Effect of a Time Delay for Concomitant Chemoradiation After Surgery for Newly Diagnosed Glioblastoma: A Single-Institution Study with Subgroup Analysis According to the Extent of Tumor Resection

2020 ◽  
Vol 133 ◽  
pp. e640-e645 ◽  
Author(s):  
Stephen Ahn ◽  
Jae-Sung Park ◽  
Jin Ho Song ◽  
Sin-Soo Jeun ◽  
Yong-Kil Hong
2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi9-vi9
Author(s):  
Stephen Ahn ◽  
Jae-Sung Park ◽  
Yong Kil Hong ◽  
Seung Ho Yang ◽  
Sin-Soo Jeun

Abstract Several studies have been conducted to determine the relationship between post-treatment total lymphocyte count (TLC) and overall survival (OS) in patients with malignant tumors including glioblastomas (GBMs). In this retrospective study, whether patients with newly diagnosed GBM experience significant lymphopenia after concomitant chemoradiation (CCRT) was evaluated, and whether TLC after this treatment is associated with OS in the treated population was examined. Using electronic medical records, all patients newly diagnosed with GBM between 2008 and 2016 at Seoul St. Mary’s Hospital were retrospectively examined. The eligible criteria included the following: 1) craniotomy with surgical resection or biopsy, 2) completion of CCRT, 3) accessible baseline and/or follow-up complete blood count (CBC). Median TLC significantly decreased after completion of CCRT, compared to TLC at baseline (1,742 versus 1,319 cells/mm3, P-value < 0.001). Patients with TLC < 1,200 cells/mm3 at 4 weeks after the completion of CCRT showed shorter survival than those with TLC ≥ 1,200 cells/mm3 with median OS of 14.5 versus 21.0 months (P-value = 0.017). Also, in multivariate analysis for OS, TLC < 1,200 cells/mm3 at 4 weeks after the completion of CCRT (HR 1.97, 95% CI 1.61 – 2.25, P-value = 0.004) were significantly associated with shorter survival. The results from the present study indicate that treatment-related total lymphocyte counts after CCRT is associated with worse survival in patients with newly diagnosed GBM.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. e14535-e14535
Author(s):  
Rachel Grossman ◽  
Felix Bokstein ◽  
Deborah T. Blumenthal ◽  
Dror Limon ◽  
Carmit Ben Harush ◽  
...  

e14535 Background: Tumor Treating Fields (TTFields), a non-invasive, loco-regional, anti-mitotic treatment comprises low-intensity alternating electric fields. In the phase III EF-14 study in newly diagnosed glioblastoma (ndGBM), TTFields in combination with temozolomide (TMZ) significantly improved survival compared to TMZ alone. In preclinical studies TTFields had a radiosensitizing effect and increased the efficacy of radiation therapy (RT). This pilot study [NCT03780569] evaluated the feasibility and safety of TTFields administered concurrently with RT and TMZ in ndGBM patients. Methods: Patients with histologically confirmed ndGBM were treated with TTFields/RT/TMZ followed by adjuvant TMZ/TTFields. TTFields (200 kHz) were delivered for ≥18 hours/day with transducer arrays removed during RT delivery. RT was administered to the tumor bed in 30 fractions (total dose 60 Gy) in combination with daily TMZ (75 mg/m2). In the adjuvant phase, patients received monthly TMZ (150–200 mg/m2 for 5 days) plus TTFields. The primary outcome was safety of the combined therapies; secondary outcomes included progression-free survival (PFS) and overall survival (OS). Adverse events (AEs) were graded per CTCAE v4.0. Results: Ten patients were enrolled at a single center between April and December 2017. Median age was 60.2 years, median Karnofsky Performance Score was 90.0, and eight (80%) patients were male. Five (50%) patients had undergone tumor resection while the remainder had biopsy only. Eight patients experienced ≥1 RT treatment delay; delays were unrelated to TTFields treatment. All patients experienced ≥1 AE. Three patients suffered from serious AEs (urinary tract infection, confusional state, and decubitus ulcer) that were considered unrelated to TTFields. The most common AE was skin toxicity, reported in eight (80%) patients; all were of low severity (CTCAE grade 1–2) and were reported as related to TTFields treatment. Median PFS from enrollment was 8.9 months; median OS was not reached at the time of study closure. Conclusions: Eighty percent of patients experienced grade 1–2 TTFields-related skin toxicity. No other TTFields-related toxicities were observed and there was no increase in RT- or TMZ-related toxicities as a result of combining TTFields with these therapies. Based on the safety and preliminary efficacy results of this pilot study, a phase 2 randomized trial (N = 60; NCT03869242) and the Phase 3 TRIDENT trial have been initiated to further investigate the efficacy of concomitant RT/TMZ/TTFields in ndGBM. Clinical trial information: NCT03780569.


