Coverage Policy Manual
Policy #: 2002033
Category: Radiology
Initiated: August 2017
Last Review: December 2023
  Brachytherapy, Brain Tumors

Description:
INTRACAVITARY BALLOON CATHETER BRAIN BRACHYTHERAPY
Intracavitary balloon catheter brain brachytherapy is localized temporary high-dose radiotherapy in the brain that requires placement of an inflatable balloon catheter in the surgical cavity, before closing the craniotomy of a resection to remove or debulk a malignant brain mass. A radiation source is then placed in the balloon to expose surrounding brain tissue to radiation, either continuously or in a series of brief treatments. After the patient completes therapy, the radiation source is permanently removed, and the balloon catheter is surgically explanted.
 
BRAIN TUMORS
Malignant Gliomas
Diffuse fibrillary astrocytoma is the most common glial brain tumor in adults. It is classified histologically into 3 grades: grade II astrocytoma, grade III anaplastic astrocytoma, and grade IV glioblastoma multiforme. Oligodendrogliomas are diffuse neoplasms closely related to diffuse fibrillary astrocytomas clinically and biologically. However, these tumors generally have better prognoses than diffuse astrocytomas, with mean survival times of 10 years vs 2 to 3 years. Also, oligodendrogliomas apparently are more chemosensitive than astrocytomas. The most aggressive and chemoresistant astrocytoma, glioblastoma multiforme has survival times of less than 2 years for most patients.
 
Treatment of primary brain tumors begins with surgery with curative intent or optimal tumor debulking, usually followed by radiotherapy and/or chemotherapy. Survival after chemoradiotherapy largely depends on the extent of residual tumor after surgery. Therefore, tumors arising in the midline, basal ganglia, or corpus callosum or those arising in the eloquent speech or motor areas of the cortex have a particularly poor outcome, because they typically cannot be extensively resected. Recurrence is common after surgery for malignant gliomas, even if followed by chemoradiotherapy because the tumors are usually diffusely infiltrating and develop resistance to chemotherapy; also, neurotoxicity limits cumulative doses of whole brain radiation. Chemotherapy regimens for gliomas usually rely on nitrosourea alkylating agents
(carmustine or lomustine), temozolomide, procarbazine, vincristine, and platinum-based agents. The most common regimen combines procarbazine, lomustine, vincristine, and single or multiagent therapy with temozolomide. A biodegradable polymer wafer impregnated with carmustine (Gliadel® Wafer; Guilford Pharmaceuticals) also can be implanted into the surgical cavity as an adjunct to surgery and radiation. It is indicated for newly diagnosed high-grade malignant glioma and for recurrent glioblastoma multiforme.   
 
Brain Metastasis From Other Primary Malignancies
Intracranial metastases are a frequent occurrence seen at autopsy in 10% to 30% of deaths from cancer. Lung cancer is the most common source of brain metastasis (relative prevalence, 48%), followed by breast cancer (15%), unknown primary (12%), melanoma (9%), and colon cancer (5%).
 
Treatment goals in these patients include local control of existing metastases, regional control to prevent the growth of undetected metastases, extending the duration of overall survival, and maintaining quality of life. Surgical resection followed by whole brain radiotherapy (WBRT) is the mainstay of treatment for patients with 1 to 3 operable brain metastases and with adequate performance status and control of extracranial disease. Resection plus WBRT extends the duration of survival compared with biopsy plus WBRT. Although adding WBRT to resection does not increase overall survival duration, it reduces local and distant recurrence of brain metastases. Thus, WBRT decreases the incidence of death from neurologic causes and may help maintain adequate quality of life, if the cumulative dose does not cause
unacceptable neurotoxicity.
 
REGULATORY STATUS
In 2001, the GliaSite® Radiation Therapy System (GliaSite® RTS; IsoRay Medical) was cleared for marketing by the U.S. Food and Drug Administration (FDA) through the 510(k) process (K003206). FDA determined that this device was substantially equivalent to separately marketed ventricular reservoirs and catheters, manual radionuclide applicator systems, and radionuclide sources.
 
