scispace - formally typeset
Search or ask a question
Journal ArticleDOI

Radiotherapy plus Concomitant and Adjuvant Temozolomide for Glioblastoma

01 May 2005-Oncology Times UK (Ovid Technologies (Wolters Kluwer Health))-Vol. 27, Iss: 9, pp 15-16
About: This article is published in Oncology Times UK.The article was published on 2005-05-01 and is currently open access. It has received 3381 citations till now. The article focuses on the topics: Temozolomide & Concomitant.

Content maybe subject to copyright    Report

Serveur Académique Lausannois SERVAL serval.unil.ch
Publisher’s version PDF
Faculty of Biology and Medicine Publication
“From [Publication Title, Author(s), Title of Article, Volume No., Page No. Copyright © (notice year)
Massachusetts Medical Society. Reprinted with permission.
Title: Radiotherapy plus concomitant and adjuvant temozolomide for
glioblastoma.
Authors: Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B,
Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U,
Curschmann J, Janzer RC, Ludwin SK, Gorlia T, Allgeier A, Lacombe D,
Cairncross JG, Eisenhauer E, Mirimanoff RO, European Organisation
for Research and Treatment of Cancer Brain Tumor and Radiotherapy
Groups., National Cancer Institute of Canada Clinical Trials Group.
Journal: The New England journal of medicine
Year: 2005 Mar 10
Volume: 352
Issue: 10
Pages: 987-96
DOI: 10.1056/NEJMoa043330

n engl j med
352;10
www.nejm.org march
10, 2005
The
new england journal
of
medicine
987
original article
Radiotherapy plus Concomitant
and Adjuvant Temozolomide for Glioblastoma
Roger Stupp, M.D., Warren P. Mason, M.D., Martin J. van den Bent, M.D.,
Michael Weller, M.D., Barbara Fisher, M.D., Martin J.B. Taphoorn, M.D.,
Karl Belanger, M.D., Alba A. Brandes, M.D., Christine Marosi, M.D.,
Ulrich Bogdahn, M.D., Jürgen Curschmann, M.D., Robert C. Janzer, M.D.,
Samuel K. Ludwin, M.D.,Thierry Gorlia, M.Sc., Anouk Allgeier, Ph.D.,
Denis Lacombe, M.D., J. Gregory Cairncross, M.D., Elizabeth Eisenhauer, M.D.,
and René O. Mirimanoff, M.D., for the European Organisation for Research
and Treatment of Cancer Brain Tumor and Radiotherapy Groups and the National
Cancer Institute of Canada Clinical Trials Group*
From the Centre Hospitalier Universitaire
Vaudois, Lausanne, Switzerland (R.S.,
R-C.J., R.O.M.); Princess Margaret Hospital,
Toronto (W.P.M.); Daniel den Hoed Oncol-
ogy Center–Erasmus University Medical
Center Rotterdam, Rotterdam, the Neth-
erlands (M.J.B.); the University of Tübin-
gen Medical School, Tübingen, Germany
(M.W.); the University of Western Ontario,
London, Ont., Canada (B.F.); the Universi-
ty Medical Center, Utrecht, the Netherlands
(M.J.B.T.); Hôpital Notre Dame du Centre
Hospitalier Universitaire, Montreal (K.B.);
Azienda-Ospedale Università, Padova, Italy
(A.A.B.); Medical University of Vienna, Vi-
enna (C.M.); Universitätskliniken, Regens-
burg, Germany (U.B.); Inselspital, Bern,
Switzerland (J.C.); Queen’s University,
Kingston, Ont., Canada (S.K.L.); the Euro-
pean Organisation for Research and Treat-
ment of Cancer Data Center, Brussels (T.G.,
A.A., D.L.); the University of Calgary, Cal-
gary, Alta., Canada (J.G.C.); and the Nation-
al Cancer Institute of Canada Clinical Trials
Group, Kingston, Ont., Canada (E.E.). Ad-
dress reprint requests to Dr. Stupp at the
Multidisciplinary Oncology Center, Centre
Hospitalier Universitaire Vaudois, 46, rue
du Bugnon, CH-1011 Lausanne, Switzer-
land, or at roger.stupp@chuv.hospvd.ch.
*Participating institutions and investiga-
tors are listed in the Appendix.
N Engl J Med 2005;352:987-96.
Copyright © 2005 Massachusetts Medical Society.
background
Glioblastoma, the most common primary brain tumor in adults, is usually rapidly fatal.
The current standard of care for newly diagnosed glioblastoma is surgical resection to
the extent feasible, followed by adjuvant radiotherapy. In this trial we compared radio-
therapy alone with radiotherapy plus temozolomide, given concomitantly with and after
radiotherapy, in terms of efficacy and safety.
methods
Patients with newly diagnosed, histologically confirmed glioblastoma were randomly
assigned to receive radiotherapy alone (fractionated focal irradiation in daily fractions of
2 Gy given 5 days per week for 6 weeks, for a total of 60 Gy) or radiotherapy plus contin-
uous daily temozolomide (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 adju-
vant temozolomide (150 to 200 mg per square meter for 5 days during each 28-day
cycle). The primary end point was overall survival.
results
A total of 573 patients from 85 centers underwent randomization. The median age was
56 years, and 84 percent of patients had undergone debulking surgery. At a median fol-
low-up of 28 months, the median survival was 14.6 months with radiotherapy plus
temozolomide and 12.1 months with radiotherapy alone. The unadjusted hazard ratio
for death in the radiotherapy-plus-temozolomide group was 0.63 (95 percent confidence
interval, 0.52 to 0.75; P<0.001 by the log-rank test). The two-year survival rate was
26.5 percent with radiotherapy plus temozolomide and 10.4 percent with radiotherapy
alone. Concomitant treatment with radiotherapy plus temozolomide resulted in grade 3
or 4 hematologic toxic effects in 7 percent of patients.
conclusions
The addition of temozolomide to radiotherapy for newly diagnosed glioblastoma result-
ed in a clinically meaningful and statistically significant survival benefit with minimal
additional toxicity.
abstract
The New England Journal of Medicine
Downloaded from nejm.org at CTR HOSPITAL UNIVERSITAIRE VAUDOIS on October 12, 2017. For personal use only. No other uses without permission.
Copyright © 2005 Massachusetts Medical Society. All rights reserved.

