ArticlePDF Available

Bevacizumab plus hypofractionated radiotherapy versus radiotherapy alone in elderly patients with glioblastoma: The randomized, open-label, phase II ARTE trial

Authors:
  • Eberhard Kals University of Tübingen, Germany

Abstract and Figures

Background: The addition of bevacizumab to temozolomide-based chemoradiotherapy (TMZ/RT→TMZ) did not prolong overall survival (OS) in patients with newly diagnosed glioblastoma in phase III trials. Elderly and frail patients are underrepresented in clinical trials, but early reports suggested preferential benefit in this population. Patients and methods: ARTE was a 2:1 randomized, multi-center, open-label, non-comparative phase II trial of hypofractionated RT (40 Gy in 15 fractions) with bevacizumab (10 mg/kg x 14 days) (arm A, N = 50) or without bevacizumab (arm B, N = 25) in patients with newly diagnosed glioblastoma aged ≥65 years. The primary objective was to obtain evidence for prolongation of median OS by the addition of bevacizumab to RT. Response was assessed by RANO criteria. Quality of life (QoL) was monitored by the EORTC QLQ-C30/BN20 modules. Exploratory studies included molecular subtyping by 450k whole methylome and gene expression analyses. Results: Median PFS was longer in arm A than in arm B (7.6 and 4.8 months, P = 0.003), but OS was similar (12.1 and 12.2 months, P = 0.77). Clinical deterioration was delayed and more patients came off steroids in arm A. Prolonged PFS in arm A was confined to tumors with the receptor tyrosine kinase (RTK) I methylation subtype (HR 0.25, P = 0.014) and proneural gene expression (HR 0.29, P = 0.025). In a Cox model of OS controlling for established prognostic factors, associations with more favorable outcome were identified for age <70 years (HR 0.52, P = 0.018) and Karnofsky performance score (KPS) 90-100% (HR 0.51, P = 0.026). Including molecular subtypes into that model identified an association of the RTK II gene methylation subtype with inferior OS (HR 1.73, P = 0.076). Conclusion: Efficacy outcomes and exploratory analyses of ARTE do not support the hypothesis that the addition of bevacizumab to RT generally prolongs survival in elderly glioblastoma patients. Molecular biomarkers may identify patients with preferential benefit from bevacizumab. Clinical trial registration number: NCT01443676.
ORIGINAL ARTICLE
Bevacizumab plus hypofractionated radiotherapy
versus radiotherapy alone in elderly patients with
glioblastoma: the randomized, open-label, phase II
ARTE trial
H.-G. Wirsching
1,2†
, G. Tabatabai
1,2
, U. Roelcke
3
, A. F. Hottinger
4,5
,F.Jo¨rger
6
, A. Schmid
7
, L. Plasswilm
8
,
D. Schrimpf
9,10
, C. Mancao
11
, D. Capper
9,10
, K. Conen
12
, T. Hundsberger
13
, F. Caparrotti
14
,
R. von Moos
15
, C. Riklin
16
, J. Felsberg
17
, P. Roth
1,2
, D. T. W. Jones
10,18
, S. Pfister
10,18
, E. J. Rushing
1,19
,
L. Abrey
20
, G. Reifenberger
17,21
, L. Held
22
, A. von Deimling
9,10
, A. Ochsenbein
7
& M. Weller
1,2
*
1
Brain Tumor Center Zurich;
2
Department of Neurology, University Hospital and University of Zurich, Zurich;
3
Department of Neurology, Brain Tumor Center Aarau,
Cantonal Hospital Aarau, Aarau; Departments of
4
Clinical Neurosciences;
5
Medical Oncology, University Hospital Lausanne, Lausanne;
6
Clinical Trials Center,
University Hospital and University of Zurich, Zurich;
7
Department of Medical Oncology, University Hospital Bern, Bern;
8
Department of Radiation Oncology,
Cantonal Hospital St. Gallen, St. Gallen, Switzerland;
9
Department of Neuropathology, Ruprecht-Karls-University Heidelberg;
10
German Cancer Research Center
(DKFZ), Heidelberg, Germany;
11
Genentech, Oncology Biomarker Development, Basel;
12
Department of Medical Oncology, University Hospital Basel, Basel;
13
Department of Neurology, Cantonal Hospital St. Gallen, St. Gallen;
14
Department of Radiation Oncology, University Hospital Geneva, Geneva;
15
Department of
Medical Oncology, Cantonal Hospital Chur, Chur;
16
Department of Medical Oncology, Cantonal Hospital Lucerne, Lucerne Switzerland;
17
Department of
Neuropathology, Heinrich Heine University Du¨ sseldorf, Du¨sseldorf;
18
Department of Pediatric Hematology and Oncology, Ruprecht-Karls-University Heidelberg,
Heidelberg, Germany;
19
Department of Neuropathology, University Hospital Zurich, Zurich;
20
F. Hoffmann-La Roche, Pharma Division, Product Development
Oncology, Basel, Switzerland;
21
German Cancer Research Center, Essen/Du¨ sseldorf, Germany;
22
Biostatistics Department, University of Zurich, Zurich, Switzerland
*Correspondence to: Prof. Michael Weller, Department of Neurology, University Hospital and University of Zurich, Frauenklinikstrasse 26, CH-8091 Zu¨rich, Switzerland.
Tel: þ41-44-255-5500; Fax: þ41-44-255-4507; E-mail: michael.weller@usz.ch
Present address: Human Biology Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Mail Stop C3-168, Seattle, WA 98109, USA.
Background: The addition of bevacizumab to temozolomide-based chemoradiotherapy (TMZ/RT !TMZ) did not prolong
overall survival (OS) in patients with newly diagnosed glioblastoma in phase III trials. Elderly and frail patients are underrepresented
in clinical trials, but early reports suggested preferential benefit in this population.
Patients and methods: ARTE was a 2 : 1 randomized, multi-center, open-label, non-comparative phase II trial of
hypofractionated RT (40 Gy in 15 fractions) with bevacizumab (10 mg/kg14 days) (arm A, N¼50) or without bevacizumab (arm
B, N¼25) in patients with newly diagnosed glioblastoma aged 65 years. The primary objective was to obtain evidence for
prolongation of median OS by the addition of bevacizumab to RT. Response was assessed by RANO criteria. Quality of life (QoL)
was monitored by the EORTC QLQ-C30/BN20 modules. Exploratory studies included molecular subtyping by 450k whole
methylome and gene expression analyses.
Results: Median PFS was longer in arm A than in arm B (7.6 and 4.8 months, P¼0.003), but OS was similar (12.1 and
12.2 months, P¼0.77). Clinical deterioration was delayed and more patients came off steroids in arm A. Prolonged PFS in arm A
was confined to tumors with the receptor tyrosine kinase (RTK) I methylation subtype (HR 0.25, P¼0.014) and proneural gene
expression (HR 0.29, P¼0.025). In a Cox model of OS controlling for established prognostic factors, associations with more
favorable outcome were identified for age <70 years (HR 0.52, P¼0.018) and Karnofsky performance score 90%–100% (HR 0.51,
P¼0.026). Including molecular subtypes into that model identified an association of the RTK II gene methylation subtype with
inferior OS (HR 1.73, P¼0.076).
Conclusion: Efficacy outcomes and exploratory analyses of ARTE do not support the hypothesis that the addition of
bevacizumab to RT generally prolongs survival in elderly glioblastoma patients. Molecular biomarkers may identify patients with
preferential benefit from bevacizumab.
V
CThe Author(s) 2018. Published by Oxford University Press on behalf of the European Society for Medical Oncology.
All rights reserved. For permissions, please email: journals.permissions@oup.com.
Annals of Oncology 29: 1423–1430, 2018
doi:10.1093/annonc/mdy120
Published online 10 April 2018
Clinical trial registration number: NCT01443676.
