Skip to main content
Erschienen in: Journal of Cancer Research and Clinical Oncology 8/2022

Open Access 26.08.2021 | Original Article – Clinical Oncology

Radiotherapy for glioblastoma patients with poor performance status

verfasst von: Christina Schröder, Dorothee Gramatzki, Erwin Vu, Matthias Guckenberger, Nicolaus Andratschke, Michael Weller, Caroline Hertler

Erschienen in: Journal of Cancer Research and Clinical Oncology | Ausgabe 8/2022

Abstract

Purpose

There is limited information on treatment recommendations for glioblastoma patients with poor performance status. Here, we aim to evaluate the association of radiotherapy on survival in glioblastoma patients presenting with poor postoperative performance status in first-line setting.

Methods

We retrospectively analyzed data of 93 glioblastoma patients presenting with poor postoperative performance status (ECOG 2–4) at the University Hospital Zurich, Switzerland, in the years 2005–2019. A total of 43 patients received radiotherapy with or without systemic therapy in the first-line setting, whereas 50 patients received no additive local or systemic treatment after initial biopsy or resection. Overall survival was calculated from primary diagnosis and from the end of radiotherapy. In addition, factors influencing survival were analyzed.

Results

Median overall survival from primary diagnosis was 6.2 months in the radiotherapy group (95% CI 6.2–14.8 weeks, range 2–149 weeks) and 2.3 months in the group without additive treatment (95% CI 1.3–7.4 weeks, range 0–28 weeks) (p < 0.001). This survival benefit was confirmed by landmark analyses. Factors associated with overall survival were extent of resection and administration of radiotherapy with or without systemic treatment. Median survival from end of radiotherapy was 3 months (95% CI 4.3–21.7 weeks, range 0–72 weeks), with 25.6% (n = 11) early termination of treatment and 83.7% (n = 36) requiring radiotherapy as in-patients. Performance status improved in 27.9% (n = 12) of patients after radiotherapy.

Conclusion

In this retrospective single-institution analysis, radiotherapy improved overall survival in patients with poor performance status, especially in patients who were amendable to neurosurgical resection.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s00432-021-03770-9.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Eighteen to 37% of glioblastoma patients present in a poor pre- or postoperative performance status at the time of diagnosis (Chang et al. 2003; Lutterbach et al. 2003; Bauchet et al. 2010; Gulati et al. 2011; Scott et al. 2012; Stark et al. 2012; Badaoui et al. 2014; McNamara et al. 2014; Zouaoui et al. 2014; Gramatzki et al. 2017). Performance status, age, mental status and extent of surgical resection have been confirmed as important prognostic factors in these patients, and low performance status at diagnosis indicates a particularly poor prognosis (Curran et al. 1993; Li et al. 2011; Scott et al. 2012).
The standard treatment of glioblastoma consists of surgery followed by radiotherapy in 30 fractions and concurrent and maintenance systemic therapy with temozolomide (TMZ) (Stupp et al. 2005; Wen et al. 2020; Weller et al. 2021). However, patients in poor performance status or elderly patients (> 70 years), are usually treated with adapted treatment regimens such as short course radiotherapy with or without chemotherapy, or alkylating chemotherapy only in O6-methylguanine-DNA methyltransferase (MGMT) promotor methylated patients (Roa et al. 2004; Malmstrom et al. 2012; Wick et al. 2012; Perry et al. 2017). Likewise, almost all clinical trials with experimental drugs preclude the enrolment of patients with poor performance status.
For elderly patients in good performance status, a survival benefit from radiotherapy after surgical resection of different extent has been confirmed (Keime-Guibert et al. 2007). However, data regarding the treatment of patients in poor performance status is scarce, especially considering that those patients are usually excluded from clinical trials, which minimizes the availability of data on potential benefit or harm for this selected patient group. A trial of 70 patients with a median Karnofsky performance status (KPS) of 60% compared the benefit of temozolomide therapy only to a historical control of patients treated with best supportive care. Temozolomide alone was characterized by acceptable tolerability especially in patients with a methylated MGMT-promotor and was associated with an improvement of the functional status. However, radiotherapy was not part of this study (Gallego Perez-Larraya et al. 2011).
To further evaluate a potential benefit of radiotherapy in patients with low performance status at diagnosis, we retrospectively analyzed the data of 43 patients with poor performance status receiving additive radiotherapy, either alone or in combination with chemotherapy, and of a control group of 50 patients receiving a diagnostic biopsy, partial or gross total resection without further tumor-targeted therapy.

Material and methods

Data source

We conducted a retrospective single center cohort study of glioblastoma patients > 18 years at diagnosis, treated at the University Hospital Zurich between April 2005 and June 2019, with poor postoperative performance status (ECOG PS 2–4) after biopsy or neurosurgical resection in the first-line setting. Electronic patient files and pathology reports were the main source of data acquisition. The study was conducted in accordance with the Declaration of Helsinki and applicable regulatory requirements and has been approved by the local ethics committee (KEK-ZH-Nr. 2009-0135/1; KEK-ZH-Nr. 2015-0437).

Disease and treatment characteristics

All tumors were classified according to the World Health Organization (WHO) 2007 criteria in the local pathology department, for patients diagnosed before 2016, and according to the WHO 2016 criteria for patients diagnosed after that time. In a second step all tumors classified by the 2007 criteria were re-classified by IDH mutation status in accordance with the WHO 2016 classification when sufficient tissue was available. IDH1/2 status was obtained by immunohistochemistry or sequencing analysis; MGMT promotor methylation status was determined by methylation-specific PCR. Extent of resection was assessed based on early postoperative imaging reports in all patients. For treatment planning, planning CT scans as well as MRI were used. The gross tumor volume (GTV) was contoured as the primary tumor (if biopsy only) or the resection cavity including contrast-enhanced regions. A safety margin of 1.5 cm was added to derive the clinical target volume (CTV). An additional planning target volume (PTV) margin was added to compensate for setup errors. For grading of toxicity, CTCAE V. 5.0 was used.

