ArticlePDF Available

Accelerated hyperfractionation plus temozolomide in glioblastoma

Authors:

Abstract and Figures

Introduction: Hyperfractionated (HFRT) or accelerated hyperfractionated radiotherapy (AHFRT) have been discussed as a potential treatment for glioblastoma based on a hypothesized reduction of late radiation injury and prevention of repopulation. HFRT and AHFRT have been examined extensively in the pre-Temozolomide era with inconclusive results. In this study we examined the role of accelerated hyperfractionation in the Temozolomide era. Materials and methods: Sixty-four patients who underwent AHFRT (62 of which received Temozolomide) were compared to 67 patients who underwent normofractionated radiotherapy (NFRT) (64 of which received TMZ) between 02/2009 and 10/2014. Follow-up data were analyzed until 01/2015. Results: Median progression-free survival (PFS) was 6 months for the entire cohort. For patients treated with NFRT median PFS was 7 months, for patients treated with AHFRT median PFS was 6 months. Median overall survival (OS) was 13 months for all patients. For patients treated with NFRT median OS was 15 months, for patients treated with AHFRT median OS was 10 months. The fractionation regimen was not a predictor of PFS or OS in univariable- or multivariable analysis. There was no difference in acute toxicity profiles between the two treatment groups. Conclusions: Univariable and multivariable analysis did not show significant differences between NFRT and AHFRT fractionation regimens in terms of PFS or OS. The benefits are immanent: the regimen does significantly shorten hospitalization time in a patient collective with highly impaired life expectancy. We propose that the role of AHFRT + TMZ should be further examined in future prospective trials.
Content may be subject to copyright.
R E S E A R C H Open Access
Accelerated hyperfractionation plus
temozolomide in glioblastoma
David Kaul
*
, Julian Florange
, Harun Badakhshi, Arne Grün, Pirus Ghadjar, Sebastian Exner and Volker Budach
Abstract
Introduction: Hyperfractionated (HFRT) or accelerated hyperfractionated radiotherapy (AHFRT) have been discussed
as a potential treatment for glioblastoma based on a hypothesized reduction of late radiation injury and prevention
of repopulation. HFRT and AHFRT have been examined extensively in the pre-Temozolomide era with inconclusive
results. In this study we examined the role of accelerated hyperfractionation in the Temozolomide era.
Materials and methods: Sixty-four patients who underwent AHFRT (62 of which received Temozolomide) were
compared to 67 patients who underwent normofractionated radiotherapy (NFRT) (64 of which received TMZ)
between 02/2009 and 10/2014. Follow-up data were analyzed until 01/2015.
Results: Median progression-free survival (PFS) was 6 months for the entire cohort. For patients treated with NFRT
median PFS was 7 months, for patients treated with AHFRT median PFS was 6 months. Median overall survival (OS)
was 13 months for all patients. For patients treated with NFRT median OS was 15 months, for patients treated with
AHFRT median OS was 10 months. The fractionation regimen was not a predictor of PFS or OS in univariable- or
multivariable analysis. There was no difference in acute toxicity profiles between the two treatment groups.
Conclusions: Univariable and multivariable analysis did not show significant differences between NFRT and AHFRT
fractionation regimens in terms of PFS or OS. The benefits are immanent: the regimen does significantly shorten
hospitalization time in a patient collective with highly impaired life expectancy. We propose that the role of
AHFRT + TMZ should be further examined in future prospective trials.
Introduction
Gliomas are the most common primary tumors of the
central nervous system (CNS) in adults representing about
one third of central nervous system tumors and 81 % of
all malignant CNS tumors reported in the United States
[1]. The most common and most malignant type of glioma
is glioblastoma (GBM), with a median overall survival
(OS) rate of 15 months after surgical resection followed by
adjuvant radiotherapy (RT) and Temozolomide (TMZ)
chemotherapy. The prevalence of GBM is highest in pa-
tients aged 50 years or older and is likely to increase with
the ongoing demographic shift toward older ages [2].
Well-known postitive prognostic factors for OS in GBM
patients are young age at diagnosis, high Karnofsky
performance score (KPS), great extent of neurosurgical
resection, O-6-methylguanine-DNA methyltransferase- gene
(MGMT) methylation as well as isocitrate dehydrogenase
(IDH) 1-mutational status [35]. Current standard of care
for newly diagnosed GBM comprises maximal safe resec-
tion, adjuvant radiotherapy with (RT) with concurrent TMZ
and post-RT TMZ chemotherapy [6, 7]. Fractionated RT to
the tumor bed in 30 fractions of 2 Gy in single doses of
2 Gy to a total accumulated dose of 60 Gy delivered over
the course of 6 weeks has been widely accepted as the
standard fractionation regimen, balancing effectiveness with
radiation toxicity. Recently some authors have suggested
hypofractionated regimens for the elderly and frail patient
population [8, 9] other authors have evaluated the role of
hypofractionation plus TMZ [10].
Other authors have examined the potential role of
hyperfractionated- (HFRT) and accelerated hyperfractio-
nated radiotherapy (AHFRT) as well as the role of protons
in GBM [11]. The use of HFRT and AHFRT is based on a
hypothesized reduction of late radiation injury and pre-
vention of tumor repopulation in treatment intervals [12].
* Correspondence: david.kaul@charite.de
Equal contributors
Klinik für Radioonkologie und Strahlentherapie, Charité Universitätsmedizin
Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin,
Germany
© 2016 Kaul et al. Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0
International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and
reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver
(http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
Kaul et al. Radiation Oncology (2016) 11:70
DOI 10.1186/s13014-016-0645-3
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Despite plausible rationales, various trials have failed
to prove the superiority of dose-escalated HFRT and
AHFRT in the pre-TMZ era [13].
In 1994, the European Organization for the Research and
Treatment of Cancer (EORTC) reported an AHFRT dose
escalation trial using doses of 4260 Gy in 2 Gy fractions
three times daily, which failed to show differences in survival
in all groups. No additional chemotherapy was used [13]. In
1999 Lutterbach et. al. showed survival rates for 1.5 Gy
thricedailyto54GycomparabletoconventionalRT,again
no chemotherapy was used [14]. In 2001 Prados et. al.
showed data for AHFRT with or without difluromethylor-
nithine (DFMO) vs. conventional irradiation with or without
DFMO with no OS benefit for the experimental groups [15].
The RTOG 8302 study tested HFRT (2 × 1.2 Gy to
doses of 64.8, 72, 76.8, or 81.6 Gy) vs. AHFRT (2 ×1.6 Gy
to doses of 48 or 54.4 Gy), all groups received concurrent
bis-chloroethyl (BCNU). Contrary to the other aforemen-
tioned studies HFRT patients who had received higher
doses of 76.8 and 81.6 Gy showed superior survival com-
pared to the AHFRT groups [16].
In summary, the data on HFRT and AHFRT mainly
stem from the pre-TMZ era and are not fully conclusive.
We therefore want to present experience from our insti-
tution on the treatment of patients with newly diagnosed
GBM with AHFRT of 2 × 1.6 Gy to 59,2 Gy and concurrent
and sequential Temozolomide following the Stupp regimen.
Apart from a potential reduction of tumor repopulation as
well as a hypothesized reduced late toxicity rate, the regi-
men does significantly shorten hospitalization time in a
group of patients with highly impaired life expectancy.
Materials and methods
Treatment decisions, patient selection and dose regimens
Starting from 01/2009 patients with resected GBM with
organs-at-risk (OAR) in close proximity to the resection
cavity were offered adjuvant radio-chemotherapy (RCTx)
with single doses of 1.6 Gy twice daily to a total dose of
59.2 Gy (19 days schedule) as an alternative to a conven-
tional fractionation with single doses of 2 Gy up to 60 Gy
(30 days schedule, NFRT). Of 131 patients 126 received
continuous daily TMZ (75 mg per square meter of body-
surface area per day, 7 days per week from the first to the
last day of radiotherapy), followed by six cycles of adjuvant
TMZ (150 mg per square meter for 5 days during each
28-day cycle).
In this study we carried out a retrospective analysis of 64
patients who underwent AHFRT plus TMZ and compared
them with 67 patients who underwent NFRT plus TMZ be-
tween 02/2009 and 10/2014. Follow-up data were analyzed
until 01/2015.
In our institution treatment decisions are based on the
votes of an interdisciplinary tumor board. Usually all
patients <70 years with a KPS >50 % are offered adjuvant
AHFRT + TMZ or NFRT + TMZ. AHFRT + TMZ is of-
fered when OARs such as the optic nerves, chiasm or
brainstem would be touched by the CTV and covered by
the PTV, and in case that the patient is willing and fit
enough to undergo treatment twice daily.
