Scalp-Sparing Radiation With Concurrent Temozolomide and Tumor Treating Fields (SPARE) for Patients With Newly Diagnosed Glioblastoma

Ryan Miller, Andrew Song, Ayesha Ali, Muneeb Niazi, Voichita Bar-Ad, Nina Martinez, Jon Glass, Iyad Alnahhas, David Andrews, Kevin Judy, James Evans, Christopher Farrell, Maria Werner-Wasik, Inna Chervoneva, Michele Ly, Joshua Palmer, Haisong Liu, Wenyin Shi, Ryan Miller, Andrew Song, Ayesha Ali, Muneeb Niazi, Voichita Bar-Ad, Nina Martinez, Jon Glass, Iyad Alnahhas, David Andrews, Kevin Judy, James Evans, Christopher Farrell, Maria Werner-Wasik, Inna Chervoneva, Michele Ly, Joshua Palmer, Haisong Liu, Wenyin Shi

Abstract

Introduction: Standard-of-care treatment for patients with newly diagnosed glioblastoma (GBM) after surgery or biopsy includes concurrent chemoradiation followed by maintenance temozolomide (TMZ) with tumor treating fields (TTFields). Preclinical studies suggest TTFields and radiotherapy work synergistically. We report the results of our trial evaluating the safety of TTFields used concurrently with chemoradiation.

Methods: This is a single-arm pilot study (clinicaltrials.gov Identifier: NCT03477110). Adult patients (age ≥ 18 years) with newly diagnosed glioblastoma and a Karnofsky performance score (KPS) of ≥ 60 were eligible. All patients received concurrent scalp-sparing radiation (60 Gy in 30 fractions) with TMZ (75 mg/m2 daily) and TTFields (200 kHz). Maintenance therapy included TMZ and continuation of TTFields. Scalp-sparing radiation treatment was used to reduce radiation dermatitis. Radiation treatment was delivered through the TTFields arrays. The primary endpoint was safety and toxicity of tri-modality treatment within 30 days of completion of chemoradiation treatment.

Results: There were 30 patients enrolled, including 20 (66.7%) men and 10 (33.3%) women, with a median age of 58 years (range 19 to 77 years). Median KPS was 90 (range 70 to 100). A total of 12 (40%) patients received a gross total resection and 18 (60%) patients had a subtotal resection. A total of 12 (40%) patients had multifocal disease at presentation. There were 20 (66.7%) patients who had unmethylated O(6)-methylguanine-DNA-methyltransferase (MGMT) promotor status and 10 (33.3%) patients who had methylated MGMT promoter status. Median follow-up was 15.2 months (range 1.7 to 23.6 months). Skin adverse events were noted in 83.3% of patients, however, these were limited to Grade 1 or 2 events, which resolved spontaneously or with topical medications. The primary end point was met; no TTFields discontinuation occurred during the evaluation period due to high grade scalp toxicity. A total of 27 (90%) patients had progression, with a median progression-free survival (PFS) of 9.3 months (95% confidence interval (CI): 8.5-11.6 months). The 1-year progression-free survival was 23% (95% CI: 12%-45%). The median overall survival (OS) was 15.8 months (95% CI: 12.5 months-infinity). The 1-year overall survival was 66% (95% CI: 51%-86%).

Conclusions: Concurrent TTFields with scalp-sparing chemoradiation is a feasible and well-tolerated treatment option with limited toxicity. A phase 3, randomized clinical trial (EF-32, clinicaltrials.gov Identifier: NCT04471844) investigating the clinical benefit of concurrent TTFields with chemoradiation treatment is currently enrolling.

Clinical trial registration: Clinicaltrials.gov, identifier NCT03477110.

Keywords: TTFields; concurrent therapy; glioblastoma; radiotherapy; scalp-sparing radiation.

Conflict of interest statement

WS: Consulting for Brainlab, Varian, and Novocure; research funding for clinical trial from Novocure and Regeneron. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Miller, Song, Ali, Niazi, Bar-Ad, Martinez, Glass, Alnahhas, Andrews, Judy, Evans, Farrell, Werner-Wasik, Chervoneva, Ly, Palmer, Liu and Shi.