Statistical analysis

Overall survival was calculated according to the Kaplan–Meier method. Overall survival from the time of surgery (biopsy or resection) to death or from end of radiotherapy to death were calculated. Additional factors were analyzed using the log-rank test. Univariate and multivariate analyses were done using Cox regression. The multivariate model was applied to all patients who had complete information on all tested co-variables. For statistical analysis of patient characteristics and performance status, the Chi-squared test and the Fisher’s exact test were used. For statistical analysis, SPSS Version 25 was used (SPSS IBM Corp., Armonk, NY, USA).

Results

Patient characteristics

We evaluated the data of 93 patients with glioblastoma and poor postoperative performance status (ECOG 2–4) that were treated at our institution between April 2005 and June 2019. Median age of patients was 69 years in the radiotherapy group and 73 in the control group (p = 0.236). Median postoperative ECOG was 2 for both the radiotherapy group and the control group. A total of 43 patients received additive radiotherapy, 18 of which after partial or gross total resection (GTR) and the remaining after tumor biopsy only. Most patients in the radiotherapy group were treated with 15 × 2.67 Gy (n = 35; 81.4%), 9.3% (n = 4) were treated with 10 × 3 Gy and 9.3% (n = 4) were treated with other fractionations. Additional concurrent or sequential systemic therapy was given in 19 patients, most frequently temozolomide during and/or after radiotherapy (n = 14; 32.6%). The patients in the control group received no tumor-specific treatment after surgical resection or biopsy (best supportive care). Further patient and treatment characteristics are summarized in Table 1 and in the supplementary file.
Table 1
Patient characteristics
 
Radiotherapy group
Control group
p value
Total
 
N
%
N
%
 
N
Age (years)
    
 Median
69
73
0.236
 
 Range
56–85
29–90
  
Sex
      
 Female
19
44.2
22
44.0
0.986
41
 Male
24
55.8
28
56.0
 
52
ECOG
      
 2
29
31.2
28
30.1
0.626
57
 3
13
14.0
20
21.5
 
33
 4
1
1.1
2
2.2
 
3
IDH
      
 Wildtype
38
88.4
45
90.0
0.129
83
 Mutated
2
4.7
0
0.0
 
2
 Unknown
3
7.0
5
10.0
8
MGMT status
      
 Methylated
7
16.3
9
18.0
0.257
16
 Unmethylated
23
53.5
15
30.0
 
38
 Unknown
13
30.2
26
52.0
39
Surgerya
      
 Biopsy only
25
58.1
26
52.0
0.011
51
 Partial resection
7
16.3
20
40.0
 
27
 Gross total resection
11
25.6
4
8.0
 
15
Tumor localisation _1
      
 Deep structures
9
20.9
13
26
0.197
22
 Frontal lobe
12
27.9
12
24
 
24
 Temporal lobe
11
25.6
7
14
 
18
 Parietal lobe
8
18.6
5
10
 
13
 Occipital lobe
3
7.0
10
20
 
13
 Infratentorial
0
0
2
4
 
2
 Multifocal
0
0
1
2
 
1
Tumor localisation _2
      
 Bihemispheric
3
7.0
10
20
0.117
13
 Left hemisphere
17
39.5
13
26
 
30
 Right hemisphere
23
53.5
25
50
 
48
 Infratentorial
0
0
2
4
 
2
Planned radiation therapy
      
 15 × 2.67 Gy
35
81.4
n.a
n.a
 
35
 10 × 3 Gy
4
9.3
n.a
n.a
 
4
 Other
4
9.3
n.a
n.a
 
4
Systemic treatment (part of first-line treatment)
      
 None
24
55.8
n.a
n.a
 
24
 TMZ before RT
3
7.0
n.a
n.a
 
3
 TMZ during and/or after RT
14
32.6
n.a
n.a
 
14
 CCNU after RT
2
4.7
n.a
n.a
 
2
 Bevacizumab after RT
7
16.3
n.a
n.a
 
7
aSign difference RT group and control group (p < 0.05, Chi-Squared test)

Adherence and practical aspects of radiation treatment

Compliance to radiotherapy was high with 32 patients (74.4%) receiving the planned dose of radiotherapy. In 11 patients (25.6%) treatment was terminated early because of a decline in performance status, either as the result of disease progression or due to pre-existing morbidity. A total of 36 patients (83.7%) received radiation treatment as in-patients for at least a part of the treatment. In 14 patients (32.6%) a specialized palliative care team was involved at some point during radiotherapy treatment. Radiotherapy was well tolerated with no ≥ G3 toxicities.

Overall survival

At the time of analysis, median follow-up of living patients was 2 weeks (95% CI 0.0–6.8) and six patients were still alive. The median survival after primary diagnosis was 27 weeks (95% CI 6.2–14.8, range 2–149) for patients treated with radiotherapy and 10 weeks (95% CI 1.3–7.4, range 0–28) for the patients in the control group (Fig. 1A) (p < 0.001). Patients that received a biopsy only had a median survival from diagnosis of 13 weeks (95% CI 2.6–23.4, range 2–149) when treated with radiotherapy (n = 25) and 7 weeks (95% CI 3.4–10.6, range 0–19) with best supportive care (n = 26) (Fig. 2A) (p < 0.001). Older patients ≥ 70 years had a median overall survival of 28 weeks (95% CI 14.1–41.9, range 3–66) when treated with radiotherapy, and 10 weeks in the best supportive care group (95% CI 6.0–14.0, range 0–28) (Fig. 2C) (p < 0.001). When separated by ECOG, patients with poor ECOG (3–4) showed a median overall survival of 22 weeks (95% CI 5.5–38.5, range 2–61) when treated with radiotherapy versus 7 weeks with best supportive care (95% CI 4.4–9.6, range 0–25 weeks) (Fig. 2E) (p < 0.001) (Table 2). In addition, a 4- and 8-week landmark analysis was performed to estimate OS without immortal time bias (Fig. 3A, B) showing similar results compared to analysis at diagnosis. Extent of surgical resection (p = 0.037) and postoperative treatment [either radiotherapy only (p = 0.002) or in combination with systemic therapy (p < 0.001)] showed statistical significance in univariate analysis and remained significant in multivariate analysis (Tables 3, 4).
Table 2
Survival data (log rank analysis)
 