Patients 70 yeas of age either receive hypofractionated
radiotherapy or TMZ only (depending on MGMT-status).
Stratification, variables and follow-up
Patients were stratified according to fractionation scheme,
age, gender, KPS, extent of surgery (biopsy, partial-, gross
total resection), MGMT-status, tumor localization (frontal,
parietal, temporal, occipital, central) and planning target
volume (PTV). Follow-up examinations, including MRI as
well as clinical and neurologic examinations were per-
formed at 68 week intervals after radiotherapy.
Treatment planning
Target delineation in GBM varies substantially between dif-
ferent institutions and several consensus statements are
available. However, an ESTRO-ACROP guideline is avail-
able since January 2016 [17]. Adjuvant RCTx was initiated
within 4 weeks after surgical resection or stereotactic bi-
opsy. Contrast agent enhanced computed tomography in a
thermoplastic mask as well as gadolinium enhanced mag-
netic resonance imaging (MRI) was performed before RT
planning.
Target volumes were based on preoperative and postop-
erative MRI. The gross tumor volume (GTV) was defined
as the summation of the postoperative surgical cavity with
or without residual tumor lesion(s) as well as tumor exten-
sion on the preoperative T1-weighted gadolinium-
enhanced imaging. The diffusion-weighted imaging (DWI)
images were also used in the estimation of GTV. The ex-
tent of peritumoral edema was not routinely included in
the clinical target volume (CTV), however, an all-round
GTV margin of 2 cm was mandatory. For the planning tar-
get volume (PTV) an additional 0.5 cm margin was added.
Intensity-modulated radiation therapy (IMRT) was applied
using a 6-MV linear accelerator with multileaf collimators.
Until 2012 treatment was performed using step-and-shoot
intensity-modulated radiation therapy (IMRT), starting in
early 2012 all patients were treated using volumetric arc
therapy (VMAT).
Toxicity
Higher grade acute toxicity (3°) was analyzed for 90 days
post treatment according to CTCAE 4.0.
Formulas and statistics
Overall survival (OS) and progression-free survival
(PFS) were calculated from the first day of irradiation
Kaul et al. Radiation Oncology (2016) 11:70 Page 2 of 7
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
using Kaplan-Meier analysis and the log-rank test.
Progression was defined retrospectively by clinical
note assessments that included integration of imaging
and clinical status. Subgroups were compared using uni-
variable analysis and the Cox proportional hazard model
for multivariable analysis. A p-value of less than 0.05 was
considered statistically significant. A p-value of less than 0.1
was considered a trend. All variables from the univariable
analysis were included in multivariable analysis. All statis-
tical analyses were performed using IBM SPSS Statistics 19
(New York, USA).
Results
Patient characteristics
Patient characteristics are shown in Table 1. One hun-
dred thirty-one patients treated for GBM were identified
in our retrospective analysis. Sixty-seven were treated
with NFRT and 64 patients were treated using AHFRT.
The two groups were well matched in terms of gender,
PTV, tumor localization, MGMT-status, extent of sur-
gery, KPS and TMZ treatment and salvage treatment.
Median age in the AHFRT group was lower than in the
NFRT group (p< 0.001).
Table 1 Patient characteristics of the 131 GBM patients analyzed
Overall Collective NFRT AHFRT p-value
(n= 131) (n= 67) (n= 64)
Median Age (min/max) [y] 61 12/80 63 43/78 59 12/80 p< 0.001 (*)
Mean PTV ± sd [ccm] 355 ±142 339 ±141.4 373 ±141.8 p= 0.17
n% n%n%
Gender m 88 67.2 % 46 68.7 % 42 65.6 % p= 0.85
f 43 32.8 % 21 31.3 % 22 34.4 %
Localization Frontal 42 32.1 % 16 23.9 % 26 40.6 % p= 0.38
Parietal 31 23.7 % 17 25.4 % 14 21.9 %
Temporal 38 29.0 % 22 32.8 % 16 25.0 %
Occipital 9 6.9 % 4 6.0 % 5 7.8 %
Central 9 6.9 % 6 9.0 % 3 4.7 %
n/a 2 1.5 % 2 3.0 % 0 0.0 %
MGMT-status unmethylated 63 48.1 % 32 47.8 % 31 48.4 % p= 0.66
methylated 43 32.8 % 23 34.3 % 20 31.3 %
n/a 25 19.1 % 12 17.9 % 13 20.3 %
Extent of surgery Biopsy 16 12.2 % 6 9.0 % 10 15.6 % p= 0.38
Partial resection 57 43.5 % 28 41.8 % 29 45.3 %
Gross tumor resection 51 38.9 % 29 43.3 % 22 34.4 %
n/a 7 5.3 % 4 6.0 % 3 4.7 %
KPS 50 % 7 5.3 % 4 6 % 3 4.7 % p= 0.3
60 % 49 37.4 % 27 40 % 22 34.4 %
70 % 47 35.9 % 24 36 % 23 35.9 %
80 % 28 21.4 % 12 18 % 16 25.0 %
Temozolomide yes 126 96.2 % 65 97.0 % 61 95.3 % p= 0.68
no 5 3.8 % 2 3.0 % 3 4.7 %
Salvage treatment Re-irradiation 20 15.3 % 12 17.9 % 8 12.5 %
Chemotherapy (tmz) 45 34.4 % 24 35.8 % 21 32.8 %
Chemotherapy (other) 6 4.6 % 3 4.5 % 3 4.7 %
Bevacizumab 11 8.4 % 5 7.5 % 6 9.4 %
Imatinib 1 0.8 % 0 0.0 % 1 1.6 %
Dendritic cell vaccination 1 0.8 % 1 1.5 % 0 0.0 %
NFRT normofractionated radiotherapy, AHFRT accelerated hyperfractionated radiotherapy, PTV planning target volume, n/a not applicable, MGMT O-6-methylguanine-DNA
methyltransferase, KPS Karnofsky performance status, tmz temozolomide
Kaul et al. Radiation Oncology (2016) 11:70 Page 3 of 7
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Progression-free survival
Median PFS was 6 months for the entire cohort (Table 2).
For patients treated with NFRT median PFS was 7 months,
for patients treated with AHFRT median PFS was
6 months. At 6 months PFS was 56.9 % in the NFRT
group and 51.7 % in the AHFRT group. At 12 months PFS
was 16.9 % in the NFRT group and 19 % in the AHFRT
group, (Fig. 1). There was no difference between both dose
regimens in univariable analysis (p=0.95).
Overall survival
Of 131 patients analyzed 107 had died at the time of
analysis (01/2015).
Median OS was 13 months for all patients (Table 3).
For patients treated with NFRT median OS was
15 months, for patients treated with AHFRT median OS
was 10 months. At 12 months OS was 66 % in the NFRT
group and 48.2 % in the AHFRT group. At 24 months
OS was 14.7 % in the NFRT group and 16.7 % in the
AHFRT group (Fig. 2). There was no difference between
both dose regimens in univariable analysis (p= 0.46).
Prognostic factors
Positive predictors of survival in univariable analysis were
female gender, higher KPS, MGMT methylation and gross
total resection. In multivariable analysis MGMT methyla-
tion and gross total resection remained significant
Table 2 Univariable analysis of potential preditive factors of progression-free survival
Univariable analysis Multivariable analysis
Variable HR 95 % CI pMedian PFS [m] HR 95 % CI p
Age (< vs. > = median of 61 years) 1.08 0.751.55 0.69 6 vs. 6 –– –
Gender (m vs. f) 0.68 0.461.01 0.05 6 vs. 9 0.57 0.350.92 0.022 (*)
KPS (< vs. > = median of 70 %) 0.5 0.340.72 <0.001 (*) 4 vs. 9 0.5 0.330.78 0.002 (*)
MGMT-status (methylated vs. unmethylated) 1.46 0.972.2 0.07 9 vs. 6 1.61 1.032.52 0.036 (*)
Localization (other vs. central) 1.51 0.763 0.24 6 vs. 5 –– –
PTV (< vs. > = median of 337 ccm) 1.13 0.791.62 0.51 7 vs. 6 –– –
Subtotal resection or biopsy vs. gross total resection 0.71 0.491.02 0.07 4 vs. 8 –– –
Fractionation regimen (NFRT vs. AHFRT) 1.01 0.951.01 0.95 7 vs. 6 –– –
(*) p-value 0.05, HR hazard ratio, CI confidence interval, PFS progression-free survival, KPS Karnofsky performance status, MGMT O-6-methylguanine-DNA methyl-
transferase, PTV planning target volume, NFRT normofractionated radiotherapy, AHFRT accelerated hyperfractionated radiotherapy
Fig. 1 Kaplan-Meier analysis of PFS rates grouped according to dose regimen. No significant differences were found between both groups
Kaul et al. Radiation Oncology (2016) 11:70 Page 4 of 7
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
predictors, the factor smaller PTVbecame significant in
multivariable analysis. Gender and lower KPS were not
significant in multivariable analysis.