Figures

Figure 1
Figure 1
Kaplan-Meier estimates of progression-free survival (A) and overall survival (B) (both represented by solid lines). The dashed lines represent the corresponding 95% confidence pointwise confidence intervals.
Figure 2
Figure 2
Kaplan-Meier estimates of progression-free survival (A) and overall survival (B) by methylation status. In both figure panels, the black line represents unmethylated MGMT promoter patients and the blue line represents methylated MGMT promoter patients.
Figure 3
Figure 3
Quality of life as measured by the EORTC Core Quality of Life questionnaire (QLQ-C30 version 3) and brain cancer-specific health-related quality of life questionnaire (QLQ-BN20). Individual scales are shown on the Y-axis. The percentage change from baseline questionnaire administration to concurrent phase questionnaire administration (at approximately Week 3) is shown on the X-axis, including the range (black line represents minimum and gray line represents maximum), as well as the median (represented by black circle).

References

    1. National Comprehensive Cancer Network . Central Nervous System Cancers (2021). Available at: (Accessed March 1, 2022).
    1. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJB, et al. . Radiotherapy Plus Concomitant and Adjuvant Temozolomide for Glioblastoma. N Engl J Med (2005) 352:987–96. doi: 10.1056/NEJMoa043330
    1. Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJB, Janzer RC, et al. . Effects of Radiotherapy With Concomitant and Adjuvant Temozolomide Versus Radiotherapy Alone on Survival in Glioblastoma in a Randomised Phase III Study: 5-Year Analysis of the EORTC-NCIC Trial. Lancet Oncol (2009) 10:459–66. doi: 10.1016/S1470-2045(09)70025-7
    1. Stupp R, Taillibert S, Kanner AA, Kesari S, Steinberg DM, Toms SA, et al. . Maintenance Therapy With Tumor-Treating Fields Plus Temozolomide vs Temozolomide Alone for Glioblastoma: A Randomized Clinical Trial. JAMA (2015) 314:2535–43. doi: 10.1001/jama.2015.16669
    1. Stupp R, Taillibert S, Kanner A, Read W, Steinberg DM, Lhermitte B, et al. . Effect of Tumor-Treating Fields Plus Maintenance Temozolomide vs Maintenance Temozolomide Alone on Survival in Patients With Glioblastoma: A Randomized Clinical Trial. JAMA (2017) 318:2306–16. doi: 10.1001/jama.2017.18718
    1. Kirson ED, Gurvich Z, Schneiderman R, Dekel E, Itzhaki A, Wasserman Y, et al. . Disruption of Cancer Cell Replication by Alternating Electric Fields. Cancer Res (2004) 64:3288–95. doi: 10.1158/0008-5472.CAN-04-0083
    1. Kirson ED, Dbalý V, Tovarys F, Vymazal J, Soustiel JF, Itzhaki A, et al. . Alternating Electric Fields Arrest Cell Proliferation in Animal Tumor Models and Human Brain Tumors. Proc Natl Acad Sci USA (2007) 104:10152–7. doi: 10.1073/pnas.0702916104
    1. Fonkem E, Wong ET. NovoTTF-100A: A New Treatment Modality for Recurrent Glioblastoma. Expert Rev Neurother (2012) 12:895–9. doi: 10.1586/ern.12.80
    1. Gutin PH, Wong ET. Noninvasive Application of Alternating Electric Fields in Glioblastoma: A Fourth Cancer Treatment Modality. Am Soc Clin Oncol Educ Book (2012) 32:126–31. doi: 10.14694/EdBook_AM.2012.32.122
    1. Bokstein F, Blumenthal D, Limon D, Harosh CB, Ram Z, Grossman R. Concurrent Tumor Treating Fields (TTFields) and Radiation Therapy for Newly Diagnosed Glioblastoma: A Prospective Safety and Feasibility Study. Front Oncol (2020) 10:411. doi: 10.3389/fonc.2020.00411
    1. Toms SA, Kim CY, Nicholas G, Ram Z. Increased Compliance With Tumor Treating Fields Therapy Is Prognostic for Improved Survival in the Treatment of Glioblastoma: A Subgroup Analysis of the EF-14 Phase III Trial. J Neurooncol (2019) 141:467–73. doi: 10.1007/s11060-018-03057-z
    1. Giladi M, Munster M, Schneiderman RS, Voloshin T, Porat Y, Blat R, et al. . Tumor Treating Fields (TTFields) Delay DNA Damage Repair Following Radiation Treatment of Glioma Cells. Radiat Oncol (2017) 12:206. doi: 10.1186/s13014-017-0941-6