From diagnosis
From RT
Survival
N
Median OS (weeks)
95% CI
p value
N
Median OS (weeks)
95% CI
p value
All patients
93
12
9.9–14.1
 
43
13
4.3–21.7
 
RT
        
 Yes
43
27
6.2–14.8
< 0.001
 
n.a
n.a
n.a
 No
50
10
1.3–7.4
 
Age
        
 < 70 with RT
22
23
5.4–40.6
< 0.001
22
12
3.1–20.9
0.308
 < 70 no RT
18
10
3.7–16.3
 
 ≤ 70 with RT
21
28
14.1–41.9
 
21
14
0.0–32.7
 ≥ 70 no RT
32
10
6.0–14.0
  
ECOG
        
 2
with RT
29
28
10.8–45.2
 
29
16
0–35.7
0.438
 2
no RT
29
11
9.3–12.7
 
 3–4
with RT
14
22
5.5–8.5
 
14
9
0–26.7
 3–4
no RT
21
7
4.4–9.6
 
Surgery
        
 Biopsy with RT
25
13
2.6–23.4
< 0.001
25
8
1.5–14.5
0.181
 Biopsy no RT
26
7
3.4–10.6
 
 PR/GTR with RT
18
36
24.2–47.8
 
18
26
1.4–50.6
 PR/GTR no RT
24
10
7.3–12.7
 
Table 3
Univariate analysis with regards to death (Cox regression analysis)
 
N
(events)
HR (95% CI)
p value
Age
   
 < 70 years
40 (37)
0.74 (0.48–1.16)
0.190
 ≥ 70 years
53 (50)
ref
 
Gender
   
 Male
52 (48)
ref
 
 Female
41 (39)
1.13 (0.74–1.74)
0.565
ECOG
   
 2
58 (54)
0.87 (0.56–1.35)
0.527
 3/4
35 (33)
ref
 
Extent of resection
   
 Biopsy
51 (48)
1.58 (1.03–2.43)
0.037
 Resection
(partial/gross total resection)
42 (39)
ref
 
MGMT promoter methylation status
   
 Unmethylated
38 (37)
ref
 
 Methylated
16 (13)
0.91 (0.49–1.70)
0.766
Treatment
   
 No tumor-specific treatment
50 (46)
ref
 
 Radiotherapy
24 (24)
0.42 (0.24–0.74)
0.002
 Radiotherapy plus systemic treatment
19 (17)
0.13 (0.06–0.26)
< 0.001
Table 4
Multivariate analysis with regards to death (Cox regression analysis)
 
N (events)
HR (95% CI)
p value
All patients
54 (51)
  
Age
   
 < 70 years
26
1.40 (0.69–2.81)
0.352
 ≥ 70 years
28
Ref
 
Gender
   
 Male
25
Ref
 
 Female
29
2.87 (1.46–5.67)
0.002
ECOG
   
 2
33
0.94 (0.52–1.70)
0.839
 3/4
21
Ref
 
Extent of resection
   
 Biopsy
25
3.93 (1.94–7.93)
< 0.001
 Resection
(partial/gross total resection)
29
Ref
 
MGMT promoter methylation status
   
 Unmethylated
38
Ref
 
 Methylated
16
0.84 (0.43–1.62)
0.600
Treatment
   
 No tumor-specific treatment
24
Ref
 
 Radiotherapy
14
0.26 (0.12–0.59)
0.001
 Radiotherapy plus systemic treatment
16
0.09 (0.03–0.25)
< 0.001

Post-radiation survival

The median survival after radiotherapy (post-radiation survival) was 13 weeks (3 months, 95% CI 4.3–21.7 weeks, range 0–72 weeks). Twelve patients (27.9%) treated with radiotherapy died within 4 weeks after radiotherapy, 10 of which had a biopsy only (83.3%). Post-radiation survival did not differ significantly when stratified for extent of resection, age group or ECOG at diagnosis (Fig. 2B, D, F) (Table 2).

Performance status

After RT, the performance status was improved at any follow-up appointment during the first 3 months in 12 patients (27.9%). The median increase in performance status was 1 with a range of 1–2. Seven patients (16.3%) of patients regained a performance status \(\le\) 1. Factors associated with an improvement in performance status within the radiation therapy group were extent of resection (biopsy only vs. partial resection vs. gross total resection, p = 0.039), combined use of systemic therapy (p = 0.001) and delivered radiotherapy dose (< 40 vs ≥ 40 Gy, p = 0.007). It has to be noted that in 75% of patients with a radiotherapy dose < 40 Gy, radiotherapy had to be terminated early. The majority of patients with an improvement in performance status had a gross total resection, received systemic therapy and all had received a radiation dose of ≥ 40 Gy. Age, MGMT status and the initial performance status were not associated with improvement in performance status after radiotherapy.