The fractionation regimen was not a predictor of
survival in univariable- or multivariable analysis.
Subgroup analysis according to predictive factors did
not reveal any specific group to benefit from either NFRT
compared to AHFRT or vice versa (Table 4).
Toxicity
All patients in both groups completed radiotherapy. All pa-
tients scheduled for concurrent chemotherapy (126/131)
completed concurrent TMZ. In the normofractionated
group seven patients did not complete post-radiotherapy
TMZ due to neutropenia or thrombocytopenia. In the
hyperfractionated group 3 patients did not complete post-
radiotherapy TMZ due to neutropenia or
thrombocytopenia.
There was no difference in acute toxicity profiles
between the two treatment groups. There were seven
grade 3 and six grade 4 events in the normofractio-
nated group (grade 3 events: 1 × headache, 2 × neuro-
logical, 3 × neutropenia, 1 × thrombocytopenia. Grade
4 events: 2 × neutropenia and 4 × thrombocytopenia).
Fig. 2 Kaplan-Meier analysis of OS rates grouped according to dose regimen. No significant differences were found between both groups
Table 3 Univariable analysis of potential preditive factors of overall survival
Univariable analysis Multivariable analysis
Variable HR 95 % CI pMedian OS [m] HR 95 % CI p
Age (< vs. > = median of 61 years) 1.18 0.81.7 0.4 14 vs. 12 –– –
Gender (m vs. f) 0.62 0.40.95 0.028 (*) 11 vs. 16 0.64 0.381.08 0.095
KPS (< vs. > = median of 70 %) 0.96 0.940.98 <0.001 (*) 9 vs. 15 –– –
MGMT-status (methylated vs. unmethylated) 1.68 1.082.61 0.021 (*) 16 vs. 11 1.89 1.1583.09 0.011 (*)
Localization (other vs. central) 1.71 0.833.56 0.15 13 vs. 13 –– –
PTV (< vs. > = median of 337 ccm) 1.37 0.932.02 0.11 14 vs. 12 1.61 12.6 0.048 (*)
Subtotal resection or biopsy vs. gross total resection 0.64 0.430.95 0.025 (*) 11 vs. 15 0.62 0.390.98 0.041 (*)
Fractionation regimen (NFRT vs. AHFRT) 1.16 0.791.71 0.46 15 vs. 10 –– –
(*) p-value 0.05, HR hazard ratio, CI confidence interval, OS overall survival, KPS Karnofsky performance status, MGMT O-6-methylguanine-DNA methyltransferase,
PTV planning target volume, NFRT normofractionated radiotherapy, AHFRT accelerated hyperfractionated radiotherapy
Kaul et al. Radiation Oncology (2016) 11:70 Page 5 of 7
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
In the hyperfractionated group there were two grade 3
events and six grade 4 events (grade 3 events: 1 × neuro-
logical, 1 × nausea/vomiting. Grade 4 events: 3 × neutro-
penia, 3 × thrombocytopenia).
Discussion
Survival
Most studies on hyperfractionation and accelerated
hyperfractionation stem from the pre-TMZ era, com-
parabilityofPFSandOSratesisthuslimited.Inour
study median OS was 13 months for all patients, 15 months
for patients treated using NFRT and 10 months for patients
treated with AHFRT. Univariable and multivariable analysis
did not show significant differences between the fraction-
ation regimens. This is worthwile to know, because an
AHFRT-regimen with 3.5 weeks overall treatment time was
capable to equalize the OS-results of the classical 6 weeks
treatment. Bearing in mind the limited prognosis of these
patients the dose-intensified treatment is a clear benefit.
One of the first studies on AHFRT in GBM was pub-
lished in 1994 by González et al. who used doses of
4260 Gy in 2 Gy fractions three times a day. Median
survival was 8.7 ± 0.7 months and no statistically significant
differences were found for the four different dose-level
groups [13].
Lutterbach et. al. published median OS rates of 8.8 months
for 1.5 Gy thrice daily to 54 Gy [14].
In 2001 Prados et al. published survival rates of pa-
tients treated with AHFRT ± DFMO vs. conventional
irradiation ± DFMO with no OS benefit for the experi-
mental groups (8.69.8 months) [15].
Werner et al. published the RTOG 8302 data in
1996, patients received HFRT (2 × 1.2 Gy to doses of
64.8, 72, 76.8, or 81.6 Gy) vs. AHFRT (2 ×1.6 Gy to
doses of 48 or 54.4 Gy), all groups received concurrent
BCNU. Contrary to the other aforementioned studies
HFRT patients who had received higher doses of 76.8
and 81.6 Gy showed superior survival compared to the
AHFRT groups. The authors found median OS rates be-
tween 10.8 and 12.7 months [16].
In 2005 Stupp et al. published data demonstrating a
survival benefit for GBM patients that received concur-
rent Temozolomide with postoperative radiation, with
median survival of 14.6 months for patients receiving
concurrent therapy versus 12.1 months for patients
who received only radiotherapy [7]. This treatment has
since become the standard of care for primary GBM
and is referred to as the Stupp regimenin everyday
clinical routine.
OS rates for all patients of 13 months as shown here
are comparable to the data published by Stupp et al. and
we did not find significant differences in OS between
AHFRT and NFRT in our patient collective.
Limitations
Our study had several limitations. Firstly, the two groups
analyzed were not perfectly matched in terms of age.
Secondly, the MGMT-status is unknown in approximately
20 % of patients in both treatment groups. Thirdly, no
analysis of chronic toxicity was performed due to the in-
trinsic uncertainties of retrospective analysis. Fourthly, the
number of patients analyzed here in both groups might
simply be too low to find significant differences in survival
between the both regimens. Fifthly, patients with GBM in
close proximity to the brainstem were more likely to re-
ceive AHFRT, potentially biasing OS rates.
Conclusions
The role of AHFRT in the TMZ era remains unclear.
The potential benefits are a reduction of tumor repopu-
lation as well as reduced late toxicity. Other benefits are
immanent; the regimen does significantly shorten
hospitalization time in a patient collective with highly im-
paired life expectancy. We propose that the role of AHFRT
+ TMZ should be further examined in future prospective
trials.
Competing interests
The authors declare that they have no competing interests.
Table 4 Subgroup analysis of potential preditive factors of
overall survival did not identify any specific subgroup to benefit
from either NFRT compared to AHFRT or vice versa
Median OS [m]
NFRT AHFRT p
Variable
Age < median of 61 years 15 12 0.66
> = median of 61 years 15 9 0.28
Gender m 14 9 0.31
f 16 14 0.98
KPS < median of 70 % 12 6 0.16
> = median of 70 % 15 13 0.67
MGMT-status methylated 16 15 0.73
unmethylated 14 9 0.09
Localization other 15 10 0.41
central 9 17 0.44
PTV < median of 337 ccm 15 12 0.82
> = median of 337 ccm 15 9 0.24
Extent of resection Subtotal resection or biopsy 13 8 0.14
gross total resection 15 13 0.6
KPS Karnofsky performance status, MGMT O-6-methylguanine-DNA
methyltransferase, PTV planning target volume, NFRT normofractionated
radiotherapy, AHFRT accelerated hyperfractionated radiotherapy
Kaul et al. Radiation Oncology (2016) 11:70 Page 6 of 7
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Authorscontributions
DK drafted the manuscript, performed statistical analysis and supervised the
discussion of the manuscript. JF helped drafting the manuscript, collected
data and helped with statistical analysis. HB planned the study and took part
in the discussion of the manuscript. AG, PG and SB took part in the
discussion of the manuscript. VB planned the study and helped drafting the
manuscript. All authors approved the final version of this manuscript.
Received: 11 February 2016 Accepted: 10 May 2016
References
1. Ostrom QT, Gittleman H, Farah P, Ondracek A, Chen Y, Wolinsky Y, Stroup
NE, Kruchko C, Barnholtz-Sloan JS. CBTRUS statistical report: Primary brain
and central nervous system tumors diagnosed in the United States in
20062010. Neuro Oncol. 2013;15 Suppl 2:ii156.
2. Paszat L, Laperriere N, Groome P, Schulze K, Mackillop W, Holowaty E. A
population-based study of glioblastoma multiforme. Int J Radiat Oncol Biol
Phys. 2001;51:1007.