In 2011, a modified GliaSite® RTS was cleared for marketing by FDA through the 510(k) process (K111931). GliaSite® RTS includes a catheter tray with a double balloon catheter and accessories used for implantation of an aqueous saline solution of molecularly bound radioactive iodine (sodium 3 [I-125] iodo-4-hydroxybenzenesulfonate; Iotrex™) as the radiation source; and an access tray with items used for afterloading and retrieving the radioactive material. One to 3 weeks after resection and balloon implantation, the Iotrex™ solution is loaded through a subcutaneous port and left in for 3 to 6 days. Prescribed radiation doses are usually 40 to 60 gray measured at 0.5 to 1.0 cm from the balloon surface. This procedure has been performed on an inpatient basis.
 
In December 2013, CESITRX (Liquid Cesium131 solution) for use with GliaSite RTS was cleared for marketing by FDA through the 510(k) process (K132996).
 
FDA product code: KXK.
 
In April 2016, IsoRay Medical filed a notice with the U.S. Securities and Exchange Commission indicating that it decided to terminate all agreements related to the patent license, supply, manufacture, and distribution of its GliaSite® Radiation Therapy System and certain ancillary products (“GliaSite® Product”) (U.S. Securities and exchange Commission). The reason cited was marginal sales. This decision affected licensing agreements with Dr. Reddy’s Laboratories and Hologic for U.S. operations and Karlheinz Goehl-Medizintechnik for international agreements.

Policy/
Coverage:
Effective August 1, 2021, for members of plans that utilize a radiation oncology benefits management program, Prior Approval is required for this service and is managed through the radiation oncology benefits management program.
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
Brachytherapy of the brain for primary, recurrent or metastatic malignancy, or benign disease does not meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness.
 
For members with contracts without primary coverage criteria, Brachytherapy of the brain for primary, recurrent or metastatic malignancy, or benign disease is considered investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage.

Rationale:
March 2006
There is an ongoing trial of GliaSite in patients with newly diagnosed brain metastases.  Phosphorus 32 is being studied in patients with glioblastoma multiforme.  
 
Selker and associates reported results of a randomized controlled trial in 2002 and concluded that there was no long-term survival advantage of increased radiation dose with (125)I seeds in newly diagnosed glioma patients.
 
Chan and associates reported 24 patients with recurrent glioblastoma multiforme.  Median survival was 9.3 months for patients with a Karnofsky performance status of 70 or greater but only 3.1 months in patients with a Karnofsky less than 70.  "Additional data are needed to fully assess the therapeutic benefit of GliaSite brachytherapy for recurrent GBM."
 
2010 Update
A literature search was conducted for studies reporting on intracavitary balloon catheter brain brachytherapy published through February 2010. For newly diagnosed glioblastoma multiforme (GBM) patients treated after surgery, one prospective (Johannesen, 1999) and one retrospective (Welsh, 2007) study were identified. Four studies focused on patients treated after surgery for recurrent glioma (Tatter, 2003) (Chan, 2005) (Payne, 2005) (Gabayan, 2006).  Additionally, there has been one study published on patients treated for newly diagnosed and resected single brain metastasis (Rogers, 2006).  An additional study on 8 patients with glioma or brain metastasis focused on computed tomography and magnetic resonance imaging after balloon placement but did not report patient outcomes (Matheus, 2004).  
 
Malignant gliomas and astrocytoma
In each of 3 reports on recurrent high-grade gliomas (Tatter, 2003) (Gabayan, 2006) (Wernicke, 2010), all patients were treated with the commercially available GliaSite® RTS device. The first (Tatter, 2003) reported on a multicenter safety and feasibility trial (n=21; 15 with GBM; 5 with anaplastic astrocytoma and one with anaplastic oligodendrogliomas [ODG]). All patients (n=11) received first-line therapy with resection and radiation, with or without systemic chemotherapy. Time from end of first-line therapy to repeated resection for recurrent disease was not reported. Although not a primary endpoint, median overall survival was 12.7 months (95% CI: 6.9–15.3), and a Kaplan-Meier curve showed estimated overall survival at 1 year was just over 50%. Investigators reported no serious device-related adverse events during brachytherapy and no symptomatic radiation necrosis during follow-up.
 
