Skip to main content

Recurrence pattern analysis after [68Ga]-DOTATATE-PET/CT -planned radiotherapy of high-grade meningiomas

Abstract

Background

The aim of the present study was to evaluate the influence of the applied safety margins of modern intensity-modulated radiotherapy (IMRT) in patients with high-grade meningiomas on local control and recurrence patterns.

Methods

Twenty patients with a neuropathological diagnosis of a high-grade meningioma (WHO°II or °III) treated with adjuvant or definitive radiotherapy between 2010 and 2015 were included in the present retrospective analysis. All patients were planned PET-based. Recurrence patterns were assessed by means of MRI and/or DOTATATE-PET/computertomography (CT).

Results

The median follow-up was 31.0 months [95% confidence interval (CI): 20.1–42.0] and the progression-free survival (PFS) after 24 months was 87.5%. Overall, four patients had a local recurrence of their meningioma. Of these, three were located in field according to the prior radiotherapy treatment region, while only one patient had a distant relapse. There were no independent factors influencing progression-free or overall survival (OS).

Conclusion

After radiotherapy (RT), patients with atypical or anaplastic meningiomas still have a defined risk of tumor recurrence. The aim of the present study was to examine mono-institutional data concerning target volume definition and recurrence patterns after radiotherapy of high-grade meningiomas as there are limited data available. Our data suggest that extended safety margins are necessary to achieve a favorable local control for high-grade meningiomas.

Background

Meningiomas account for 20–30% of all primary intracranial neoplasms and represent the most common intracranial tumors in adults [1,2,3,4]. High-grade meningiomas show an excessive mitotic index on histopathological examination [5]. Additional criteria for the diagnosis of atypical meningiomas are brain invasion or three of the five following histopathological aspects: prominent nucleoli, high cellularity, small cells, spontaneous necrosis or sheeting, i.e. loss of whorling or fascicular architecture [6, 7]. Overall, high-grade meningiomas (WHO (World Health Organisation) °II and °III) show a significantly more aggressive behavior and poorer outcome as compared to low-grade meningiomas [8,9,10,11,12,13,14].

A multimodal treatment approach with a combination of surgery and radiotherapy (RT) is nowadays considered to be the treatment of choice [8, 15]. Due to the frequent adhesion to neurological structures such as the optical nerve, optical chiasm or brainstem, gross total resections remain challenging [14]. Therefore, postoperative RT is recommended for most cases of atypical meningiomas WHO grade II and all anaplastic meningiomas WHO grade III. A large multicenter analysis with more than 2000 patients by Wang et al. concludes that adjuvant radiotherapy after subtotal resection of an adjuvant meningioma significantly improves the overall survival while it does not after gross total resection [16]. As Condra et al. pointed out, high-grade meningiomas have a poorer outcome as compared to low-grade meningiomas and benefit from a more aggressive treatment including postoperative radiotherapy [17], as RT is known to significantly improve local control [12]. Over the past decades, radiotherapy techniques improved substantially, as new modalities such as intensity modulated radiotherapy (IMRT) or the image-guided application of radiotherapy (IGRT) using robotic patient positioning couches increasing the precision and accuracy of radiotherapy [18]. Thus, radiotherapy has become a more valuable treatment option in the management of high-grade meningiomas.

While the indication for postoperative RT in high-grade meningiomas appears undoubted, the appropriate target volume definition and radiation dose remain a matter of debate. We present a mono-institutional retrospective analysis on the recurrence patterns of high-grade meningiomas after RT with a special emphasis on radiation dose and safety margins of target volumes.

Materials and methods

Patient selection

Patients with histopathologically proven atypical or anaplastic meningioma who underwent RT at our department from 05/2010 to 09/2015 were included in this retrospective study. Even if patients did not have prior surgery within 12 weeks before radiotherapy, there was a former histopathological result confirming a high-grade meningioma. Patients were excluded if they had prior radiosurgical treatment or EBRT at the same site. Altogether 20 patients with atypical or anaplastic meningioma were included in this analysis. All of them were initially PET-positive and therefore were planned DOTATATE-based. Fourteen patients had prior surgery, while six patients were treated with definitive radiotherapy. Patients’ and tumor characteristics are shown in Table 1.