Discussion

This retrospective single-institution analysis addressed the value of radiotherapy compared to best supportive care in glioblastoma patients with poor postoperative performance status in the first-line setting.
Performance status is a well-documented prognostic factor for overall survival in glioblastoma patients, the most important factors being age (< 50 vs. ≥ 50 years) (Curran et al. 1993; Li et al. 2011) or type of surgery (Gross total resection/partial resection vs. biopsy) (Scott et al. 2012). However, with regard to the optimal treatment recommendation of patients in poor performance status, reliable data is sparse (Curran et al. 1993; Lutterbach et al. 2003; Li et al. 2011; Scott et al. 2012; Stark et al. 2012; McNamara et al. 2014; Gately et al. 2016; Glynn, Rangaswamy et al. 2019). The median overall survival of patients with poor performance status is short with 2.3–6.6 months from the time of diagnosis, although the definition of poor performance status is inconsistent. In a cohort published by Scott et al., patients with biopsy only and a KPS < 70% had a median OS of 2.3 months. Patients with more extensive surgeries had a better overall survival, independent of performance status (Scott et al. 2012). Marina et al. analyzed data of patients with a KPS < 50% in a cohort with about half of patients biopsied only, and 64% of patients receiving radiotherapy, and reported a median overall survival from the date of the most excessive surgery of 2.3 month, too (Marina et al. 2011).
In our cohort, the median survival from diagnosis was 27 weeks in all patients treated with radiotherapy, and 13 weeks in the biopsy only followed by radiotherapy group. Survival from the end of radiotherapy was median 13 weeks for all patients treated with radiotherapy and 8 weeks for biopsy followed by radiotherapy. This indicates patients treated with biopsy only are at increased risk of early death irrespective of post-biopsy radiotherapy. Admittedly, our radiotherapy group comprised more patients with gross total resection compared to the best supportive care group. Yet, without post-surgery radiotherapy, survival does not increase with gross total resection compared to biopsy only, underlining the role of radiotherapy (Fig. 2A, B), and the median overall survival from diagnosis was significantly better in the radiotherapy group than in the control group (10 weeks) irrespective of performance status or extent of surgical resection. Landmark analyses at 4 and 8 weeks confirmed the survival benefit in the radiotherapy group (Fig. 3).
In patients with poor performance status, survival is only one aspect to consider. Another important question is whether impaired patients may benefit from radiation therapy in terms of performance status improvement. Very few data are available on this topic, and especially prospective data is rare. Retrospective data and data from a phase II trial suggest that KPS might improve by 10–30% in up to 70% patients with radiotherapy, but overall patient numbers are small (Gallego Perez-Larraya et al. 2011; Marina et al. 2011; Reyngold et al. 2012). In this analysis, 30% of patients had an improvement in performance status of 1–2 after radiotherapy, consistent with previous data. One fifth of our patient population regained an ECOG ≥ 1, allowing for self-care, and therefore representing a factor of autonomy for the patient.
Another factor to consider is treatment-induced toxicity. Radiation-induced toxicity for glioblastoma patients is usually low, e.g., 8% ≥ G3 toxicity in the radiation arm (Malmstrom et al. 2012; Wick et al. 2012) in a group of elderly, therefore more vulnerable, patients. This is confirmed in our study with no case of grade 3 or higher radiation-induced toxicity. Treatment with TMZ, either as monotherapy or with radiation, causes mostly hematologic ≤ G3 toxicity in up to 25% of patients (Stupp et al. 2005; Gallego Perez-Larraya et al. 2011; Malmstrom et al. 2012).
Short course radiation treatment is the standard of care in elderly and poor-performance status patients with unmethylated MGMT-promotor (Guedes de Castro et al. 2017; Weller et al. 2017), which limits the total treatment time to 3 weeks and 15 radiotherapy sessions. All patients in our study were treated with hypo-fractionated radiotherapy; however, it was interesting that the benefit of radiotherapy was restricted to patients treated with 40.05 Gy in 15 fractions, whereas no benefit was observed for lower-dose radiotherapy. Best-supportive care or shorter fractionated radiotherapy should, therefore, be considered for patients not eligible for a full-course radiotherapy of 15 fractions over three weeks.
Despite absolute overall survival is short in glioblastoma patients with poor performance status, as reported above, radiotherapy was well-tolerated as delivered over maximum three weeks and appears favorable considering that median overall survival is prolonged by a factor of almost three. However, careful and balanced patient information about the absolute and relative benefits of surgery and postoperative radiotherapy and chemotherapy is mandatory.
Limitations of this study include the retrospective nature of this small population with heterogeneous treatment, including combination of chemotherapeutics with radiotherapy as part of the first-line treatment. However, there was no significant imbalance with regard to MGMT promotor methylation as predictive marker in the radiotherapy plus systemic treatment group, which could have indicated a better response to additional chemotherapy. Also, lack of patient-reported outcomes in this retrospective analysis precludes a statement on subjective benefits and quality of life in this specific patient population, therefore omitting a relevant patient-centered outcome surrogate. Still, the inclusion of relevant numbers of patients biopsied only, in both radiotherapy and control group (25 vs. 26 patients, respectively), provides more information on this patient group with poor predictors of outcome that may benefit with regard to the performance status improvement.
In conclusion, this retrospective single-institution analysis observed a significant and clinically relevant overall survival benefit of radiotherapy in glioblastoma patients with poor performance status. However, considering the short absolute overall survival, especially in patients not eligible for neurosurgical resection, the potential benefit of prolonged survival must be carefully weighted against the large portion of extended lifespan being spent under treatment. Better prognostic and predictive factors for patient selection are urgently needed. Most importantly, quality-of-life and patient-reported outcome need to be considered in addition to sole overall survival and should be incorporated not only into clinical studies, but also as routine practice, including patients in reduced performance status who are usually excluded from study populations (Coomans et al. 2019; Hertler et al. 2020).

Declarations

Conflict of interest

The authors declare no conflict of interest or financial interest related to this publication.

Ethical approval

The study was approved by the Ethics Committee and, wherever applicable, patients consented to retrospective data collection including use of encoded data for publication. (KEK-ZH-Nr. 2009-0135/1; KEK-ZH-Nr. 2015-0437).