The largest group was reported in a retrospective multi-institutional analysis (n=95; 80 with GBM, 9 with anaplastic astrocytoma, 4 with anaplastic ODG, and one each with a mixed anaplastic tumor or gliosarcoma) (Gabayan, 2006).  All patients received external-beam radiation after initial resection, and 55 (58%) also received systemic chemotherapy. Time from end of front-line therapy to repeated resection for recurrent disease was not reported. Fifteen patients (16%) who had previously been treated with external-beam radiation following maximal debulking surgery were treated with GliaSite® (average dose of 60 Gy) upon tumor recurrence. Median overall survival from the time of GliaSite® placement was 9.1 months (95% CI: 7.8–10.4), and overall survival at 1 year was 31.1% (95% CI: 21.2–41.0%). Only 2 patients experienced Radiation Therapy Oncology Group (RTOG) grade 3 toxicity attributable to radiation and none experienced grades 4 or 5. However, 10 adverse events were attributed to surgery. The authors concluded that survival benefit was modest and that these data were similarly inconclusive to previous feasibility studies.
 
The retrospective analysis on GliaSite® (Gabayan, 2006) did not report important prognostic factors available from the Gliadel® randomized trial (e.g., median interval from first operation; cumulative radiation dose and proportion given WBRT vs. local radiation vs. both in first-line therapy and completeness of the second resections). The authors concluded that it is a challenge to assess survival value from these studies without better comparative evidence on demonstrably similar patient groups, preferably from a randomized comparative trial.
 
Glioblastoma multiforme
Johannesen et al. reported a phase I/II study on 44 newly diagnosed glioblastoma multiforme (GBM) patients implanted with intracavitary balloon catheters at resection (Johannesen, 1999). Two to 3 days after surgery, high dose-rate 192-Ir sources were inserted twice daily for 15 minutes over 5 to 6 days, using remote after-loading devices designed and fabricated by the investigators. Cumulative radiation doses were 60 (n=33) or 72 Gy (n=11). Median survival was 11.7 months (range: 2.7 to 50.9 months) for all patients, 12.8 months for those treated with 60 Gy, and 9.9 months for those treated with 72 Gy. Overall survival at 1 year was 46%. Relapses occurred in 89% of patients at a median follow-up time of 8.3 months after treatment (range: 1.2 to 34.7 months). These outcomes are similar to those of conventional WBRT after resection, although investigators emphasized the shorter treatment time (1 vs. 5 to 6 weeks) with balloon catheter brachytherapy. While the authors asserted that hospital stays were shorter (median, 21 days) and quality of life over the first 6 months was better than after conventional WBRT, they did not report data to support these claims.
 
In the multicenter, retrospective study performed by Welsh et al. data were compiled from 8 centers on 20 patients with GBM patients with median age and Karnofsky performance status of 59 and 89, respectively (welsh, 2007). Following maximal tumor debulking patients were treated with GliaSite® (median dose 60 Gy) prior to external-beam radiation (median dose 110 Gy). In this cohort, average survival was 11.4 months (range 4-29); 4 months longer than historical controls (95% CI: 0.23–4.9). RTOG grade 3 central nervous system toxicity was observed in 3 patients (14%). It is noteworthy that 50% of treatment failures had the balloons placed 2 cm from the margin of the tumor. While this study may suggest that administration of increased doses (up to 100 Gy) using GliaSite® is feasible and relatively well tolerated, the authors acknowledge that putative survival advantage must be interpreted with caution. Additional studies using GliaSite® in conjunction with external beam radiation following surgery for newly diagnosed GBM would be required to adequately assess safety and efficacy.
 
In an additional clinical trial on recurrent GBM performed at Johns Hopkins Medical Center, investigators reported results to be inconclusive (Chan, 2005). Front-line therapy included surgery followed by external-beam radiation. Time from primary resection (or from end of primary treatment) to recurrence was not reported. Median overall survival was 23.3 months from diagnosis of the primary tumor, and 9.1 months  from GliaSite® RTS treatment. Kaplan-Meier analyses showed overall survival at 1 year to be approximately 33%. GliaSite® was relatively well-tolerated in this cohort with few serious adverse events. Acute adverse effects were reportedly mild; 1 patient experienced mild nausea and vomiting and 10 experienced mild to moderate headaches. Late complications included 1 case of global aphasia and 2 incidents of symptomatic necrosis.
 
Finally, Payne published a case report of one GBM patient undergoing implantation with 2 brachytherapy balloons (Payne, 2005); however, there are no additional studies assessing the 2-balloon approach.
 
