumor volume was 10 cc with a range of 1.2 to 59.2 cc. The median overall survival time and progression-free survival time were 10 and 5 months, respectively. However, no prognostic factor was significant enough to influence the survival time. In a retrospective study of 19 patients with recurrent GBMs, Sirin et al. also showed that the median overall survival time and progression-free survival time were only 9.3 and 5.7 months, respectively [33]. In the bevacizumab era, three studies reported the combination of LINAC SRS, and bevacizumab improved the overall survival time ranging from 11.2 to 14.4 months [35, 39, 40]. In a retrospective study of 48 patients with recurrent GBMs, Cuneo et al. reported that the median progression-free survival time in recurrent patients who received adjuvant bevacizumab and LINAC SRS was 5.2 months vs. 2.1 months for patients who received LINAC SRS alone. The median overall survival times for patients who received a combination of adjuvant bevacizumab/LINAC SRS and LINAC SRS alone were 11.2 and 3.9 months, respectively. The authors concluded that the combination of salvage radiosurgery and bevacizumab to treat recurrent malignant gliomas seemed to be associated with improved outcomes. Younger age and higher KPS were still significant prognostic factors associated with overall survival time in patients with recurrent GBMs.

3.2 The effect of gamma knife radiosurgery in patients with recurrent GBMs

From 1996 to 2019, a total of 1247 patients with recurrent GBMs in 23 published studies were treated with Gamma Knife SRS [13, 17, 18, 20, 22, 24, 27, 29, 31, 36, 37, 38, 41, 45, 47, 50, 52, 53, 55, 57, 58, 59, 61]. The median age ranged from 43 to 61 years. The median prescribed marginal dose varied from 6 to 20 Gy. The median targeted tumor volume ranged from 1.35 to 21.4 cc. The median overall survival time ranged from 7 to 30 months, whereas the median progression-free survival time ranged from 3.8 to 14.9 months.

In a retrospective study of 189 patients with recurrent high-grade gliomas treated with Gamma Knife SRS, Larson et al. first reported that the median overall survival time in 66 patients with recurrent GBMs was 10 months [17]. Younger age, smaller tumor volume, higher KPS, and unifocal tumors were significant prognostic factors associated with patients’ overall survival times. Several studies reported the impact of a combination of Gamma Knife SRS and adjuvant chemoagents on overall survival times [17, 37, 47, 55, 59]. In 2002, Larson et al. reported a prospective phase II study on patients who received a combination of Gamma Knife SRS, and marimastat had a median overall survival of 8.7 months, whereas the median survival time in patients who received only Gamma Knife SRS alone was 10.1 months. Marimastat did not offer an advantage for patients with recurrent GBMs. However, in a retrospective study of 57 patients with recurrent GBMs, Kim et al. showed that the combination of adjuvant temozolomide and Gamma Knife SRS significantly improved the medium overall survival time from 9.2 to 15.5 months [47]. In the bevacizumab era, two studies reported that the median survival time was approximately 13 months after the combined treatment of Gamma Knife SRS and adjuvant bevacizumab [55, 59].

3.3 The effect of Cyberknife radiosurgery in patients with recurrent GBMs

From 2009 to 2017, a total of 318 patients with recurrent GBMs in eight published studies were treated with Cyberknife SRS [30, 34, 42, 44, 46, 48, 51, 54]. The median age ranged from 37 to 59.9 years. The median prescribed marginal dose ranged from 15 to 30 Gy. The median targeted tumor volume ranged from 2 to 24 cc. The median overall survival time after Cyberknife SRS ranged from 5.3 to 12 months, whereas the median progression-free survival time ranged from 4 to 7.9 months.

