Does Pretreatment Tumor Growth Hold Prognostic Information for Patients with Glioblastoma?

Does Pretreatment Tumor Growth Hold Prognostic Information for Patients with Glioblastoma?

Accepted Manuscript Does pre-treatment tumor growth hold prognostic information for glioblastoma patients? Anne Line Stensjøen, M.D., PhD, Erik Magnus...

8MB Sizes 0 Downloads 8 Views

Accepted Manuscript Does pre-treatment tumor growth hold prognostic information for glioblastoma patients? Anne Line Stensjøen, M.D., PhD, Erik Magnus Berntsen, M.D., PhD, Vilde E. Mikkelsen, Sverre H. Torp, M.D., PhD, Asgeir S. Jakola, M.D., PhD, Øyvind Salvesen, PhD, Ole Solheim, M.D., PhD PII:

S1878-8750(17)30324-8

DOI:

10.1016/j.wneu.2017.03.012

Reference:

WNEU 5381

To appear in:

World Neurosurgery

Received Date: 18 November 2016 Revised Date:

1 March 2017

Accepted Date: 2 March 2017

Please cite this article as: Stensjøen AL, Berntsen EM, Mikkelsen VE, Torp SH, Jakola AS, Salvesen Ø, Solheim O, Does pre-treatment tumor growth hold prognostic information for glioblastoma patients?, World Neurosurgery (2017), doi: 10.1016/j.wneu.2017.03.012. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

Title: Does pre-treatment tumor growth hold prognostic information for glioblastoma patients? Authors: Anne Line Stensjøen1,2 M.D., PhD, Erik Magnus Berntsen1,2 M.D., PhD, Vilde E. Mikkelsen3,

RI PT

Sverre H. Torp3,4 M.D., PhD, Asgeir S. Jakola5,6,7 M.D., PhD, Øyvind Salvesen8 PhD, Ole Solheim5,9,10 M.D., PhD Affiliations:

Department of Circulation and Medical Imaging, Faculty of Medicine, NTNU - Norwegian

SC

1

University of Science and Technology, Trondheim, Norway

Department of Radiology, St. Olavs University Hospital, Trondheim, Norway

3

Department of Laboratory Medicine, Children´s and Women´s Health, Faculty of Medicine,

M AN U

2

NTNU - Norwegian University of Science and Technology, Trondheim, Norway 4

Department of Pathology and Medical Genetics, St. Olavs University Hospital, Trondheim,

TE D

Norway

Department of Neurosurgery, St. Olavs University Hospital, Trondheim, Norway

6

Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden

7

Institute of Neuroscience and Physiology, University of Gothenburg, Sahlgrenska Academy,

EP

5

8

AC C

Gothenburg, Sweden.

Department of Public Health and General Practice, Faculty of Medicine, NTNU - Norwegian

University of Science and Technology, Trondheim, Norway 9

Department of Neuroscience, Faculty of Medicine, NTNU - Norwegian University of Science

and Technology, Trondheim, Norway

1

ACCEPTED MANUSCRIPT

10

National Competence Centre for Ultrasound and Image Guided Therapy, St. Olavs University

RI PT

Hospital, Trondheim, Norway

Corresponding author: Anne Line Stensjøen, MR Center, Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology,

SC

Postbox 8905, N-7491 Trondheim, Norway. Phone: 0047-40465213, [email protected]

Abbreviations: GTR Gross total resection IDH Isocitrate dehydrogenase

TE D

KPS Karnofsky performance status

M AN U

Keywords: Glioblastoma, Longevity, Magnetic Resonance Imaging, Prognosis, tumor growth

MRI Magnetic resonance imaging

AC C

EP

PI Proliferation index

2

ACCEPTED MANUSCRIPT

Abstract Background

RI PT

Glioblastomas are highly aggressive and heterogeneous tumors, both in terms of patient outcome and molecular profile. Magnetic resonance imaging of tumor growth could potentially reveal new insights about tumor biology non-invasively. The aim of this exploratory retrospective study

SC

was to investigate the prognostic potential of pre-treatment growth rate of glioblastomas, after controlling for known prognostic factors.

M AN U

Methods

A growth model derived from clinical pre-treatment post-contrast T1-weighted MRI images was used to divide 106 glioblastoma patients into two groups. The “faster growth” group had tumors growing faster than expected based on their volume at diagnosis, while the “slower growth”

TE D

group had tumors growing slower than expected. Associations between tumor growth and survival were examined using multivariable cox regression and logistic regression. Results

EP

None of the known prognostic factors were significantly associated with tumor growth. An extended multivariable cox model showed that during the first twelve months of follow up, there

AC C

was no significant difference in survival between faster and slower growing tumors. However, beyond 12 months follow up, there was a significant, independent survival benefit in having a tumor with slower pre-treatment growth. In a multiple logistic regression model including patients receiving both radiotherapy and chemotherapy (n=82), slower pre-treatment growth of the tumor was shown to be a significant predictor of two-year survival (Odds Ratio 4.4). Conclusion 3

ACCEPTED MANUSCRIPT

Pre-treatment glioblastoma growth harbors prognostic information. Patients with slower growing

AC C

EP

TE D

M AN U

SC

RI PT

tumors have higher odds of survival beyond two years, adjusted for other prognostic factors.

4

ACCEPTED MANUSCRIPT

Introduction The clinical course in patients with glioblastoma is heterogeneous and prognostication is difficult 2

Despite advances in imaging and molecular biology, the most

RI PT

in individual patients.1,

established and strongest prognostic factors are still age at diagnosis and preoperative Karnofsky performance status (KPS). In addition, treatment associated factors such as surgical extent of

SC

resection, radiotherapy, and temozolomide treatment are linked to survival.3-5

It is frequently observed that some glioblastomas grow rapidly after the diagnostic MRI scan,

M AN U

before therapy is initiated, while others are seemingly more stable. For low-grade gliomas, tumor growth rate is an independent prognostic marker.6,

7

Still, few studies have investigated the

prognostic potential of growth for untreated glioblastomas. A small study including treated gliomas indicated an association between slower growth rates and longer survival for all

TE D

astrocytomas, but did not evaluate survival in glioblastoma patients alone.8 Other studies have proposed that mathematical models of glioblastoma growth kinetics can predict IDH1 status,9 and may have a prognostic role.10

EP

In this exploratory study, we sought to investigate the prognostic potential of measured growth in untreated glioblastomas. We also sought to assess potential associations between tumor growth

AC C

and established prognostic factors.

