Skip to main content
Top
Published in: Journal of Neuro-Oncology 3/2013

01-07-2013 | Clinical Study

Immediate post-operative brachytherapy prior to irradiation and temozolomide for newly diagnosed glioblastoma

Authors: J. Dawn Waters, Brent Rose, David D. Gonda, Daniel J. Scanderbeg, Michelle Russell, John F. Alksne, Kevin Murphy, Bob S. Carter, Joshua Lawson, Clark C. Chen

Published in: Journal of Neuro-Oncology | Issue 3/2013

Login to get access

Abstract

To determine whether immediate post-operative brachytherapy can be safely applied to newly diagnosed glioblastomas to retard tumor progression prior to initiation of external beam radiation therapy (EBRT) and temozolomide. Between 1996 and 2011, eleven patients underwent implantation of GliaSite (n = 9) or MammoSite (n = 2) at the time of surgical resection. Brachytherapy was carried out on post-operative day 2–3, with 45–60 Gy delivered to a 1 cm margin. All patients underwent subsequent standard radiation/temozolomide treatment 4–5 weeks post-irradiation. There were no wound related complications. Toxicity was observed in two patients (2/11 or 18 %), including one post-operative seizure and one case of cerebral edema that resolved after a course of steroid treatment. Immediate post-operative and pre-irradiation/temozolomide magnetic resonance imaging assessment was available for 9 of the 11 patients. Two of these nine patients (22 %) developed new regions of contrast enhancement prior to irradiation/temozolomide. This compares favorably to historical data where 53 % of patient suffer such tumor progression. While there was a trend toward improved 6 month progression free survival in the brachytherapy/temozolomide/radiation treated patients, the overall survival of these patients were comparable to historical controls. This case series demonstrates the safety of immediate post-operative brachytherapy when applied prior to EBRT and temozolomide in the treatment of newly diagnosed glioblastomas.
Literature
2.
go back to reference Walker MD et al (1978) Evaluation of BCNU and/or radiotherapy in the treatment of anaplastic gliomas. A cooperative clinical trial. J Neurosurg 49(3):333–343PubMedCrossRef Walker MD et al (1978) Evaluation of BCNU and/or radiotherapy in the treatment of anaplastic gliomas. A cooperative clinical trial. J Neurosurg 49(3):333–343PubMedCrossRef
3.
go back to reference Stupp R et al (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. New Engl J Med 352(10):987–996PubMedCrossRef Stupp R et al (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. New Engl J Med 352(10):987–996PubMedCrossRef
4.
go back to reference Pennington C et al (2006) A pilot study of brain tumour growth between radiotherapy planning and delivery. Clin Oncol 18(2):104–108CrossRef Pennington C et al (2006) A pilot study of brain tumour growth between radiotherapy planning and delivery. Clin Oncol 18(2):104–108CrossRef
5.
go back to reference Pirzkall A et al (2009) Tumor regrowth between surgery and initiation of adjuvant therapy in patients with newly diagnosed glioblastoma. Neuro Oncol 11(6):842–852PubMedCrossRef Pirzkall A et al (2009) Tumor regrowth between surgery and initiation of adjuvant therapy in patients with newly diagnosed glioblastoma. Neuro Oncol 11(6):842–852PubMedCrossRef
6.
go back to reference Dempsey JF et al (1998) Dosimetric properties of a novel brachytherapy balloon applicator for the treatment of malignant brain-tumor resection-cavity margins. Int J Radiat Oncol Biol Phys 42(2):421–429PubMedCrossRef Dempsey JF et al (1998) Dosimetric properties of a novel brachytherapy balloon applicator for the treatment of malignant brain-tumor resection-cavity margins. Int J Radiat Oncol Biol Phys 42(2):421–429PubMedCrossRef
7.
go back to reference Edmundson GK et al (2002) Dosimetric characteristics of the MammoSite RTS, a new breast brachytherapy applicator. Int J Radiat Oncol Biol Phys 52(4):1132–1139PubMedCrossRef Edmundson GK et al (2002) Dosimetric characteristics of the MammoSite RTS, a new breast brachytherapy applicator. Int J Radiat Oncol Biol Phys 52(4):1132–1139PubMedCrossRef