The first report on Cyberknife SRS for patients with recurrent GBMs was published by Villavicencio et al. [30]. The median overall survival time in a total of 26 patients with recurrent GBMs was 7 months. No prognostic factor associated with the overall survival time was identified. In 2015, Pinzi et al. reported a retrospective study of more than 100 patients who had recurrent high-grade glioma treated with Cyberknife SRS [48]. Among 88 patients with recurrent GBMs, the median survival time was 10 months after treatment with Cyberknife SRS. Adjuvant second-line chemotherapy and/or surgery were the significant prognostic factors associated with overall survival times. The effect of adjuvant chemoagents, including bevacizumab, temozolomide, and anti-epidermal factor (125)-mAB 425, on the overall survival times was evaluated in four studies [34, 42, 44, 46]. In 2012, Conti et al. compared the effect of a combination of temozolomide and Cyberknife SRS with that of Cyberknife SRS alone on the overall survival times of patients with recurrent GBMs [34]. The progression-free survival time and median survival time in patients who received the adjuvant temozolomide and Cyberknife SRS were 7 and 12 months, respectively. The patients who received Cyberknife SRS alone had a progression-free survival time and median survival time of only 4 and 7 months, respectively. In the bevacizumab era, Yazici et al. revealed that the median survival time in 37 patients with recurrent GBMs was 10.6 months, whereas the median progression-free survival time was 7.9 months [44]. A tumor volume less than 24 cc was the only significant prognostic factor associated with overall survival times.

Advertisement

4. Discussion

4.1 The role of SRS for recurrent GBMs

GBM is an incurable disease with local progression in the majority of patients. The management of recurrent GBMs is a clinically challenging problem, and treatment options are limited [7, 10]. Although reoperation with gross-total removal of the tumor has been shown to improve the overall survival time in patients with recurrent GBMs, surgery may not be preferred for patients with tumors in the eloquent area, older age, or lower performance status [8]. Re-irradiation offers an alternative option for treating recurrent GBMs. In a systematic review and meta-analysis of re-irradiation with external beam radiotherapy for recurrent GBMs, Kazmi et al. showed that the 6- and 12-month overall survival times from the time of re-irradiation were 70 and 34%, respectively, whereas the 6- and 12-month progression-free survival times were 40 and 16%, respectively [11]. The overall toxicity rate was low, ranging from 4 to 10%.

SRS has the ability to combine surgical and radio-oncological treatments to deliver a high dose of focused radiation on the focal tumor lesion and spare the adjacent normal anatomical structures. For recurrent GBMs, the majority of tumors tend to grow within 2 cm of the contrast-enhancing lesion border, and SRS seems to be a reasonable tool to add radiation boost for the focal lesion followed by the standard treatment of initial radiation with 60 Gy in 30 fractions [4, 13]. In our present review, despite the different SRS modalities with the median prescribed dose ranging from 6 to 30 Gy, the overall survival time from the treatment of SRS ranged from 3.9 to 17.9 months, where the progression-free survival time from the treatment of SRS ranged from 2.1 to 14.9 months. Severe prognostic factors, such as small tumor volume, younger age, higher KPS score, and lower RPA class, were mostly suggested to be significantly associated with the overall survival time in patients with recurrent GBMs treated with SRS. These results showed that re-irradiation with the SRS modality are an alternative and feasible method to manage patients with recurrent GBMs.

4.2 The impact of SRS and adjuvant temozolomide for recurrent GBMs

Since 2005, temozolomide, which is an alkylating agent, is the most important FDA-approved chemoagent for the standard treatment of patients with newly diagnosed GBMs [1, 4]. The median overall survival time significantly improved from 12.1 to 14.6 months after the patients with newly diagnosed GBMs received combined treatments with radiotherapy and adjuvant temozolomide. However, the disease frequently progresses within 6–9 months, and the 2-year survival rate is less than 25% [62]. The failure of temozolomide treatment has been found to be associated with the expression of MGMT protein [63, 64, 65]. Among the GBM patients with a methylated MGMT promoter, the median overall survival time was 21.7 months after treatment with radiotherapy and temozolomide, whereas the median survival time was 15.3 months in the unmethylated group treated with radiotherapy alone [64].