5

ACCEPTED MANUSCRIPT

Material and methods Patients

RI PT

We retrospectively evaluated all patients with histopathologically verified glioblastoma who were diagnosed at our hospital between January 2004 and May 2014. The inclusion criteria in this study were (1) patients ≥18 years, (2) histopathological diagnosis of glioblastoma, (3) no

SC

previous history of brain tumor, (4) at least two post-contrast T1-weighted MRI scans before surgery. Patients were excluded if the time interval between the two MRI examinations was less

M AN U

than 14 days, if the tumor was non-contrast enhancing, or if they had gliomatosis cerebri according to radiological criteria.11 Tumor growth data concerning the same patients have previously been reported.12 The study was approved by the Regional Ethics Committee (Central) as part of a larger project (references 2011/974 and 2013/1348), and adhered with the

TE D

Declaration of Helsinki. Most patients had provided informed consent to be included in a related glioma outcome study (reference 2011/974), and the regional ethics committee waived informed

EP

consent for retrospective evaluation of patient data for the remaining patients.

Magnetic resonance imaging

AC C

Two preoperative, post-contrast, T1-weighted magnetic resonance scans of each patient were retrieved from the hospital’s radiology database. These were images obtained as a part of the routine preoperative investigation using 1T, 1.5T or 3T magnetic resonance scanners. Scan parameters and their impact on image segmentations have been reported previously.12 The first scan, hereafter referred to as the diagnostic scan, was obtained at one of 15 local hospitals. The second scan, hereafter referred to as the preoperative scan, was obtained at St. Olavs University Hospital, usually the day before surgery, and used for intraoperative neuronavigation. 6

ACCEPTED MANUSCRIPT

Tumor growth The tumor volume at each MRI scan was segmented on the post contrast T1-weighted images by

RI PT

one of the authors (A.L.S.) using the software BrainVoyager QX (Brain Innovation, Maastricht, the Netherlands), and verified by a neuroradiologist (E.M.B.). The segmentation method has previously been described in detail.12 Tumor volume was defined based on the border of the contrast-enhancing rim of the tumor, and included all non-enhancing tumor tissue enclosed by

SC

this (i.e. necrosis).

M AN U

As a quality control, tumor growth was inspected on preoperative T2/FLAIR images in addition to the post-contrast T1-weighted images. Lesions on T2 images beyond the contrast enhancing tumor were classified into edema and non-enhancing tumor, based on the criteria of the VASARI features.13 Significant non-enhancing tumor compartments were evaluated for growth by a neuroradiologist (E.M.B), using diameter measurements and the RANO criteria.14 Edema was

TE D

evaluated visually for significant volume changes. The results of this quality control can be found in Supplementary Figure 1. All the tumors with a growing non-enhancing tumor

Growth groups

EP

compartment had concurrent extensive growth of the contrast enhancing tumor.

AC C

In a previous study, tumor volumes and scan interval were used to estimate glioblastoma growth dynamics, based on the assumption that all tumors follow the same growth pattern.12 Tumor growth rates correlated with tumor volume, indicating that point estimates of growth rate will be a poor representation of tumor biology. Since we also found that the volume of necrosis increases faster in lager tumors, and since intracranial space is limited, we further assumed that glioblastomas eventually approach a plateau phase with slower growth. Based on these assumptions we found that glioblastoma growth was best fitted by a gompertzian growth model, 7

ACCEPTED MANUSCRIPT

with a declining growth rate over time.15 Figure 1 shows the gompertzian growth curve with parameters fitted from the observed tumor growth. Under the assumption that all glioblastomas

and predict future growth of a tumor with one known volume.

RI PT

follow the same gompertzian growth pattern, this curve can be used to estimate previous growth

In the present study, the gompertzian growth model was used to divide the patients into two groups, based on how their tumor growth related to the growth model (Figure 1). The “faster

SC

growth” group was defined as tumors with larger volume increase than expected from the model,

M AN U

while a “slower growth” group included tumors with a smaller volume increases than expected. Examples of MRI scans of tumors in the two groups are presented in Figure 2. This dichotomization is used in the following survival analyses.

Clinical data collection

TE D

Clinical characteristics, treatment data, and survival was either retrieved from a prospectively collected research database, or retrospectively retrieved from hospital records. The clinical data retrieved were sex, age at diagnosis, KPS, treatment and overall survival. KPS was mostly

EP

retrieved from the research database, but in 25 cases with missing data, KPS was estimated after retrospective review of hospital records. Four different treatment variables were recorded;

AC C

Complete resection of enhancing tumor (GTR) (yes/no), chemotherapy treatment (yes/no), radiotherapy (yes/no) and corticosteroid treatment (yes/no). Due to the difficulties in evaluating contrast enhancement in the rim of the resection cavity, GTR calculated from an ellipsoid formula on post-operative MRI scans was defined as residual volume of less than 0.175 ml, as done by others.16 A more detailed categorization of extent of resection was not performed, since the benefit of subtotal resection is not settled.17, 18 Chemotherapy treated patients were defined as patients who received temozolomide within the first six months after surgery, either before 8

ACCEPTED MANUSCRIPT

radiotherapy, concomitant, adjuvant or any combination of these. Radiotherapy treated patients were defined as patients who received radiotherapy in any regime during follow-up. Corticosteroid treatment was defined as any corticosteroids given before or between the two

RI PT

retrieved MRI examinations. Overall survival was defined as the time from first surgery to death. Patients were censored if alive at the end of the study (June 2016), which was 24.5 months after

SC

the surgery of the last included patient.

Histopathology and molecular markers

M AN U

A medical student (V.E.M), in close collaboration with an experienced neuropathologist (S.H.T.), reviewed all tissue samples and confirmed the glioblastoma diagnosis in all included patients, according to the 2007 WHO classification.3 All cases underwent immunohistochemical analyses. Paraffin sections, 4 µm thick, were incubated with monoclonal antibodies using

TE D

standard immunohistochemical analyses using an automated immunohistostainer (DAKO Techmate 500). In 12 cases for IDH1 and 9 cases for Ki-67/MIB-1, formalin-fixed paraffinembedded frozen sections were used instead of paraffin sections. For determination of

EP

proliferative activity, the sections were incubated with Ki-67/MIB-1 (dilution 1:800 or 1:50; Dako, Glostrup, Denmark). A Ki-67/MIB-1 proliferation index (PI) was calculated as a

AC C

percentage of distinct immunoreactive tumor cell nuclei in areas with highest labeling density (“hot spots”). IDH1 mutant protein status was examined by incubation with the IDH1 R132H antibody (dilution 1:100, Dianova, Hamburg, Germany). In each staining experiment, positive controls were included, and in the negative controls the primary antibodies were omitted. According to the recent revision of the WHO classification criteria, immunohistochemistry against IDH1 R132H is considered adequate for evaluation of IDH mutation status in patients

9

ACCEPTED MANUSCRIPT

older than 55 years, while mutation status should be confirmed by IDH gene sequencing in younger patients.19 Gene sequencing was not available for this cohort.