8.
go back to reference Tatter SB et al (2003) An inflatable balloon catheter and liquid 125I radiation source (GliaSite Radiation Therapy System) for treatment of recurrent malignant glioma: multicenter safety and feasibility trial. J Neurosurg 99(2):297–303PubMedCrossRef Tatter SB et al (2003) An inflatable balloon catheter and liquid 125I radiation source (GliaSite Radiation Therapy System) for treatment of recurrent malignant glioma: multicenter safety and feasibility trial. J Neurosurg 99(2):297–303PubMedCrossRef
9.
go back to reference Gutin PH et al (1984) Brachytherapy of recurrent malignant brain tumors with removable high-activity iodine-125 sources. J Neurosurg 60(1):61–68PubMedCrossRef Gutin PH et al (1984) Brachytherapy of recurrent malignant brain tumors with removable high-activity iodine-125 sources. J Neurosurg 60(1):61–68PubMedCrossRef
10.
go back to reference Gutin PH et al (1991) External irradiation followed by an interstitial high activity iodine-125 implant “boost” in the initial treatment of malignant gliomas: NCOG study 6G–82-2. Int J Radiat Oncol Biol Phys 21(3):601–606PubMedCrossRef Gutin PH et al (1991) External irradiation followed by an interstitial high activity iodine-125 implant “boost” in the initial treatment of malignant gliomas: NCOG study 6G–82-2. Int J Radiat Oncol Biol Phys 21(3):601–606PubMedCrossRef
11.
go back to reference Prados MD et al (1992) Interstitial brachytherapy for newly diagnosed patients with malignant gliomas: the UCSF experience. Int J Radiat Oncol Biol Phys 24(4):593–597PubMedCrossRef Prados MD et al (1992) Interstitial brachytherapy for newly diagnosed patients with malignant gliomas: the UCSF experience. Int J Radiat Oncol Biol Phys 24(4):593–597PubMedCrossRef
12.
go back to reference Leibel SA et al (1989) Survival and quality of life after interstitial implantation of removable high-activity iodine-125 sources for the treatment of patients with recurrent malignant gliomas. Int J Radiat Oncol Biol Phys 17(6):1129–1139PubMedCrossRef Leibel SA et al (1989) Survival and quality of life after interstitial implantation of removable high-activity iodine-125 sources for the treatment of patients with recurrent malignant gliomas. Int J Radiat Oncol Biol Phys 17(6):1129–1139PubMedCrossRef
13.
go back to reference Scharfen CO et al (1992) High activity iodine-125 interstitial implant for gliomas. Int J Radiat Oncol Biol Phys 24(4):583–591CrossRef Scharfen CO et al (1992) High activity iodine-125 interstitial implant for gliomas. Int J Radiat Oncol Biol Phys 24(4):583–591CrossRef
14.
go back to reference Laperriere NJ et al (1998) Randomized study of brachytherapy in the initial management of patients with malignant astrocytoma. Int J Radiat Oncol Biol Phys 41(5):1005–1011PubMedCrossRef Laperriere NJ et al (1998) Randomized study of brachytherapy in the initial management of patients with malignant astrocytoma. Int J Radiat Oncol Biol Phys 41(5):1005–1011PubMedCrossRef
15.
go back to reference Gabayan AJ et al (2006) GliaSite brachytherapy for treatment of recurrent malignant gliomas: a retrospective multi-institutional analysis. Neurosurgery 58(4):701–709; discussion 701–709PubMedCrossRef Gabayan AJ et al (2006) GliaSite brachytherapy for treatment of recurrent malignant gliomas: a retrospective multi-institutional analysis. Neurosurgery 58(4):701–709; discussion 701–709PubMedCrossRef
16.
go back to reference Sanai N et al (2011) An extent of resection threshold for newly diagnosed glioblastomas. J Neurosurg 115(1):3–8PubMedCrossRef Sanai N et al (2011) An extent of resection threshold for newly diagnosed glioblastomas. J Neurosurg 115(1):3–8PubMedCrossRef
17.
go back to reference Lacroix M et al (2001) A multivariate analysis of 416 patients with glioblastoma multiforme: prognosis, extent of resection, and survival. J Neurosurg 95(2):190–198PubMedCrossRef Lacroix M et al (2001) A multivariate analysis of 416 patients with glioblastoma multiforme: prognosis, extent of resection, and survival. J Neurosurg 95(2):190–198PubMedCrossRef