Due to the blood-brain barrier, temozolomide rechallenge is considered to be a reasonable option in patients with recurrent GBMs. In this review, the combination of SRS and temozolomide was employed in three studies [34, 42, 47]. Cyberknife SRS was performed in two studies, and the other study used the Gamma Knife SRS. The median overall survival time ranged from 9 to 15.5 months, and the median progression-free survival time was approximately 7 months after the time of SRS treatment. In 2012, Conti et al. analyzed the effect of adjuvant temozolomide in recurrent GBM patients treated with Cyberknife SRS [34]. The median overall survival time significantly improved from 7 to 12 months, whereas the median progression-free survival time improved from 4 to 7 months. Based on 57 recurrent GBM patients, Kim et al. also showed that the improved median overall survival time and progression-free survival time were 15.5 and 6 months, respectively [47]. Otherwise, in a retrospective review of 61 patients who received Gamma Knife SRS as a salvage treatment at the time of the first progression, Kim et al. showed that the median overall survival time was 14 months in the methylated MGMT promoter group and 9 months in the unmethylated group [53]. Methylation of the MGMT promoter was significantly corrected with better overall survival times and progression-free survival times. The results mentioned above indicated that the combination of salvage SRS and adjuvant temozolomide may offer an important treatment option to improve the overall survival times in patients with recurrent GBMs.

4.3 The impact of SRS and adjuvant bevacizumab for recurrent GBMs

Bevacizumab is a recombinant human monoclonal antibody that acts against the vascular endothelial growth factor to prevent the growth and maintenance of tumor blood vessels. In 2009, bevacizumab was approved by the USFDA for the treatment of patients with recurrent GBMs [66, 67]. The use of bevacizumab demonstrated a radiological response of up to 40% [68]. However, in a large prospective phase III trial, the use of adjuvant bevacizumab revealed only improvement in the progression-free survival times from 1.5 to 4.2 months but not in the overall survival times [69]. In a systematic review and meta-analysis, Diaz et al. showed that the survival advantage of bevacizumab at recurrence was limited to 4 months [70]. Although bevacizumab may reduce steroid requirements, there was no additional benefit in the health-related quality of life. The role of bevacizumab in combination with other cytotoxic chemoagents remains unclear.

The role of adjuvant bevacizumab in patients with recurrent GBMs treated with SRS has been reported in nine studies, which were included in our review [35, 37, 39, 40, 44, 46, 49, 55, 59]. The median overall survival time ranged from 5.3 to 17.9 months, whereas the median progression-free survival time ranged from 3.9 to 14.9 months. The comparison of SRA with or without adjuvant bevacizumab was investigated in two studies [35, 37]. Among 49 patients with recurrent GBMs, Cuneo et al. showed that the median overall survival time was 11.2 months in patients receiving SRS and adjuvant bevacizumab and 3.9 months in patients receiving SRS therapy alone [35]. The progression-free survival time also improved from 2.1 to 5.2 months. In a case-controlled study of patients with recurrent GBMs treated with SRS and adjuvant bevacizumab plus temozolomide or irinotecan, Park et al. also showed that the median overall survival time and progression-free survival time improved from 12.2 to 17.9 months and 6.7 to 14.9 months, respectively [37]. In a retrospective study and review of the literature, Morris et al. reported that the dual role of bevacizumab and radiosurgery had a benefit in the overall survival times (11.2–17.9 months) and progression-free survival times (3.9–14.9 months). These results showed the potential therapeutic effect of adjuvant bevacizumab in combination with other treatment modalities, such as cytotoxic chemoagents or salvage SRS, in patients with recurrent GBMs.

4.4 The future of SRS for recurrent GBMs

With the advance of molecular diagnostic techniques, newly diagnosed GBMs should be classified based on the mutant status of isocitrate dehydrogenase 1 defined by the updated guidelines of the World Health Organization in 2016 [71]. These molecular profiles influence the overall survival time and the possible therapeutic effects of chemoagents. Similar to the recurrent GBMs, several main molecules, such as MLH1 [72], CASP8 [73], MSH2 [74], and P53 [74], were found to be different from primary GBMs [75]. The molecular features, intra-tumor heterogeneity, immunogenicity, and microenvironment around the tumor contribute to the clinical prognostic outcomes in patients with recurrent GBMs [7, 10]. Reoperation, re-chemotherapy, and re-irradiation currently remain as the standard treatments for most patients with recurrent GBMs [2, 7, 10, 11]. A growing body of literature, including our current review, demonstrates the tolerability and efficacy of salvage SRS for recurrent GBMs, which did not inhibit re-irradiation, followed by a total of 60 Gy typically applied in the first-line treatment [59]. Although younger age is commonly considered as an important independent prognostic factor that is associated with survival, the selected criteria of salvage SRS for better outcomes need to be investigated in further large prospective studies. In the future, individualized precise multi-modality treatment will play an important role in patients with recurrent GBMs, including the combination of cytotoxic chemotherapy, angiogenesis inhibitors, or immunotherapy [76]. Salvage SRS with a combination of other treatment modalities may offer an alternative therapeutic method to manage patients with recurrent GBMs.