Data availability

No data are available.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
Zurück zum Zitat Badaoui N, Meyronet D, Cartalat-Carel S, Guyotat J, Jouanneau E, d’Hombres A, Sunyach MP, Jouvet A, Louis-Tisserand G, Archinet A, Frappaz D, Bauchet L, Honnorat J, Ducray F (2014) Patterns of care and survival of glioblastoma patients: a comparative study between 2004 and 2008 in Lyon, France. Rev Neurol (paris) 170(3):222–227CrossRef Badaoui N, Meyronet D, Cartalat-Carel S, Guyotat J, Jouanneau E, d’Hombres A, Sunyach MP, Jouvet A, Louis-Tisserand G, Archinet A, Frappaz D, Bauchet L, Honnorat J, Ducray F (2014) Patterns of care and survival of glioblastoma patients: a comparative study between 2004 and 2008 in Lyon, France. Rev Neurol (paris) 170(3):222–227CrossRef
Zurück zum Zitat Bauchet L, Mathieu-Daude H, Fabbro-Peray P, Rigau V, Fabbro M, Chinot O, Pallusseau L, Carnin C, Laine K, Schlama A, Thiebaut A, Patru MC, Bauchet F, Lionnet M, Wager M, Faillot T, Taillandier L, Figarella-Branger D, Capelle L, Loiseau H, Frappaz D, Campello C, Kerr C, Duffau H, Reme-Saumon M, Tretarre B, Daures JP, Henin D, Labrousse F, Menei P, Honnorat J, N Societe Francaise de, Club de Neuro-Oncologie of the Societe Francaise de N, Societe Francaise de and F Association des Neuro-Oncologues d’Expression (2010) Oncological patterns of care and outcome for 952 patients with newly diagnosed glioblastoma in 2004. Neuro Oncol 12(7):725–735CrossRef Bauchet L, Mathieu-Daude H, Fabbro-Peray P, Rigau V, Fabbro M, Chinot O, Pallusseau L, Carnin C, Laine K, Schlama A, Thiebaut A, Patru MC, Bauchet F, Lionnet M, Wager M, Faillot T, Taillandier L, Figarella-Branger D, Capelle L, Loiseau H, Frappaz D, Campello C, Kerr C, Duffau H, Reme-Saumon M, Tretarre B, Daures JP, Henin D, Labrousse F, Menei P, Honnorat J, N Societe Francaise de, Club de Neuro-Oncologie of the Societe Francaise de N, Societe Francaise de and F Association des Neuro-Oncologues d’Expression (2010) Oncological patterns of care and outcome for 952 patients with newly diagnosed glioblastoma in 2004. Neuro Oncol 12(7):725–735CrossRef
Zurück zum Zitat Chang SM, Parney IF, McDermott M, Barker FG 2nd, Schmidt MH, Huang W, Laws ER Jr, Lillehei KO, Bernstein M, Brem H, Sloan AE, Berger M, Glioma Outcomes I (2003) Perioperative complications and neurological outcomes of first and second craniotomies among patients enrolled in the Glioma outcome project. J Neurosurg 98(6):1175–1181CrossRef Chang SM, Parney IF, McDermott M, Barker FG 2nd, Schmidt MH, Huang W, Laws ER Jr, Lillehei KO, Bernstein M, Brem H, Sloan AE, Berger M, Glioma Outcomes I (2003) Perioperative complications and neurological outcomes of first and second craniotomies among patients enrolled in the Glioma outcome project. J Neurosurg 98(6):1175–1181CrossRef
Zurück zum Zitat Coomans M, Dirven L, Baumert van den Bent Bottomley Brandes Chinot Coens Gorlia Herrlinger Keime-Guibert Malmstrom Martinelli Stupp Talacchi Weller Wick Reijneveld Taphoorn KANBGMAAAOCTUFAFRAMWJCMJB, E. Q. o. L. Group and E. B. T. G. the (2019) The added value of health-related quality of life as a prognostic indicator of overall survival and progression-free survival in glioma patients: a meta-analysis based on individual patient data from randomised controlled trials. Eur J Cancer 116:190–198CrossRef Coomans M, Dirven L, Baumert van den Bent Bottomley Brandes Chinot Coens Gorlia Herrlinger Keime-Guibert Malmstrom Martinelli Stupp Talacchi Weller Wick Reijneveld Taphoorn KANBGMAAAOCTUFAFRAMWJCMJB, E. Q. o. L. Group and E. B. T. G. the (2019) The added value of health-related quality of life as a prognostic indicator of overall survival and progression-free survival in glioma patients: a meta-analysis based on individual patient data from randomised controlled trials. Eur J Cancer 116:190–198CrossRef
Zurück zum Zitat Curran WJ Jr, Scott CB, Horton J, Nelson JS, Weinstein AS, Fischbach AJ, Chang CH, Rotman M, Asbell SO, Krisch RE et al (1993) Recursive partitioning analysis of prognostic factors in three Radiation Therapy Oncology Group malignant glioma trials. J Natl Cancer Inst 85(9):704–710CrossRef Curran WJ Jr, Scott CB, Horton J, Nelson JS, Weinstein AS, Fischbach AJ, Chang CH, Rotman M, Asbell SO, Krisch RE et al (1993) Recursive partitioning analysis of prognostic factors in three Radiation Therapy Oncology Group malignant glioma trials. J Natl Cancer Inst 85(9):704–710CrossRef
Zurück zum Zitat Gallego Perez-Larraya J, Ducray F, Chinot O, Catry-Thomas I, Taillandier L, Guillamo JS, Campello C, Monjour A, Cartalat-Carel S, Barrie M, Huchet A, Beauchesne P, Matta M, Mokhtari K, Tanguy ML, Honnorat J, Delattre JY (2011) Temozolomide in elderly patients with newly diagnosed glioblastoma and poor performance status: an ANOCEF phase II trial. J Clin Oncol 29(22):3050–3055CrossRef Gallego Perez-Larraya J, Ducray F, Chinot O, Catry-Thomas I, Taillandier L, Guillamo JS, Campello C, Monjour A, Cartalat-Carel S, Barrie M, Huchet A, Beauchesne P, Matta M, Mokhtari K, Tanguy ML, Honnorat J, Delattre JY (2011) Temozolomide in elderly patients with newly diagnosed glioblastoma and poor performance status: an ANOCEF phase II trial. J Clin Oncol 29(22):3050–3055CrossRef