RI PT

Statistical methods Exploratory statistical analyses were performed using Stata/IC 13.1 for Windows (32-bit) and R version 3.1.2. The statistical significance level was set to P <.05. Descriptive statistics were

SC

assessed in Stata, and possible associations between the growth groups and the different variables were investigated using Wilcoxon Rank-sum tests or Fisher’s exact/Pearson Chi square

M AN U

tests, as appropriate. Quantitative variables were treated as continuous in all analyses. The outcome in all cox proportional hazards models were overall survival. In R, unadjusted cox proportional hazards models were fitted for the following variables: Age, KPS, Ki-67/MIB-1 PI, Preoperative tumor volume, GTR, Chemotherapy, radiation, corticosteroids and Slower growth.

TE D

To determine the independent prognostic value of glioblastoma growth, a multivariable cox proportional hazards model was fitted, including previously established prognostic factors and significant factors from unadjusted models, as reported in Supplementary Table 2. The

EP

proportional hazards assumption was tested for all variables using Schoenfeld residuals.20 Violation of this assumption lead to extension of the cox proportional hazards model, to account

AC C

for time-varying effects of the growth variable.21

10

ACCEPTED MANUSCRIPT

Results Participants

RI PT

262 patients were evaluated for inclusion in the study, and 106 of these were included in the analyses. Reasons for exclusion are reported in Figure 3. All patients were followed until death

SC

or the end of the study.

Descriptive data

M AN U

Of the 106 patients, 34 were female (32%). The mean age at diagnosis was 62.9 ±11.7 years. The median preoperative KPS was 80 (range 40 to 100), and 18 of the patients (17%) had a KPS below 70. Median Ki-67/MIB-1 PI was 13.2% (range 1.4-57.3%). Two patients had tumors immunopositive for the IDH1 R132H mutation. The distributions of the treatments received are

TE D

presented in Table 1. Biopsy only had been performed in 17 cases (16 %). There was no missing data for any of the variables. As seen in Table 1, there were no significant associations between

Outcome data

EP

patient characteristics and established prognostic factors and fast versus slow growth.

AC C

Median overall survival for the patients in this study was 12.6 months (95% CI 10.1-15.4 months). One-year survival was 52.8%, while two-year survival was 19.8%.

The prognostic potential of glioblastoma growth Figure 4 shows the unadjusted Kaplan-Meier plot for the two growth groups. The Kaplan-Meier plot showed that the two survival curves crossed, indicating that the proportional hazards assumption was violated for the growth variable. This was confirmed in both unadjusted and multivariable cox regression models, as testing using Schoenfeld residuals showed that the 11

ACCEPTED MANUSCRIPT

violation was significant (see Supplementary Table 1 and 2, with results from cox regression models). This would make inferences from standard cox models inaccurate. To account for the non-proportional hazard functions of the growth groups, an extended cox model was developed.

RI PT

This model allowed different hazard ratios between the growth groups in the first 12 months of follow-up and the later follow up for all patients. The results from this analysis are shown in Table 2. In the first 12 months of follow-up, growth group was not significantly linked to

SC

survival. However, after 12 months follow-up, the hazard rate was significantly lower in patients

M AN U

with tumors with slower growth (hazard ratio 0.41 (95% CI 0.21-0.78), P=0.007).

Tumor growth as a predictor of two-year survival

The extended cox regression model suggested that slower tumor growth might be a predictor of longevity. Of patients who survived more than two years, 15 (71.4%) had a slower growth than

TE D

expected, while 6 (28.6%) had a faster growth. All eight patients (7.5%) who had survived more than 3 years at the time of analysis belonged to the slower growth group. Associations between two-year survival and clinical variables are shown in Supplementary Table 3. We explored

EP

predictors of two-year survival in a multiple logistic regression model, including known prognostic factors. Patients who had not received either temozolomide chemotherapy or

AC C

radiotherapy were excluded, as this corresponded perfectly with not surviving beyond two years. For the remaining 82 patients, the logistic regression model showed that slower growth was a significant predictor of two-year survival, after controlling for age, KPS and GTR. Tumors with slower growth had 4 times higher odds of survival beyond two years, compared to patients with faster growing tumors (Table 3).

12

ACCEPTED MANUSCRIPT

Results using different growth pattern In the review process, we were asked to supplement our findings with another way of stratifying tumor growth. We then used a linear radial growth curve, which was fitted as previously

RI PT

described.12 The tumors were stratified according to whether they grew slower or faster than expected, based on this growth pattern, and these groups were used for additional survival analyses. The results of these analyses were similar to the results using the gompertzian growth

AC C

EP

TE D

M AN U

SC

pattern (Supplementary Table 4-6).

13

ACCEPTED MANUSCRIPT

Discussion Key results

RI PT

In this exploratory study, we found that slow radiological tumor growth in glioblastomas was associated with survival. While there was no significant difference in survival between faster and slower growing tumors during the first twelve months of follow up, there was a significant

SC

survival benefit in having a tumor with slower growth beyond 12 months. In a multiple logistic regression model including the patients who received both chemotherapy and radiotherapy,

M AN U

slower growth of the tumor was shown to be a significant predictor of two-year survival. None of the clinical variables, including established prognostic factors, were significantly associated with radiological tumor growth. Pre-treatment tumor growth harbors prognostic properties in glioblastoma, and holds potential to capture differences in biological properties in vivo.