3. Lacroix M, Abi-Said D, Fourney DR, Gokaslan ZL, Shi W, DeMonte F, Lang FF,
McCutcheon IE, Hassenbusch SJ, Holland E, et al. A multivariate analysis of
416 patients with glioblastoma multiforme: prognosis, extent of resection,
and survival. J Neurosurg. 2001;95:1908.
4. Bauchet L, Mathieu-Daude H, Fabbro-Peray P, Rigau V, Fabbro M, Chinot O,
Pallusseau L, Carnin C, Laine K, Schlama A, et al. Oncological patterns of
care and outcome for 952 patients with newly diagnosed glioblastoma in
2004. Neuro Oncol. 2010;12:72535.
5. Leu S, von Felten S, Frank S, Vassella E, Vajtai I, Taylor E, Schulz M, Hutter G,
Hench J, Schucht P, et al. IDH/MGMT-driven molecular classification of
low-grade glioma is a strong predictor for long-term survival. Neuro Oncol.
2013;15:46979.
6. Keles GE, Anderson B, Berger MS. The effect of extent of resection on time
to tumor progression and survival in patients with glioblastoma multiforme
of the cerebral hemisphere. Surg Neurol. 1999;52:3719.
7. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ,
Belanger K, Brandes AA, Marosi C, Bogdahn U, et al. Radiotherapy plus
concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med.
2005;352:98796.
8. Malmstrom A, Gronberg BH, Marosi C, Stupp R, Frappaz D, Schultz H,
Abacioglu U, Tavelin B, Lhermitte B, Hegi ME, et al. Temozolomide versus
standard 6-week radiotherapy versus hypofractionated radiotherapy in
patients older than 60 years with glioblastoma: the Nordic randomised,
phase 3 trial. Lancet Oncol. 2012;13:91626.
9. Roa W, Kepka L, Kumar N, Sinaika V, Matiello J, Lomidze D, Hentati D,
Guedes de Castro D, Dyttus-Cebulok K, Drodge S, et al. International Atomic
Energy Agency Randomized Phase III Study of Radiation Therapy in Elderly
and/or Frail Patients With Newly Diagnosed Glioblastoma Multiforme. J Clin
Oncol. 2015;33:414550.
10. Chen C, Damek D, Gaspar LE, Waziri A, Lillehei K, Kleinschmidt-DeMasters
BK, Robischon M, Stuhr K, Rusthoven KE, Kavanagh BD. Phase I trial of
hypofractionated intensity-modulated radiotherapy with temozolomide
chemotherapy for patients with newly diagnosed glioblastoma multiforme.
Int J Radiat Oncol Biol Phys. 2011;81:106674.
11. Mizumoto M, Tsuboi K, Igaki H, Yamamoto T, Takano S, Oshiro Y, Hayashi Y,
Hashii H, Kanemoto A, Nakayama H, et al. Phase I/II trial of hyperfractionated
concomitant boost proton radiotherapy for supratentorial glioblastoma
multiforme. Int J Radiat Oncol Biol Phys. 2010;77:98105.
12. Withers HR, Peters LJ, Thames HD, Fletcher GH. Hyperfractionation. Int J
Radiat Oncol Biol Phys. 1982;8:18079.
13. Gonzalez DG, Menten J, Bosch DA, van der Schueren E, Troost D, Hulshof
MC, Bernier J. Accelerated radiotherapy in glioblastoma multiforme: a dose
searching prospective study. Radiother Oncol. 1994;32:98105.
14. Lutterbach J, Weigel P, Guttenberger R, Hinkelbein W. Accelerated
hyperfractionated radiotherapy in 149 patients with glioblastoma
multiforme. Radiother Oncol. 1999;53:4952.
15. Prados MD, Wara WM, Sneed PK, McDermott M, Chang SM, Rabbitt J, Page
M, Malec M, Davis RL, Gutin PH, et al. Phase III trial of accelerated
hyperfractionation with or without difluromethylornithine (DFMO) versus
standard fractionated radiotherapy with or without DFMO for newly
diagnosed patients with glioblastoma multiforme. Int J Radiat Oncol Biol
Phys. 2001;49:717.
16. Werner-Wasik M, Scott CB, Nelson DF, Gaspar LE, Murray KJ, Fischbach JA,
Nelson JS, Weinstein AS, Curran WJ, Jr. Final report of a phase I/II trial of
hyperfractionated and accelerated hyperfractionated radiation therapy with
carmustine for adults with supratentorial malignant gliomas. Radiation
Therapy Oncology Group Study 8302. Cancer. 1996;77:153543.
17. Niyazi M, Brada M, Chalmers AJ, Combs SE, Erridge SC, Fiorentino A, Grosu
AL, Lagerwaard FJ, Minniti G, Mirimanoff RO, et al. ESTRO-ACROP guideline
target delineation of glioblastomas. Radiother Oncol. 2016;118:3542.
We accept pre-submission inquiries
Our selector tool helps you to find the most relevant journal
We provide round the clock customer support
Convenient online submission
Thorough peer review
Inclusion in PubMed and all major indexing services
Maximum visibility for your research
Submit your manuscript at
www.biomedcentral.com/submit
Submit your next manuscript to BioMed Central
and we will help you at every step:
Kaul et al. Radiation Oncology (2016) 11:70 Page 7 of 7
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... The current treatment standard for primary GB is the combination of radiotherapy and TMZ according to the Stupp protocol [38] plus an eventual adjuvant therapy with tumor treating fields which may add a few months to the PFS and OS [4]. To the best of our knowledge, there are no prospective randomized trials investigating the efficacy of HFRT versus NFRT in combination with TMZ, with only a couple of monocentric retrospective studies reporting comparable outcomes [39,40]. To further explore any differences that might exist between HFRT and NFRT, we performed a retrospective multicenter analysis of GB patients which allowed us to model the effects of both schemes on PFS and OS while accounting for known prognostic factors. ...
... The analysis indicates that HFRT yielded comparable outcomes to NFRT. This confirms previous findings that accelerated HFRT achieves treatment effects comparable to NFRT in a shorter time frame [39,40]. The lower α/β ratio of normal brain tissue for late reactions, for which an α/β value of 2 Gy is widely accepted, favors a hyperfractionated acceleration instead of a hypofractionated one. ...
... However, since prophylactic steroid administration was also performed, it cannot be considered an accurate surrogate parameter of toxicity. Previous data from Kaul et al. [39] and Lewitzki et al. [40] reported good tolerance of simultaneous TMZ and HFRT. ...
Article
Full-text available
Background and purpose The standard treatment of glioblastoma patients consists of surgery followed by normofractionated radiotherapy (NFRT) with concomitant and adjuvant temozolomide chemotherapy. Whether accelerated hyperfractionated radiotherapy (HFRT) yields comparable results to NFRT in combination with temozolomide has only sparsely been investigated. The objective of this study was to compare NFRT with HFRT in a multicenter analysis. Materials and methods A total of 484 glioblastoma patients from four centers were retrospectively pooled and analyzed. Three-hundred-ten and 174 patients had been treated with NFRT (30 × 1.8 Gy or 30 × 2 Gy) and HFRT (37 × 1.6 Gy or 30 × 1.8 Gy twice/day), respectively. The primary outcome of interest was overall survival (OS) which was correlated with patient-, tumor- and treatment-related variables via univariable and multivariable Cox frailty models. For multivariable modeling, missing covariates were imputed using multiple imputation by chained equations, and a sensitivity analysis was performed on the complete-cases-only dataset. Results After a median follow-up of 15.7 months (range 0.8–88.6 months), median OS was 16.9 months (15.0–18.7 months) in the NFRT group and 14.9 months (13.2–17.3 months) in the HFRT group (p = 0.26). In multivariable frailty regression, better performance status, gross-total versus not gross-total resection, MGMT hypermethylation, IDH mutation, smaller planning target volume and salvage therapy were significantly associated with longer OS (all p < 0.01). Treatment differences (HFRT versus NFRT) had no significant effect on OS in either univariable or multivariable analysis. Conclusions Since HFRT with temozolomide was not associated with worse OS, we assume HFRT to be a potential option for patients wishing to shorten their treatment time.
... The current treatment standard for primary GB is the combination of radiotherapy and TMZ according to the Stupp protocol [37] plus an eventual adjuvant therapy with tumor treating fields which may add a few months to the PFS and OS [4]. To the best of our knowledge, there are no prospective randomized trials investigating the efficacy of HFRT versus NFRT in combination with TMZ, with only a couple of monocentric retrospective studies reporting comparable outcomes [38,39]. To further explore any differences that might exist between HFRT and NFRT, we performed a retrospective multicenter analysis of GB patients which allowed us to model the effects of both schemes on PFS and OS while accounting for known prognostic factors. ...