n engl j med
352;10
www.nejm.org march
10
,
2005
The
new england journal
of
medicine
988
lioblastoma is the most frequent
primary malignant brain tumor in adults.
Median survival is generally less than one
year from the time of diagnosis, and even in the
most favorable situations, most patients die within
two years.
1-3
Standard therapy consists of surgical
resection to the extent that is safely feasible, fol-
lowed by radiotherapy; in the United States, adju-
vant carmustine, a nitrosourea drug, is commonly
prescribed.
4,5
Cooperative-group trials have inves-
tigated the addition of various chemotherapeutic
regimens to radiotherapy,
6-9
but no randomized
phase 3 trial of nitrosourea-based adjuvant chemo-
therapy has demonstrated a significant survival ben-
efit as compared with radiotherapy alone, although
there were more long-term survivors in the chemo-
therapy groups in some studies.
10
A meta-analysis
based on 12 randomized trials suggested a small
survival benefit of chemotherapy, as compared
with radiotherapy alone (a 5 percent increase in
survival at two years, from 15 percent to 20 per-
cent).
11
The meta-analysis included 37 percent of
patients with prognostically more favorable, lower-
grade gliomas.
Temozolomide, an oral alkylating agent, has
demonstrated antitumor activity as a single agent
in the treatment of recurrent glioma.
12-14
The ap-
proved conventional schedule is a daily dose of 150
to 200 mg per square meter of body-surface area for
5 days of every 28-day cycle. Daily therapy at a dose
of 75 mg per square meter for up to seven weeks is
safe; this level of exposure to temozolomide
15
de-
pletes the DNA-repair enzyme O
6
-methylguanine-
DNA methyltransferase (MGMT).
16
This effect may
be important because low levels of MGMT in tumor
tissue are associated with longer survival among pa-
tients with glioblastoma who are receiving nitroso-
urea-based adjuvant chemotherapy.
17,18
A pilot phase 2 trial demonstrated the feasibility
of the concomitant administration of temozolo-
mide with fractionated radiotherapy, followed by up
to six cycles of adjuvant temozolomide, and suggest-
ed that this treatment had promising clinical activ-
ity (two-year survival rate, 31 percent).
19
The Euro-
pean Organisation for Research and Treatment of
Cancer (EORTC) Brain Tumor and Radiotherapy
Groups and the National Cancer Institute of Canada
(NCIC) Clinical Trials Group therefore initiated a
randomized, multicenter, phase 3 trial to compare
this regimen with radiotherapy alone in patients
with newly diagnosed glioblastoma.
patients
Patients 18 to 70 years of age with newly diagnosed
and histologically confirmed glioblastoma (World
Health Organization [WHO] grade IV astrocytoma)
were eligible for the study. Eligible patients had a
WHO performance status of 2 or less and adequate
hematologic, renal, and hepatic function (absolute
neutrophil count, ≥1500 per cubic millimeter; plate-
let count, ≥100,000 per cubic millimeter; serum cre-
atinine level, ≤1.5 times the upper limit of normal
in the laboratory where it was measured; total se-
rum bilirubin level, ≤1.5 times the upper limit of
normal; and liver-function values, <3 times the up-
per limit of normal for the laboratory). Patients who
were receiving corticosteroids had to receive a stable
or decreasing dose for at least 14 days before ran-
domization. All patients provided written informed
consent, and the study was approved by the ethics
committees of the participating centers.
study design and treatment
Within six weeks after the histologic diagnosis of
glioblastoma, we randomly assigned eligible pa-
tients to receive standard focal radiotherapy alone
(the control group) or standard radiotherapy plus
concomitant daily temozolomide, followed by adju-
vant temozolomide. Randomization was performed
at the EORTC Data Center, and patients were strat-
ified according to WHO performance status, wheth-
er or not they had previously undergone debulking
surgery, and the treatment center.
20
The assigned
treatment had to begin within one week after ran-
domization.
Radiotherapy consisted of fractionated focal ir-
radiation at a dose of 2 Gy per fraction given once
daily five days per week (Monday through Friday)
over a period of six weeks, for a total dose of 60 Gy.
Radiotherapy was delivered to the gross tumor vol-
ume with a 2-to-3-cm margin for the clinical target
volume. Radiotherapy was planned with dedicated
computed tomography (CT) and three-dimensional
planning systems; conformal radiotherapy was de-
livered with linear accelerators with nominal energy
of 6 MV or more, and quality assurance was per-
formed by means of individual case reviews.
21
Concomitant chemotherapy consisted of tem-
ozolomide (marketed as Temodal in Europe and
Canada and Temodar in the United States; Scher-
ing-Plough) at a dose of 75 mg per square meter per
g
methods
The New England Journal of Medicine
Downloaded from nejm.org at CTR HOSPITAL UNIVERSITAIRE VAUDOIS on October 12, 2017. For personal use only. No other uses without permission.
Copyright © 2005 Massachusetts Medical Society. All rights reserved.