18.
go back to reference Kesari S et al (2011) DNA damage response and repair: insights into strategies for radiation sensitization of gliomas. Future Oncol 7(11):1335–1346PubMedCrossRef Kesari S et al (2011) DNA damage response and repair: insights into strategies for radiation sensitization of gliomas. Future Oncol 7(11):1335–1346PubMedCrossRef
19.
go back to reference Yoshimura J et al (2012) The effects of temozolomide delivered by prolonged intracerebral microinfusion against the rat brainstem GBM allograft model. Childs Nerv Syst 28(5):707–713PubMedCrossRef Yoshimura J et al (2012) The effects of temozolomide delivered by prolonged intracerebral microinfusion against the rat brainstem GBM allograft model. Childs Nerv Syst 28(5):707–713PubMedCrossRef
20.
go back to reference Kil WJ et al (2008) In vitro and in vivo radiosensitization induced by the DNA methylating agent temozolomide. Clin Cancer Res 14(3):931–938PubMedCrossRef Kil WJ et al (2008) In vitro and in vivo radiosensitization induced by the DNA methylating agent temozolomide. Clin Cancer Res 14(3):931–938PubMedCrossRef
21.
go back to reference Fowler JF (1989) The linear-quadratic formula and progress in fractionated radiotherapy. British J Radiol 62(740):679–694CrossRef Fowler JF (1989) The linear-quadratic formula and progress in fractionated radiotherapy. British J Radiol 62(740):679–694CrossRef
22.
go back to reference Wen PY et al (2010) Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol 28(11):1963–1972PubMedCrossRef Wen PY et al (2010) Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol 28(11):1963–1972PubMedCrossRef
24.
go back to reference Lamborn KR et al (2008) Progression-free survival: an important end point in evaluating therapy for recurrent high-grade gliomas. Neuro Oncol 10(2):162–170CrossRef Lamborn KR et al (2008) Progression-free survival: an important end point in evaluating therapy for recurrent high-grade gliomas. Neuro Oncol 10(2):162–170CrossRef
25.
go back to reference Curran WJ Jr et al (1993) Recursive partitioning analysis of prognostic factors in three Radiation Therapy Oncology Group malignant glioma trials. J Natl Cancer Inst 85(9):704–710PubMedCrossRef Curran WJ Jr et al (1993) Recursive partitioning analysis of prognostic factors in three Radiation Therapy Oncology Group malignant glioma trials. J Natl Cancer Inst 85(9):704–710PubMedCrossRef
26.
go back to reference Mehta MP et al (1994) Stereotactic radiosurgery for glioblastoma multiforme: report of a prospective study evaluating prognostic factors and analyzing long-term survival advantage. Int J Radiat Oncol Biol Phys 30(3):541–549PubMedCrossRef Mehta MP et al (1994) Stereotactic radiosurgery for glioblastoma multiforme: report of a prospective study evaluating prognostic factors and analyzing long-term survival advantage. Int J Radiat Oncol Biol Phys 30(3):541–549PubMedCrossRef
27.
go back to reference Shenouda G et al (1997) Radiosurgery and accelerated radiotherapy for patients with glioblastoma. Can J Neurol Sci 24(2):110–115PubMed Shenouda G et al (1997) Radiosurgery and accelerated radiotherapy for patients with glioblastoma. Can J Neurol Sci 24(2):110–115PubMed
28.
go back to reference Ballman KV et al (2007) The relationship between six-month progression-free survival and 12-month overall survival end points for phase II trials in patients with glioblastoma multiforme. Neuro Oncol 9(1):29–38PubMedCrossRef Ballman KV et al (2007) The relationship between six-month progression-free survival and 12-month overall survival end points for phase II trials in patients with glioblastoma multiforme. Neuro Oncol 9(1):29–38PubMedCrossRef
29.
go back to reference Combs SE et al (2005) Efficacy of fractionated stereotactic reirradiation in recurrent gliomas: long-term results in 172 patients treated in a single institution. J Clin Oncol 23(34):8863–8869PubMedCrossRef Combs SE et al (2005) Efficacy of fractionated stereotactic reirradiation in recurrent gliomas: long-term results in 172 patients treated in a single institution. J Clin Oncol 23(34):8863–8869PubMedCrossRef