Zurück zum Zitat Gately L, Collins A, Murphy M, Dowling A (2016) Age alone is not a predictor for survival in glioblastoma. J Neurooncol 129(3):479–485CrossRef Gately L, Collins A, Murphy M, Dowling A (2016) Age alone is not a predictor for survival in glioblastoma. J Neurooncol 129(3):479–485CrossRef
Zurück zum Zitat Glynn AM, Rangaswamy G, O’Shea J, Dunne M, Grogan R, MacNally S, Fitzpatrick D, Faul C (2019) Glioblastoma Multiforme in the over 70’s: to treat or not to treat with radiotherapy? Cancer Med 8:4669–4677CrossRef Glynn AM, Rangaswamy G, O’Shea J, Dunne M, Grogan R, MacNally S, Fitzpatrick D, Faul C (2019) Glioblastoma Multiforme in the over 70’s: to treat or not to treat with radiotherapy? Cancer Med 8:4669–4677CrossRef
Zurück zum Zitat Gramatzki D, Kickingereder P, Hentschel B, Felsberg J, Herrlinger U, Schackert G, Tonn JC, Westphal M, Sabel M, Schlegel U, Wick W, Pietsch T, Reifenberger G, Loeffler M, Bendszus M, Weller M (2017) Limited role for extended maintenance temozolomide for newly diagnosed glioblastoma. Neurology 88(15):1422–1430CrossRef Gramatzki D, Kickingereder P, Hentschel B, Felsberg J, Herrlinger U, Schackert G, Tonn JC, Westphal M, Sabel M, Schlegel U, Wick W, Pietsch T, Reifenberger G, Loeffler M, Bendszus M, Weller M (2017) Limited role for extended maintenance temozolomide for newly diagnosed glioblastoma. Neurology 88(15):1422–1430CrossRef
Zurück zum Zitat Guedes de Castro D, Matiello J, Roa W, Ghosh S, Kepka L, Kumar N, Sinaika V, Lomidze D, Hentati D, Rosenblatt E, Fidarova E (2017) Survival outcomes with short-course radiation therapy in elderly patients with glioblastoma: data from a randomized phase 3 trial. Int J Radiat Oncol Biol Phys 98(4):931–938CrossRef Guedes de Castro D, Matiello J, Roa W, Ghosh S, Kepka L, Kumar N, Sinaika V, Lomidze D, Hentati D, Rosenblatt E, Fidarova E (2017) Survival outcomes with short-course radiation therapy in elderly patients with glioblastoma: data from a randomized phase 3 trial. Int J Radiat Oncol Biol Phys 98(4):931–938CrossRef
Zurück zum Zitat Gulati S, Jakola AS, Nerland US, Weber C, Solheim O (2011) The risk of getting worse: surgically acquired deficits, perioperative complications, and functional outcomes after primary resection of glioblastoma. World Neurosurg 76(6):572–579CrossRef Gulati S, Jakola AS, Nerland US, Weber C, Solheim O (2011) The risk of getting worse: surgically acquired deficits, perioperative complications, and functional outcomes after primary resection of glioblastoma. World Neurosurg 76(6):572–579CrossRef
Zurück zum Zitat Hertler C, Seiler A, Gramatzki D, Schettle M, Blum D (2020) Sex-specific and gender-specific aspects in patient-reported outcomes. ESMO Open 5(Suppl 4):e000837CrossRef Hertler C, Seiler A, Gramatzki D, Schettle M, Blum D (2020) Sex-specific and gender-specific aspects in patient-reported outcomes. ESMO Open 5(Suppl 4):e000837CrossRef
Zurück zum Zitat Keime-Guibert F, Chinot O, Taillandier L, Cartalat-Carel S, Frenay M, Kantor G, Guillamo JS, Jadaud E, Colin P, Bondiau PY, Menei P, Loiseau H, Bernier V, Honnorat J, Barrie M, Mokhtari K, Mazeron JJ, Bissery A, Delattre JY, N-O Association of French-Speaking (2007) Radiotherapy for glioblastoma in the elderly. N Engl J Med 356(15):1527–1535CrossRef Keime-Guibert F, Chinot O, Taillandier L, Cartalat-Carel S, Frenay M, Kantor G, Guillamo JS, Jadaud E, Colin P, Bondiau PY, Menei P, Loiseau H, Bernier V, Honnorat J, Barrie M, Mokhtari K, Mazeron JJ, Bissery A, Delattre JY, N-O Association of French-Speaking (2007) Radiotherapy for glioblastoma in the elderly. N Engl J Med 356(15):1527–1535CrossRef
Zurück zum Zitat Li J, Wang M, Won M, Shaw EG, Coughlin C, Curran WJ Jr, Mehta MP (2011) Validation and simplification of the Radiation Therapy Oncology Group recursive partitioning analysis classification for glioblastoma. Int J Radiat Oncol Biol Phys 81(3):623–630CrossRef Li J, Wang M, Won M, Shaw EG, Coughlin C, Curran WJ Jr, Mehta MP (2011) Validation and simplification of the Radiation Therapy Oncology Group recursive partitioning analysis classification for glioblastoma. Int J Radiat Oncol Biol Phys 81(3):623–630CrossRef
Zurück zum Zitat Lutterbach J, Sauerbrei W, Guttenberger R (2003) Multivariate analysis of prognostic factors in patients with glioblastoma. Strahlenther Onkol 179(1):8–15CrossRef Lutterbach J, Sauerbrei W, Guttenberger R (2003) Multivariate analysis of prognostic factors in patients with glioblastoma. Strahlenther Onkol 179(1):8–15CrossRef
Zurück zum Zitat Malmstrom A, Gronberg BH, Marosi C, Stupp R, Frappaz D, Schultz H, Abacioglu U, Tavelin B, Lhermitte B, Hegi ME, Rosell J, Henriksson R, G. Nordic Clinical Brain Tumour Study (2012) 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 13(9):916–926CrossRef Malmstrom A, Gronberg BH, Marosi C, Stupp R, Frappaz D, Schultz H, Abacioglu U, Tavelin B, Lhermitte B, Hegi ME, Rosell J, Henriksson R, G. Nordic Clinical Brain Tumour Study (2012) 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 13(9):916–926CrossRef