n engl j med
352;10
www.nejm.org march
10, 2005
radiotherapy with or without temozolomide for glioblastoma
989
day, given 7 days per week from the first day of ra-
diotherapy until the last day of radiotherapy, but for
no longer than 49 days. After a 4-week break, pa-
tients were then to receive up to six cycles of adju-
vant temozolomide according to the standard 5-day
schedule every 28 days. The dose was 150 mg per
square meter for the first cycle and was increased to
200 mg per square meter beginning with the second
cycle, so long as there were no hematologic toxic ef-
fects. Because continuous daily temozolomide can
cause lymphocytopenia, with a possible increased
risk of opportunistic infections, patients in the ra-
diotherapy-plus-temozolomide group were to re-
ceive prophylaxis against
Pneumocystis carinii
pneu-
monia, consisting of either inhaled pentamidine
or oral trimethoprim–sulfamethoxazole,
22
during
concomitant treatment with radiotherapy plus tem-
ozolomide. Antiemetic prophylaxis with metoclo-
pramide or a 5-hydroxytryptamine
3
antagonist was
recommended before the initial doses of concomi-
tant temozolomide and was required during the ad-
juvant five-day courses of temozolomide.
surveillance and follow-up
The baseline examination included CT or magnetic
resonance imaging (MRI), full blood counts and
blood chemistry tests, and a physical examination
that included the Mini–Mental State Examination
(MMSE) and a quality-of-life questionnaire. During
radiotherapy (with or without temozolomide), pa-
tients were to be seen every week. Twenty-one to 28
days after the completion of radiotherapy and every
3 months thereafter, patients underwent a compre-
hensive evaluation, including administration of the
MMSE and the quality-of-life questionnaire and
radiologic assessment of the tumor. During adju-
vant temozolomide therapy, patients underwent a
monthly clinical evaluation and a comprehensive
evaluation at the end of cycles 3 and 6. Tumor pro-
gression was defined according to the modified
WHO criteria as an increase in tumor size by 25 per-
cent, the appearance of new lesions, or an increased
need for corticosteroids.
23
When there was tumor
progression or after two years of follow-up, patients
were treated at the investigator’s discretion, and the
type of second-line therapy was recorded. Toxic ef-
fects were graded according to the National Cancer
Institute Common Toxicity Criteria, version 2.0,
with a score of 1 indicating mild adverse effects, a
score of 2 moderate adverse effects, a score of 3 se-
vere adverse effects, and a score of 4 life-threatening
adverse effects.
statistical analysis
The primary end point was overall survival; second-
ary end points were progression-free survival, safety,
and the quality of life. Overall survival and pro-
gression-free survival were analyzed by the Kaplan–
Meier method, with use of two-sided log-rank
statistics. This study had 80 percent power at a sig-
nificance level of 0.05 to detect a 33 percent in-
crease in median survival (hazard ratio for death,
0.75), assuming that 382 deaths occurred. All analy-
ses were conducted on an intention-to-treat basis.
The Cox proportional-hazards model was fitted to
adjust for stratification factors and other confound-
ing variables. Toxic effects are reported separately
for the radiotherapy period, defined as extending
from day 1 of radiotherapy until 28 days after the
last day of radiotherapy, or until the first day of ad-
juvant temozolomide therapy. The adjuvant-therapy
period was defined as extending from the first day
of adjuvant temozolomide therapy until 35 days
after day 1 of the last cycle of temozolomide. Find-
ings with respect to the quality of life are not re-
ported here.
organization of the trial
The concept of the trial was developed by Dr. Stupp
in collaboration with the EORTC Data Center, the
EORTC Brain Tumor and Radiotherapy Groups,
and the NCIC Clinical Trials Group, represented by
Drs. Cairncross and Eisenhauer. The radiotherapy
design and quality assurance were supervised by
Dr. Mirimanoff. The trial was sponsored by the
EORTC Brain Tumor and Radiotherapy Groups
(trial 22981/26981) in Europe and the NCIC Clini-
cal Trials Group (trial CE.3) in Canada. The trial was
supported by an unrestricted educational grant
from Schering-Plough, which also provided the
study drug; however, Schering-Plough was not in-
volved in trial design or analysis. All data were col-
lected by the EORTC and NCIC data centers and re-
viewed by Drs. Stupp and Mirimanoff. The analysis
was performed by the EORTC statistician, Mr. Gor-
lia. Histologic specimens were reviewed centrally
(according to the revised WHO classification sys-
tem
24
) by a panel of three neuropathologists in Eu-
rope (Robert C. Janzer in Lausanne, Switzerland
[chair]; Peter Wesseling in Nijmegen, the Nether-
lands; and Karima Mohktari in Paris) and a single
neuropathologist in Canada (Samuel Ludwin, King-
ston, Ont.). The article was written by Dr. Stupp with
support from a medical writer and coauthors; all au-
thors reviewed the manuscript.
The New England Journal of Medicine
Downloaded from nejm.org at CTR HOSPITAL UNIVERSITAIRE VAUDOIS on October 12, 2017. For personal use only. No other uses without permission.
Copyright © 2005 Massachusetts Medical Society. All rights reserved.