30.
go back to reference Bristow RE et al (2002) Survival effect of maximal cytoreductive surgery for advanced ovarian carcinoma during the platinum era: a meta-analysis. J Clin Oncol 20(5):1248–1259PubMedCrossRef Bristow RE et al (2002) Survival effect of maximal cytoreductive surgery for advanced ovarian carcinoma during the platinum era: a meta-analysis. J Clin Oncol 20(5):1248–1259PubMedCrossRef
31.
go back to reference Chen AM et al (2007) Phase I trial of gross total resection, permanent iodine-125 brachytherapy, and hyperfractionated radiotherapy for newly diagnosed glioblastoma multiforme. Int J Radiat Oncol Biol Phys 69(3):825–830PubMedCrossRef Chen AM et al (2007) Phase I trial of gross total resection, permanent iodine-125 brachytherapy, and hyperfractionated radiotherapy for newly diagnosed glioblastoma multiforme. Int J Radiat Oncol Biol Phys 69(3):825–830PubMedCrossRef
32.
go back to reference Koot RW et al (2000) Brachytherapy: results of two different therapy strategies for patients with primary glioblastoma multiforme. Cancer 88(12):2796–2802PubMedCrossRef Koot RW et al (2000) Brachytherapy: results of two different therapy strategies for patients with primary glioblastoma multiforme. Cancer 88(12):2796–2802PubMedCrossRef
33.
go back to reference Welsh J et al (2007) GliaSite brachytherapy boost as part of initial treatment of glioblastoma multiforme: a retrospective multi-institutional pilot study. Int J Radiat Oncol Biol Phys 68(1):159–165PubMedCrossRef Welsh J et al (2007) GliaSite brachytherapy boost as part of initial treatment of glioblastoma multiforme: a retrospective multi-institutional pilot study. Int J Radiat Oncol Biol Phys 68(1):159–165PubMedCrossRef
34.
go back to reference Hegi ME et al (2005) MGMT gene silencing and benefit from temozolomide in glioblastoma. New Engl J Med 352(10):997–1003PubMedCrossRef Hegi ME et al (2005) MGMT gene silencing and benefit from temozolomide in glioblastoma. New Engl J Med 352(10):997–1003PubMedCrossRef
35.
go back to reference Noushmehr H et al (2010) Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 17(5):510–522PubMedCrossRef Noushmehr H et al (2010) Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 17(5):510–522PubMedCrossRef
36.
go back to reference Souhami L et al (2004) Randomized comparison of stereotactic radiosurgery followed by conventional radiotherapy with carmustine to conventional radiotherapy with carmustine for patients with glioblastoma multiforme: report of Radiation Therapy Oncology Group 93–05 protocol. Int J Radiat Oncol Biol Phys 60(3):853–860CrossRef Souhami L et al (2004) Randomized comparison of stereotactic radiosurgery followed by conventional radiotherapy with carmustine to conventional radiotherapy with carmustine for patients with glioblastoma multiforme: report of Radiation Therapy Oncology Group 93–05 protocol. Int J Radiat Oncol Biol Phys 60(3):853–860CrossRef
37.
go back to reference Fulton DS et al (1992) Increasing radiation dose intensity using hyperfractionation in patients with malignant glioma. Final report of a prospective phase I-II dose response study. J Neurooncol 14(1):63–72PubMedCrossRef Fulton DS et al (1992) Increasing radiation dose intensity using hyperfractionation in patients with malignant glioma. Final report of a prospective phase I-II dose response study. J Neurooncol 14(1):63–72PubMedCrossRef
38.
go back to reference Lustig RA et al (2007) Imaging response in malignant glioma, RTOG 90–06. Am J Clin Oncol 30(1):32–37PubMedCrossRef Lustig RA et al (2007) Imaging response in malignant glioma, RTOG 90–06. Am J Clin Oncol 30(1):32–37PubMedCrossRef
39.