Table 1 Patient characteristics, n = 20 (PTVhom = PTV homogenous, PTVext = PTVextended, PTVboost = PTV simultaneous integrated boost, CTVhom = CTV homogenous, CTVext = CTV extended)

All patients gave their written informed consent for the treatment. This retrospective analysis was approved by the ethics committee of the LMU Munich on record number 545–16. There was no experimental research on humans or animals performed or reported. The declaration of Helsinki has been obeyed in all points.

Treatment and follow-up

A [68Ga]-DOTATATE-PET/CT and a Gadolinium-enhanced magnetic resonance imaging (MRI) of the brain were performed and fused with the treatment planning CT to delineate the target volume. To ensure reproducibility patients were immobilized with a thermoplastic mask system. For critically located lesions, a double-layered thermoplastic mask system was used to minimize setup uncertainties. Treatment planning was performed using the Oncentra® treatment planning system (OTP MasterPlan®, Elekta, Crawley, UK) for 3D-conformal RT and Hyperion® for IMRT which employs constrained optimization using a Monte Carlo dose algorithm [19]. Intensity-modulated radiotherapy (IMRT) was used if adjacent critical organs at risk structures were present. Organs at risk (OAR) constraints were chosen according to conservative estimates given by QUANTEC [20] - the constraints for maximum point doses were 54Gy for optic pathway structures including optic chiasm [21] and 54 Gy for the brainstem [22]. The mean cochlear dose was optimized to be lower than 45 Gy. Planning target volume (PTV) was defined as gross tumor volume (GTV) for patients with macroscopic tumor or resection cavity for patients with prior surgery plus a 15 mm isotropic margin for clinical target volume (CTV) with an additional 3–5 mm PTV expansion. Individual adaptions were made and are shown in Table 1. GTV included the contrast enhancing lesion in T1w + Gd MRI and was adapted to the DOTATATE-enhancement to detect tumoral dural tails or bone infiltration. All patients with prior resection were planned on the resection cavity,

The outcome was evaluated on a regular basis (first time three months after RT, later once per year) using an MRI of the brain and/or DOTATATE-PET-CT in case of suspicious MRI findings. Similarly to the study of Lee et al. recurrence of the tumor was defined as “in-field” if more than 80% of the tumor were located within the prescribed 95%-isodose [23]. A “marginal” recurrence was present if 20–80% were inside the 95%-isodose surface. And any other recurrence was defined as “ex-field”. If there was multifocal recurrence, the tumor volume that was most distant to the initial tumor site was taken as a reference.

Statistics

Overall survival (OS) as well as progression-free survival (PFS) and local progression-free survival (LPFS) were measured from the beginning of RT to progression, death or respectively the date of last follow-up. For the latter, patients who died because of medical co-morbidities or had a distant relapse were censored. The Kaplan-Meier method was used for survival analysis. 95% confidence intervals (CI) were calculated using the associated estimated standard errors. Survival estimates were compared using the log-rank test. A Cox regression analysis was performed to identify factors influencing overall survival or progression-free survival. P-values were considered as significant at ≤0.05.

Results

A summary of baseline patient, tumor and treatment characteristics is shown in Table 1. Median age was 59.7 years (range 26–79 years). Sixteen patients had a meningioma grade II and four patients had a meningioma grade III. Fourteen patients underwent surgery before RT (within ≤12 weeks). Four patients had a relevant time delay to the start of RT due to various reasons (refusal, comorbidities). Two patients had no surgery prior to RT, as the tumor volume was too extensive. Surgery resulted in a gross total resection in 10 cases, subtotal in three and a debulking resection in one case. Grading for the extent of tumor resection was determined according to the study of Simpson et al. [24]. Nine patients had a Simpson-grade I resection, one patient a grade II resection, three had a grade IV resection and one had a Simpson-grade V resection. Nine of the patients having surgery within 12 weeks before radiotherapy had undergone at least one other previous resection before the surgery that led to radiotherapy. Tumor localization was frontal (70%), frontoparietal (15%), frontotemporal (5%), occipital (5%) and parietooccipital (5%).

Concerning radiotherapy, median number of fractions was 30 (28–33) with a median dose per fraction of 2.0 Gy (1.8–2.14 Gy) and a median total radiation dose of 60.0 Gy (59.4–60.0 Gy) (Table 1). Moderate hypofractionation (single dose 2.14 Gy) was only exceptionally used as simultaneous integrated boost (SIB) dose in two cases (59.92 Gy cumulative SIB dose). All patients had a pre-radiotherapeutic [68Ga]-DOTATATE-PET-CT. Mean tumor maximum standardized uptake volume (SUVmax) was 9.76 (0–25.3). Fig. 1 shows a patient with a right frontal meningioma where the additional [68Ga]-DOTATATE-PET-CT shows an impressive infiltration of the skull base which was not unequivocally visualized by postoperative MRI.