... This confirms previous findings that accelerated HFRT achieves treatment effects comparable to NFRT in a shorter time frame [38,39]. The lower α/β ratio of normal brain tissue for late reactions, for which an α/β value of 2Gy is widely accepted, favors a hyperfractionated acceleration instead of a hypofractionated one. ...
... However, since prophylactic steroid administration was also performed, it cannot be considered an accurate surrogate parameter of toxicity. Previous data from Kaul et al. [38] and Lewitzki et al. [39] reported good tolerance of simultaneous temozolomide and HFRT. ...
Preprint
Full-text available
Background and purpose The standard treatment of glioblastoma (GB) patients consists of surgery followed by normofractionated radiotherapy (NFRT) with concomitant and adjuvant temozolomide chemotherapy. Whether accelerated hyperfractionated radiotherapy (HFRT) yields comparable results to NFRT in combination with temozolomide has only sparsely been investigated. The objective of this study was to compare NFRT with HFRT in a multicenter analysis. Materials and methods A total of 484 GB patients from four centers were retrospectively pooled and analyzed. 310 and 174 patients had been treated with NFRT (30×1.8Gy or 30×2Gy) and HFRT (37×1.6Gy or 30×1.8Gy twice/day), respectively. The primary outcome of interest was overall survival (OS) which was correlated with patient-, tumor- and treatment-related variables via univariable and multivariable Cox frailty models. For multivariable modeling, missing covariates were imputed using multiple imputation by chained equations, and a sensitivity analysis was performed on the complete-cases-only dataset. Results After a median follow-up of 15.7 months (range 0.8-88.6 months), median OS was 16.9 months (15.0-18.7 months) in the NFRT group and 14.9 months (13.2-17.3 months) in the HFRT group (p=0.26). In multivariable frailty regression, better performance status, complete versus not complete resection, MGMT hypermethylation, IDH mutation, smaller planning target volume, no steroid administration, and salvage therapy were significantly associated with longer OS (all p<0.01). Treatment differences (HFRT versus NFRT) had no significant effect on OS in either univariable or multivariable analysis. Conclusions This analysis suggests that HFRT and temozolomide is a safe option for patients wishing to shorten their treatment time and does not affect OS.
... On the other hand, Malmström et al. [Malmström et al., 2012] reported hypofractionated RT worked better for the elderly GBM patients (age >70). Meanwhile, some previous studies reported no significant differences occurred between conventional RT and hyperfractionated RT [Kaul et al., 2016, Prados et al., 2001, Cardinale et al., 2006, Floyd et al., 2004, Gonzalez et al., 1994, Shibamoto et al., 1997 or hypofractionated RT [Phillips et al., 2003, Roa et al., 2004 about GBM. As shown in Supplementary Figure 10, the virtual patient received 84Gy of BED via hyperfractionated RT showed somewhat higher survival probabilities, but in case for 60Gy of BED had no significant difference. ...
... As shown in Supplementary Figure 10, the virtual patient received 84Gy of BED via hyperfractionated RT showed somewhat higher survival probabilities, but in case for 60Gy of BED had no significant difference. As the later BED is closer to that of practical RT, we confirmed that estimation of our best RSF model is consistent with the growing evidence for no significant differences between conventional RT and the other fractionated RT [Kaul et al., 2016, Prados et al., 2001, Cardinale et al., 2006, Floyd et al., 2004, Gonzalez et al., 1994, Shibamoto et al., 1997. ...
Preprint
We propose predictive models that estimate GBM patients' health status of one-year after treatments (Classification task), predict the long-term prognosis of GBM patients at an individual level (Survival task). We used total of 467 GBM patients' clinical profile consists of 13 features and two follow-up dates. For baseline models of random forest classifier(RFC) and random survival forest model (RSF), we introduced generalized linear model (GLM), support vector machine (SVM) and Cox proportional hazardous model (COX), accelerated failure time model (AFT) respectively. After preprocessing and prefixing stratified 5-fold data set, we generated best performing models for model types using recursive feature elimination process. Total 10, 4, and 13 features were extracted for best performing one-year survival/progression status RFC models and RSF model via the recursive feature elimination process. In classification task, AUROC of best performing RFC recorded 0.6990 (for one-year survival status classification) and 0.7076 (for one-year progression classification) while that of second best baseline models (GLM in both cases) recorded 0.6691 and 0.6997 respectively. About survival task, the highest C-index of 0.7157 and the lowest IBS of 0.1038 came from the best performing RSF model while that of second best baseline models were 0.6556 and 0.1139 respectively. A simplified linear correlation (extracted from LIME and virtual patient group analysis) between each feature and prognosis of GBM patient were consistent with proven medical knowledge. Our machine learning models suggest that the top three prognostic factors for GBM patient survival were MGMT gene promoter, the extent of resection, and age. To the best of our knowledge, this study is the very first study introducing a interpretable and medical knowledge consistent GBM prognosis predictive models.
... In a previous analysis of concomitant TMZ with hyperfractionated radiochemotherapy performed in our institution there was a survival advantage and good tolerance of simultaneous radiochemotherapy [38]. Also Kaul et al. reported good tolerance of simultaneous TMZ and HFRT [39]. ...
... Although limited to monocentric data, there is clinical evidence for these radiobiological considerations. Kaul et al. [39] published data of 129 patients with GBM treated with a similar protocol of 1.6 Gy twice daily to 59.2 Gy in the HFRT arm. There was comparable efficacy and tolerability of HFRT and NFRT. ...
Article
Full-text available
Background: Current standard of treatment for newly diagnosed patients with glioblastoma (GBM) is surgical resection with adjuvant normofractionated radiotherapy (NFRT) combined with temozolomide (TMZ) chemotherapy. Hyperfractionated accelerated radiotherapy (HFRT) which was known as an option from randomized controlled trials before the temozolomide era has not been compared to the standard therapy in a randomized setting combined with TMZ. Methods: Data of 152 patients with newly diagnosed GBM treated from 10/2004 until 7/2018 at a single tertiary care institution were extracted from a clinical database and retrospectively analyzed. Thirty-eight patients treated with NFRT of 60 Gy in 30 fractions (34 with simultaneous and 2 with sequential TMZ) were compared to 114 patients treated with HFRT of 54.0 Gy in 30 fraction of 1.8 Gy twice daily (109 with simultaneous and 3 with sequential TMZ). The association between treatment protocol and other variables with overall survival (OS) was assessed using univariable and multivariable Cox regression analysis; the latter was performed using variables selected by the LASSO method. Results: Median overall survival (OS) was 20.3 month for the entire cohort. For patients treated with NFRT median OS was 24.4 months compared to 18.5 months in patients treated with HFRT (p = 0.131). In univariable regression analysis the use of dexamethasone during radiotherapy had a significant negative impact on OS in both patient groups, HR 2.21 (95% CI 1.47-3.31, p = 0.0001). In multivariable analysis adjusted for O6-methylguanine-DNA methyl-transferase (MGMT) promotor methylation status, salvage treatment and secondary GBM, the use of dexamethasone was still a negative prognostic factor, HR 1.95 (95% CI 1.21-3.13, p = 0.006). Positive MGMT-methylation status and salvage treatment were highly significant positive prognostic factors. There was no strong association between treatment protocol and OS (p = 0.504). Conclusions: Our retrospective analysis supports the hypothesis of equivalence between HFRT and the standard protocol of treatment for GBM. For those patients who are willing to obtain the benefit of shortening the course of radiochemotherapy, HFRT may be an alternative with comparable efficacy although it was not yet tested in a large prospective randomized study against the current standard. The positive influence of salvage therapy and negative impact of concomitant use of corticosteroids should be addressed in future prospective trials. To confirm our results, we plan to perform a pooled analysis with other tertiary clinics in order to achieve better statistical reliability.
... [10] After the use of TMZ with RT became standard in GB management, RT dose-escalation studies have been accelerated again to improve survival further. [13,[16][17][18]32,33,40,41] Kaul et al. compared 64 patients who underwent accelerated hyperfractionated RT with 67 patients who underwent conventional RT, and they found no significant difference between the two groups. [40] Similarly, Badiyan et al. failed to show improvement in survival rate in patients who received high-dose RT with TMZ compared to those who received the standard-dose RT. [41] On the other hand, Massaccessi et al. performed a dose-escalation study by using the simultaneous integral boost (SIB) IMRT technique, and they reported that 70 Gy delivered in 25 fractions with TMZ was well tolerated. ...