Brain metastasis from other primary solid malignancies
A prospective, multicenter, Phase II trial enrolled 71 patients with 1 to 3 brain metastases from a solid tumor of distant origin (Rogers, 2006). Patients enrolled received either GliaSite® (n=62); or, standard brachytherapy with lotrex solution (n=54). Outcomes were analyzed without an intention-to-treat model. Primary malignancies included non-small-cell lung  and gastrointestinal cancers, melanoma, renal carcinoma, and others. While most patients (57%) had only brain metastases, many (43%) also had extracranial metastases. Prior therapies varied widely, and included no treatment, surgery, surgery and radiation or surgery in addition to chemotherapy followed by radiation. The investigators estimated local control at 1 year was 79%, and the median duration of local control was greater than 16.5 months. Median overall survival was 10 months, overall survival at 1 year was 40%, and median duration of functional independence was 10 months. Symptomatic imaging changes led to repeated operation in 13 patients, 9 of whom had radiation necrosis, 2 had mixed tumor and necrosis, and 2 had tumor recurrence only. A total of 9 grade 3 and one grade 4 toxicities were reported in the treated population.
 
Investigators indirectly compared the rate of local control in the GliaSite® treated population: 79% with historical data showing 80–90% local control after resection plus WBRT and only 40% after resection only. However, an accompanying editorial cautions that the rate of new metastases elsewhere in the brain was 50% by 1 year after treatment, and attributes this to omission of WBRT (Barker, 2006). The editorial also stresses the need for direct comparative evidence to determine whether neurocognitive function and quality of life are adequately maintained for longer durations with initially focal treatment and WBRT at recurrence or with focal treatment immediately combined with WBRT.
 
Wernicke and colleagues conducted a single institution, dose escalation study to investigate safety and feasibility of GliaSite® following surgical resection of localized brain tumors (Wernicke, 2010) The balloon was implanted during surgery; then 2 to 3 weeks later aqueous solution of 125-I was introduced for times ranging from 68 to 120 hours. Median total dose was 52 Gy. Median survival for this cohort was 14 months. There were no reports of RTOG Grade 3 or 4 toxicities. Similarly to the other studies cited, results from this trial suggest that the GliaSite® radiation therapy system is relatively safe and well-tolerated in patients with localized brain tumors. However, further studies would be required to assess efficacy.
 
Another study (Chino, 2008) examined feasibility of outpatient GliaSite® brachytherapy in 37 patients. Rather than overnight hospitalization, patients were released after the treatment sessions. Although the study was small and ultimately inconclusive, the outpatient approach did not appear to increase adverse events and seemed to be generally well tolerated.
 
Safety considerations
Overall, adverse events with GliaSite® are not greatly varied from those observed with other brain brachytherapy techniques; however, in 2008, Adkison and colleagues reported a case where linens of a patient with the GliaSite® implant were contaminated with radiation (Adkison, 2008). Recovery studies confirmed that systemic absorption is greater than anticipated. Adkison concluded that precaution with a Foley catheter should be taken in patients with urinary incontinence. Some cases of brain hemorrhage have been reported, so careful coagulation control is critical (Gerber, 2007).  
 
National Comprehensive Cancer Network (NCCN)
The NCCN guidelines for glioblastoma state, “The role of focal radiation techniques in this diffusely infiltrative disease remains undefined.”
 
Ongoing Trials
A search on ClinicalTrials.gov did not reveal any ongoing studies. One study testing GliaSite® in conjunction with temozolomide for primary brain tumors was terminated due to loss of funding.
 
Summary
To date, no standard medical care is established for primary brain malignancies or brain metastases of solid tumors and, there are no clinical data available to provide convincing evidence that intracavitary balloon brachytherapy extends the duration of survival, time to relapse, quality of life, or progression.
 
2011 Update
This policy was updated with a search of the MEDLINE database through August 2011. The search did not identify any new literature that would prompt a change in the coverage statement.
 
2012 Update
A literature search conducted through September 2012 did not reveal any new information that would prompt a change in the coverage statement.
 
In 2011, Gobitti and colleagues reported on 15 patients treated with GliaSite® brachytherapy after surgical resection of recurrent grade 3 or 4 gliomas (10 with GBM, 4 anaplastic astrocytoma, and 1 anaplastic xanthoastrocytoma) (Gobitti, 2011). Patients were followed from 1-30 months. Only 2 patients survived to 30 months follow-up. Eleven patients experienced local tumor recurrence. After GliaSite® brachytherapy, median overall survival was 13 months and median disease-free survival was 7 months. Late radiation necrosis was experienced by 3 patients; 2 subsequently died of further complications. One patient had hemiparesis and dysphagia, which resolved over 6 months. The authors concluded that re-intervention followed by GliaSite brachytherapy should not be offered as a standard treatment for recurrent high-grade glioma, because of the high rate of late complications, treatment-related deaths, and high treatment costs.
   