Fig. 1
figure 1

Example of a patient with a right frontal meningioma s/p subtotal resection while postoperative MRI does not unequivocally show a tumor residual but radiologists describe postoperative granulation tissue. The PET/CT scan nicely visualizes the bone involvement, on the right the IMRT plan including PTV and 95%/90%/80%/50%/30%-isodoses, respectively

The median cumulative margin from gross tumor volume (GTV) to planning target volume (PTV) was 20 mm at maximum, in median 15 mm to generate the CTV and additional 4 mm in median for PTV. In five patients, the pre-operative volume was mainly used to define the adapted clinical target volume, in all other patients either remaining macroscopic tumor or the resection cavity was used. Six patients had a concept with a simultaneous integrated boost (SIB). We considered the high-dose volume as the PTVboost in case of a SIB and the surrounding PTV-volume as PTVextended (PTVext). In cases without SIB the PTV was named PTVhomogeneous (PTVhom). For PTVboost the GTV was expanded 3 mm in median.

Four of the 20 patients had a relapse of their meningioma. Regarding recurrence patterns, two patients (10%) with a recurrence after radiotherapy had an in-field recurrence concerning the PTVhom. One patient (5%) had radiotherapy with a simultaneous integrated boost concept whose recurrence was located in field of the PTVext but a marginal recurrence regarding the PTVboost. Another patient developed a distant relapse of the tumor, which may be considered as a second meningioma without any relation to the primary lesion. Regarding the in-field relapses, all patients had macroscopic tumor at the time of RT: in one case, there was a debulking resection prior to radiotherapy, in another case there was a relapse of the tumor after surgery and in the third case, there was no resection possible, as the tumor, volume was considered too large. One recurrence occurred in a patient with an anaplastic meningioma, the other three occurred in patients with atypical meningioma. There is no statistical significance, which is due to the small number of only four patients having a recurrence. In two cases, recurrence was diagnosed by MRI and DOTATATE-PET/CT. In the two other cases, recurrences were also diagnosed by MRT and PET, but surgery was performed afterwards so that there is also a histopathological proof for the recurrence.

Median follow-up was 31.0 months [95%-CI: 20.1–42.0]. Median overall survival was 64.7 months [95%-CI: 52.6–76.8] (Fig. 2). Two patients died shortly after treatment, one of them had a relapse of the tumor. Both deaths were not related to the meningioma. Local progression free survival was 87.5% after 24 months and 70% after 36 months (Fig. 2).

Fig. 2
figure 2

Local progression-free survival (LPFS) and overall survival (OS) [Kaplan-Meier method]

Patients presenting with a local recurrence had a median margin from GTV to CTV of 12.5 mm plus another 4 mm margin for PTV (either PTVext or PTVhom). In one patient with a local relapse who had a SIB concept, the GTV was planned as boost volume without any additional safety margin.

In one case with a recurrence, there were relatively small safety margins from GTV to PTV (5 mm in total), the other three patients presenting with local relapse adequate margins of at least 15 mm were identified. Therefore, the detected tumor relapses would not have been avoided by using larger margins. Only in the case of the SIB-concept, a larger safety margin to the PTVboost would have been advantageous but was not possible because of the surrounding organs at risk (optical system).

Overall, radiotherapeutic toxicity was low. Despite mild radiodermatitis Grade I-II (n = 9), there were no higher graded toxicities such as radiation necrosis. One patient suffered from a transitory severe hyperglycemia associated to steroid medication given due to increased intracranial pressure.

On univariate analysis for categorical variables (Table 2), no significant factors influencing OS or PFS were identified (age < 50 years, WHO grade, SIB or non-SIB-concept, type of resection (total vs. subtotal/biopsy), histology or Simpson grade of resection). For continuous variables such as age, SUVmax, time between DOTATATE-PET and treatment beginning, timing of RT (post-operative versus salvage), size of the PTVhom/ext no influence on PFS or OS could be detected as well. All patients were DOTATATE-planned and there was a slight trend that patients with a high SUVmax were more likely to have a relapse of the tumor reflecting macroscopic tumor at the beginning of the treatment.