... [13,[16][17][18]32,33,40,41] Kaul et al. compared 64 patients who underwent accelerated hyperfractionated RT with 67 patients who underwent conventional RT, and they found no significant difference between the two groups. [40] Similarly, Badiyan et al. failed to show improvement in survival rate in patients who received high-dose RT with TMZ compared to those who received the standard-dose RT. [41] On the other hand, Massaccessi et al. performed a dose-escalation study by using the simultaneous integral boost (SIB) IMRT technique, and they reported that 70 Gy delivered in 25 fractions with TMZ was well tolerated. [16] In addition, a study in Japan reported that the median survival was extended to 21.6 months in patients treated with hyperfractionated concomitant boost proton RT of 96.6 GyE in 56 fractions. ...
... Similarly, studies using HFRT were evaluated that again showed no specific benefit. 10 The other approach for dose escalation was using HFRT. This had the dual advantage of increased cell kill by higher dose per fraction and reducing the effect of accelerated repopulation by condensed overall treatment time. ...
Article
Full-text available
Rasla Parween Despite multimodality treatment in high-grade glioma (HGG) involving maximal safe resection and adjuvant chemoradiotherapy, the prognosis remains dismal. In this study, we aimed to evaluate a method of biological enhancement by combining dose escalation with a condensed overall treatment time, aiming for maximal cytoreduction as a surrogate for improved outcomes. Hypofractionation has the dual advantage of enhanced cell kill with reduced overall treatment time. To this effect, we have employed a study involving hypofractionated simultaneous integrated boost (SIB) versus conventional treatment. As a secondary objective, we evaluated volumetric modulated arc therapy (VMAT) and intensity modulated radiotherapy (IMRT) in terms of optimal delivery technique for SIB boost. Forty patients were randomized into two arms, the study arm received 60 Gy in 25 fractions and the standard arm received 60 Gy in 30 fractions with concurrent and adjuvant temozolomide. The patients were assessed radiologically for tumor cytoreduction and acute toxicity parameters weekly during treatment, 6 weeks post-treatment, and 3 monthly follow-up. All patients were planned using VMAT and IMRT techniques in the study arm for the comparison of treatment time and dosimetric efficiency. However, the treatment was performed through VMAT technique. Data were analyzed using simple descriptive statistics including Student's t-tests, proportion tests, and Pearson correlation for association. The total sample size was estimated at 40, with 20 samples per group, providing a statistical power of 81% and a significance level (p-value) of 0.05. It was observed that tumor cytoreduction was significantly enhanced in a subgroup of patients in the study arm with smaller volume residual disease (p = 0.04) that was found at 6 weeks post-treatment evaluation. The tolerance, toxicity, and compliance were comparable in both arms. During the dosimetric evaluation, it was determined that VMAT had a significantly lower hot spot compared to the IMRT plan (64.22 Gy vs. 64.75 Gy, p = 0.02). It was also observed that the delivery with VMAT was faster and involved a lesser number of monitor units (555.7 MU vs. 679.6MU, p = 0.001). The hypofractionated SIB radiotherapy using the VMAT technique can provide a feasible method of biological dose enhancement without compromising toxicity and might have the future potential to improve local control in HGG.
... In our study, the OS rate was 7.2 months (239.5 days), like data from the United States [median survival of 8 months and five-year relative survival of 6.8% (Ostrom et al., 2021)] and Germany [median OS of 9.5 and 13 months (Ening et al., 2015;Kaul et al., 2016)]. Median inpatient interval in our patients was 18.5 days (9-167), higher than in other studies [mean of 3 days (Fabrini et al., 2009), and median of 5 days (1-37) (Tully et al., 2016)]. ...
Article
Full-text available
Glioblastoma (GBM) is the most frequent tumor of the central nervous system, and its heterogeneity is a challenge in treatment. This study examined tumoral heterogeneity involving PDGFRA, KIT, and KDR gene amplification (GA) in 4q12 and its association with clinical parameters. Specimens from 22 GBM cases with GA for the 4q12 amplicon detected by FISH were investigated for homogeneous or heterogeneous coamplification patterns, diffuse or focal distribution of cells harboring GA throughout tumor sections, and pattern of clustering of fluorescence signals. Sixteen cases had homogenously amplification for all three genes (45.5%), for PDGFRA and KDR (22.7%), or only for PDGFRA (4.6%); six cases had heterogeneous GA patterns, with subpopulations including GA for all three genes and for two genes - PDGFRA and KDR (13.6%), or GA for all three and for only one gene - PDGFRA (9.1%) or KIT (4.6%). In 6 tumors (27.3%), GA was observed in focal tumor areas, while in the remaining 16 tumors (72.7%) it was diffusely distributed throughout the pathological specimen. Amplification was universally expressed as double minutes and homogenously stained regions. Coamplification of all three genes PDGFRA, KIT, and KDR, age ≥ 60 years, and total tumor resection were statistically associated with poor prognosis. FISH proved effective for detailed interpretation of molecular heterogeneity. The study uncovered an even more diverse range of amplification patterns involving the 4q12 oncogenes in GBM than previously described, thus highlighting a complex tumoral heterogeneity to be considered when devising more effective therapies. Supplementary Information The online version contains supplementary material available at 10.1007/s12017-023-08749-y.
... Recent studies have utilized ablative hypofractionated RT (AHFRT) rather than the conventional fractionated RT (CFRT). Compared with CFRT, AHFRT significantly inhibits tumor growth and reduces recruitment of MDSCs, most likely through downregulation of VEGF and decreasing tumor hypoxia [107]. However, in the context of brain cancers, high doses of radiation can have detrimental side effects. ...
Article
Full-text available
Cancer cells “hijack” host immune cells to promote growth, survival, and metastasis. The immune microenvironment of high-grade gliomas (HGG) is a complex and heterogeneous system, consisting of diverse cell types such as microglia, bone marrow-derived macrophages (BMDMs), myeloid-derived suppressor cells (MDSCs), dendritic cells, natural killer (NK) cells, and T-cells. Of these, MDSCs are one of the major tumor-infiltrating immune cells and are correlated not only with overall worse prognosis but also poor clinical outcomes. Upon entry from the bone marrow into the peripheral blood, spleen, as well as in tumor microenvironment (TME) in HGG patients, MDSCs deploy an array of mechanisms to perform their immune and non-immune suppressive functions. Here, we highlight the origin, function, and characterization of MDSCs and how they are recruited and metabolically reprogrammed in HGG. Furthermore, we discuss the mechanisms by which MDSCs contribute to immunosuppression and resistance to current therapies. Finally, we conclude by summarizing the emerging approaches for targeting MDSCs alone as a monotherapy or in combination with other standard-of-care therapies to improve the current treatment of high-grade glioma patients.
Article
Background and purpose Current glioblastoma (GBM) therapies prolong survival, but overall prognosis is still poor. Irradiation of the subventricular zone (SVZ) has recently been discussed as a promising concept as this tissue harbors stem cells which seem to play a role in the initiation and recurrence of GBM. In this study, we retrospectively examined the relationship of SVZ irradiation dose and survival in a large, homogeneous GBM patient cohort. Materials and methods We included 200 GBM patients who had been treated at our institution with trimodal therapy (surgery, radiotherapy and chemotherapy) between 2009 and 2020. The SVZ was delineated, and dose-volume histograms were calculated and extracted. Tumors were classified according to their contact with the SVZ. The Kaplan-Meier method was used for survival analysis, and univariable and multivariable Cox regression (MVA) were used to determine prognostic effects on progression-free survival (PFS) and overall survival (OS). Results Median PFS of the study group was 7.2 months; median OS was 15.1 months. In MVA (with mean dose to the ipsilateral SVZ as a continuous covariable), PFS was significantly lower for patients with a Karnofsky performance status(KPS) <70% and without MGMT promoter methylation. Factors prognostic for shorter OS were old age, lower KPS, unmethylated MGMT status, SVZ contact and biopsy instead of subtotal- or gross total resection. There was no significant correlation between survival and SVZ dose. Conclusion In this cohort, an increased mean dose to the ipsilateral or contralateral SVZ did not correlate with improved survival in irradiated GBM patients in MVA. Patients whose tumor directly involved the SVZ showed worse OS in MVA.