Key words:glioblastoma, elderly, bevacizumab, radiotherapy, molecular subtype
Introduction
Almost 50% of all patients with glioblastoma are older than
65 years, <20% of these survive the first year [1]. Many elderly
patients with glioblastoma are deemed too frail to tolerate com-
bined post-operative temozolomide chemoradiotherapy (TMZ/
RT !TMZ) [2,3], rendering monotherapy regimens of hypo-
fractionated RT [4] or TMZ alone preferred treatment options
for some of these patients [57].
The vascular endothelial growth factor (VEGF)-targeting anti-
body bevacizumab (Avastin
TM
) was considered an optimal can-
didate drug to improve overall survival (OS) of elderly
glioblastoma patients based on results of early, uncontrolled clin-
ical reports that suggested preferential benefit from bevacizumab
in frail and elderly patients (supplementary Note S1, available at
Annals of Oncology online). Yet, bevacizumab failed to prolong
OS in two phase III trials and there was only a trend for more
benefit from bevacizumab in patients with unfavorable prognos-
tic factors, although frail elderly patients were underrepresented
in these trials since being eligible for TMZ/RT !TMZ was an in-
clusion criterion in both trials [8,9].
Patients and methods
Study design
ARTE (NCT01443676) was designed as a two-arm, randomized, non-
comparative, open-label phase II trial. The primary objective was to de-
termine the efficacy and tolerability of the combination of bevacizumab
and radiotherapy in elderly patients with newly diagnosed glioblastoma.
The primary end point was median OS. Major inclusion criteria were: age
65 years or older, newly diagnosed supratentorial glioblastoma, eligible
for first infusion of bevacizumab 28 and 49 days after surgery for glio-
blastoma, Karnofsky performance score (KPS) of 60 or more, stable or
decreasing corticosteroid dose within 5 days before enrolment, availabil-
ity of paraffin-embedded tissue for central pathology review and deter-
mination of O
6
-methylguanine DNA methyltransferase (MGMT)
promoter methylation status, and adequate hematological, renal and
liver function. An amendment (November 2013) requested the absence
of MGMT promoter methylation when it became clear that MGMT pro-
moter methylation predicted larger benefit from temozolomide alone
than from RT alone in patients with glioblastoma aged 65 years [5,6].
All patients gave written informed consent, and the study was approved
by the local ethical committees and competent authorities (KEK-ZH No.
2011-0135).
Central pathology review and molecular analyses
Histological diagnoses were reviewed centrally (ER) based on the 2007
World Health Organization (WHO) classification [10]. Molecular assess-
ments included IDH mutation, MGMT promoter methylation, genome-
wide CpG methylation and nCounter gene expression arrays (supplementary
Note S2, available at Annals of Oncology online).
Study treatment, assessments and end points
Patients were allocated to treatment arms using a web-based randomiza-
tion system without stratification in a 2 : 1 distribution. RT was adminis-
tered to the gross tumor volume plus a 2 cm margin over 3 weeks, in 15
fractions of 2.66 Gy, to a total 40.0 Gy. Bevacizumab was administered
intravenously at 10 mg/kg bodyweight every 2 weeks. Dose delays were
foreseen for adverse reactions associated with bevacizumab that were
considered likely to evolve into serious or life-threatening events.
Bevacizumab was stopped for disease progression or unacceptable tox-
icity, including life-threatening events or subjective patient-related fac-
tors. Details on clinical assessments and end points are provided in
supplementary Note S3, available at Annals of Oncology online.
Statistical analyses
The initial sample size was 40 patients for arm A and 20 patients for arm
B. Sample size calculations were based on the experimental arm only for
the null hypothesis that, based on NOA-08 [6], median OS was
10 months, for 80% power to detect an improvement of 2 months using
a two-sided alpha level of 10%. Per amendment, the study design was
modified to aim for enrollment of 60 patients with tumors without
MGMT promoter methylation. Details on the applied statistical tests are
summarized in supplementary Note S4, available at Annals of Oncology
online.
Results
Patient characteristics
At 9 sites in Switzerland, 75 patients aged 65 years or older were
randomized from March 2013 to August 2015 (supplementary
Figure S1, available at Annals of Oncology online). Arms A and B
were overall well balanced (Table 1). More patients in arm A had
cognitive impairment (42% versus 15%, P¼0.032). The MGMT
promoter methylation status was available for 72 patients (96%),
55 tumors (76%) had an unmethylated MGMT promoter.
Genome-wide methylation arrays revealed 16 tumors (28%) with
receptor tyrosine kinase (RTK) I, 23 tumors (40%) with RTK II
and 16 tumors (28%) with mesenchymal (MES) methylation pat-
terns. Gene expression subtyping revealed 15 proneural (PN)
(29%), 19 classical (CL) (37%) and 17 MES (33%) glioblastomas.
At the time of databank closure (19 August 2016), 74 deaths were
documented, of which 70 were attributed to tumor progression,
1 was of unknown cause and 3 from pulmonary embolism, myo-
cardial infarction and gastrointestinal perforation, respectively.
One patient was still on study treatment, no patient was lost to
follow up for OS.
Safety and tolerability
Adverse events are summarized in supplementary Table S1, avail-
able at Annals of Oncology online. Grade 3–5 events were overall
infrequent. More adverse events occurred in arm A than in arm
B, but there was also a longer treatment period in arm A. The rate
of thromboembolic events was similar in both groups (19%
Original article Annals of Oncology
1424 | Wirsching et al. Volume 29 | Issue 6 | 2018
versus 20%), but more severe and life-threatening thrombo-
embolic events occurred in arm A (16% versus 8%), including
one death each from pulmonary embolism and myocardial in-
farction. Bevacizumab was discontinued for reasons other than
progression in four patients, including death in one patient, pul-
monary embolism in two patients and withdrawal of consent in
one patient.
Efficacy estimates by treatment arm
The median time from surgery to randomization was 25 days
(range 16–41) in arm A and 24 days (range 16–37) in arm B
(P¼0.60). Forty-seven of 50 patients in arm A and 23 of 25
patients in arm B completed the intended dose of 40 Gy of hypo-
fractionated RT. The median number of bevacizumab adminis-
trations in arm A was 12 (range 0–86). Median PFS was
7.6 months in arm A and 4.8 months in arm B (Figure 1A; supple-
mentary Table S2, available at Annals of Oncology online,
P¼0.003) and PFS at 6 months (PFS6) was 78% and 28% in
arms A and B (supplementary Table S2, available at Annals of
Oncology online, P<0.001). Median PFS was also more favorable
in arm A when analyses were restricted to the per protocol popu-
lation, i.e. to patients with tumors with an unmethylated MGMT
promoter and confirmation of the histopathological diagnosis by
central neuropathology review (supplementary Figure S2, avail-
able at Annals of Oncology online). Median PFS in the per proto-
col population was 7.6 months in arm A and 4.7 months in arm B
(supplementary Table S3, available at Annals of Oncology online,
P¼0.071). Twenty-four of 48 patients in arm A (50%), but 18 of
25 patients (72%) in arm B received any salvage therapy at pro-
gression (P¼0.071). Median OS in the intent to treat population
was 12.1 months in arm A and 12.2 months in arm B (Figure 1B;
supplementary Table S2, available at Annals of Oncology online,
P¼0.77), and survival rates at 12 months (OS12) were 54% and
56% in arms A and B (supplementary Table S2, available at
Annals of Oncology online, P¼0.85). Similarly, no apparent
benefit from bevacizumab with respect to OS emerged in the per
protocol population (supplementary Table S3, available at
Annals of Oncology online).
Central radiology review and response assessment
Serial MRI of 66 patients was available for post hoc central review.
At study entry, all but two patients, both from arm A, had
contrast-enhancing lesions, which qualified for measurable dis-
ease. Response measures were balanced between treatment arms
(P¼0.15; supplementary Table S4, available at Annals of
Oncology online). The locally determined time point of progres-
sion was centrally confirmed in 54 patients. It was antedated in
arm A by one scan in eight patients, two scans in two patients and
four scans in one patient, all of which qualified for pseudores-
ponse by RANO criteria because of progression on T2-FLAIR
sequences. Among patients from arm B, progression was ante-
dated one scan in one patient. Central review confirmed longer
PFS in arm A than in arm B (median PFS 7.0 versus 4.7 months,
P¼0.029).