Zurück zum Zitat Marina O, Suh JH, Reddy CA, Barnett GH, Vogelbaum MA, Peereboom DM, Stevens GH, Elinzano H, Chao ST (2011) Treatment outcomes for patients with glioblastoma multiforme and a low Karnofsky Performance Scale score on presentation to a tertiary care institution. Clinical article. J Neurosurg 115(2):220–229CrossRef Marina O, Suh JH, Reddy CA, Barnett GH, Vogelbaum MA, Peereboom DM, Stevens GH, Elinzano H, Chao ST (2011) Treatment outcomes for patients with glioblastoma multiforme and a low Karnofsky Performance Scale score on presentation to a tertiary care institution. Clinical article. J Neurosurg 115(2):220–229CrossRef
Zurück zum Zitat McNamara MG, Lwin Z, Jiang H, Chung C, Millar BA, Sahgal A, Laperriere N, Mason WP (2014) Conditional probability of survival and post-progression survival in patients with glioblastoma in the temozolomide treatment era. J Neurooncol 117(1):153–160CrossRef McNamara MG, Lwin Z, Jiang H, Chung C, Millar BA, Sahgal A, Laperriere N, Mason WP (2014) Conditional probability of survival and post-progression survival in patients with glioblastoma in the temozolomide treatment era. J Neurooncol 117(1):153–160CrossRef
Zurück zum Zitat Perry JR, Laperriere N, O’Callaghan CJ, Brandes AA, Menten J, Phillips C, Fay M, Nishikawa R, Cairncross JG, Roa W, Osoba D, Rossiter JP, Sahgal A, Hirte H, Laigle-Donadey F, Franceschi E, Chinot O, Golfinopoulos V, Fariselli L, Wick A, Feuvret L, Back M, Tills M, Winch C, Baumert BG, Wick W, Ding K, Mason WP, Trial I (2017) Short-course radiation plus temozolomide in elderly patients with glioblastoma. N Engl J Med 376(11):1027–1037CrossRef Perry JR, Laperriere N, O’Callaghan CJ, Brandes AA, Menten J, Phillips C, Fay M, Nishikawa R, Cairncross JG, Roa W, Osoba D, Rossiter JP, Sahgal A, Hirte H, Laigle-Donadey F, Franceschi E, Chinot O, Golfinopoulos V, Fariselli L, Wick A, Feuvret L, Back M, Tills M, Winch C, Baumert BG, Wick W, Ding K, Mason WP, Trial I (2017) Short-course radiation plus temozolomide in elderly patients with glioblastoma. N Engl J Med 376(11):1027–1037CrossRef
Zurück zum Zitat Reyngold M, Lassman AB, Chan TA, Yamada Y, Gutin PH, Beal K (2012) Abbreviated course of radiation therapy with concurrent temozolomide for high-grade glioma in patients of advanced age or poor functional status. J Neurooncol 110(3):369–374CrossRef Reyngold M, Lassman AB, Chan TA, Yamada Y, Gutin PH, Beal K (2012) Abbreviated course of radiation therapy with concurrent temozolomide for high-grade glioma in patients of advanced age or poor functional status. J Neurooncol 110(3):369–374CrossRef
Zurück zum Zitat Roa W, Brasher PM, Bauman G, Anthes M, Bruera E, Chan A, Fisher B, Fulton D, Gulavita S, Hao C, Husain S, Murtha A, Petruk K, Stewart D, Tai P, Urtasun R, Cairncross JG, Forsyth P (2004) Abbreviated course of radiation therapy in older patients with glioblastoma multiforme: a prospective randomized clinical trial. J Clin Oncol 22(9):1583–1588CrossRef Roa W, Brasher PM, Bauman G, Anthes M, Bruera E, Chan A, Fisher B, Fulton D, Gulavita S, Hao C, Husain S, Murtha A, Petruk K, Stewart D, Tai P, Urtasun R, Cairncross JG, Forsyth P (2004) Abbreviated course of radiation therapy in older patients with glioblastoma multiforme: a prospective randomized clinical trial. J Clin Oncol 22(9):1583–1588CrossRef
Zurück zum Zitat Scott JG, Bauchet L, Fraum TJ, Nayak L, Cooper AR, Chao ST, Suh JH, Vogelbaum MA, Peereboom DM, Zouaoui S, Mathieu-Daude H, Fabbro-Peray P, Rigau V, Taillandier L, Abrey LE, DeAngelis LM, Shih JH, Iwamoto FM (2012) Recursive partitioning analysis of prognostic factors for glioblastoma patients aged 70 years or older. Cancer 118(22):5595–5600CrossRef Scott JG, Bauchet L, Fraum TJ, Nayak L, Cooper AR, Chao ST, Suh JH, Vogelbaum MA, Peereboom DM, Zouaoui S, Mathieu-Daude H, Fabbro-Peray P, Rigau V, Taillandier L, Abrey LE, DeAngelis LM, Shih JH, Iwamoto FM (2012) Recursive partitioning analysis of prognostic factors for glioblastoma patients aged 70 years or older. Cancer 118(22):5595–5600CrossRef
Zurück zum Zitat Stark AM, van de Bergh J, Hedderich J, Mehdorn HM, Nabavi A (2012) Glioblastoma: clinical characteristics, prognostic factors and survival in 492 patients. Clin Neurol Neurosurg 114(7):840–845CrossRef Stark AM, van de Bergh J, Hedderich J, Mehdorn HM, Nabavi A (2012) Glioblastoma: clinical characteristics, prognostic factors and survival in 492 patients. Clin Neurol Neurosurg 114(7):840–845CrossRef
Zurück zum Zitat 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, R. European Organisation for, T. Treatment of Cancer Brain, G. Radiotherapy, G. National Cancer Institute of Canada Clinical Trials (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352(10):987–996CrossRef 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, R. European Organisation for, T. Treatment of Cancer Brain, G. Radiotherapy, G. National Cancer Institute of Canada Clinical Trials (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352(10):987–996CrossRef