Advertisement

5. Conclusion

Our review suggests that salvage SRS is an important treatment protocol for managing patients with recurrent GBMs. The irradiation doses provided by SRS may improve the clinical outcome of patients with recurrent GBMs, which is not hampered by the standard case of 60 Gy prescribed for newly diagnosed GBMs. The dual role of salvage SRS and other cytotoxic chemoagents, such as temozolomide and bevacizumab, also seems to be effective in the management of recurrent GBMs. Further application of salvage SRS combined with other chemoagents or a new treatment modality needs to be investigated.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Preusser M, de Ribaupierre S, Wöhrer A, Erridge SC, Hegi M, Weller M, et al. Current concepts and management of glioblastoma. Annals of Neurology. 2011;70(1):9-21
  2. 2. Weller M, Cloughesy T, Perry JR, Wick W. Standards of care for treatment of recurrent glioblastoma—Are we there yet? Neuro-Oncology. 2013;15(1):4-27
  3. 3. Ostrom QT, Cioffi G, Gittleman H, Patil N, Waite K, Kruchko C, et al. CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2012–2016. Neuro-Oncology. 2019;21(Suppl. 5):v1-v100
  4. 4. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. The New England Journal of Medicine. 2005;352(10):987-996
  5. 5. Sanai N, Polley MY, McDermott MW, Parsa AT, Berger MS. An extent of resection threshold for newly diagnosed glioblastomas. Journal of Neurosurgery. 2011;115(1):3-8
  6. 6. Hou LC, Veeravagu A, Hsu AR, Tse VC. Recurrent glioblastoma multiforme: A review of natural history and management options. Neurosurgical Focus. 2006;20(4):E5
  7. 7. Mallick S, Benson R, Hakim A, Rath GK. Management of glioblastoma after recurrence: A changing paradigm. Journal of the Egyptian National Cancer Institute. 2016;28(4):199-210
  8. 8. Bloch O, Han SJ, Cha S, Sun MZ, Aghi MK, McDermott MW, et al. Impact of extent of resection for recurrent glioblastoma on overall survival: Clinical article. Journal of Neurosurgery. 2012;117(6):1032-1038
  9. 9. Lu VM, Goyal A, Graffeo CS, Perry A, Burns TC, Parney IF, et al. Survival benefit of maximal resection for glioblastoma reoperation in the temozolomide era: A meta-analysis. World Neurosurgery. 2019;127:31-37
  10. 10. Chaul-Barbosa C, Marques DF. How we treat recurrent glioblastoma today and current evidence. Current Oncology Reports. 2019;21(10):94
  11. 11. Kazmi F, Soon YY, Leong YH, Koh WY, Vellayappan B. Re-irradiation for recurrent glioblastoma (GBM): A systematic review and meta-analysis. Journal of Neuro-Oncology. 2019;142(1):79-90
  12. 12. Jayamanne D, Wheeler H, Brazier D, Newey A, Kastelan M, Guo L, et al. Predicting patterns of failure in temporal lobe GBMs: Possible implications on radiotherapy treatment portals. Radiotherapy and Oncology. 2018;13(1):133
  13. 13. Niranjan A, Kano H, Iyer A, Kondziolka D, Flickinger JC, Lunsford LD. Role of adjuvant or salvage radiosurgery in the management of unresected residual or progressive glioblastoma multiforme in the pre-bevacizumab era. Journal of Neurosurgery. 2015;122(4):757-765
  14. 14. Chamberlain MC, Barba D, Kormanik P, Shea WM. Stereotactic radiosurgery for recurrent gliomas. Cancer. 1994;74(4):1342-1347
  15. 15. Hall WA, Djalilian HR, Sperduto PW, Cho KH, Gerbi BJ, Gibbons JP, et al. Stereotactic radiosurgery for recurrent malignant gliomas. Journal of Clinical Oncology. 1995;13(7):1642-1648