Table 1. Patient characteristics
Arm A N550 Arm B N525 P
Age at diagnosis (years)
Median 70 70
Range 65–87 65–79 0.93
Gender, N(%)
Male 32 (64) 16 (64)
Female 18 (36) 9 (36) 1.00
KPS at study entry, N(%)
90–100 25 (50) 16 (64)
70–80 21 (42) 6 (24)
60 4 (8) 3 (12) 0.30
MMSE at study entry (points)
27 26 (52) 17 (68)
<27 19 (38) 3 (12) 0.032
No data 5 (10) 5 (20)
Steroids at study entry, N(%)
Yes 22 (44) 12 (44)
No 27 (54) 13 (56) 0.80
No data 1 (2) 0 (0)
CE-T1 (cm
2
)
Median 5.9 3.8
Range 0.0–32.2 0.5–17.8 0.12
Tumor burden on T2 (cm
2
)
Median 15.3 12.1
Range 2.3–43.2 1.8–39.0 0.13
Central histopathology, N(%)
Glioblastoma 46 (92) 23 (96)
Anaplastic astrocytoma 1 (2) 0 (0)
Anaplastic ependymoma 0 (0) 1 (4) 1.00
No data 3 (6) 1 (4)
IDH,N(%)
Mutated 1 (2) 0 (0)
Wildtype 39 (78) 19 (76) 1.00
No data 10 (20) 6 (24)
MGMT promoter, N(%)
Methylated 10 (20) 6 (24)
Unmethylated 37 (74) 18 (72) 0.72
No data 3 (6) 1 (4)
Gene methylation, N(%)
a
RTK I 11 (22) 5 (20) 0.67
RTK II 15 (30) 8 (32) 0.73
MES 11 (22) 5 (20) 0.88
MYCN 1 (2) 0 (0.0) n.a.
IDH mut 1 (2) 0 (0.0) n.a.
No data 11 (22) 7 (28) n.a.
Gene expression, N(%)
b
PN 9 (18) 6 (24) 0.39
CL 11 (22) 8 (32) 0.20
MES 15 (30) 2 (8) 0.033
No data 15 (30) 9 (36) n.a.
a
Jones et al. [11].
b
Verhaak et al. [12].
P-values <0.05 may be considered statistically significant.
CE-T1, contrast enhancement on T1-weighted images; CI, confidence
interval; CL, classical; IDH, isocitrate dehydrogenase; KPS, Karnofsky per-
formance score; MMSE, mini-mental status examination; MES, mesenchy-
mal; MGMT, O
6
-methylguanine DNA methyltransferase; MYCN, N-myc
proto-oncogene; PN, proneural; RTK, receptor tyrosine kinase.
Annals of Oncology Original article
Volume 29 | Issue 6 | 2018 doi:10.1093/annonc/mdy120 | 1425
Quality of life, cognitive functioning and steroid
use
Adherence of the study population to quality of life measure-
ments at each scheduled study visit was in the range of 65%–96%
in arm A and 44%–80% in arm B. Median deterioration-free
survival from baseline was 5.7 months in arm A and 2.8 months
in arm B (Figure 1C; supplementary Table S3, available at Annals
of Oncology online, P<0.001). Before progression, no differences
Figure 1. Clinical outcome by treatment arm. Progression-free survival (A) and overall survival (B) were analyzed in the intention-to-treat
population in patients treated with bevacizumab and radiotherapy (arm A) versus radiotherapy alone (arm B) utilizing the log-rank test. (C)
Deterioration-free survival in the intention-to-treat population in arm A versus arm B. (D) Individual functional and symptom scores from
EORTC QLQ-C30/BN20 modules before tumor progression were analyzed in a generalized linear mixed model that controlled for time treat-
ment interactions. Percent effects and 95%confidence intervals are depicted. (E) Cognitive functioning before tumor progression was
assessed by mini-mental status examination (MMSE). Boxes define the median and interquartile range, whiskers define the range. Differences
in treatment arms were compared by Student’s t-test for each timepoint using the Holm–Sidak method to correct for multiple comparisons.
Original article Annals of Oncology
1426 | Wirsching et al. Volume 29 | Issue 6 | 2018
were detected for individual scales in a generalized linear mixed
model, except for less favorable values in arm A for global health
(P¼0.048) and pain (P¼0.027) (Figure 1D).
Cognitive functioning was additionally assessed by serial
MMSE. It remained stable before progression in both groups and
no difference between arms was detected at individual visits
(Figure 1E), or in a linear regression model (P¼0.43 for differen-
ces in slopes). Six individuals with a decline in MMSE of 5 points
or more before progression were identified, all of which were
treated in arm A. Two of these six patients qualified for pseudor-
esponse by RANO.
Among 34 patients who were on steroids at study entry, 21 of
22 patients (95%) in arm A and 8 of 12 patients (66%) in arm B
came off steroids before progression (P¼0.024). The median
time on steroids from study entry was 1.6 months in arm A and
2.6 months in arm B (P¼0.38) (censoring patients at
progression).
Exploratory analyses of treatment effects
Treatment effects on PFS were noted throughout most clinically
defined subgroups and were pronounced in patients with poor
general condition at baseline (KPS 80%, HR 0.16, 95% CI
0.06–0.45, P<0.001) and in patients with cognitive impairment
at baseline (MMSE <27 points, HR 0.08, 95% CI 0.02–0.41,
P¼0.002). In contrast, no improved PFS was noted in patients
with better cognitive functioning (MMSE 27, HR 0.85, 95% CI
0.42–1.72, P¼0.66; supplementary Figure S3, available at Annals
of Oncology online).
Exploratory analyses of treatment effects with respect to OS
identified no subgroup with benefit from bevacizumab, albeit a
tendency to prolonged OS was noted in patients with poor gen-
eral condition (KPS 60%–80%, HR 0.55, 95% CI 0.24–1.24,
P¼0.15) and patients who did not receive any second line ther-
apy (HR 0.44, 95% CI 0.17–1.15, P¼0.092, supplementary
Figure S3, available at Annals of Oncology online).
Utilizing median PFS or OS as a cut-off, receiver operating
characteristics analyses failed to identify prognostic cut-offs for
baseline planimetric estimates of contrast enhancement on T1-
weighted images, or for tumor sizes on T2-weighted images,
including separate analysis of the entire study cohort, arm A or
arm B (data not shown).
Clinical characteristics and outcome by gene
methylation and gene expression
Unsupervised clustering of gene methylation similarities reflected
the methylation classifier-based assignment of glioblastoma sub-
types (Figure 2A). Groups defined by methylation subtypes did
not differ with respect to established prognostic factors, including
age (P¼0.44), KPS (P¼0.80), steroid intake at study entry
(P¼0.41) or MGMT promoter methylation (P¼0.91) and no
differences in PFS or OS were detected (supplementary Figure S4,
available at Annals of Oncology online).
Longer PFS in arm A than in arm B was detected solely in the
subgroup of RTK I glioblastomas (HR 0.25, 95% CI 0.07–0.82,
P¼0.014; Figures 2B, S3 and S4) and there was a trend towards a
higher rate of radiographic complete responses of RTK I com-
pared with RTK II or MES glioblastomas (supplementary Table
S4, available at Annals of Oncology online). No gene methylation
subtype with prolonged OS in arm A versus arm B was identified
(Figure 2C; supplementary Figures S3 and S4, available at Annals
of Oncology online).