go back to reference Coughlin C et al (2000) Phase II, two-arm RTOG trial (94–11) of bischloroethyl-nitrosourea plus accelerated hyperfractionated radiotherapy (64.0 or 70.4 Gy) based on tumor volume (>20 or ≤20 cm (2), respectively) in the treatment of newly-diagnosed radiosurgery-ineligible glioblastoma multiforme patients. Int J Radiat Oncol Biol Phys 48(5):1351–1358PubMedCrossRef Coughlin C et al (2000) Phase II, two-arm RTOG trial (94–11) of bischloroethyl-nitrosourea plus accelerated hyperfractionated radiotherapy (64.0 or 70.4 Gy) based on tumor volume (>20 or ≤20 cm (2), respectively) in the treatment of newly-diagnosed radiosurgery-ineligible glioblastoma multiforme patients. Int J Radiat Oncol Biol Phys 48(5):1351–1358PubMedCrossRef
40.
go back to reference Tsien C et al (2009) Phase I three-dimensional conformal radiation dose escalation study in newly diagnosed glioblastoma: radiation Therapy Oncology Group Trial 98–03. Int J Radiat Oncol Biol Phys 73(3):699–708PubMedCrossRef Tsien C et al (2009) Phase I three-dimensional conformal radiation dose escalation study in newly diagnosed glioblastoma: radiation Therapy Oncology Group Trial 98–03. Int J Radiat Oncol Biol Phys 73(3):699–708PubMedCrossRef
41.
go back to reference Patel S et al (2000) Permanent iodine-125 interstitial implants for the treatment of recurrent glioblastoma multiforme. Neurosurgery 46(5):1123–1128; discussion 1128–1130PubMedCrossRef Patel S et al (2000) Permanent iodine-125 interstitial implants for the treatment of recurrent glioblastoma multiforme. Neurosurgery 46(5):1123–1128; discussion 1128–1130PubMedCrossRef
42.
go back to reference Larson DA et al (2004) Permanent iodine 125 brachytherapy in patients with progressive or recurrent glioblastoma multiforme. Neuro Oncol 6(2):119–126PubMedCrossRef Larson DA et al (2004) Permanent iodine 125 brachytherapy in patients with progressive or recurrent glioblastoma multiforme. Neuro Oncol 6(2):119–126PubMedCrossRef
43.
go back to reference Selker RG et al (2002) The Brain Tumor Cooperative Group NIH Trial 87-01: a randomized comparison of surgery, external radiotherapy, and carmustine versus surgery, intersitial radiotherapy boost, external radiation therapy, and carmustine. Neurosurgery 51(2):345–355; discussion 355–357 Selker RG et al (2002) The Brain Tumor Cooperative Group NIH Trial 87-01: a randomized comparison of surgery, external radiotherapy, and carmustine versus surgery, intersitial radiotherapy boost, external radiation therapy, and carmustine. Neurosurgery 51(2):345–355; discussion 355–357
44.
go back to reference Mclendon RE et al (2007) Tumor resection cavity administered iodine-131-labeled antitenascin 81C6 radioimmunotherapy in patients with malignant glioma: neuropathology aspects. Nucl Med Biol 34(4):405–413 Mclendon RE et al (2007) Tumor resection cavity administered iodine-131-labeled antitenascin 81C6 radioimmunotherapy in patients with malignant glioma: neuropathology aspects. Nucl Med Biol 34(4):405–413
45.
go back to reference Sneed PK et al (1996) Demonstration of brachytherapy boost dose-response relationships in glioblastoma multiforme. Int J Radiat Oncol Biol Phys 35(1):37–44 Sneed PK et al (1996) Demonstration of brachytherapy boost dose-response relationships in glioblastoma multiforme. Int J Radiat Oncol Biol Phys 35(1):37–44
46.
go back to reference Wen PY et al (1994) Long term results of stereotactic brachytherapy used in the initial treatment of patients with glioblastomas. Cancer 73(12):3029–3036 Wen PY et al (1994) Long term results of stereotactic brachytherapy used in the initial treatment of patients with glioblastomas. Cancer 73(12):3029–3036
Metadata
Title
Immediate post-operative brachytherapy prior to irradiation and temozolomide for newly diagnosed glioblastoma
Authors
J. Dawn Waters
Brent Rose
David D. Gonda
Daniel J. Scanderbeg
Michelle Russell
John F. Alksne
Kevin Murphy
Bob S. Carter
Joshua Lawson
Clark C. Chen
Publication date
01-07-2013
Publisher
Springer US
Published in
Journal of Neuro-Oncology / Issue 3/2013
Print ISSN: 0167-594X
Electronic ISSN: 1573-7373
DOI
https://doi.org/10.1007/s11060-013-1139-x

Other articles of this Issue 3/2013

Journal of Neuro-Oncology 3/2013 Go to the issue