TE D

Growth as indicator of tumor biology

While molecular classification of lower grade gliomas has been successful in terms of predicting survival,2 more heterogeneous and conflicting results are reported for glioblastoma in

EP

comprehensive genetic studies.2, 22 The Cancer Genome Atlas identified four clusters of genetic alterations in glioblastomas, but the prognostic value seems modest.23 A limitation of molecular

AC C

studies is the heterogeneity of genetic profile within the same tumors and over time.24, 25 In contrast to molecular and genetic markers, radiological assessment of glioblastomas holds the potential to readily investigate both the entire tumor at once, and changes over time. Growth rates and response to treatment can be assessed from conventional structural MR sequences.14, 26 More advanced imaging modalities have also been shown to harbor biological information, including perfusion and diffusion MRI and PET-imaging.27, 28 However, so far studies exploring

14

ACCEPTED MANUSCRIPT

potential prognostic features from preoperative MRIs have been static in nature, i.e. assessing data at one time point.29-31

RI PT

Tumor growth is not associated with clinical parameters While advanced age is a strong negative prognostic factor in glioblastoma32 that is also linked to molecular profiles, we did not find a significant association between preoperative growth and

SC

age. This might indicate that the poor survival in the elderly may reflect response to therapy, treatment nihilism, or diagnostic lead-time bias, more than the speed of growth of the disease.

M AN U

Further, we did not find an association between KPS and preoperative tumor growth. The prognostic value of functional status may in part reflect the tradition for avoiding interventions in functionally dependent patients, i.e. with lesions in eloquent regions, and does not always reflect the extent or the aggressiveness of the disease.

TE D

Ki-67/MIB-1 PI was not a prognostic marker in this study, and while some authors have previously shown it to be a weak prognostic marker,33 several others have not.34, 35 Ki-67/MIB-1 was not associated with radiological growth groups in our study. As with other molecular

EP

markers, Ki-67 PI will be based on a sample of the heterogeneous tumor tissue, and might not reflect the biology of the most aggressive part of the tumor. Furthermore, Ki-67/MIB-1 PIs in

37

AC C

glioblastomas display a wide range of values, which overlap those of low-grade astrocytomas,36, so one may wonder whether this proliferation marker fully unveil the proliferative activity in

human glioblastomas.

Speed of growth and long-term survival Previously suggested prognostic or predictive factors for long-term survival for glioblastoma include age and KPS, in addition to MGMT hypermethylation and IDH1/2-mutations.38,

15

39

ACCEPTED MANUSCRIPT

Gerber et al. found that patients with long term survival had very heterogeneous genetic profiles,40 which could indicate that molecular profiling will not be helpful in identifying these patients. The present study shows that radiological growth might improve prognostication, as

RI PT

long term survival beyond 2 years is more unlikely if fast radiological growth is measured preoperatively.

The long term survival in this study was not linked to IDH-mutations, as eight patients had a

SC

survival of more than 3 years, and only one of these harbored a tumor with an IDH1 R132H mutation. Although it has been reported that IDH mutations in glioblastoma is linked to

M AN U

survival,41 two recent studies reported that IDH-mutations are not essential for long time survival.40, 42

While longevity in this study was linked to tumor growth, survival the first year was not. This may reflect that numerous factors, including extent of disease at diagnosis, treatment given and

TE D

treatment responses affect survival in the short term, while a less aggressive biology of disease is necessary for long term survival. However, the lack of significant survival differences the first year of follow-up may also be due to lack of statistical power, or it may illustrate that the

EP

association between growth and survival is complex, with several interfering factors.

AC C

Potential implications

The findings in this study, even if confirmed in later studies, will probably be difficult to use directly in a clinical setting. The measurement of growth rates requires two pre-treatment MRI scans, which are often not obtained as part of clinical routine. Still, our results may suggest that in search of biological subgroups with different prognoses or clinical courses, radiological assessments including growth rates may be a way to advance. As a research field this is much more unexplored than for example tumor genetics and molecular biology. 16

ACCEPTED MANUSCRIPT

While our study shows that long term survival is unlikely with fast growing tumors, clinicians should not withhold treatment from an otherwise stable patient with a fast growing tumor. However, tumor growth might be included in a larger decision algorithm, or as suggested by

RI PT

Wang et al., as part of a personalized mathematical model.10 Tumor growth has been used to quantify the invasiveness and proliferative potential of each tumor, which again was reported predictive of IDH1 status and response to radiotherapy.9, 43

SC

Generalizability

M AN U

Patients were included consecutively in a department with a defined geographical catchment region. This enhances external validity of our findings. As previously reported, the patients included in this study had no significant differences in the age and gender distribution compared to excluded patients.12

TE D

It is expected that the incidence of IDH immunoreactive tumors would be low in a glioblastoma patient cohort without any previous history of brain tumors, and this was indeed the case in our study, with only two positive cases. This means that the results presented in this thesis to a large

EP

degree reflects the growth of IDH-wildtype glioblastomas, and should not necessarily be generalized to IDH-mutated glioblastomas.

AC C

It has previously been shown that the growth rate of glioblastomas correlates with tumor volume, and thus could be expected to change over time.12 A simple point estimate of tumor growth rate would potentially have been easier to implement in the clinic, but would not be an accurate description of tumor biology. In the present study, we instead used a previously described model of glioblastoma growth, and classified the tumors according to whether they grew faster or slower than predicted by the model. Assuming that the model is a correct representation of group-level glioblastoma growth, this classification gives us a more reliable representation of 17

ACCEPTED MANUSCRIPT

tumor biology. It should be noted that this growth model still lacks validation in other data sets. While the growth measures underlying this model are similar to the ones recently reported by Ellingson et al,26 independent testing of the assumption of gompertzian growth is needed.

conclusions did not change if this pattern was assumed.

SC

Limitations

RI PT

Another possible growth pattern for glioblastomas is the linear radial growth pattern,12 and our

The main limitation is the exploratory nature of our study with few pre-planned statistical tests.

M AN U

This data driven post hoc methodology was a necessity due to the novelty of the study approach, but limits the strength of our conclusions.

Only two of the included patients had a positive immunohistochemistry for the IDH1 R132H mutation. It has been shown that immunohistochemistry is less reliable for detection of IDH

TE D

mutations in patients younger than 55 years.19 However, among the patients who survived more than three years in our study, none were younger than 55, indicating that the probability of false negative IDH status is very low.44

EP

An important molecular marker for glioblastoma patients is MGMT promoter status.45 The prognostic role of this marker is controversial. While some have found that it is a prognostic

AC C

marker,46 others report that it is only predictive for response to temozolomide, at least in IDH wildtype glioblastomas.47,

48

Unfortunately, this genetic marker was not available for this

retrospective cohort. If MGMT status is predictive of temozolomide response, statistical dependence between temozolomide treatment and MGMT status would be expected, and we adjusted for temozolomide treatment in the regression model. In future studies, it will be

18

ACCEPTED MANUSCRIPT

important to evaluate whether the prognostic role of glioblastoma growth is independent of MGMT status.