There was an association of methylation subtypes with gene ex-
pression subtypes (P¼0.011), including RTK I methylation in
57% of PN, RTK II methylation in 73% of CL and MES methyla-
tion in 53% of glioblastomas with MES gene expression (Figure
2A). No differences in PFS and OS were detected between gene
expression subtypes (Figure S5). PFS was specifically longer
among bevacizumab-treated patients with IDH wildtype PN glio-
blastomas (HR 0.29, 95% CI 0.08–0.98, P¼0.025) (Figure 2D;
supplementary Figures S3 and S5, available at Annals of Oncology
online). No gene expression subtype with differences in OS be-
tween treatment arms was identified (Figure 2E; supplementary
Figures S3 and S5, available at Annals of Oncology online).
Outcome analyses of the MES gene expression subtype were pre-
cluded by imbalance between treatment arms (Table 1;supple-
mentary Figure S5, available at Annals of Oncology online).
Multivariate modeling of outcome
Cox proportional hazards modeling for PFS revealed treatment
with bevacizumab and higher KPS as relevant prognostic factors,
whereas no role for age, steroid intake at study entry or MGMT
promoter methylation emerged (Table 2). Applying the same
model to OS revealed age and KPS as relevant prognostic factors
(Table 2). Univariate analyses are summarized in supplementary
Table S5, available at Annals of Oncology online. Inclusion of gene
methylation and gene expression subtypes into this model as add-
itional single variables identified an association of the RTK II sub-
type with inferior PFS and OS (supplementary Table S6, available
at Annals of Oncology online).
We also considered the possibility that second line treatment
masked effects from study treatment on OS. However, in our
model the inclusion of any salvage therapy versus best supportive
care at recurrence did not render treatment arm a prognostic fac-
tor (HR 1.01, 95% CI 0.55–1.87, P¼0.96), and neither were sur-
gery at recurrence (HR for arm A versus arm B 0.89, 95% CI
0.34–2.30, P¼0.80), or cross-over to receive bevacizumab at re-
currence in arm B (HR for arm A versus arm B 0.63, 95% CI
0.29–1.32, P¼0.22) prognostic. In arm B, 13 patients (52%)
received bevacizumab at first or second recurrence, including two
patients (8%) who received bevacizumab at first and second re-
currence (supplementary Figure S1, available at Annals of
Oncology online).
Discussion
The ARTE trial was designed to provide evidence for a benefit
from first-line bevacizumab antiangiogenic therapy when added
to RT in elderly glioblastoma patients considered non-eligible for
combined modality treatment, based on standards of care when
the trial was initiated [13]. The amendment of the trial to include
only patients with an unmethylated MGMT promoter was moti-
vated ethically when it became clear that temozolomide and not
RT was the preferred first-line monotherapy for elderly glioblast-
oma patients with a methylated MGMT promoter considered
Annals of Oncology Original article
Volume 29 | Issue 6 | 2018 doi:10.1093/annonc/mdy120 | 1427
Figure 2. Molecular glioblastoma subtypes. (A) Unsupervised hierarchical clustering by gene methylation patterns. Columns represent
patients and rows CpG probes. Pearson’s correlation was carried out as distance measure and average linkage utilizing the 5000 probes with
highest standard deviations. Progression-free survival (B, D) and overall survival (C, E) by treatment arm in patients with RTK I gene methyla-
tion (B, C), and in patients with PN gene expression (D, E) glioblastoma subtypes. One patient with IDH mutated glioblastoma was excluded
from outcome analysis in (C) and (E).
Original article Annals of Oncology
1428 | Wirsching et al. Volume 29 | Issue 6 | 2018
non-eligible for combined modality treatment [5,6]. Given that
benefit from RT is not associated with absence or presence of
MGMT promoter methylation in this patient population, we
focused our analyses on the intention-to-treat population.
We found indeed a small increase in OS of 2 months over the
historical benchmark of the NOA-08 trial, but a similar increase
was also seen in the patients treated with RT alone, suggesting an
effect of patient selection or improved standards of care with
time [14], although cross-over to receive bevacizumab in arm B
contributing to OS cannot be ruled out. Overall the outcome
data reported in ARTE compare well with contemporary patient
populations (supplementary Table S7, available at Annals of
Oncology online). That the addition of bevacizumab increased
PFS, but apparently not OS is consistent with the phase III trials
concluded while ARTE was ongoing [8,9]. The benefit in PFS
with bevacizumab according to central review was consistent
with the benefit according to investigator assessment, albeit the
timepoint of progression was antedated in 22% of patients in arm
A due to more rigorous application of imaging criteria.
Early uncontrolled reports of bevacizumab therapy in glio-
blastoma patients suggested that benefit may be pronounced in
elderly and frail patients [1517]. Accordingly, an OS benefit
from bevacizumab in patients who did not qualify for second line
therapy was reported from secondary analyses of the AVAGlio
trial [18]. We also noted a tendency towards prolonged OS in
patients with lower KPS and in patients who did not receive fur-
ther treatment at tumor progression, albeit these subgroup analy-
ses suffer from the lack of statistical power.
In line with reports from the AVAGlio trial [8,19], patient-
reported quality of life was maintained under bevacizumab at least
until progression and more patients came off steroids with bevaci-
zumab. In contrast, an association of bevacizumab treatment with
inferior QoL and worse cognitive functioning was reported from
the RTOG-0825 trial [9], fostering speculation on putative neuro-
toxicity of bevacizumab. In the ARTE trial, bevacizumab was over-
all well tolerated, but indeed more neuropsychiatric adverse events
and cognitive decline before progression were noted in arm A.
Although neurotoxicity from bevacizumab cannot be ruled out,
other causes potentially contributing to cognitive impairment in
individual patients in arm A included the prolonged treatment
period, imbalances in cognitive functioning at baseline and unrec-
ognized tumor progression, as determined by the post hoc radi-
ology review. The largest fraction of adverse events in both
treatment arms were classifiedas other neurologic events, compris-
ing mostly sensorimotor symptoms likely related to tumor burden.
Secondary analyses of outcome measures also included stratifi-
cation by molecular markers. Genome-wide analysis of gene
methylation identified an association of the RTK I methylation
subtype and the proneural gene expression subtype with PFS
benefit from bevacizumab. Although sample size was small, these
results confirm the previously reported link of preferential benefit
from bevacizumab in patients with tumors with a proneural gene
expression pattern [20]. This may provide an important path for-
ward for patient selection in future clinical trials exploring
VEGF-targeted therapies.
In conclusion, ARTE did not confirm the hypothesis that the
combination of bevacizumab with hypofractionated RT prolongs
OS in elderly glioblastoma patients, but exploratory analyses
yielded novel molecular genetics and imaging biomarkers that
may help to enrich patient populations for future trials on novel
anti-VEGF compounds.
Acknowledgements
Nadine Bosbach and Dr Gabriela Senti, Clinical Trial Center
Zurich, for administrative support.
Funding
The study was supported by a grant from Roche Pharma (Basel,
Switzerland). No grant numbers apply.
Disclosure
AFH, HGW, LP, MW, PR, RvM and TH received speaker hono-
raria for advisory boards from Roche. MW received financial
support for research to his institution by Roche. CM and LA are
employees and stock owners of Roche / Genentech. The remain-
der authors declare no potential conflict of interest.
References
1. Ostrom QT, Gittleman H, Fulop J et al. CBTRUS statistical report: pri-
mary brain and central nervous system tumors diagnosed in the United
States in 2008–2012. Neuro Oncol 2015; 17(Suppl 4): iv1–iv62.
Table 2. Multivariate analyses of associations with PFS and OS
PFS OS
Hazard ratio and 95%CI PHazard ratio and 95%CI P
Treatment arm:arm A versus arm B 0.36 (0.20–0.65) 0.001 1.09 (0.63–1.89) 0.75
Age at study entry:65–69 versus 70þyears 1.13 (0.66–1.92) 0.651 0.52 (0.30–0.89) 0.018
KPS:90%–100% versus 60%–80% 0.50 (0.28–0.89) 0.018 0.51 (0.28–0.92) 0.026
Steroids at study entry:no versus yes 1.10 (0.66–1.84) 0.704 0.87 (0.52–1.44) 0.59
MGMT promoter: methylated versus unmethylated
a
0.69 (0.37–1.30) 0.250 0.79 (0.42–1.49) 0.46
a
Note that enrollment was restricted per amendment 1 to unmethylated patients (see supplementary Figure S2, available at Annals of Oncology online).