n engl j med
352;10
www.nejm.org march
10
,
2005
The
new england journal
of
medicine
990
patients
From August 2000 until March 2002, 573 patients
from 85 institutions in 15 countries were randomly
assigned to receive radiotherapy (286 patients) or
radiotherapy plus temozolomide (287 patients).
Nearly 50 percent of the patients were enrolled at
17 institutions. The characteristics of the patients
in the two groups were well balanced at baseline
(Table 1). The median age was 56 years, and 84 per-
cent of patients had undergone debulking surgery.
Slightly more patients in the radiotherapy group
than in the radiotherapy-plus-temozolomide group
were receiving corticosteroids at the time of ran-
domization (75 percent vs. 67 percent). Histologic
slides were submitted for 85 percent of patients,
and central pathological review confirmed the diag-
nosis of glioblastoma in 93 percent of the reviewed
cases; 3 percent had anaplastic astrocytoma or oli-
goastrocytoma (WHO grade III), and in 1 percent
submitted material was insufficient for a definitive
diagnosis.
disposition of patients and delivery
of treatment
The median time from diagnosis to the start of ther-
apy was 5 weeks (range, 2.0 to 12.9) in the radio-
therapy group and 5 weeks (range, 1.7 to 10.7) in
the radiotherapy-plus-temozolomide group. Table 2
summarizes the details of treatment. Unplanned
interruptions in radiotherapy were usually brief
(median, four days) and interruptions due to the
toxicity of therapy occurred in only 3 percent of the
radiotherapy group and 4 percent of the radiother-
apy-plus-temozolomide group. The other reasons
were mainly administrative (e.g., holidays, radio-
therapy equipment maintenance, or technical prob-
lems). One patient randomly assigned to radiother-
apy alone received radiotherapy plus temozolomide.
Among the 287 patients who were assigned to re-
ceive concomitant radiotherapy plus temozolo-
mide, 85 percent completed both radiotherapy and
temozolomide as planned. Thirty-seven patients
(13 percent) prematurely discontinued temozolo-
mide because of toxic effects (in 14 patients), dis-
ease progression (in 11), or other reasons (in 12).
After radiotherapy, 223 patients in the radio-
therapy-plus-temozolomide group (78 percent)
started adjuvant temozolomide and received a me-
dian of 3 cycles (range, 0 to 7); 47 percent of pa-
tients completed 6 cycles. The main reason for not
results
* This characteristic was used as a stratification factor at the time of random-
ization.
A performance status of 0 denotes asymptomatic, 1 symptomatic and fully
ambulatory, and 2 symptomatic and in bed less than 50 percent of the day.
The maximum score on the Mini–Mental State Examination (MMSE) is 30,
and scores above 26 are considered to indicate normal mental status.
§ Anaplastic astrocytoma included oligoastrocytoma.
Table 1. Demographic Characteristics of the Patients at Baseline.
Characteristic
Radiotherapy
(N=286)
Radiotherapy
plus Temozolo-
mide (N=287)
Age — yr
Median 57 56
Range 23–71 19–70
Age — no. (%)*
<50 yr 81 (28) 90 (31)
50 yr 205 (72) 197 (69)
Sex — no. (%)
Male 175 (61) 185 (64)
Female 111 (39) 102 (36)
WHO performance status — no. (%)*†
0 110 (38) 113 (39)
1 141 (49) 136 (47)
2 35 (12) 38 (13)
Extent of surgery — no. (%)*
Biopsy 45 (16) 48 (17)
Debulking 241 (84) 239 (83)
Complete resection 113 (40) 113 (39)
Partial resection 128 (45) 126 (44)
Time from diagnosis to radiotherapy — wk
Median 5 5
Range 2.0–12.9 1.7–10.7
Baseline MMSE score — no. (%)‡
30 91 (32) 100 (35)
27–29 97 (34) 96 (33)
≤26 86 (30) 81 (28)
Data missing 12 (4) 10 (3)
Corticosteroid therapy — no. (%)
Yes 215 (75) 193 (67)
No 70 (24) 94 (33)
Data missing 1 (<1) 0
Slides available for pathological review
— no. (%)
246 (86) 239 (83)
Findings on pathological review — no. (%)
Glioblastoma 229 (93) 221 (92)
Anaplastic astrocytoma§ 9 (4) 7 (3)
Inconclusive material 3 (1) 3 (1)
Other 5 (2) 8 (3)
The New England Journal of Medicine
Downloaded from nejm.org at CTR HOSPITAL UNIVERSITAIRE VAUDOIS on October 12, 2017. For personal use only. No other uses without permission.
Copyright © 2005 Massachusetts Medical Society. All rights reserved.