RI PT

In this study, we have investigated glioblastoma growth as seen on contrast enhanced T1weighted images. It is likely that some glioblastomas have a growth characterized more by a diffuse spread of cells, than growth of the tumor bulk.10 This means that the total tumor burden could increase, while the visible tumor on contrast enhanced T1-weighted images stays constant.

SC

Unfortunately, this diffuse growth of the tumor is difficult to detect or differentiate from edema

M AN U

using standard anatomical imaging methods.49 In addition, changes in T2/FLAIR volume may have several different reasons, such as effects from corticosteroid use, and it is difficult to differentiate such changes from true tumor growth.14 When visually inspecting the T2/FLAIR images of all patients, we found that none of the patients had a significant growth of the T2/FLAIR compartment, without concurrent growth of the contrast enhancing tumor. It is

TE D

therefore unlikely that we would have detected any additional tumors with rapid, diffuse spread, by segmenting the T2/FLAIR components.

Our inclusion criteria reduced the number of eligible patients from 262 to 106, which could have

EP

introduced selection bias. However, as previously reported, there were no significant differences

AC C

in age and sex between included and excluded patients.12 The range of KPS scores in our patient sample corresponds to the values found in various international population based studies.50, 51 We did not examine the survival in the excluded patients. However, the median overall survival in the included patients was 12.6 months (95% CI 10.1-15.4 months), which is only slightly higher than the overall survival of 10.1 months (95% CI 9.4-11.0 months) reported in a registry-based study of unselected glioblastoma patients in Norway.52

19

ACCEPTED MANUSCRIPT

Conclusion Pre-treatment tumor growth harbors prognostic properties in glioblastoma, and holds potential to

RI PT

capture differences in biological properties in vivo. For patients receiving both chemotherapy and radiotherapy, having a slower growing tumor gave higher odds of survival beyond two years.

SC

Due to the exploratory nature of this study, our findings should be subject to validation studies.

Acknowledgements

M AN U

We would like to thank M.Sc. Lisa Millgård Sagberg for her contribution to the collection of clinical data.

Funding

EP

or not-for-profit sectors.

TE D

This research did not receive any specific grant from funding agencies in the public, commercial,

Conflicts of interest

AC C

Ole Solheim is an unpaid member of a national advisory committee on treatment guidelines for brain tumors. Asgeir S. Jakola is an unpaid member of the Swedish National Brain Tumor Trialist Group. The other authors disclose no potential conflicts of interest.

20

ACCEPTED MANUSCRIPT

References 1.

Parks C, Heald J, Hall G, Kamaly-Asl I. Can the prognosis of individual patients with glioblastoma be predicted using an online calculator? Neuro Oncol. 2013;15(8):1074-

2.

RI PT

1078, doi: 10.1093/neuonc/not033.

Eckel-Passow JE, Lachance DH, Molinaro AM, et al. Glioma Groups Based on 1p/19q, IDH, and TERT Promoter Mutations in Tumors. N Engl J Med. 2015;372(26):2499-2508, doi: 10.1056/NEJMoa1407279.

Louis D, Ohgaki H, Wiestler O, Cavenee W. WHO classification of tumours of the

SC

3.

central nervous system. 4th ed. Geneva, Switzerland: Distributed by WHO Press, World

4.

M AN U

Health Organization; 2007.

Lacroix M, Abi-Said D, Fourney DR, et al. A multivariate analysis of 416 patients with glioblastoma multiforme: prognosis, extent of resection, and survival. J Neurosurg. 2001;95(2):190-198, doi: 10.3171/jns.2001.95.2.0190.

5.

Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987-996, doi:

6.

TE D

10.1056/NEJMoa043330.

Pallud J, Blonski M, Mandonnet E, et al. Velocity of tumor spontaneous expansion predicts long-term outcomes for diffuse low-grade gliomas. Neuro Oncol. 2013;15(5):595-606, doi: 10.1093/neuonc/nos331. Rees J, Watt H, Jager HR, et al. Volumes and growth rates of untreated adult low-grade

EP

7.

gliomas indicate risk of early malignant transformation. Eur J Radiol. 2009;72(1):54-64,

8.

AC C

doi: 10.1016/j.ejrad.2008.06.013. Blankenberg FG, Teplitz RL, Ellis W, et al. The influence of volumetric tumor doubling time, DNA ploidy, and histologic grade on the survival of patients with intracranial astrocytomas. AJNR Am J Neuroradiol. 1995;16(5):1001-1012.

9.

Baldock AL, Yagle K, Born DE, et al. Invasion and proliferation kinetics in enhancing gliomas predict IDH1 mutation status. Neuro Oncol. 2014;16(6):779-786, doi: 10.1093/neuonc/nou027.

10.

Wang CH, Rockhill JK, Mrugala M, et al. Prognostic significance of growth kinetics in newly diagnosed glioblastomas revealed by combining serial imaging with a novel 21

ACCEPTED MANUSCRIPT

biomathematical model. Cancer Res. 2009;69(23):9133-9140, doi: 10.1158/00085472.CAN-08-3863. 11.

Osborn AG. Astrocytomas. In: Osborn AG, ed. Osborn’s Brain: Imaging, Pathology, and Anatomy. Salt Lake City, UT: Amirsys, Inc; 2012 435-472. Stensjøen AL, Solheim O, Kvistad KA, Håberg AK, Salvesen Ø, Berntsen EM. Growth

RI PT

12.

dynamics of untreated glioblastomas in vivo. Neuro Oncol. 2015;17(10):1402-1411, doi: 10.1093/neuonc/nov029. 13.

The Cancer Imaging Archive. VASARI Research Project. 2015;

SC

https://wiki.cancerimagingarchive.net/display/Public/VASARI+Research+Project Accessed 18. sep, 2016.

Wen PY, Macdonald DR, Reardon DA, et al. Updated Response Assessment Criteria for

M AN U

14.

High-Grade Gliomas: Response Assessment in Neuro-Oncology Working Group. J Clin Oncol. 2010;28(11):1963-1972, doi: 10.1200/jco.2009.26.3541. 15.

Laird AK. Dynamics of Tumor Growth. Br J Cancer. 1964;18(3):490-502.

16.

Stummer W, Pichlmeier U, Meinel T, et al. Fluorescence-guided surgery with 5aminolevulinic acid for resection of malignant glioma: a randomised controlled

2045(06)70665-9. 17.

TE D

multicentre phase III trial. Lancet Oncol. 2006;7(5):392-401, doi: 10.1016/S1470-

Kreth FW, Thon N, Simon M, et al. Gross total but not incomplete resection of glioblastoma prolongs survival in the era of radiochemotherapy. Ann Oncol.