2014 Update
 
A literature search conducted through June 2014 did not reveal any new information that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
In 2013, Waters et al reported on a retrospective review of 11 patients with newly diagnosed glioblastomas who received brain brachytherapy 2 to 3 days postsurgical resection before external beam radiation therapy and temozolomide (Waters, 2013). Brachytherapy was delivered at 45 to 60 Gy with GliaSite in 9 patients and with MammoSite in 2 patients. While progression-free survival trended toward improvement at 6 months, OS did not differ from historical controls
 
2017 Update
A literature search was conducted using the MEDLINE database through November 2017. There was no information identified that would prompt a change in the coverage statement.
  
2018 Update
A literature search was conducted through November 2018.  There was no new information identified that would prompt a change in the coverage statement.  
 
2021 Update
Annual policy review completed with a literature search using the MEDLINE database through November 2021. No new literature was identified that would prompt a change in the coverage statement.
 
2022 Update
Annual policy review completed with a literature search using the MEDLINE database through November 2022. No new literature was identified that would prompt a change in the coverage statement.
 
2023 Update
Annual policy review completed with a literature search using the MEDLINE database through November 2023. No new literature was identified that would prompt a change in the coverage statement.

CPT/HCPCS:
64999Unlisted procedure, nervous system
77316Brachytherapy isodose plan; simple (calculation[s] made from 1 to 4 sources, or remote afterloading brachytherapy, 1 channel), includes basic dosimetry calculation(s)
77317Brachytherapy isodose plan; intermediate (calculation[s] made from 5 to 10 sources, or remote afterloading brachytherapy, 2 12 channels), includes basic dosimetry calculation(s)
77318Brachytherapy isodose plan; complex (calculation[s] made from over 10 sources, or remote afterloading brachytherapy, over 12 channels), includes basic dosimetry calculation(s)
77761Intracavitary radiation source application; simple
77762Intracavitary radiation source application; intermediate
77763Intracavitary radiation source application; complex
77770Remote afterloading high dose rate radionuclide interstitial or intracavitary brachytherapy, includes basic dosimetry, when performed; 1 channel
77771Remote afterloading high dose rate radionuclide interstitial or intracavitary brachytherapy, includes basic dosimetry, when performed; 2 12 channels
77772Remote afterloading high dose rate radionuclide interstitial or intracavitary brachytherapy, includes basic dosimetry, when performed; over 12 channels
A9527Iodine i 125, sodium iodide solution, therapeutic, per millicurie

References: Adkison JB, Thomadsen B, Howard SP.(2008) Systemic iodine 125 activity after GliaSite brachytherapy: safety considerations. Brachytherapy 2008; 7(1):43-6.

Barker F(2006) Brain metastasis. J Neurosurg 2006; 105(3):371-2.

Brachytherapy for malignant gliomas. Hayes Directory, Dec 2006.

Burton EC, Prados MD.(2000) Malignant gliomas. Curr Treat Options Oncol 2000; 1:459-68.

Butowski NA, Sneed PK, Chang SM.(2006) Diagnosis and treatment of recurrent high-grade astrocytoma. J Clin Oncol 2006; 24:1273-80.

Chan TA, Weingart JD, et al.(2005) Treatment of recurrent glioblastoma multiforme with GliaSite brachytherapy. Int J Radiat Oncol Biol Phys 2005; 62:1133-9.

Chino K, Silvain D, Grace A et al.(2008) Feasibility and safety of outpatient brachytherapy in 37 patients with brain tumors using the GliaSite Radiation Therapy System. Med Phys 2008; 35(7):3383-8. Med Phys 2008; 35(7):3383-8.

Davey P.(2002) Brain metastases: treatment options to improve outcomes. CNS Drugs 2002; 16:325-38.

FDA.(2001) 510(k) for GliaSite Radiation Therapy System. www.fda.gov; 2001.