Citations
More filters
Journal ArticleDOI
TL;DR: Patients with glioblastoma containing a methylated MGMT promoter benefited from temozolomide, whereas those who did not have a methylation of theMGMT promoter did notHave such a benefit and were assigned to only radiotherapy.
Abstract: background Epigenetic silencing of the MGMT (O 6 -methylguanine–DNA methyltransferase) DNArepair gene by promoter methylation compromises DNA repair and has been associated with longer survival in patients with glioblastoma who receive alkylating agents. methods We tested the relationship between MGMT silencing in the tumor and the survival of patients who were enrolled in a randomized trial comparing radiotherapy alone with radiotherapy combined with concomitant and adjuvant treatment with temozolomide. The methylation status of the MGMT promoter was determined by methylation-specific polymerase-chain-reaction analysis. results The MGMT promoter was methylated in 45 percent of 206 assessable cases. Irrespective of treatment, MGMT promoter methylation was an independent favorable prognostic factor (P<0.001 by the log-rank test; hazard ratio, 0.45; 95 percent confidence interval, 0.32 to 0.61). Among patients whose tumor contained a methylated MGMT promoter, a survival benefit was observed in patients treated with temozolomide and radiotherapy; their median survival was 21.7 months (95 percent confidence interval, 17.4 to 30.4), as compared with 15.3 months (95 percent confidence interval, 13.0 to 20.9) among those who were assigned to only radiotherapy (P=0.007 by the log-rank test). In the absence of methylation of the MGMT promoter, there was a smaller and statistically insignificant difference in survival between the treatment groups. conclusions Patients with glioblastoma containing a methylated MGMT promoter benefited from temozolomide, whereas those who did not have a methylated MGMT promoter did not have such a benefit.