  16. 16. Shrieve DC, Alexander E 3rd, Wen PY, Fine HA, Kooy HM, Black PM, et al. Comparison of stereotactic radiosurgery and brachytherapy in the treatment of recurrent glioblastoma multiforme. Neurosurgery. 1995;36(2):275-282 discussion 82-4
  17. 17. Larson DA, Gutin PH, McDermott M, Lamborn K, Sneed PK, Wara WM, et al. Gamma knife for glioma: Selection factors and survival. International Journal of Radiation Oncology, Biology, Physics. 1996;36(5):1045-1053
  18. 18. Kondziolka D, Flickinger JC, Bissonette DJ, Bozik M, Lunsford LD. Survival benefit of stereotactic radiosurgery for patients with malignant glial neoplasms. Neurosurgery. 1997;41(4):776-783 discussion 83-5
  19. 19. Cho KH, Hall WA, Gerbi BJ, Higgins PD, McGuire WA, Clark HB. Single dose versus fractionated stereotactic radiotherapy for recurrent high-grade gliomas. International Journal of Radiation Oncology, Biology, Physics. 1999;45(5):1133-1141
  20. 20. Larson DA, Prados M, Lamborn KR, Smith V, Sneed PK, Chang S, et al. Phase II study of high central dose Gamma knife radiosurgery and marimastat in patients with recurrent malignant glioma. International Journal of Radiation Oncology, Biology, Physics. 2002;54(5):1397-1404
  21. 21. 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(10):2168-2173
  22. 22. Hsieh PC, Chandler JP, Bhangoo S, Panagiotopoulos K, Kalapurakal JA, Marymont MH, et al. Adjuvant gamma knife stereotactic radiosurgery at the time of tumor progression potentially improves survival for patients with glioblastoma multiforme. Neurosurgery. 2005;57(4):684-692 discussion 684
  23. 23. Mahajan A, McCutcheon IE, Suki D, Chang EL, Hassenbusch SJ, Weinberg JS, et al. Case-control study of stereotactic radiosurgery for recurrent glioblastoma multiforme. Journal of Neurosurgery. 2005;103(2):210-217
  24. 24. 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(9):2046-2051
  25. 25. Schwer AL, Damek DM, Kavanagh BD, Gaspar LE, Lillehei K, Stuhr K, et al. A phase I dose-escalation study of fractionated stereotactic radiosurgery in combination with gefitinib in patients with recurrent malignant gliomas. International Journal of Radiation Oncology, Biology, Physics. 2008;70(4):993-1001
  26. 26. Biswas T, Okunieff P, Schell MC, Smudzin T, Pilcher WH, Bakos RS, et al. Stereotactic radiosurgery for glioblastoma: Retrospective analysis. Radiotherapy and Oncology. 2009;4:11
  27. 27. Kida Y, Yoshimoto M, Hasegawa T. Radiosurgery for intracranial gliomas. Progress in Neurological Surgery. 2009;22:122-128
  28. 28. Patel M, Siddiqui F, Jin JY, Mikkelsen T, Rosenblum M, Movsas B, et al. Salvage reirradiation for recurrent glioblastoma with radiosurgery: Radiographic response and improved survival. Journal of Neuro-Oncology. 2009;92(2):185-191
  29. 29. Pouratian N, Crowley RW, Sherman JH, Jagannathan J, Sheehan JP. Gamma knife radiosurgery after radiation therapy as an adjunctive treatment for glioblastoma. Journal of Neuro-Oncology. 2009;94(3):409-418
  30. 30. Villavicencio AT, Burneikiene S, Romanelli P, Fariselli L, McNeely L, Lipani JD, et al. Survival following stereotactic radiosurgery for newly diagnosed and recurrent glioblastoma multiforme: A multicenter experience. Neurosurgical Review. 2009;32(4):417-424