Annals of Oncology Original article
Volume 29 | Issue 6 | 2018 doi:10.1093/annonc/mdy120 | 1429
2. Stupp R, Mason WP, van den Bent MJ et al. Radiotherapy plus concomi-
tant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005;
352(10): 987–996.
3. Perry JR, Laperriere N, O’Callaghan CJ et al. Short-course radiation plus
temozolomide in elderly patients with glioblastoma. N Engl J Med 2017;
376(11): 1027–1037.
4. Roa W, Brasher PM, Bauman G et al. Abbreviated course of radiation
therapy in older patients with glioblastoma multiforme: a prospective
randomized clinical trial. JCO 2004; 22(9): 1583–1588.
5. Malmstrom A, Gronberg BH, Marosi C 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(9): 916–926.
6. Wick W, Platten M, Meisner C et al. Temozolomide chemotherapy alone
versus radiotherapy alone for malignant astrocytoma in the elderly: the
NOA-08 randomised, phase 3 trial. Lancet Oncol 2012; 13(7): 707–715.
7. Weller M, van den Bent M, Tonn JC et al. European Association for Neuro-
Oncology (EANO) guideline on the diagnosis and treatment of adult astro-
cytic and oligodendroglial gliomas. Lancet Oncol 2017; 18(6): e315–e329.
8. Chinot OL, Wick W, Mason W et al. Bevacizumab plus radiotherapy-
temozolomide for newly diagnosed glioblastoma. N Engl J Med 2014;
370(8): 709–722.
9. Gilbert MR, Dignam JJ, Armstrong TS et al. A randomized trial of bevacizumab
for newly diagnosed glioblastoma. N Engl J Med 2014; 370(8): 699–708.
10. Louis DN, Ohgaki H, Wiestler OD et al. The 2007 WHO classification of
tumours of the central nervous system. Acta Neuropathol 2007; 114(2):
97–109.
11. Jones DT, Capper D, Sill M et al. Next-generation neuropathology:
improving diagnostic accuracy for brain tumors using DNA methylation
array-based molecular profiling. Neuro Oncol 2014; 16: iii4A.
12. Verhaak RG, Hoadley KA, Purdom E et al. Integrated genomic analysis
identifies clinically relevant subtypes of glioblastoma characterized by
abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010; 17:
98–110.
13. Weller M, van den Bent M, Hopkins K et al. EANO guideline for the
diagnosis and treatment of anaplastic gliomas and glioblastoma. Lancet
Oncol 2014; 15(9): e395–e403.
14. Gramatzki D, Dehler S, Rushing EJ et al. Glioblastoma in the Canton of
Zurich, Switzerland revisited: 2005 to 2009. Cancer 2016; 122(14):
2206–2215.
15. Nghiemphu PL, Liu W, Lee Y et al. Bevacizumab and chemotherapy for
recurrent glioblastoma: a single-institution experience. Neurology 2009;
72(14): 1217–1222.
16. Lai A, Tran A, Nghiemphu PL et al. Phase II study of bevacizumab plus
temozolomide during and after radiation therapy for patients with newly
diagnosed glioblastoma multiforme. JCO 2011; 29(2): 142–148.
17. Kreisl TN, Kim L, Moore K et al. Phase II trial of single-agent bevacizu-
mab followed by bevacizumab plus irinotecan at tumor progression in
recurrent glioblastoma. JCO 2009; 27(5): 740–745.
18. Chinot OL, Nishikawa R, Mason W et al. Upfront bevacizumab may ex-
tend survival for glioblastoma patients who do not receive second-line
therapy: an exploratory analysis of AVAglio. Neuro Oncol 2016; 18(9):
1313–1318.
19. Taphoorn MJ, Henriksson R, Bottomley A et al. Health-related quality of
life in a randomized phase III study of bevacizumab, temozolomide, and
radiotherapy in newly diagnosed glioblastoma. JCO 2015; 33(19):
2166–2175.
20. Sandmann T, Bourgon R, Garcia J et al. Patients with proneural glio-
blastoma may derive overall survival benefit from the addition of bevaci-
zumab to first-line radiotherapy and temozolomide: retrospective
analysis of the AVAglio trial. JCO 2015; 33(25): 2735–2744.
Original article Annals of Oncology
1430 | Wirsching et al. Volume 29 | Issue 6 | 2018
... 9,10 Among patients over 80 years, the median OS ranges from 2 to 3.5 months. 4,11 Despite increasing awareness, elderly patients continue to be underrepresented in clinical trials [12][13][14] and even such trials that focus on geriatric patients include only a minority of patients beyond the age of 80. [15][16][17][18][19] The standard treatment for glioblastoma patients up to the age of approximately 70 years consists of maximum safe tumor resection, followed by combined radiochemotherapy with temozolomide. 20 This treatment is typically well tolerated in younger patients but may impose a higher risk for toxicity in the elderly. ...
... One patient received single-agent bevacizumab as a first-line treatment and 1 patient was enrolled in the ARTE trial and was treated with short-course radiotherapy and bevacizumab. 15 Fourteen patients (14%) received second-line treatment after tumor progression: 2 patients (2%) underwent re-resection, 1 patient proceeded with bevacizumab, and the other one without any further treatment. The most commonly administered second-line treatment was temozolomide (n = 6; 6%). ...
... Of note, approximately a third of our patients were treated before prospective studies focusing on elderly glioblastoma patients were available. [15][16][17][18][19] At least some patients, particularly with MGMT-methylated tumors, may have benefitted from temozolomide as mono-or combination therapy. 26,40 Moreover, among patients who received chemotherapy with temozolomide only, 50% had a KPS ≥70% and might have qualified for combination therapy. ...
Article
Full-text available
Background Incidence rates of glioblastoma in very old patients are rising. The standard of care for this cohort is only partially defined and survival remains poor. The aims of this study were to reveal current practice of tumor-specific therapy and supportive care, and to identify predictors for survival in this cohort. Methods Patients aged 80 years or older at the time of glioblastoma diagnosis were retrospectively identified in 6 clinical centers in Switzerland and France. Demographics, clinical parameters, and survival outcomes were annotated from patient charts. Cox proportional hazards modeling was performed to identify parameters associated with survival. Results Of 107 patients, 45 were diagnosed by biopsy, 30 underwent subtotal resection, and 25 had gross total resection. In 7 patients, the extent of resection was not specified. Postoperatively, 34 patients did not receive further tumor-specific treatment. Twelve patients received radiotherapy with concomitant temozolomide, but only 2 patients had maintenance temozolomide therapy. Fourteen patients received temozolomide alone, 35 patients received radiotherapy alone, 1 patient received bevacizumab, and 1 took part in a clinical trial. Median progression-free survival (PFS) was 3.3 months and median overall survival (OS) was 4.2 months. Among patients who received any postoperative treatment, median PFS was 3.9 months and median OS was 7.2 months. Karnofsky performance status (KPS) ≥70%, gross total resection, and combination therapy were associated with better outcomes. The median time spent hospitalized was 30 days, accounting for 23% of the median OS. End-of-life care was mostly provided by nursing homes (n = 20; 32%) and palliative care wards (n = 16; 26%). Conclusions In this cohort of very old patients diagnosed with glioblastoma, a large proportion was treated with best supportive care. Treatment beyond surgery and, in particular, combined modality treatment were associated with longer OS and may be considered for selected patients even at higher ages.
... MGMT methylation data from fourteen studies, involving a total of 1231 patients with differing treatment regimens were included in the analysis for association of OS and MGMT status. The MGMT methylation status was determined in 10 out of 14 studies by methylation specific PCR [15,26,28,30,[33][34][35]38,[42][43][44]. Pyrosequencing was used in one study [40], and 3 studies did not report the methodology of MGMT methylation assessment [15,25,39]. ...