6,018 citations

Journal ArticleDOI
TL;DR: Mutations of NADP(+)-dependent isocitrate dehydrogenases encoded by IDH1 and IDH2 occur in a majority of several types of malignant gliomas.
Abstract: Background A recent genomewide mutational analysis of glioblastomas (World Health Organization [WHO] grade IV glioma) revealed somatic mutations of the isocitrate dehydrogenase 1 gene (IDH1) in a fraction of such tumors, most frequently in tumors that were known to have evolved from lower-grade gliomas (secondary glioblastomas). Methods We determined the sequence of the IDH1 gene and the related IDH2 gene in 445 central nervous system (CNS) tumors and 494 non-CNS tumors. The enzymatic activity of the proteins that were produced from normal and mutant IDH1 and IDH2 genes was determined in cultured glioma cells that were transfected with these genes. Results We identified mutations that affected amino acid 132 of IDH1 in more than 70% of WHO grade II and III astrocytomas and oligodendrogliomas and in glioblastomas that developed from these lower-grade lesions. Tumors without mutations in IDH1 often had mutations affecting the analogous amino acid (R172) of the IDH2 gene. Tumors with IDH1 or IDH2 mutations h...

4,853 citations

Journal ArticleDOI
TL;DR: The authors found that approximately 5% of patients with malignant gliomas have a family history of glioma and most of these familial cases are associated with rare genetic syndromes, such as neurofibromatosis types 1 and 2, the Li−Fraumeni syndrome (germ-line p53 mutations associated with an increased risk of several cancers), and Turcot's syndrome (intestinal polyposis and brain tumors).
Abstract: Approximately 5% of patients with malignant gliomas have a family history of gliomas. Some of these familial cases are associated with rare genetic syndromes, such as neurofibromatosis types 1 and 2, the Li−Fraumeni syndrome (germ-line p53 mutations associated with an increased risk of several cancers), and Turcot’s syndrome (intestinal polyposis and brain tumors). 10 However, most familial cases have

3,823 citations

Journal ArticleDOI
TL;DR: This comprehensive review highlights the physicochemical properties of cisplatin and related platinum-based drugs, and discusses its uses (either alone or in combination with other drugs) for the treatment of various human cancers.