18.

EP

2013;24(12):3117-3123, doi: 10.1093/annonc/mdt388. Stummer W, Reulen HJ, Meinel T, et al. Extent of resection and survival in glioblastoma

AC C

multiforme: identification of and adjustment for bias. Neurosurgery. 2008;62(3):564-576; discussion 564-576, doi: 10.1227/01.neu.0000317304.31579.17.

19.

Louis DN, Perry A, Reifenberger G, et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol.

2016;131(6):803-820, doi: 10.1007/s00401-016-1545-1.

20.

Schoenfeld D. Partial residuals for the proportional hazards regression model. Biometrika. 1982;69(1):239-241, doi: 10.1093/biomet/69.1.239.

21.

Kleinbaum D, Klein M. Survival Analysis - A Self-Learning Text. Third edition ed: Springer; 2012. 22

ACCEPTED MANUSCRIPT

22.

Labussière M, Boisselier B, Mokhtari K, et al. Combined analysis of TERT, EGFR, and IDH status defines distinct prognostic glioblastoma classes. Neurology. 2014;83(13):1200-1206, doi: 10.1212/wnl.0000000000000814.

23.

Verhaak RG, Hoadley KA, Purdom E, et al. Integrated genomic analysis identifies

RI PT

clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 2010;17(1):98-110, doi: 10.1016/j.ccr.2009.12.020. 24.

Patel AP, Tirosh I, Trombetta JJ, et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science. 2014;344(6190):1396-1401, doi:

25.

SC

10.1126/science.1254257.

Johnson BE, Mazor T, Hong C, et al. Mutational Analysis Reveals the Origin and

10.1126/science.1239947. 26.

M AN U

Therapy-Driven Evolution of Recurrent Glioma. Science. 2014;343(6167):189-193, doi:

Ellingson BM, Nguyen HN, Lai A, et al. Contrast-enhancing tumor growth dynamics of preoperative, treatment-naive human glioblastoma. Cancer. 2016;122(11):1718-1727, doi: 10.1002/cncr.29957.

27.

Kalpathy-Cramer J, Gerstner ER, Emblem KE, Andronesi OC, Rosen B. Advanced

TE D

Magnetic Resonance Imaging of the Physical Processes in Human Glioblastoma. Cancer Res. 2014;74(17):4622-4637, doi: 10.1158/0008-5472.can-14-0383. 28.

Kamson DO, Juhasz C, Buth A, et al. Tryptophan PET in pretreatment delineation of newly-diagnosed gliomas: MRI and histopathologic correlates. J Neurooncol.

29.

EP

2013;112(1):121-132, doi: 10.1007/s11060-013-1043-4. Mazurowski MA, Desjardins A, Malof JM. Imaging descriptors improve the predictive

AC C

power of survival models for glioblastoma patients. Neuro Oncol. 2013;15(10):13891394, doi: 10.1093/neuonc/nos335.

30.

Gutman DA, Cooper LA, Hwang SN, et al. MR imaging predictors of molecular profile and survival: multi-institutional study of the TCGA glioblastoma data set. Radiology.

2013;267(2):560-569, doi: 10.1148/radiol.13120118.

31.

Jain R, Poisson LM, Gutman D, et al. Outcome prediction in patients with glioblastoma by using imaging, clinical, and genomic biomarkers: focus on the nonenhancing component of the tumor. Radiology. 2014;272(2):484-493, doi: 10.1148/radiol.14131691.

23

ACCEPTED MANUSCRIPT

32.

Gulati S, Jakola AS, Johannesen TB, Solheim O. Survival and treatment patterns of glioblastoma in the elderly: a population-based study. World Neurosurg. 2012;78(5):518526, doi: 10.1016/j.wneu.2011.12.008.

33.

Thuy MN, Kam JK, Lee GC, et al. A novel literature-based approach to identify genetic

RI PT

and molecular predictors of survival in glioblastoma multiforme: Analysis of 14,678 patients using systematic review and meta-analytical tools. J Clin Neurosci. 2015;22(5):785-799, doi: 10.1016/j.jocn.2014.10.029. 34.

Moskowitz SI, Jin T, Prayson RA. Role of MIB1 in predicting survival in patients with

35.

SC

glioblastomas. J Neurooncol. 2006;76(2):193-200, doi: 10.1007/s11060-005-5262-1. Bouvier-Labit C, Chinot O, Ochi C, Gambarelli D, Dufour H, Figarella-Branger D.

M AN U

Prognostic significance of Ki67, p53 and epidermal growth factor receptor immunostaining in human glioblastomas. Neuropathol Appl Neurobiol. 1998;24(5):381388. 36.

Torp SH. Diagnostic and prognostic role of Ki67 immunostaining in human astrocytomas using four different antibodies. Clin Neuropathol. 2002;21(6):252-257.

37.

Skjulsvik AJ, Mork JN, Torp MO, Torp SH. Ki-67/MIB-1 immunostaining in a cohort of

38.

TE D

human gliomas. Int J Clin Exp Pathol. 2014;7(12):8905-8910. Hartmann C, Hentschel B, Simon M, et al. Long-term survival in primary glioblastoma with versus without isocitrate dehydrogenase mutations. Clin Cancer Res. 2013;19(18):5146-5157, doi: 10.1158/1078-0432.CCR-13-0017. Das P, Puri T, Jha P, et al. A clinicopathological and molecular analysis of glioblastoma

EP

39.

multiforme with long-term survival. J Clin Neurosci. 2011;18(1):66-70, doi:

40.

AC C

10.1016/j.jocn.2010.04.050.

Gerber NK, Goenka A, Turcan S, et al. Transcriptional diversity of long-term glioblastoma survivors. Neuro Oncol. 2014;16(9):1186-1195, doi: 10.1093/neuonc/nou043.

41.

Cheng HB, Yue W, Xie C, Zhang RY, Hu SS, Wang Z. IDH1 mutation is associated with

improved overall survival in patients with glioblastoma: a meta-analysis. Tumour Biol. 2013;34(6):3555-3559, doi: 10.1007/s13277-013-0934-5.

24

ACCEPTED MANUSCRIPT

42.

Amelot A, De Cremoux P, Quillien V, et al. IDH-Mutation Is a Weak Predictor of LongTerm Survival in Glioblastoma Patients. PLoS One. 2015;10(7):e0130596. doi: 10.1371/journal.pone.0130596.

43.