  31. 31. Elliott RE, Parker EC, Rush SC, Kalhorn SP, Moshel YA, Narayana A, et al. Efficacy of gamma knife radiosurgery for small-volume recurrent malignant gliomas after initial radical resection. World Neurosurgery. 2011;76(1–2):128-140 discussion 61-2
  32. 32. Maranzano E, Anselmo P, Casale M, Trippa F, Carletti S, Principi M, et al. Treatment of recurrent glioblastoma with stereotactic radiotherapy: Long-term results of a mono-institutional trial. Tumori. 2011;97(1):56-61
  33. 33. Sirin S, Oysul K, Surenkok S, Sager O, Dincoglan F, Dirican B, et al. Linear accelerator-based stereotactic radiosurgery in recurrent glioblastoma: A single center experience. Vojnosanitetski Pregled. 2011;68(11):961-966
  34. 34. Conti A, Pontoriero A, Arpa D, Siragusa C, Tomasello C, Romanelli P, et al. Efficacy and toxicity of Cyberknife re-irradiation and “dose dense” temozolomide for recurrent gliomas. Acta Neurochirurgica. 2012;154(2):203-209
  35. 35. Cuneo KC, Vredenburgh JJ, Sampson JH, Reardon DA, Desjardins A, Peters KB, et al. Safety and efficacy of stereotactic radiosurgery and adjuvant bevacizumab in patients with recurrent malignant gliomas. International Journal of Radiation Oncology, Biology, Physics. 2012;82(5):2018-2024
  36. 36. Koga T, Maruyama K, Tanaka M, Ino Y, Saito N, Nakagawa K, et al. Extended field stereotactic radiosurgery for recurrent glioblastoma. Cancer. 2012;118(17):4193-4200
  37. 37. Park KJ, Kano H, Iyer A, Liu X, Niranjan A, Flickinger JC, et al. Salvage gamma knife stereotactic radiosurgery followed by bevacizumab for recurrent glioblastoma multiforme: A case-control study. Journal of Neuro-Oncology. 2012;107(2):323-333
  38. 38. Skeie BS, Enger PØ, Brøgger J, Ganz JC, Thorsen F, Heggdal JI, et al. Gamma knife surgery versus reoperation for recurrent glioblastoma multiforme. World Neurosurgery. 2012;78(6):658-669
  39. 39. Cabrera AR, Cuneo KC, Desjardins A, Sampson JH, McSherry F, Herndon JE 2nd, et al. Concurrent stereotactic radiosurgery and bevacizumab in recurrent malignant gliomas: A prospective trial. International Journal of Radiation Oncology, Biology, Physics. 2013;86(5):873-879
  40. 40. Clark GM, McDonald AM, Nabors LB, Fathalla-Shaykh H, Han X, Willey CD, et al. Hypofractionated stereotactic radiosurgery with concurrent bevacizumab for recurrent malignant gliomas: The University of Alabama at Birmingham experience. Neurooncology Practice. 2014;1(4):172-177
  41. 41. Dodoo E, Huffmann B, Peredo I, Grinaker H, Sinclair G, Machinis T, et al. Increased survival using delayed gamma knife radiosurgery for recurrent high-grade glioma: A feasibility study. World Neurosurgery. 2014;82(5):e623-e632
  42. 42. Greenspoon JN, Sharieff W, Hirte H, Overholt A, Devillers R, Gunnarsson T, et al. Fractionated stereotactic radiosurgery with concurrent temozolomide chemotherapy for locally recurrent glioblastoma multiforme: A prospective cohort study. OncoTargets and Therapy. 2014;7:485-490
  43. 43. Martínez-Carrillo M, Tovar-Martín I, Zurita-Herrera M, Del Moral-Ávila R, Guerrero-Tejada R, Saura-Rojas E, et al. Salvage radiosurgery for selected patients with recurrent malignant gliomas. BioMed Research International. 2014;2014:657953
  44. 44. Yazici G, Cengiz M, Ozyigit G, Eren G, Yildiz F, Akyol F, et al. Hypofractionated stereotactic reirradiation for recurrent glioblastoma. Journal of Neuro-Oncology. 2014;120(1):117-123