3,467 citations

Journal ArticleDOI
TL;DR: The recognition that contrast enhancement is nonspecific and may not always be a true surrogate of tumor response and the need to account for the nonenhancing component of the tumor mandate that new criteria be developed and validated to permit accurate assessment of the efficacy of novel therapies.
Abstract: Currently, the most widely used criteria for assessing response to therapy in high-grade gliomas are based on two-dimensional tumor measurements on computed tomography (CT) or magnetic resonance imaging (MRI), in conjunction with clinical assessment and corticosteroid dose (the Macdonald Criteria). It is increasingly apparent that there are significant limitations to these criteria, which only address the contrast-enhancing component of the tumor. For example, chemoradiotherapy for newly diagnosed glioblastomas results in transient increase in tumor enhancement (pseudoprogression) in 20% to 30% of patients, which is difficult to differentiate from true tumor progression. Antiangiogenic agents produce high radiographic response rates, as defined by a rapid decrease in contrast enhancement on CT/MRI that occurs within days of initiation of treatment and that is partly a result of reduced vascular permeability to contrast agents rather than a true antitumor effect. In addition, a subset of patients treated with antiangiogenic agents develop tumor recurrence characterized by an increase in the nonenhancing component depicted on T2-weighted/fluid-attenuated inversion recovery sequences. The recognition that contrast enhancement is nonspecific and may not always be a true surrogate of tumor response and the need to account for the nonenhancing component of the tumor mandate that new criteria be developed and validated to permit accurate assessment of the efficacy of novel therapies. The Response Assessment in Neuro-Oncology Working Group is an international effort to develop new standardized response criteria for clinical trials in brain tumors. In this proposal, we present the recommendations for updated response criteria for high-grade gliomas.

3,077 citations


Cites methods from "Radiotherapy plus Concomitant and A..."

  • ...Pseudoprogression and Radiation Effects Standard therapy for glioblastoma involves maximal safe tumor resection followed by radiotherapy with concurrent and adjuvant temozolomide.(24,25) Twenty to 30% of patients undergoing their first postradiation MRI show increased contrast enhancement that eventually subsides without any change in therapy (Fig 2)....

    [...]

References
More filters
Journal ArticleDOI
TL;DR: This work suggests "new" response criteria for phase II studies of supratentorial malignant glioma and favor rigorous criteria similar to those in medical oncology, with important modifications, to minimize misinterpretations of response.
Abstract: We suggest "new" response criteria for phase II studies of supratentorial malignant glioma and favor rigorous criteria similar to those in medical oncology, with important modifications. Four response categories are proposed: complete response (CR), partial response (PR), stable disease (SD), and progressive disease (PD). Response in this scheme is based on major changes in tumor size on the enhanced computed tomographic (CT) or magnetic resonance imaging (MRI) scan. Scan changes are interpreted in light of steroid use and neurologic findings. We advocate careful patient selection, emphasize pitfalls in the assessment of response, and suggest guidelines to minimize misinterpretations of response.

2,307 citations

Journal ArticleDOI
TL;DR: Methylation of the MGMT promoter in gliomas is a useful predictor of the responsiveness of the tumors to alkylating agents and an independent and stronger prognostic factor than age, stage, tumor grade, or performance status.
Abstract: Background The DNA-repair enzyme O 6-methylguanine-DNA methyltransferase (MGMT) inhibits the killing of tumor cells by alkylating agents. MGMT activity is controlled by a promoter; methylation of the promoter silences the gene in cancer, and the cells no longer produce MGMT. We examined gliomas to determine whether methylation of the MGMT promoter is related to the responsiveness of the tumor to alkylating agents. Methods We analyzed the MGMT promoter in tumor DNA by a methylation-specific polymerase-chain-reaction assay. The gliomas were obtained from patients who had been treated with carmustine (1,3-bis(2-chloroethyl)-1-nitrosourea, or BCNU). The molecular data were correlated with the clinical outcome. Results The MGMT promoter was methylated in gliomas from 19 of 47 patients (40 percent). This finding was associated with regression of the tumor and prolonged overall and disease-free survival. It was an independent and stronger prognostic factor than age, stage, tumor grade, or performance status. Con...