Swanson KR, Harpold HLP, Peacock DL, et al. Velocity of Radial Expansion of

RI PT

Contrast-enhancing Gliomas and the Effectiveness of Radiotherapy in Individual

Patients: a Proof of Principle. Clin Oncol (R Coll Radiol). 2008;20(4):301-308, doi: 10.1016/j.clon.2008.01.006. 44.

Chen L, Voronovich Z, Clark K, et al. Predicting the likelihood of an isocitrate

SC

dehydrogenase 1 or 2 mutation in diagnoses of infiltrative glioma. Neuro Oncol. 2014;16(11):1478-1483, doi: 10.1093/neuonc/nou097.

Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene silencing and benefit from

M AN U

45.

temozolomide in glioblastoma. N Engl J Med. 2005;352(10):997-1003, doi: 10.1056/NEJMoa043331. 46.

Rivera AL, Pelloski CE, Gilbert MR, et al. MGMT promoter methylation is predictive of response to radiotherapy and prognostic in the absence of adjuvant alkylating chemotherapy for glioblastoma. Neuro-Oncology. 2010;12(2):116-121, doi:

47.

TE D

10.1093/neuonc/nop020.

Wick W, Meisner C, Hentschel B, et al. Prognostic or predictive value of MGMT promoter methylation in gliomas depends on IDH1 mutation. Neurology. 2013;81(17):1515-1522, doi: 10.1212/WNL.0b013e3182a95680. Stupp R, Brada M, van den Bent MJ, Tonn JC, Pentheroudakis G. High-grade glioma:

EP

48.

ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Annals of

49.

AC C

Oncology. 2014;25(suppl_3):iii93-iii101, doi: 10.1093/annonc/mdu050. Kelly PJ, Daumas-Duport C, Kispert DB, Kall BA, Scheithauer BW, Illig JJ. Imagingbased stereotaxic serial biopsies in untreated intracranial glial neoplasms. J Neurosurg.

1987;66(6):865-874, doi: 10.3171/jns.1987.66.6.0865.

50.

Brandes AA, Franceschi E, Ermani M, et al. Pattern of care and effectiveness of treatment for glioblastoma patients in the real world: Results from a prospective population-based registry. Could survival differ in a high-volume center? Neurooncol Pract. 2014;1(4):166-171, doi: 10.1093/nop/npu021.

25

ACCEPTED MANUSCRIPT

51.

Yang P, Wang Y, Peng X, et al. Management and survival rates in patients with glioma in China (2004-2010): A retrospective study from a single-institution. J Neurooncol. 2013;113(2):259-266, doi: 10.1007/s11060-013-1103-9. Rønning PA, Helseth E, Meling TR, Johannesen TB. A population-based study on the

EP

TE D

M AN U

SC

2012;14(9):1178-1184, doi: 10.1093/neuonc/nos153.

RI PT

effect of temozolomide in the treatment of glioblastoma multiforme. Neuro Oncol.

AC C

52.

26

ACCEPTED MANUSCRIPT

Figure captions Figure 1: The sigmoid curve shows the expected growth pattern of glioblastomas on a

RI PT

logarithmic time scale, based on growth data from all patients. The points are the observed preoperative total tumor volumes for all patients. Tumors above the curve had a faster growth than expected, while tumors below the curve had a slower growth than expected. The black points are the tumors of patients surviving less than two years, while the red points represent the

SC

tumors of patients who survived at least two years. The curve is drawn from day 10 with a tumor

M AN U

volume of 0.226 mL, corresponding to an arbitrary starting volume of 0.135 mL at day 0. Figure 2: Examples of tumors in the two growth groups. Panel A shows a tumor growing more slowly than expected, with a volume increase from 13.2 mL to 14.3 mL in 17 days. This tumor was expected to have grown to 17.8 mL in 17 days. Panel B shows a tumor growing faster than expected, with a volume increase from 0.7 mL to 11.2 mL in 31 days. This tumor was expected

TE D

to have grown to 2.2 mL in 31 days.

Figure 3: Patient selection diagram with exclusion criteria.

EP

Figure 4: Survival in glioblastoma patients in two different growth groups. The “faster growth” group was defined as tumors with larger volume increase than expected from a mathematical

AC C

model, while a “slower growth” group included tumors with a smaller volume increases than expected.

27

ACCEPTED MANUSCRIPT

Fast growth

(n=53)

(n=53)

Age (years)

63

64

Gender (n of women)

16

18

KPS

80

70

Preoperative volume (mL)

24.0

28.8

Ki-67/MIB-1 (%)

13.7

12.3

IDH1+ (n)

1

Corticosteroids (n)

45

GTR (n)

15

Chemotherapy (n)

40

TE D

Slow growth

M AN U

Table 1: Distribution of different variables between growth groups.

Radiation (n)

49

P-value

Rank-sum

RI PT

.08

Statistical test

χ2

.17

Rank-sum

.62

Rank-sum

SC

.68

.91

Rank-sum

1

1

Fisher exact

41

.32

χ2

15

1

χ2

43

.48

χ2

47

.51

χ2

EP

Numbers in italic are median values. For treatments, the numbers indicate how many patients who recieved each treatment. Abbreviations: KPS, Karnofsky performance status; GTR,

AC C

Gross total resection.

ACCEPTED MANUSCRIPT

Hazard ratio (95% CI)

P-value

Age

1.02 (1.00-1.04)

.11

KPS (10 points)

0.76 (0.62-0.93)

.008

Preoperative volume

1.00 (0.99-1.01)

GTR

0.72 (0.43-1.21)

Chemotherapy

0.27 (0.14-0.53)

Radiation

0.15 (0.06-0.39)

RI PT

Table 2: Results of an extended multivariable Cox regression model

Slower growth (<12 months)

1.55 (0.88-2.74)

Slower growth (≥12 months)

.98

.22

<.001

<.001

SC

M AN U

Variable

0.41 (0.21-0.78)

.13 .007

Abbreviations: CI, confidence interval; KPS, Karnofsky performance status; GTR, Gross

AC C

EP

TE D

total resection.

ACCEPTED MANUSCRIPT Table 3: Odds ratios for survival above two years from a multiple logistic regression *

Variable

Odds ratio (95% CI)

P-value

Age

0.97 (0.92-1.01)

.16

KPS (10 points)

1.44 (0.87-2.38)

.16

GTR

1.32 (0.40-4.38)

.65

Slower growth

4.42 (1.33-14.74)

.015

RI PT

analysis

SC

Abbreviations: CI, confidence interval; KPS, Karnofsky performance status; GTR, Gross

M AN U

total resection.