... A forest plot demonstrating the association of MGMT methylation status with OS [15,25,26,28,30,[33][34][35]38,39,40,[42][43][44]. Abbreviations: SE: standard error; CI: confidence interval, ref=reference, bev= bevacizumab, niv= nivolumab < 60= < 60 years, > 60= >60 years. ...
... Size of the red square indicates the relative weight of the study as it contributes to the results of the overall comparison. The diamond at the bottom of the forest plot shows the result when all the individual Figure 2. A forest plot demonstrating the association of MGMT methylation status with OS [15,25,26,28,30,[33][34][35][38][39][40][42][43][44]. Abbreviations: SE: standard error; CI: confidence interval, bev= bevacizumab, niv= nivolumab < 60= < 60 years, > 60= >60 years. ...
Article
Full-text available
Background: Glioblastoma (GBM) is a highly aggressive cancer with poor prognosis that needs better treatment modalities. Moreover, there is a lack of reliable biomarkers to predict the response and outcome of current or newly designed therapies. While several molecular markers have been proposed as potential biomarkers for GBM, their uptake into clinical settings is slow and impeded by marker heterogeneity. Detailed assessment of prognostic and predictive value for biomarkers in well-defined clinical trial settings, if available, is scattered throughout the literature. Here we conducted a systematic review and meta-analysis to evaluate the prognostic and predictive significance of clinically relevant molecular biomarkers in GBM patients. Material and methods: A comprehensive literature search was conducted to retrieve publications from 3 databases (Pubmed, Cochrane and Embase) from January 2010 to December 2021, using specific terms. The combined hazard ratios (HR) and confidence intervals (95% CI) were used to evaluate the association of biomarkers with overall survival (OS) in GBM patients. Results: Twenty-six out of 1831 screened articles were included in this review. Nineteen articles were included in the meta-analyses, and 7 articles were quantitatively summarised. Fourteen studies with 1231 GBM patients showed a significant association of MGMT methylation with better OS with the pooled HR of 1.66 (95% CI 1.32-2.09, p < 0.0001, random effect). Five studies including 541 GBM patients analysed for the prognostic significance of IDH1 mutation showed significantly better OS in patients with IDH1 mutation with a pooled HR of 2.37 (95% CI 1.81-3.12; p < 0.00001]. Meta-analysis performed on 5 studies including 575 GBM patients presenting with either amplification or high expression of EGFR gene did not reveal any prognostic significance with a pooled HR of 1.31 (95% CI 0.96-1.79; p = 0.08). Conclusions: MGMT promoter methylation and IDH1 mutation are significantly associated with better OS in GBM patients. No significant associations were found between EGFR amplification or overexpression with OS.
... /10.5772/intechopen.106408 Bevacizumab is not recommended for people with newly diagnosed glioblastoma due to high rates of adverse events and no improvement in overall survival [79][80][81]. In select cases, bevacizumab may be cautiously used to treat tumor-related edema and to avoid the side-effects of steroids. ...
Chapter
Full-text available
Glioblastoma (GBM) is the third most common primary intracranial tumor and the commonest primary malignant brain tumor in adults. The peak incidence is between 65 and 84 years old. The incidence of GBM increases starkly with age—from 1.3/100,000 between the ages of 35–44 to 15.3/100,000 between the ages of 75–84 years. Elderly patients with GBM have increased comorbidities, lower functional status, aggressive tumor biology, and an overall worse outcome as compared with their younger counterparts. Age is an independent and powerful prognosticator of GBM outcomes, even if the performance status is controlled. Elderly patients with GBM represent a vulnerable heterogeneous cohort. Surgical resection in elderly patients offers a better outcome and improved quality of life as compared with biopsy alone and nowadays can be safely tolerated by elderly patients in specialized centers. The standard of care treatment of glioblastoma based on the Stupp’s protocol excluded patients over the age of 70. Thus, the standard of care treatment in elderly patients with GBM remains controversial. Selected elderly patients with excellent performance status may be treated with Stupp’s protocol. Elderly patients with lower functional status may be treated with a hypofractionated treatment regimen with concomitant and adjuvant temozolomide. Frail patients with MGMT methylated tumor can be treated with temozolomide monotherapy alone. It is also not unreasonable to treat elderly frail patients with MGMT unmethylated GBM with hypofractionated RT alone. Thus, treatment of elderly patients with GBM needs a multidisciplinary approach based on the extent of the tumor, MGMT methylation status, performance status, and even the social situation unique to the elderly patient. This chapter seeks to bring a comprehensive and updated review on the treatment of glioblastoma in the elderly population.
Article
Full-text available
Glioblastoma (GB) stands out as the most prevalent and lethal form of brain cancer. Although great efforts have been made by clinicians and researchers, no significant improvement in survival has been achieved since the Stupp protocol became the standard of care (SOC) in 2005. Despite multimodality treatments, recurrence is almost universal with survival rates under 2 years after diagnosis. Here, we discuss the recent progress in our understanding of GB pathophysiology, in particular, the importance of glioma stem cells (GSCs), the tumor microenvironment conditions, and epigenetic mechanisms involved in GB growth, aggressiveness and recurrence. The discussion on therapeutic strategies first covers the SOC treatment and targeted therapies that have been shown to interfere with different signaling pathways (pRB/CDK4/RB1/P16ink4, TP53/MDM2/P14arf, PI3k/Akt-PTEN, RAS/RAF/MEK, PARP) involved in GB tumorigenesis, pathophysiology, and treatment resistance acquisition. Below, we analyze several immunotherapeutic approaches (i.e., checkpoint inhibitors, vaccines, CAR-modified NK or T cells, oncolytic virotherapy) that have been used in an attempt to enhance the immune response against GB, and thereby avoid recidivism or increase survival of GB patients. Finally, we present treatment attempts made using nanotherapies (nanometric structures having active anti-GB agents such as antibodies, chemotherapeutic/anti-angiogenic drugs or sensitizers, radionuclides, and molecules that target GB cellular receptors or open the blood–brain barrier) and non-ionizing energies (laser interstitial thermal therapy, high/low intensity focused ultrasounds, photodynamic/sonodynamic therapies and electroporation). The aim of this review is to discuss the advances and limitations of the current therapies and to present novel approaches that are under development or following clinical trials.
Article
Full-text available
Glioblastoma multiforme (GBM) is the most common type of glioma, with a median survival of 14.6 months post-diagnosis. Understanding the molecular profile of such tumors allowed the development of specific targeted therapies toward GBM, with a major role attributed to tyrosine kinase receptor inhibitors and immune checkpoint inhibitors. Targeted therapeutics are drugs that work by specific binding to GBM-specific or overexpressed markers on the tumor cellular surface and therefore contain a recognition moiety linked to a cytotoxic agent, which produces an antiproliferative effect. In this review, we have summarized the available information on the targeted therapeutics used in clinical trials of GBM and summarized current obstacles and advances in targeted therapy concerning specific targets present in GBM tumor cells, outlined efficacy endpoints for major classes of investigational drugs, and discussed promising strategies towards an increase in drug efficacy in GBM.
Article
Full-text available
ABSTRACTS Glioblastoma multiforme (GBM) is a malignant type of glioma. This malignant brain tumor is a devastating disease and is often fatal. The spectrum of illness and poor prognosis associated with brain tumors extract a terrible toll on patients and their families. The inoperability of these tumors and resistance to radiation and chemotherapy contribute to the fatal outcome of this disease. Thus, scientists are hunting for the new drug candidate and safer chemoprevention, especially the phytochemicals that possess potent anti-tumor properties. We have summarized the cellular and biochemical impacts of different phytochemicals that can successfully encounter GBM via induction of apoptosis and active interference in different cell and molecular pathways associated with GBM in brain tumors. The in silico predictive model determining the blood–brain barrier permeability of the compound and their potential druggability are discussed in the review.