  45. 45. Bir SC, Connor DE Jr, Ambekar S, Wilden JA, Nanda A. Factors predictive of improved overall survival following stereotactic radiosurgery for recurrent glioblastoma. Neurosurgical Review. 2015;38(4):705-713
  46. 46. Hasan S, Chen E, Lanciano R, Yang J, Hanlon A, Lamond J, et al. Salvage fractionated stereotactic radiotherapy with or without chemotherapy and immunotherapy for recurrent glioblastoma multiforme: A single institution experience. Frontiers in Oncology. 2015;5:106
  47. 47. Kim HR, Kim KH, Kong DS, Seol HJ, Nam DH, Lim DH, et al. Outcome of salvage treatment for recurrent glioblastoma. Journal of Clinical Neuroscience. 2015;22(3):468-473
  48. 48. Pinzi V, Orsi C, Marchetti M, Milanesi IM, Bianchi LC, DiMeco F, et al. Radiosurgery reirradiation for high-grade glioma recurrence: A retrospective analysis. Neurological Sciences. 2015;36(8):1431-1440
  49. 49. Bokstein F, Blumenthal DT, Corn BW, Gez E, Matceyevsky D, Shtraus N, et al. Stereotactic radiosurgery (SRS) in high-grade glioma: Judicious selection of small target volumes improves results. Journal of Neuro-Oncology. 2016;126(3):551-557
  50. 50. Frischer JM, Marosi C, Woehrer A, Hainfellner JA, Dieckmann KU, Eiter H, et al. Gamma knife radiosurgery in recurrent glioblastoma. Stereotactic and Functional Neurosurgery. 2016;94(4):265-272
  51. 51. Holt DE, Bernard ME, Quan K, Clump DA, Engh JA, Burton SA, et al. Salvage stereotactic radiosurgery for recurrent glioblastoma multiforme with prior radiation therapy. Journal of Cancer Research and Therapeutics. 2016;12(4):1243-1248
  52. 52. Imber BS, Kanungo I, Braunstein S, Barani IJ, Fogh SE, Nakamura JL, et al. Indications and efficacy of gamma knife stereotactic radiosurgery for recurrent glioblastoma: 2 decades of institutional experience. Neurosurgery. 2017;80(1):129-139
  53. 53. Kim BS, Kong DS, Seol HJ, Nam DH, Lee JI. MGMT promoter methylation status as a prognostic factor for the outcome of gamma knife radiosurgery for recurrent glioblastoma. Journal of Neuro-Oncology. 2017;133(3):615-622
  54. 54. Sutera PA, Bernard ME, Gill BS, Quan K, Engh JA, Burton SA, et al. Salvage stereotactic radiosurgery for recurrent gliomas with prior radiation therapy. Future Oncology. 2017;13(29):2681-2690
  55. 55. Abbassy M, Missios S, Barnett GH, Brewer C, Peereboom DM, Ahluwalia M, et al. Phase I trial of radiosurgery dose escalation plus bevacizumab in patients with recurrent/progressive glioblastoma. Neurosurgery. 2018;83(3):385-392
  56. 56. Gigliotti MJ, Hasan S, Karlovits SM, Ranjan T, Wegner RE. Re-irradiation with stereotactic radiosurgery/radiotherapy for recurrent high-grade gliomas: Improved survival in the modern era. Stereotactic and Functional Neurosurgery. 2018;96(5):289-295
  57. 57. Guseynova K, Liscak R, Simonova G, Novotny J Jr. Gamma knife radiosurgery for local recurrence of glioblastoma. Neuro Endocrinology Letters. 2018;39(4):281-287
  58. 58. Sharma M, Schroeder JL, Elson P, Meola A, Barnett GH, Vogelbaum MA, et al. Outcomes and prognostic stratification of patients with recurrent glioblastoma treated with salvage stereotactic radiosurgery. Journal of Neurosurgery. 2018:1-11
  59. 59. Morris SL, Zhu P, Rao M, Martir M, Zhu JJ, Hsu S, et al. Gamma knife stereotactic radiosurgery in combination with bevacizumab for recurrent glioblastoma. World Neurosurgery. 2019;127:e523-e533
  60. 60. Sanghavi S, Skrupky R, Badie B, Robins I, Tome W, Mehta MP. Recurrent malignant gliomas treated with radiosurgery. Journal of Radiosurgery. 1999;2(3):119-125