2014 ◽  
Vol 32 (15_suppl) ◽  
pp. 2082-2082
Author(s):  
Henry S. Friedman ◽  
James Emmett Herndon ◽  
Frances McSherry ◽  
Arliene Ravelo ◽  
Eric S Lipp ◽  
...  

2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi108-vi108
Author(s):  
Lindsay J Lipinski ◽  
Ajay Abad ◽  
Robert A Fenstermaker ◽  
Andrew J Fabiano ◽  
Dheerendra Prasad ◽  
...  

Abstract Glioblastoma is an aggressive tumor that is clinically and pathologically heterogeneous, and as such, remains challenging to treat. Response to initial standard treatment is widely variable and patient specific. We describe a cohort of 11 newly diagnosed patients enrolled in the 3D-PREDICT study at a single institution who underwent maximal safe surgical resection with tumor tissue collected prospectively for ex vivo cell culture assays against a panel of commonly used agents, including temozolomide. Nine of eleven patients received concurrent radiation + temozolomide followed by adjuvant temozolomide as per current standard of care. Two patients progressed immediately following concurrent chemoradiation; pseudoprogression was ruled out with short interval repeat imaging and clinical deterioration with pathology confirming recurrent tumor on re-resection in one patient. Median follow-up was 10 months (range 2-18). Outcomes were assessed retrospectively using Kaplan-Meier time-to-event curves, separating into two groups the temozolomide responders (n=4) and temozolomide nonresponders (n=7), as defined by the previously validated assay. The event was reached when radiographic tumor recurrence/progression occurred. Due to limited sample size, statistical significance was not reached, but a trend toward longer time to recurrence was noted among the temozolomide responder group. Continued experience with this tool may help clinicians predict which patients with newly diagnosed glioblastoma will respond well to initial treatment with temozolomide and those that may be more appropriate for clinical trial enrollment, independent of MGMT promoter methylation status. Longer term studies with a larger number of patients will help to determine the true significance of this drug response prediction assay.


2022 ◽  
Vol 10 (1) ◽  
Author(s):  
Carine Jiguet-Jiglaire ◽  
Sebastien Boissonneau ◽  
Emilie Denicolai ◽  
Victoria Hein ◽  
Romain Lasseur ◽  
...  

AbstractWe previously identified matrix metalloproteinase 2 (MMP2) and MMP9 plasma levels as candidate biomarkers of bevacizumab activity in patients with recurrent glioblastoma. The aim of this study was to assess the predictive value of MMP2 and MMP9 in a randomized phase III trial in patients with newly diagnosed glioblastoma and to explore their tumor source. In this post hoc analysis of the AVAglio trial (AVAGlio/NCT00943826), plasma samples from 577 patients (bevacizumab, n = 283; placebo, n = 294) were analyzed for plasma MMP9 and MMP2 levels by enzyme-linked immunosorbent assay. A prospective local cohort of 38 patients with newly diagnosed glioblastoma was developed for analysis of tumor characteristics by magnetic resonance imaging and measurement of plasma and tumor levels of MMP9 and MMP2. In this AVAglio study, MMP9, but not MMP2, was correlated with bevacizumab efficacy. Patients with low MMP9 derived a significant 5.2-month overall survival (OS) benefit with bevacizumab (HR 0.51, 95% CI 0.34–0.76, p = 0.0009; median 13.6 vs. 18.8 months). In multivariate analysis, a significant interaction was seen between treatment and MMP9 (p = 0.03) for OS. In the local cohort, we showed that preoperative MMP9 plasma levels decreased after tumor resection and were correlated with tumor levels of MMP9 mRNA (p = 0.03). However, plasma MMP9 was not correlated with tumor size, invasive pattern, or angiogenesis. Using immunohistochemistry, we showed that MMP9 was expressed by inflammatory cells but not by tumor cells. After cell sorting, we showed that MMP9 was expressed by CD45+ immune cells. Finally, using flow cytometry, we showed that MMP9 was expressed by tumor-infiltrating neutrophils. In conclusion, circulating MMP9 is predictive of bevacizumab efficacy and is released by tumor-infiltrating neutrophils.


Sign in / Sign up

Export Citation Format

Share Document