Article
Full-text available
Glioblastoma multiforme (GBM) is the most frequent and malignant primary central nervous system tumor in adults. The gold-standard management of GBM includes post-operative radiotherapy (RT) with concurrent and secondary temozolomide (TMZ) treatment. The present meta-analysis study examined the efficacy of the early administration of bevacizumab prior to standard RT plus TMZ in managing patients with GBM and unfavorable prognostic factors. Between 1983 and 2020, the present study looked for comparative articles involving standard RT plus TMZ and RT/TMZ combined with bevacizumab treatment in patients with GBM. The primary outcomes involved in this study include progression-free survival and overall survival. The present study suggested that bevacizumab administration plus standard RT/TMZ (BEV group) treatment was associated with increased survival of patients with GBM compared with those treated with standard RT/TMZ (CG/Control group) treatment only.
Article
Full-text available
Purpose of Review To summarize the mechanisms of tumor angiogenesis and resistance to antiangiogenic therapy, and the influence on tumor microenvironment. Recent Findings Several clinical trials have investigated the activity of anti-VEGF monoclonal antibodies and tyrosine kinase inhibitors in glioblastoma, shedding the light on their limitations in terms of disease control and survival. We have outlined the mechanisms of resistance to antiangiogenic therapy, including vessel co-option, hypoxic signaling in response to vessel destruction, modulation of glioma stem cells, and trafficking of tumor-associated macrophages in tumor microenvironment. Moreover, novel generation of antiangiogenic compounds for glioblastoma, including small interfering RNAs and nanoparticles, as a delivery vehicle, could enhance selectivity and reduce side effects of treatments. Summary There is still a rationale for the use of antiangiogenic therapy, but a better understanding of vascular co-option, vascular mimicry, and dynamic relationships between immunosuppressive microenvironment and blood vessel destruction is crucial to develop next-generation antiangiogenic compounds.
Article
The intent of this article is to provide a comprehensive, educational review of adult-type diffuse gliomas, bringing together the role of the multidisciplinary team and giving the reader biologic, surgical, radiologic, and oncologic insight into these tumors. The reader will be guided through subsections that explain the molecular pathophysiology, diagnosis, treatment, prognosis and natural history of the disease. The 2021 World Health Organization Classification will be explained through a combination of concise historic review of glioma diagnosis and contemporary knowledge of the molecular pathways. Accompanying illustrations will act as powerful tools for comprehension. These concepts will be underpinned by frequent reference to neuroradiology and a grounding of normal brain composition.Learning Objective: To understand the pathophysiologic concepts that underly the World Health Organization Classification and natural history of adult-type diffuse gliomas and how these relate to imaging and treatment strategies
Article
Objective To fully investigate the efficacy and safety of bevacizumab for glioblastoma. Methods Databases were searched for phase II/III randomized controlled trials treated with bevacizumab. Results Bevacizumab significantly improved the PFS in glioblastoma patients, but did not prolong OS. PFS was significantly prolonged in both first-line and second-line treatment. Bevacizumab plus temozolomide was correlated with improved PFS for patients with different MGMT methylation status. Bevacizumab could increase the risk of hypertension, proteinuria, thromboembolic, and infection. Hypertension should be well concerned. Conclusions Bevacizumab-containing regimen can significantly improve PFS, but did not prolong OS.
Article
Full-text available
The AVAglio (Avastin in Glioblastoma) and RTOG-0825 randomized, placebo-controlled phase III trials in newly diagnosed glioblastoma reported prolonged progression-free survival (PFS), but not overall survival (OS), with the addition of bevacizumab to radiotherapy plus temozolomide. To establish whether certain patient subgroups derived an OS benefit from the addition of bevacizumab to first-line standard-of-care therapy, AVAglio patients were retrospectively evaluated for molecular subtype, and bevacizumab efficacy was assessed for each patient subgroup. A total of 349 pretreatment specimens (bevacizumab arm, n = 171; placebo arm, n = 178) from AVAglio patients (total, N = 921) were available for biomarker analysis. Samples were profiled for gene expression and isocitrate dehydrogenase 1 (IDH1) mutation status and classified into previously identified molecular subtypes. PFS and OS were assessed within each subtype. A multivariable analysis accounting for prognostic covariates revealed that bevacizumab conferred a significant OS advantage versus placebo for patients with proneural IDH1 wild-type tumors (17.1 v 12.8 months, respectively; hazard ratio, 0.43; 95% CI, 0.26 to 0.73; P = .002). This analysis also revealed an interaction between the proneural subtype biomarker and treatment arm (P = .023). The group of patients with mesenchymal and proneural tumors derived a PFS benefit from bevacizumab compared with placebo; however, this translated to an OS benefit in the proneural subset only. Retrospective analysis of AVAglio data suggests that patients with IDH1 wild-type proneural glioblastoma may derive an OS benefit from first-line bevacizumab treatment. The predictive value of the proneural subtype observed in AVAglio should be validated in an independent data set. © 2015 by American Society of Clinical Oncology.
Article
The European Association for Neuro-Oncology guideline provides recommendations for the clinical care of adult patients with astrocytic and oligodendroglial gliomas, including glioblastomas. The guideline is based on the 2016 WHO classification of tumours of the central nervous system and on scientific developments since the 2014 guideline. The recommendations focus on pathological and radiological diagnostics, and the main treatment modalities of surgery, radiotherapy, and pharmacotherapy. In this guideline we have also integrated the results from contemporary clinical trials that have changed clinical practice. The guideline aims to provide guidance for diagnostic and management decisions, while limiting unnecessary treatments and costs. The recommendations are a resource for professionals involved in the management of patients with glioma, for patients and caregivers, and for health-care providers in Europe. The implementation of this guideline requires multidisciplinary structures of care, and defined processes of diagnosis and treatment.
Article
Background Glioblastoma is associated with a poor prognosis in the elderly. Survival has been shown to increase among patients 70 years of age or younger when temozolomide chemotherapy is added to standard radiotherapy (60 Gy over a period of 6 weeks). In elderly patients, more convenient shorter courses of radiotherapy are commonly used, but the benefit of adding temozolomide to a shorter course of radiotherapy is unknown. Methods We conducted a trial involving patients 65 years of age or older with newly diagnosed glioblastoma. Patients were randomly assigned to receive either radiotherapy alone (40 Gy in 15 fractions) or radiotherapy with concomitant and adjuvant temozolomide. Results A total of 562 patients underwent randomization, 281 to each group. The median age was 73 years (range, 65 to 90). The median overall survival was longer with radiotherapy plus temozolomide than with radiotherapy alone (9.3 months vs. 7.6 months; hazard ratio for death, 0.67; 95% confidence interval [CI], 0.56 to 0.80; P<0.001), as was the median progression-free survival (5.3 months vs. 3.9 months; hazard ratio for disease progression or death, 0.50; 95% CI, 0.41 to 0.60; P<0.001). Among 165 patients with methylated O⁶-methylguanine–DNA methyltransferase (MGMT) status, the median overall survival was 13.5 months with radiotherapy plus temozolomide and 7.7 months with radiotherapy alone (hazard ratio for death, 0.53; 95% CI, 0.38 to 0.73; P<0.001). Among 189 patients with unmethylated MGMT status, the median overall survival was 10.0 months with radiotherapy plus temozolomide and 7.9 months with radiotherapy alone (hazard ratio for death, 0.75; 95% CI, 0.56 to 1.01; P=0.055; P=0.08 for interaction). Quality of life was similar in the two trial groups. Conclusions In elderly patients with glioblastoma, the addition of temozolomide to short-course radiotherapy resulted in longer survival than short-course radiotherapy alone. (Funded by the Canadian Cancer Society Research Institute and others; ClinicalTrials.gov number, NCT00482677.)