  61. 61. Park JL, Suh JH, Barnett GH, Reddy CA, Peereboom DM, Stevens GHJ, et al. Survival after stereotactic radiosurgery for recurrent glioblastoma multiforme. Journal of Radiosurgery. 2000;3(4):169-175
  62. 62. Wen PY, Kesari S. Malignant gliomas in adults. The New England Journal of Medicine. 2008;359(5):492-507
  63. 63. Silber JR, Bobola MS, Blank A, Chamberlain MC. O6-methylguanine-DNA methyltransferase in glioma therapy: Promise and problems. Biochimica et Biophysica Acta. 2012;1826(1):71-82
  64. 64. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. The New England Journal of Medicine. 2005;352(10):997-1003
  65. 65. Hsieh CT, Su IC, Huang CT, Chang CJ, Wang JS. The prognostic value of O6-methylguanine-DNA methyltransferase gene promoter methylation detected by gel-based methylation-specific polymerase chain reaction in patients with glioblastoma multiforme: A systematic review. International Journal of Clinical and Experimental Medicine. 2016;9(6):10899-10906
  66. 66. Kreisl TN, Kim L, Moore K, Duic P, Royce C, Stroud I, et al. Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. Journal of Clinical Oncology. 2009;27(5):740-745
  67. 67. Friedman HS, Prados MD, Wen PY, Mikkelsen T, Schiff D, Abrey LE, et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. Journal of Clinical Oncology. 2009;27(28):4733-4740
  68. 68. Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishikawa R, et al. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. The New England Journal of Medicine. 2014;370(8):709-722
  69. 69. Wick W, Gorlia T, Bendszus M, Taphoorn M, Sahm F, Harting I, et al. Lomustine and bevacizumab in progressive glioblastoma. The New England Journal of Medicine. 2017;377(20):1954-1963
  70. 70. Diaz RJ, Ali S, Qadir MG, De La Fuente MI, Ivan ME, Komotar RJ. The role of bevacizumab in the treatment of glioblastoma. Journal of Neuro-Oncology. 2017;133(3):455-467
  71. 71. Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. The 2016 World Health Organization classification of tumors of the central nervous system: A summary. Acta Neuropathologica. 2016;131(6):803-820
  72. 72. Stark AM, Doukas A, Hugo HH, Mehdorn HM. The expression of mismatch repair proteins MLH1, MSH2 and MSH6 correlates with the Ki67 proliferation index and survival in patients with recurrent glioblastoma. Neurological Research. 2010;32(8):816-820
  73. 73. Martinez R, Setien F, Voelter C, Casado S, Quesada MP, Schackert G, et al. CpG island promoter hypermethylation of the pro-apoptotic gene caspase-8 is a common hallmark of relapsed glioblastoma multiforme. Carcinogenesis. 2007;28(6):1264-1268
  74. 74. Stark AM, Witzel P, Strege RJ, Hugo HH, Mehdorn HM. p53, mdm2, EGFR, and msh2 expression in paired initial and recurrent glioblastoma multiforme. Journal of Neurology, Neurosurgery, and Psychiatry. 2003;74(6):779-783
  75. 75. Campos B, Olsen LR, Urup T, Poulsen HS. A comprehensive profile of recurrent glioblastoma. Oncogene. 2016;35(45):5819-5825
  76. 76. Seystahl K, Gramatzki D, Roth P, Weller M. Pharmacotherapies for the treatment of glioblastoma—Current evidence and perspectives. Expert Opinion on Pharmacotherapy. 2016;17(9):1259-1270

Written By

Cheng-Ta Hsieh and Da-Tong Ju

Submitted: 21 September 2019 Reviewed: 13 January 2020 Published: 20 February 2020