Article
Full-text available
Introduction There is no standard of care for recurrent high-grade glioma. Treatment strategies include re-resection, re-irradiation, systemic agents, intratumoral thermotherapy using magnetic iron-oxide nanoparticles (“nanotherapy”), and tumor treating fields. Only a small number of patients are eligible for re-resection, and since many patients receive a full course of radiation therapy, there is fear of re-irradiation-induced morbidity. Modern radiation techniques have resulted in greater acceptance of re-irradiation. In this work we retrospectively analyzed patients who had undergone re-irradiation of high-grade glioma at XXX. Materials and Methods All patients treated with re-irradiation for recurrent high-grade glioma in our department from January 1997 to February 2014 were analyzed in this study. In total, 198 patients were included. The primary endpoint was overall survival after recurrence. Results One hundred ninety-eight patients were identified. Median time from first RT to re-irradiation was 14 months. Median follow-up from the first day of re-irradiation to last contact or death was 7 months. Median overall survival after relapse was 7 months for the overall cohort. For glioblastoma (GBM), median overall survival after relapse was 6 months and for III° gliomas 14 months. Treatment was generally well tolerated. Common Terminology Criteria for Adverse Events (CTCAE) grade 3 toxicity was observed in 5.1% patients and grade 4 toxicity in 2.5%. No patient developed grade 5 toxicity. The likelihood of developing severe toxicity (CTCAE grade 3/4) was not significantly higher in the group of patients who received re-irradiation in the first 14 months after initial RT. Patients who received a higher Biologically effective dose to the tumor also did not have a significantly higher rate of severe acute toxicity. Conclusions The prognosis of recurrent high-grade glioma remains dismal. Re-irradiation is often tolerable even after early recurrence (< 14 months) and with higher doses (e.g. 49.4 Gy/3.8 Gy) in selected patients.
Article
Full-text available
Purpose: The optimal radiotherapy regimen for elderly and/or frail patients with newly diagnosed glioblastoma remains to be established. This study compared two radiotherapy regimens on the outcome of these patients. Patients and methods: Between 2010 and 2013, 98 patients (frail = age ≥ 50 years and Karnofsky performance status [KPS] of 50% to 70%; elderly and frail = age ≥ 65 years and KPS of 50% to 70%; elderly = age ≥ 65 years and KPS of 80% to 100%) were prospectively randomly assigned to two arms in a 1:1 ratio, stratified by age (< and ≥ 65 years old), KPS, and extent of surgical resection. Arm 1 received short-course radiotherapy (25 Gy in five daily fractions over 1 week), and arm 2 received commonly used radiotherapy (40 Gy in 15 daily fractions over 3 weeks). Results: The short-course radiotherapy was noninferior to commonly used radiotherapy. The median overall survival time was 7.9 months (95% CI, 6.3 to 9.6 months) in arm 1 and 6.4 months (95% CI, 5.1 to 7.6 months) in arm 2 (P = .988). Median progression-free survival time was 4.2 months (95% CI, 2.5 to 5.9) in arm 1 and 4.2 months (95% CI, 2.6 to 5.7) in arm B (P = .716). With a median follow-up time of 6.3 months, the quality of life between both arms at 4 weeks after treatment and 8 weeks after treatment was not different. Conclusion: There were no differences in overall survival time, progression-free survival time, and quality of life between patients receiving the two radiotherapy regimens. In view of the reduced treatment time, the short 1-week radiotherapy regimen may be recommended as a treatment option for elderly and/or frail patients with newly diagnosed glioblastoma.
Article
Full-text available
The objective of the CBTRUS Statistical Report: Primary Brain and Central Nervous System Tumors Diagnosed in the United States in 2007–2011 is to provide a comprehensive summary of the current descriptive epidemiology of primary brain and central nervous system (CNS) tumors in the United States population. CBTRUS obtained the latest available data on all newly diagnosed primary brain and CNS tumors from the CDC, National Program of Cancer Registries (NPCR), and the NCI, SEER program for diagnosis years 2007–2011. Incidence counts and rates of primary malignant and non-malignant brain and CNS tumors are documented by histology, gender, age, race, and Hispanic ethnicity. Mortality and relative survival rates for selected malignant histologies calculated using SEER data for the period 1995–2011 are also presented.
Article
Full-text available
This report, an audit requested by the French government, describes oncological patterns of care, prognostic factors, and survival for patients with newly diagnosed and histologically confirmed glioblastoma multiforme (GBM) in France. The French Brain Tumor DataBase, which is a national multidisciplinary (neurosurgeons, neuropathologists, radiotherapists, neurooncologists, epidemiologists, and biostatisticians) network, prospectively collected initial data for the cases of GBM in 2004, and a specific data card was used to retrospectively collect data on the management and follow-up care of these patients between January 1, 2004, and December 1, 2006. We recorded 952 cases of GBM (male/female ratio 1.6, median age 63.9 years, mean preoperative Karnofsky performance status [KPS] 79). Surgery consisted of resection (RS; n = 541) and biopsy (n = 411); 180 patients did not have subsequent oncological treatment. After surgery, first-line treatment (n = 772) consisted of radiotherapy (RT) and temozolomide (TMZ) concomitant +/− adjuvant in 314 patients, RT alone in 236 patients, chemotherapy (CT) alone in 157 patients, and other treatment modalities in 65 patients. Median overall survival was 286 days (95% CI, 266–314) and was significantly affected by age, KPS, and tumor location. Median survival (days, 95% CI) associated with these main strategies, when analyzed by a surgical group, were as follows: RS + RT-TMZ(n=224): 476 (441–506), biopsy + RT-TMZ(n=90): 329 (301–413), RS + RT(n=147): 363 (331–431), biopsy + RT(n=89): 178 (153–237), RS + CT(n=61): 245 (190–361), biopsy + CT(n=96): 244 (198–280), and biopsy only(n=118): 55 (46–71). This study illustrates the usefulness of a national brain tumor database. To our knowledge, this work is the largest report of recent GBM management in Europe.
Article
Background and purpose: Target delineation in glioblastoma (GBM) varies substantially between different institutions and several consensus statements are available. This guideline aims to develop a joint European consensus on the delineation of the clinical target volume in patients with a glioblastoma (GBM). Material and methods: A literature search was conducted in PubMed that evaluated adults with GBM. Both MeSH terms and text words were used and the following search strategy was applied: ("Glioblastoma/radiotherapy" [MeSH] OR "glioblastoma" OR "malignant glioma" OR high-grade glioma) AND ((delineation) OR (target volume) OR (CTV) OR (PTV) OR (margin) OR (recurrence pattern) OR (contouring) OR (organs at risk)). In parallel, abstracts from ESTRO and ASTRO 2010-2015 were analysed and separately reviewed. The ACROP committee identified 14 European experts in close interaction with the ESTRO clinical committee who discussed and analysed the body of evidence concerning GBM target delineation. Results: Several key issues were identified and are discussed including (i) pre-treatment steps and immobilization, (ii) target delineation and the use of standard and novel imaging techniques, and (iii) technical aspects of treatment including planning techniques, and fractionation. Based on the EORTC recommendation focusing on the resection cavity and residual enhancing regions on T1-sequences with the addition of a 20mm margin, special situations are presented with corresponding potential adaptations depending on the specific clinical situation. Conclusions: Currently, based on the EORTC consensus, a single clinical target volume definition based on postoperative T1/T2 FLAIR abnormalities is recommended, using isotropic margins without the need to cone down. A PTV margin based on the individual mask system and IGRT procedures available is advised, usually of the order of 3-5mm.
Article
Background Low-grade gliomas (LGGs) are rare brain neoplasms, with survival spanning up to a few decades. Thus, accurate evaluations on how biomarkers impact survival among patients with LGG require long-term studies on samples prospectively collected over a long period.Methods The 210 adult LGGs collected in our databank were screened for IDH1 and IDH2 mutations (IDHmut), MGMT gene promoter methylation (MGMTmet), 1p/19q loss of heterozygosity (1p19qloh), and nuclear TP53 immunopositivity (TP53pos). Multivariate survival analyses with multiple imputation of missing data were performed using either histopathology or molecular markers. Both models were compared using Akaike's information criterion (AIC). The molecular model was reduced by stepwise model selection to filter out the most critical predictors. A third model was generated to assess for various marker combinations.ResultsMolecular parameters were better survival predictors than histology (ΔAIC = 12.5, P< .001). Forty-five percent of studied patients died. MGMTmet was positively associated with IDHmut (P< .001). In the molecular model with marker combinations, IDHmut/MGMTmet combined status had a favorable impact on overall survival, compared with IDHwt (hazard ratio [HR] = 0.33, P< .01), and even more so the triple combination, IDHmut/MGMTmet/1p19qloh (HR = 0.18, P< .001). Furthermore, IDHmut/MGMTmet/TP53pos triple combination was a significant risk factor for malignant transformation (HR = 2.75, P< .05).Conclusion By integrating networks of activated molecular glioma pathways, the model based on genotype better predicts prognosis than histology and, therefore, provides a more reliable tool for standardizing future treatment strategies.