Zurück zum Zitat Weller M, van den Bent M, Tonn JC, Stupp R, Preusser M, Cohen-Jonathan-Moyal E, Henriksson R, Le Rhun E, Balana C, Chinot O, Bendszus M, Reijneveld JC, Dhermain F, French P, Marosi C, Watts C, Oberg I, Pilkington G, Baumert BG, Taphoorn MJB, Hegi M, Westphal M, Reifenberger G, Soffietti R, Wick W (2017) European Association for Neuro-Oncology (EANO) guideline on the diagnosis and treatment of adult astrocytic and oligodendroglial gliomas. Lancet Oncol 18(6):e315–e329CrossRef Weller M, van den Bent M, Tonn JC, Stupp R, Preusser M, Cohen-Jonathan-Moyal E, Henriksson R, Le Rhun E, Balana C, Chinot O, Bendszus M, Reijneveld JC, Dhermain F, French P, Marosi C, Watts C, Oberg I, Pilkington G, Baumert BG, Taphoorn MJB, Hegi M, Westphal M, Reifenberger G, Soffietti R, Wick W (2017) European Association for Neuro-Oncology (EANO) guideline on the diagnosis and treatment of adult astrocytic and oligodendroglial gliomas. Lancet Oncol 18(6):e315–e329CrossRef
Zurück zum Zitat Weller M, van den Bent M, Preusser M, Le Rhun E, Tonn JC, Minniti G, Bendszus M, Balana C, Chinot O, Dirven L, French P, Hegi ME, Jakola AS, Platten M, Roth P, Ruda R, Short S, Smits M, Taphoorn MJB, von Deimling A, Westphal M, Soffietti R, Reifenberger G, Wick W (2021) EANO guidelines on the diagnosis and treatment of diffuse gliomas of adulthood. Nat Rev Clin Oncol 18(3):170–186CrossRef Weller M, van den Bent M, Preusser M, Le Rhun E, Tonn JC, Minniti G, Bendszus M, Balana C, Chinot O, Dirven L, French P, Hegi ME, Jakola AS, Platten M, Roth P, Ruda R, Short S, Smits M, Taphoorn MJB, von Deimling A, Westphal M, Soffietti R, Reifenberger G, Wick W (2021) EANO guidelines on the diagnosis and treatment of diffuse gliomas of adulthood. Nat Rev Clin Oncol 18(3):170–186CrossRef
Zurück zum Zitat Wen PY, Weller M, Lee EQ, Alexander BM, Barnholtz-Sloan JS, Barthel FP, Batchelor TT, Bindra RS, Chang SM, Chiocca EA, Cloughesy TF, DeGroot JF, Galanis E, Gilbert MR, Hegi ME, Horbinski C, Huang RY, Lassman AB, Le Rhun E, Lim M, Mehta MP, Mellinghoff IK, Minniti G, Nathanson D, Platten M, Preusser M, Roth P, Sanson M, Schiff D, Short SC, Taphoorn MJB, Tonn JC, Tsang J, Verhaak RGW, von Deimling A, Wick W, Zadeh G, Reardon DA, Aldape KD, van den Bent MJ (2020) Glioblastoma in adults: a Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions. Neuro Oncol 22(8):1073–1113CrossRef Wen PY, Weller M, Lee EQ, Alexander BM, Barnholtz-Sloan JS, Barthel FP, Batchelor TT, Bindra RS, Chang SM, Chiocca EA, Cloughesy TF, DeGroot JF, Galanis E, Gilbert MR, Hegi ME, Horbinski C, Huang RY, Lassman AB, Le Rhun E, Lim M, Mehta MP, Mellinghoff IK, Minniti G, Nathanson D, Platten M, Preusser M, Roth P, Sanson M, Schiff D, Short SC, Taphoorn MJB, Tonn JC, Tsang J, Verhaak RGW, von Deimling A, Wick W, Zadeh G, Reardon DA, Aldape KD, van den Bent MJ (2020) Glioblastoma in adults: a Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions. Neuro Oncol 22(8):1073–1113CrossRef
Zurück zum Zitat Wick W, Platten M, Meisner C, Felsberg J, Tabatabai G, Simon M, Nikkhah G, Papsdorf K, Steinbach JP, Sabel M, Combs SE, Vesper J, Braun C, Meixensberger J, Ketter R, Mayer-Steinacker R, Reifenberger G, Weller M, N. O. A. S. G. o. N.-o. W. G. o. G. C. Society (2012) Temozolomide chemotherapy alone versus radiotherapy alone for malignant astrocytoma in the elderly: the NOA-08 randomised, phase 3 trial. Lancet Oncol 13(7):707–715CrossRef Wick W, Platten M, Meisner C, Felsberg J, Tabatabai G, Simon M, Nikkhah G, Papsdorf K, Steinbach JP, Sabel M, Combs SE, Vesper J, Braun C, Meixensberger J, Ketter R, Mayer-Steinacker R, Reifenberger G, Weller M, N. O. A. S. G. o. N.-o. W. G. o. G. C. Society (2012) Temozolomide chemotherapy alone versus radiotherapy alone for malignant astrocytoma in the elderly: the NOA-08 randomised, phase 3 trial. Lancet Oncol 13(7):707–715CrossRef
Zurück zum Zitat Zouaoui S, Darlix A, Fabbro-Peray P, Mathieu-Daude H, Rigau V, Fabbro M, Bessaoud F, Taillandier L, Ducray F, Bauchet F, Wager M, Faillot T, Capelle L, Loiseau H, Kerr C, Menei P, Duffau H, Figarella-Branger D, Chinot O, Tretarre B, Bauchet L (2014) Oncological patterns of care and outcomes for 265 elderly patients with newly diagnosed glioblastoma in France. Neurosurg Rev 37(3):415–423CrossRef Zouaoui S, Darlix A, Fabbro-Peray P, Mathieu-Daude H, Rigau V, Fabbro M, Bessaoud F, Taillandier L, Ducray F, Bauchet F, Wager M, Faillot T, Capelle L, Loiseau H, Kerr C, Menei P, Duffau H, Figarella-Branger D, Chinot O, Tretarre B, Bauchet L (2014) Oncological patterns of care and outcomes for 265 elderly patients with newly diagnosed glioblastoma in France. Neurosurg Rev 37(3):415–423CrossRef
Metadaten
Titel
Radiotherapy for glioblastoma patients with poor performance status
verfasst von
Christina Schröder
Dorothee Gramatzki
Erwin Vu
Matthias Guckenberger
Nicolaus Andratschke
Michael Weller
Caroline Hertler
Publikationsdatum
26.08.2021
Verlag
Springer Berlin Heidelberg
Erschienen in
Journal of Cancer Research and Clinical Oncology / Ausgabe 8/2022
Print ISSN: 0171-5216
Elektronische ISSN: 1432-1335
DOI
https://doi.org/10.1007/s00432-021-03770-9

Weitere Artikel der Ausgabe 8/2022

Journal of Cancer Research and Clinical Oncology 8/2022 Zur Ausgabe

Review – Clinical Oncology

Role of ANO1 in tumors and tumor immunity

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.