    1. Kim EH, Kim YH, Song HS, Jeong YK, Lee JY, Sung J, et al. . Biological Effect of an Alternating Electric Field on Cell Proliferation and Synergistic Antimitotic Effect in Combination With Ionizing Radiation. Oncotarget (2016) 7:62267–79. doi: 10.18632/oncotarget.11407
    1. Li T, Shukla G, Peng C, Lockamy V, Liu H, Shi W. Dosimetric Impact of a Tumor Treating Fields Device for Glioblastoma Patients Undergoing Simultaneous Radiation Therapy. Front Oncol (2018) 8:51. doi: 10.3389/fonc.2018.00051
    1. Song A, Bar-Ad V, Martinez N, Glass J, Andrews DA, Judy K, et al. . Initial Experience With Scalp Sparing Radiation With Concurrent Temozolomide and Tumor Treatment Fields (SPARE) for Patients With Newly Diagnosed Glioblastoma. J Neuro-Oncol (2020) 147:653–61. doi: 10.1007/s11060-020-03466-z
    1. Cabrera AR, Kirkpatrick JP, Fiveash JB, Shih HA, Koay EJ, Lutz S, et al. . Radiation Therapy for Glioblastoma: Executive Summary of an American Society for Radiation Oncology Evidence-Based Clinical Practice Guideline. Pract Radiat Oncol (2016) 6:217–25. doi: 10.1016/j.prro.2016.03.007
    1. Lacouture ME, Davis ME, Elzinga G, Butowski N, Tran D, Villano JL, et al. . Characterization and Management of Dermatologic Adverse Events With the NovoTTF-100a System, a Novel Anti-Mitotic Electric Field Device for the Treatment of Recurrent Glioblastoma. Semin Oncol (2014) Suppl 4:S1–S14. doi: 10.1053/j.seminoncol.2014.03.011
    1. Wen PY, Macdonald DR, Reardon DA, Cloughesy TF, Sorensen G, Galanis E, et al. . Updated Response Assessment Criteria for High-Grade Gliomas: Response Assessment in Neuro-Oncology Working Group. J Clin Oncol (2010) 28:1963–72. doi: 10.1200/JCO.2009.26.3541
    1. Basch E, Reeve BB, Mitchell SA, Clauser SB, Minasian LM, Dueck AC, et al. . Development of the National Cancer Institute’s Patient-Reported Outcomes Version of the Common Terminology Criteria for Adverse Events (PRO- CTCAE). J Natl Cancer Inst (2014) 106:dju244. doi: 10.1093/jnci/dju244
    1. Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT, Vogelbaum MA, et al. . A Randomized Trial of Bevacizumab for Newly Diagnosed Glioblastoma. N Engl J Med (2014) 370:699–708. doi: 10.1056/NEJMoa1308573
    1. Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishikawa R, et al. . Bevacizumab Plus Radiotherapy-Temozolomide for Newly Diagnosed Glioblastoma. N Engl J Med (2014) 370:709–22. doi: 10.1056/NEJMoa1308345
    1. Glas M, Scheffler B, Lazaridis L, Herrlinger U, Pierscianek D, Sure U, et al. . P01.060 PriCoTTF: A Phase I/II Trial of Tumor Treating Fields Prior and Concomitant to Radiotherapy in Newly Diagnosed Glioblastoma. Neuro Onco (2018) 20(Suppl 3):iii243. doi: 10.1093/neuonc/noy139.102
    1. Shi W, Kleinberg L, Jeyapalan SA, Goldlust SA, Nagpal S, Reardon DA, et al. . Phase III TRIDENT Trial: Radiation and Temozolomide +/- Tumor Treating Fields in Newly Diagnosed Glioblastoma. J Clin Oncol (2020) 38(15_suppl):TPS2580-TPS2580. doi: 10.1200/JCO.2020.38.15_suppl.TPS2580
    1. Alnahhas I, Alsawas M, Rayi A, Palmer JD, Raval R, Ong S, et al. . Characterizing Benefit From Temozolomide in MGMT Promoter Unmethylated and Methylated Glioblastoma: A Systematic Review and Meta-Analysis. Neuro-Oncol Adv (2020) 3:vdab095. doi: 10.1093/noajnl/vdaa082
    1. Syed M, Liermann J, Verma V, Bernhardt D, Bougatf N, Paul A, et al. . Survival and Recurrence Patterns of Multifocal Glioblastoma After Radiation Therapy. Cancer Manag Res (2018) 10:4229–35. doi: 10.2147/CMAR.S165956
    1. Onken J, Staub-Bartelt F, Vajkoczy P, Misch M. Acceptance and Compliance of TTFields Treatment Among High Grade Glioma Patients. J Neurooncol (2018) 139:177–84. doi: 10.1007/s11060-018-2858-9

Source: PubMed

3
Se inscrever