AC C

EP

TE D

*Including only patients receiving both chemotherapy and radiotherapy (n=82)

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT

Highlights

RI PT SC M AN U TE D EP

• •

Using a gompertzian growth model, tumors were divided into slower and faster growing Established prognostic factors were not associated with glioblastoma growth groups Slower pre-treatment growth was a significant predictor of two-year survival

AC C



ACCEPTED MANUSCRIPT Supplementary table 1: Unadjusted cox regression models for each variable, with tests of proportional hazard assumption. Unadjusted cox regression models Variable

Hazard ratio

Schoenfeld residuals test

p-value

Chi-square statistic 3.05

.08

4.28

.04

1.93

.17

0.19

.67

0.06

.80

<.001

1.13

.29

0.05 (0.02-0.12)

SC

1.03 (1.01-1.05)

.009

KPS (10 points)

0.68 (0.58-0.80)

<.001

Preoperative volume

1.01 (1.00-1.01)

.02

Ki-67/MIB-1 PI

1.00 (0.98-1.02)

.74

GTR

0.80 (0.51-1.26)

.35

Chemotherapy

0.14 (0.08-0.23)

Radiation

<.001

0.04

.84

Corticosteroids

1.55 (0.90-2.65)

.11

0.14

.71

Slower growth

0.74 (0.48-1.12)

.15

7.79

.005

M AN U

Age

RI PT

(95% CI)

p-value

Abbreviations: CI, confidence interval; KPS, Karnofsky performance status; PI, Proliferation

AC C

EP

TE D

Index, GTR, Gross total resection.

ACCEPTED MANUSCRIPT Supplementary table 2: Multivariable cox regression analysis with test of proportional hazards assumption (Schoenfeld residuals test) Multivariable cox regression model Variable

Hazard ratio

Schoenfeld residuals test

p-value

Chi-

(95% CI)

p-value

square

.13

KPS (10 points)

0.77 (0.63-0.93)

.008

Preoperative volume

1.00 (0.99-1.01)

.88

GTR

0.72 (0.43-1.21)

.22

Chemotherapy

0.27 (0.14-0.52)

<.001

Radiation

0.16 (0.06-0.41)

<.001

Slower growth

0.86 (0.56-1.33)

.50

<.01

.93

2.10

.14

.74

.39

1.85

.17

.23

.63

.20

.66

12.23

<.001

SC

1.02 (1.00-1.04)

M AN U

Age

RI PT

statistic

Abbreviations: CI, confidence interval; KPS, Karnofsky performance status; GTR, Gross total

AC C

EP

TE D

resection.

ACCEPTED MANUSCRIPT Supplementary Table 3: Distribution of clinical variables across survival groups with measures of association Variable

Two-year

P-value

Statistical Test

survival Yes

(n=85)

(n=21)

Age (years)

65

62

.12

Rank-sum

KPS (score)

70

90

.003

Rank-sum

Preoperative volume (mL)

30.5

16.2

.10

Rank-sum

Ki-67/MIB1 (%)

12.5

15.9

.26

Rank-sum

Male

58

14

Female

27

7

14

6

71

15

23

0

62

21

10

0

75

21

62

14

23

7

Slow growth

38

15

Fast growth

47

6

Yes Chemotherapy (n)

No Yes

Radiation (n)

No

No Yes

EP

Growth rate (n)

TE D

Yes GTR (n)

.89

χ2

.22

Fisher’s exact

.006

Fisher’s exact

.21

Fisher’s exact

.57

χ2

.03

χ2

M AN U

Corticosteroids (n) No

SC

Gender (n)

RI PT

No

AC C

Numbers in italic are median values. Abbreviations: KPS, Karnofsky performance status; GTR, Gross total resection.

ACCEPTED MANUSCRIPT

Results with patients divided in groups based on linear radial growth Supplementary table 4: Results of an ordinary multivariable Cox regression model with growth groups based on assumption of linear radial growth pattern. Multivariable cox regression model Hazard ratio

p-value

Chi-

p-value

RI PT

Variable

Schoenfeld residuals test

(95% CI)

square

statistic

1.02 (1.00-1.04)

.13

KPS (10 points)

0.77 (0.63-0.94)

.009

Preoperative volume

1.00 (0.99-1.01)

.82

GTR

0.73 (0.43-1.22)

.23

Chemotherapy

0.27 (0.14-0.52)

<.001

Radiation

0.16 (0.06-0.42)

Slower growth

0.85 (0.55-1.31)

.02

.90

1.95

.16

SC

Age

.60

2.05

.15

.24

.62

<.001

.24

.62

.46

8.53

.004

M AN U

.27

Abbreviations: CI, confidence interval; KPS, Karnofsky performance status; GTR, Gross total

TE D

resection.

Supplementary table 5: Results of an extended multivariable Cox regression model with growth groups based on assumption of linear radial growth pattern. Hazard ratio (95% CI)

P-value

1.02 (1.00-1.04)

.12

0.76 (0.63-0.93)

.008

Preoperative volume

1.00 (0.99-1.01)

.96

GTR

0.72 (0.43-1.22)

.22

Chemotherapy

0.27 (0.14-0.52)

<.001

Radiation

0.15 (0.06-0.39)

<.001

Slower growth (<12 months)

1.37 (0.77-2.42)

.28

Slower growth (≥12 months)

0.48 (0.25-0.90)

.02

Age

AC C

KPS (10 points)

EP

Variable

Abbreviations: CI, confidence interval; KPS, Karnofsky performance status; GTR, Gross total resection.

ACCEPTED MANUSCRIPT Supplementary table 6: Odds ratios for survival above two years from a multiple logistic

Variable

Odds ratio (95% CI)

P-value

Age

0.97 (0.92-1.02)

.18

KPS (10 points)

1.49 (0.90-2.47)

.12

GTR

1.25 (0.38-4.09)

.71

Slower growth

3.90 (1.18-12.87)

.03

RI PT

regression analysis*, with growth groups based on assumption of linear radial growth.

Abbreviations: CI, confidence interval; KPS, Karnofsky performance status; GTR, Gross total resection.

AC C

EP

TE D

M AN U

SC

*Including only patients receiving both chemotherapy and radiotherapy (n=82)

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT

Conflicts of interest Ole Solheim is an unpaid member of a national advisory committee on treatment guidelines for brain tumors. Asgeir S. Jakola is an unpaid member of the Swedish National Brain Tumor

AC C

EP

TE D

M AN U

SC

RI PT

Trialist Group. The other authors disclose no potential conflicts of interest.