2,109 citations

Journal ArticleDOI
TL;DR: A new general procedure for treatment assignment is described which concentrates on minimizing imbalance in the distributions of treatment numbers within the levels of each individual prognostic factor.
Abstract: In controlled clinical trials there are usually several prognostic factors known or thought to influence the patient's ability to respond to treatment. Therefore, the method of sequential treatment assignment needs to be designed so that treatment balance is simultaneously achieved across all such patients factor. Traditional methods of restricted randomization such as "permuted blocks within strata" prove inadequate once the number of strata, or combinations of factor levels, approaches the sample size. A new general procedure for treatment assignment is described which concentrates on minimizing imbalance in the distributions of treatment numbers within the levels of each individual prognostic factor. The improved treatment balance obtained by this approach is explored using simulation for a simple model of a clinical trial. Further discussion centers on the selection, predictability and practicability of such a procedure.

2,078 citations

Journal ArticleDOI
TL;DR: An analysis of prognostic factors indicates that the initial performance status, age, the use of only a surgical biopsy, parietal location, the presence of seizures, or the involvement of cranial nerves II, III, IV, and VI are all of significance.
Abstract: A controlled, prospective, randomized study evaluated the use of 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) and/or radiotherapy in the treatment of patients who were operated on and had histological confirmation of anaplastic glioma. A total of 303 patients were randomized into this study, of whom 222 (73%) were within the Valid Study Group (VSG), having met the protocol criteria of neuropathology, corticosteroid control, and therapeutic approach. Patients were divided into four random groups, and received BCNU (80 mg/sq m/day on 3 successive days every 6 to 8 weeks), and/or radiotherapy (5000 to 6000 rads to the whole brain through bilateral opposing ports), or best conventional care but no chemotherapy or radiotherapy. Analysis was performed on all patients who received any amount of therapy (VSG) and on the Adequately Treated Group (ATG), who had received 5000 or more rads radiotherapy, two or more courses of chemotherapy, and had a minimum survival of 8 or more weeks (the interval that would have been required to have received either the radiotherapy or chemotherapy). Median survival of patients in the VSG was, best conventional care: 14 weeks (ATG: 17.0 weeks); BCNU: 18.5 weeks (ATG: 25.0 weeks); radiotherapy: 35 weeks (ATG: 37.5 weeks); and BCNU plus radiotherapy: 34.5 weeks (ATG: 40.5 weeks). All therapeutic modalities showed some statistical superiority compared to best conventional care. There was no significant difference between the four groups in relation to age distribution, sex, location of tumor, diagnosis, tumor characteristics, signs or symptoms, or the amount of corticosteroid used. An analysis of prognostic factors indicates that the initial performance status (Karnofsky rating), age, the use of only a surgical biopsy, parietal location, the presence of seizures, or the involvement of cranial nerves II, III, IV, and VI are all of significance. Toxicity included acceptable, reversible thrombocytopenia and leukopenia.

1,642 citations

Journal ArticleDOI
TL;DR: It is suggested that it is best to use radiotherapy in the post-surgical treatment of malignant glioma and to continue the search for an effective chemotherapeutic regimen to use in addition to radiotherapy.
Abstract: Within three weeks of definitive surgical intervention, 467 patients with histologically proved malignant glioma were randomized to receive one of four treatment regimens: semustine (MeCCNU), radiotherapy, carmustine (BCNU) plus radiotherapy, or semustine plus radiotherapy. We analyzed the data for the total randomized population and for the 358 patients in whom the initial protocol specifications were met (the valid study group). Observed toxicity included acceptable skin reactions secondary to radiotherapy and reversible leukopenia and thrombocytopenia due to chemotherapy. Radiotherapy used alone or in combination with a nitrosourea significantly improved survival in comparison with semustine alone. The group receiving carmustine plus radiotherapy had the best survival, but the difference in survival between the groups receiving carmustine plus radiotherapy and semustine plus radiotherapy was not statistically significant. The combination of carmustine plus radiotherapy produced a modest benefi...

1,582 citations

Related Papers (4)