Gabayan AJ, Green SB, Sanan A et al.(2006) GliaSite brachytherapy for treatment of recurrent malignant gliomas: a retrospective multi-institutional analysis. Neurosurgery 2006; 58(4):701-9; discussion 01-9.

Gerber DE, Grossman SA, Chan TA et al.(2007) Brachytherapy 2008; 7(1):43-6. J Radiother Pract 2007; 6:53-7.

Gobitti C, Borsatti E, Arcicasa M et al.(2011) Treatment of recurrent high-grade gliomas with GliaSite brachytherapy: a prospective mono-institutional Italian experience. Tumori 2011; 97(5):614-9.

Johannesen TB, Watne K, Lote K et al.(1999) Intracavity fractionated balloon brachytherapy in glioblastoma. Acta Neurochir (Wien) 1999; 141(2):127-33.

Kolotas C, Birn G, et al.(1999) CT guided interstitial high dose rate brachytherapy for recurrent malignant gliomas. Br J Rad 1999; 72:805-8.

Koot RW, Maarouf M, et al.(2000) Brachytherapy: Results of two different therapy strategies for patients with primary glioblastoma multiforme. Cancer 2000; 88:2796-802.

Matheus MG, Castillo M, Ewend M et al.(2004) J Neurosurg 2006; 105(3):375-84. AJNR Am J Neuroradiol 2004; 25(7):1211-7.

McDermott MW, Berger MS, et al.(2004) Stereotactic radiosurgery and interstitial brachytherapy for glial neoplasms. J Neuro-Oncol 2004; 69:83-100.

National Comprehensive Cancer Network (NCCN) Clinical Practice Guidelines in Oncology. Central nervous system cancers (V.1.2010). Available online at http://www.nccn.org/professionals/physician_gls/PDF/cns.pdf. Last accessed March 2010.

Nieder C, Grosu AL, Molls M.(2000) A comparison of treatment results for recurrent malignant gliomas. Cancer Treat Rev 2000; 26:397-409.

Norden AD, Wen PY, Kesari S.(2006) Brain metastases. Curr Opin Int Med 2006; 5:42-49.

Patel S, Breneman JC, et al.(2000) Permanent iodine-125 interstitial implants for treatment of recurrent glioblastoma multiforme. Neurosurgery 2000; 46:1123-8.

Payne JT, St Clair WH, Given CA, 2nd et al.(2005) Double balloon GliaSite in the management of recurrent glioblastoma multiforme. South Med J 2005; 98(9):957-8.

Reardon DA, Rich JN, et al.(2006) Recent advances in the treatment of malignant astrocytoma. J Clin Oncol 2006; 24:1253-65.

Rogers LR, Rock JP, Sills AK et al.(2006) Results of a phase II trial of the GliaSite radiation therapy system for the treatment of newly diagnosed, resected single brain metastases. J Neurosurg 2006; 105(3):375-84.

Rostomily RC, Halligan J, et al.(2001) Permanent low-activity (125) I seed placement for the treatment of pediatric brain tumors: preliminary experience. Pediatr Neurosurg 2001; 34:198-205.

Selker RG, Shapiro WR, et al.(2002) The Brain Tumor Cooperative Group NIH Trial 87-01: a randomized comparison of surgery, external radiotherapy, and carmustine versus surgery, interstitial radiotherapy boost, external radiation therapy, and carmustine. Neurosurgery 2002; 51:343-55.

Tatter SB, Shaw EG, Rosenblum ML 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 2003; 99(2):297-303.

U.S. Securities and Exchange Commission. Form 8-K: IsoRay, Inc. Commission File No. 0001-33407; 2016.

Waters JD, Rose B, Gonda DD et al.(2013) Immediate post-operative brachytherapy prior to irradiation and temozolomide for newly diagnosed glioblastoma. J Neurooncol 2013; 113(3):467-77.

Welsh J, Sanan A, Gabayan AJ 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 2007; 68(1):159-65.

Wernicke AG, Sherr DL, Schwartz TH et al.(2010) The role of dose escalation with intracavitary brachytherapy in the treatment of localized CNS malignancies: outcomes and toxicities of a prospective study. Brachytherapy 2010; 9(1):91-9.


Group specific policy will supersede this policy when applicable. This policy does not apply to the Wal-Mart Associates Group Health Plan participants or to the Tyson Group Health Plan participants.
CPT Codes Copyright © 2024 American Medical Association.