Article
Most patients with glioblastoma are older than 60 years, but treatment guidelines are based on trials in patients aged only up to 70 years. We did a randomised trial to assess the optimum palliative treatment in patients aged 60 years and older with glioblastoma. Patients with newly diagnosed glioblastoma were recruited from Austria, Denmark, France, Norway, Sweden, Switzerland, and Turkey. They were assigned by a computer-generated randomisation schedule, stratified by centre, to receive temozolomide (200 mg/m(2) on days 1-5 of every 28 days for up to six cycles), hypofractionated radiotherapy (34·0 Gy administered in 3·4 Gy fractions over 2 weeks), or standard radiotherapy (60·0 Gy administered in 2·0 Gy fractions over 6 weeks). Patients and study staff were aware of treatment assignment. The primary endpoint was overall survival. Analyses were done by intention to treat. This trial is registered, number ISRCTN81470623. 342 patients were enrolled, of whom 291 were randomised across three treatment groups (temozolomide n=93, hypofractionated radiotherapy n=98, standard radiotherapy n=100) and 51 of whom were randomised across only two groups (temozolomide n=26, hypofractionated radiotherapy n=25). In the three-group randomisation, in comparison with standard radiotherapy, median overall survival was significantly longer with temozolomide (8·3 months [95% CI 7·1-9·5; n=93] vs 6·0 months [95% CI 5·1-6·8; n=100], hazard ratio [HR] 0·70; 95% CI 0·52-0·93, p=0·01), but not with hypofractionated radiotherapy (7·5 months [6·5-8·6; n=98], HR 0·85 [0·64-1·12], p=0·24). For all patients who received temozolomide or hypofractionated radiotherapy (n=242) overall survival was similar (8·4 months [7·3-9·4; n=119] vs 7·4 months [6·4-8·4; n=123]; HR 0·82, 95% CI 0·63-1·06; p=0·12). For age older than 70 years, survival was better with temozolomide and with hypofractionated radiotherapy than with standard radiotherapy (HR for temozolomide vs standard radiotherapy 0·35 [0·21-0·56], p<0·0001; HR for hypofractionated vs standard radiotherapy 0·59 [95% CI 0·37-0·93], p=0·02). Patients treated with temozolomide who had tumour MGMT promoter methylation had significantly longer survival than those without MGMT promoter methylation (9·7 months [95% CI 8·0-11·4] vs 6·8 months [5·9-7·7]; HR 0·56 [95% CI 0·34-0·93], p=0·02), but no difference was noted between those with methylated and unmethylated MGMT promoter treated with radiotherapy (HR 0·97 [95% CI 0·69-1·38]; p=0·81). As expected, the most common grade 3-4 adverse events in the temozolomide group were neutropenia (n=12) and thrombocytopenia (n=18). Grade 3-5 infections in all randomisation groups were reported in 18 patients. Two patients had fatal infections (one in the temozolomide group and one in the standard radiotherapy group) and one in the temozolomide group with grade 2 thrombocytopenia died from complications after surgery for a gastrointestinal bleed. Standard radiotherapy was associated with poor outcomes, especially in patients older than 70 years. Both temozolomide and hypofractionated radiotherapy should be considered as standard treatment options in elderly patients with glioblastoma. MGMT promoter methylation status might be a useful predictive marker for benefit from temozolomide. Merck, Lion's Cancer Research Foundation, University of Umeå, and the Swedish Cancer Society.
Article
Background: Efforts to improve local control and survival by increasing the dose of once-daily radiation therapy beyond 70 Gray (Gy) for patients with malignant gliomas has yet been unsuccessful. Hyperfractionated radiation therapy (HF) should allow for delivery of a higher total dose without increasing normal tissue late effects, whereas accelerated hyperfractionated radiation therapy (AHF) may minimize tumor repopulation by shortening overall treatment time. The Radiation Therapy Oncology Group (RTOG) conducted a randomized Phase I/II study of escalating doses of HF and AHF either carmustine (bis-chlorethyl nitrosourea [BCNU]) fro adults with supratentorial glioblastoma multiforme (GBM) or anaplastic astrocytoma (AA). Primary study endpoints were overall survival and acute and chronic treatment-related toxicity. Methods: From 1983 to 1989, 786 patients with supratentorial gliomas (81% with GBM and 19% with AA) were stratified by histology, age, and performance status and randomized to receive partial brain irradiation, utilizing either HF (1.2 Gy twice daily to doses of 64.8, 72, 76.8, or 81.6 Gy) of AHF (1.6 Gy twice daily to doses of 48 or 54.4 Gy). All patients received carmustine. The distinction of pronistic factors was similar on all arms. Results: There were 747 eligible and analyzable patients among 786 enrolled patients (95%). Two patients had a Grade 5 and 65 patients had a Grade 4 chemotherapy toxicity. Two patients in the 81.6 Gy arm experienced late Grade 4 radiation toxicity and there was 1 late radiation-associated death in the 54.4 Gy arm. The rate of Grade 3 of worse radiation toxicity at 5 years, calculated by the delivered does level, was 3% in the lowest total dose arms (48 and 54.4 Gy), 4% in the intermediate dose arms (64.8 and 72 Gy), and 5% in the highest dose arms (76.8 and 81.6 Gy) (p = 0.54). Survival rates at 2 and 5 years were: 21% and 11%, and 4%, respectively, for GBM patients. There were no significant differences between the treatment arms with regard to median survival time (MST), when analyzed by the originally assigned dose. The MST for all patients varied between 10.8 months and 12.7 months (P = 0.59); between 9.6 months and 11 months for patients with GBM (P = 0.43); and between 30.4 months and 85.8 months for patients with AA (P = 0.78). Analysis of the survival rates for all patients by dose received rather than by dose assigned revealed a 14% 5-year survival rate for the lower HF doses (64.8 and 73 Gy), 11% for the higher doses (76.8 and 81.6 Gy), and 10% for the AHF doses (48 and 54.4 Gy) (P = 0.1). The subgroup a AA patients had a better MST (49.9 months) in the lower received HF doses than in the higher HF doses (34.6 months) (P = 0.35). In contrast, GM patients who received the higher HF doses had survival superior to the patients in the AHF arms (MST of 11.6 months and 10.2 months, respectively, P = 0.04). Conclusions: The use of HF with BCNU and dose escalation up to 81.6 Gy is both feasible and tolerable, although late toxicity increases slightly with increasing dose. The best MST with the least toxicity were observed for AA in the lower received HF doses (72 and 64.8 Gy). Accordingly, 72 Gy in two 1.2 Gy fractions was used as the investigational arm of a completed Phase III trial (RTOG 90-06). In contrast, for GBM patients, longer survival times were noted in the higher received HF doses (78.6 and 81.6 Gy), suggesting the role for further dose escalation. The low toxicity rate with AHF arms suggest that further dose escalation is possible and is currently occurring in RTOG 94-11.
Article
To determine the maximal tolerated biologic dose intensification of radiotherapy using fractional dose escalation with temozolomide (TMZ) chemotherapy in patients with newly diagnosed glioblastoma multiforme. Patients with newly diagnosed glioblastoma multiforme after biopsy or resection and with adequate performance status, bone marrow, and organ function were eligible. The patients underwent postoperative intensity-modulated radiotherapy (IMRT) with concurrent and adjuvant TMZ. All patients received a total dose of 60 Gy to the surgical cavity and residual tumor, with a 5-mm margin. IMRT biologic dose intensification was achieved by escalating from 3 Gy/fraction (Level 1) to 6 Gy/fraction (Level 4) in 1-Gy increments. Concurrent TMZ was given at 75 mg/m(2)/d for 28 consecutive days. Adjuvant TMZ was given at 150-200 mg/m(2)/d for 5 days every 28 days. Dose-limiting toxicity was defined as any Common Terminology Criteria for Adverse Events, version 3, Grade 3-4 nonhematologic toxicity, excluding Grade 3 fatigue, nausea, and vomiting. A standard 3+3 Phase I design was used. A total of 16 patients were accrued (12 men and 4 women, median age, 69 years; range, 34-84. The median Karnofsky performance status was 80 (range, 60-90). Of the 16 patients, 3 each were treated at Levels 1 and 2, 4 at Level 3, and 6 at Level 4. All patients received IMRT and concurrent TMZ according to the protocol, except for 1 patient, who received 14 days of concurrent TMZ. The median number of adjuvant TMZ cycles was 7.5 (range, 0-12). The median survival was 16.2 months (range, 3-33). One patient experienced vision loss in the left eye 7 months after IMRT. Four patients underwent repeat surgery for suspected tumor recurrence 6-12 months after IMRT; 3 had radionecrosis. The maximal tolerated IMRT fraction size was not reached in our study. Our results have shown that 60 Gy IMRT delivered in 6-Gy fractions within 2 weeks with concurrent and adjuvant TMZ is tolerable in selected patients with a T(1)-weighted enhancing tumor <6 cm.