Article
BACKGROUND: A population-based analysis of patients with glioma diagnosed between 1980 and 1994 in the Canton of Zurich in Switzerland confirmed the overall poor prognosis of glioblastoma. To explore changes in outcome, registry data were reevaluated for patients diagnosed between 2005 and 2009. METHODS: Patients with glioblastoma who were diagnosed between 2005 and 2009 were identified by the Zurich and Zug Cancer Registry. The prognostic significance of epidemiological and clinical data, isocitrate dehydrogenase 1 (IDH1)(R132H) mutation status, and O6 methylguanine DNA methyltransferase (MGMT) promoter methylation status was analyzed using the Kaplan-Meier method and the Cox proportional hazards model. RESULTS: A total of 264 patients with glioblastoma were identified, for an annual incidence of 3.9 compared with the previous incidence of 3.7. The mean age of the patients at the time of diagnosis was 59.5 years in the current cohort compared with 61.3 years previously. The overall survival (OS) rate was 46.4% at 1 year, 22.5% at 2 years, and 14.4% at 3 years in the current study compared with 17.7% at 1 year, 3.3% at 2 years, and 1.2% at 3 years as reported previously. The median OS for all patients with glioblastoma was 11.5 months compared with 4.9 months in the former patient population. The median OS was 1.9 months for best supportive care, 6.2 months for radiotherapy alone, 6.7 months for temozolomide alone, and 17.0 months for radiotherapy plus temozolomide. Multivariate analysis revealed age, Karnofsky performance score, extent of tumor resection, first-line treatment, year of diagnosis, and MGMT promoter methylation status were associated with survival in patients with IDH1(R132H) -nonmutant glioblastoma. CONCLUSIONS: The OS of patients newly diagnosed with glioblastoma in the Canton of Zurich in Switzerland markedly improved from 1980 through 1994 to 2005 through 2009. Cancer 2016. © 2016 American Cancer Society.
Article
The fourth edition of the World Health Organization (WHO) classification of tumours of the central nervous system, published in 2007, lists several new entities, including angiocentric glioma, papillary glioneuronal tumour, rosette-forming glioneuronal tumour of the fourth ventricle, papillary tumour of the pineal region, pituicytoma and spindle cell oncocytoma of the adenohypophysis. Histological variants were added if there was evidence of a different age distribution, location, genetic profile or clinical behaviour; these included pilomyxoid astrocytoma, anaplastic medulloblastoma and medulloblastoma with extensive nodularity. The WHO grading scheme and the sections on genetic profiles were updated and the rhabdoid tumour predisposition syndrome was added to the list of familial tumour syndromes typically involving the nervous system. As in the previous, 2000 edition of the WHO ‘Blue Book', the classification is accompanied by a concise commentary on clinico-pathological characteristics of each tumour type. The 2007 WHO classification is based on the consensus of an international Working Group of 25 pathologists and geneticists, as well as contributions from more than 70 international experts overall, and is presented as the standard for the definition of brain tumours to the clinical oncology and cancer research communities world-wide
Article
Background: A population-based analysis of patients with glioma diagnosed between 1980 and 1994 in the Canton of Zurich in Switzerland confirmed the overall poor prognosis of glioblastoma. To explore changes in outcome, registry data were reevaluated for patients diagnosed between 2005 and 2009. Methods: Patients with glioblastoma who were diagnosed between 2005 and 2009 were identified by the Zurich and Zug Cancer Registry. The prognostic significance of epidemiological and clinical data, isocitrate dehydrogenase 1 (IDH1)(R132H) mutation status, and O6 methylguanine DNA methyltransferase (MGMT) promoter methylation status was analyzed using the Kaplan-Meier method and the Cox proportional hazards model. Results: A total of 264 patients with glioblastoma were identified, for an annual incidence of 3.9 compared with the previous incidence of 3.7. The mean age of the patients at the time of diagnosis was 59.5 years in the current cohort compared with 61.3 years previously. The overall survival (OS) rate was 46.4% at 1 year, 22.5% at 2 years, and 14.4% at 3 years in the current study compared with 17.7% at 1 year, 3.3% at 2 years, and 1.2% at 3 years as reported previously. The median OS for all patients with glioblastoma was 11.5 months compared with 4.9 months in the former patient population. The median OS was 1.9 months for best supportive care, 6.2 months for radiotherapy alone, 6.7 months for temozolomide alone, and 17.0 months for radiotherapy plus temozolomide. Multivariate analysis revealed age, Karnofsky performance score, extent of tumor resection, first-line treatment, year of diagnosis, and MGMT promoter methylation status were associated with survival in patients with IDH1(R132H) -nonmutant glioblastoma. Conclusions: The OS of patients newly diagnosed with glioblastoma in the Canton of Zurich in Switzerland markedly improved from 1980 through 1994 to 2005 through 2009. Cancer 2016. © 2016 American Cancer Society.
Article
Background: In this post-hoc, exploratory analysis, we examined outcomes for patients enrolled in the AVAglio trial of front-line bevacizumab or placebo plus radiotherapy/temozolomide who received only a single line of therapy. Methods: Patients with newly diagnosed glioblastoma received protocol-defined treatment until progressive disease (PD). Co-primary endpoints were investigator-assessed progression-free survival (PFS) and overall survival (OS). After confirmed PD, patients were treated at the investigators' discretion. PFS/OS were assessed in patients with a PFS event who did not receive post-PD therapy (Group 1) and patients with a PFS event who received post-PD therapy plus patients who did not have a PFS event at the final data cutoff (Group 2). Kaplan-Meier methodology was used. A multivariate Cox proportional hazards model for known prognostic variables was generated. Results: Baseline characteristics were balanced. In patients with a PFS event who did not receive post-PD therapy (Group 1; n = 225 [24.4% of the intent-to-treat population]), the addition of bevacizumab to radiotherapy/temozolomide resulted in a 3.6-month extension in both median PFS (hazard ratio [HR]: 0.62, P = .0016) and median OS (HR: 0.67, P = .0102). Multivariate analyses supported this OS benefit (HR: 0.66). In the remaining patients (Group 2; n = 696), a 5.2-month PFS extension was observed in bevacizumab-treated patients (HR: 0.61, P < .0001); OS was comparable between the treatment arms (HR: 0.88, P = .1502). No significant differences in safety were observed between the 2 groups. Conclusion: This exploratory analysis suggests that the addition of bevacizumab to standard glioblastoma treatment prolongs PFS and OS for patients with PD who receive only one line of therapy.
Article
As glioblastoma progresses, patients experience a decline in health-related quality of life (HRQoL). Delaying this decline is an important treatment goal. In newly diagnosed glioblastoma, progression-free survival was prolonged when bevacizumab was added to radiotherapy plus temozolomide (RT/TMZ) versus placebo plus RT/TMZ (phase III AVAglio study; hazard ratio, 0.64; 95% CI, 0.55 to 0.74; P < .001). To ensure that addition of bevacizumab to standard-of-care therapy was not associated with HRQoL detriment, HRQoL assessment was a secondary objective. Patients completed European Organisation for Research and Treatment of Cancer Quality of Life Questionnaires C30 and BN20 at each tumor assessment (Appendix Table A1, online only). Raw scores were converted to a 100-point scale and mean changes from baseline scores were evaluated (stable: < 10-point change; clinically relevant deterioration/improvement: ≥ 10-point change). Deterioration-free survival was the time to deterioration/progression/death; time to deterioration was the time to deterioration/death. Most evaluable patients who had not progressed (> 74%) completed all HRQoL assessments for at least 1 year of treatment, and almost all completed at least one HRQoL assessment at baseline (98.3% and 97.6%, bevacizumab and placebo arms, respectively). Mean changes from baseline did not reach a clinically relevant difference between arms for most items. HRQoL declined at progression in both arms. The addition of bevacizumab to RT/TMZ resulted in statistically longer (P < .001) deterioration-free survival across all items. Time to deterioration was not statistically longer in the placebo plus RT/TMZ arm (v bevacizumab) for any HRQoL item. The addition of bevacizumab to standard-of-care treatment for newly diagnosed glioblastoma had no impact on HRQoL during the progression-free period. © 2015 by American Society of Clinical Oncology.