Table 2 Cox regression analysis on potentially prognostic factors and their impact on overall and progression-free survival, n = 20, HR = hazard ratio

Discussion

The aim of the present study was to examine mono-institutional data concerning target volume definition and recurrence patterns after radiotherapy of high-grade meningiomas as there are limited data available. Table 3 shows an overview of the existing data in comparison to our study.

Table 3 Dose and Safety Margins (*excessive mitotic figures without other features of frank malignancy)

Compared to the existing literature this analysis shows a favorable local control rate for high-grade meningioma after radiotherapy with a total dose of 60 Gy and a safety margin of 15 mm for CTV and additional 4 mm for PTV. In the present analysis, most recurrences were observed in field in patients with macroscopic tumors. A local dose escalation could probably improve the local control rates and should be evaluated in further studies. In comparison with other studies we could not identify factors which influenced overall or progression-free survival.

In contrast to other studies, all treatments of the present analysis were planned by using an MRI and a [68Ga]-DOTATATE-PET/CT fusion. Usually, target delineation is mainly based on contrast-enhanced MRIs (pre- and postoperative) only [25, 26]. Additional [68Ga]-DOTATATE-PET/CT scanning can provide valuable information on bone infiltration or dural tails [27].

Goyal et al. noticed that atypical meningioma benefit from gross total resection (Simpson Grades I-III), as gross total resection is associated with better local control rates, but he concludes that the role of postoperative RT remains unclear [28]. In contrast, Milosevic et al. recommend immediate RT after initial surgery for high-grade meningiomas [29]. Choi et al. conclude with the fact that postoperative radiotherapy could improve local control in patients with high-grade meningiomas after incomplete surgical resection and emphasize that a gross total resection is the most important factor for local control [30]. In contrast to these studies, Champeaux et al. failed to demonstrate a significant improvement in different clinical outcomes after RT for meningioma grade II [31]. We could not identify a difference between patients treated post-operative or salvage, which is likely due to our small number of patients.

Goldsmith et al. suggested a radiation dose of 60 Gy for high-grade meningiomas [32]. This suggestion is based on their retrospective analysis of 140 patients whereas 23 of them had a malignant type of meningioma and were treated in median with 54 Gy (range 44.62 to 69.26Gy) [32]. Katz et al. performed an analysis with an accelerated hyperfractionated RT for patients with atypical and anaplastic meningioma [33]. Thirty-six patients were treated with 60 Gy in 1.5 Gy single dose twice per day [33]. Local control rate was significantly poorer with 45% in comparison to less aggressive treatment schedules but caused significantly higher toxicity [33]. This study concluded that 50–60 Gy delivered with once-daily fractionation seems feasible as it looks unlikely that more aggressive RT could improve outcome [33]. Combs et al. suggest on the one hand that PTV has to be enlarged to the resection cavity plus a safety margin of 1–2 cm and on the other hand advocate for a dose escalation to 60–66 Gy [8].

The results of the EORTC-trial 22,042–26,042 are eagerly awaited and will highlight the role of dose escalation in this setting. In this trial, patients with Simpson grades 1 to 3 receive standard postoperative RT, while patients with a higher grade receive an additional boost. There are also few data indicating that hypofractionation might be an effective option. Maranzano et al. describe their long-term results of moderate hypofractionated stereotactic radiotherapy for intracranial meningiomas [34]. In this study 77 patients were treated with a median volume of 23 cm3 while dose was prescribed either in 15 × 3 Gy or 14 × 3 Gy [34]. The authors conclude that moderate hypofractionation had a good outcome and was tolerated well [34] whereas a longer follow up and further trials should be performed to corroborate these findings.

However, any kind of dose escalation or hypofractionation should be done carefully, considering the adjacent organs at risk and their dose tolerance. Bostrom et al. report about a patient developing severe radiation necrosis due to the RT in several lesions (some of them treated hypofractionated) [35].

Considering the planning process and the safety margins an analysis by Press et al. found that meningioma grade II treated with conformal IMRT and limited safety margins < 1 cm, did not lead to a higher rate of recurrence [36]. Six of 46 patients had a tumor relapse [36]. The study confirms the findings of the present analysis, as most relapses occurred “in-field”. Press et al. found 5 of 6 recurrences within the prior radiation volume and one was combined in-field and marginal [36]. A safety margin of 5 mm for CTV and 3 mm for PTV was applied in this patient cohort [36]. Press et al. postulate that as none of their patients relapsed marginally safety margins of < 1 cm might be sufficient [36]. In contrast to the present study, the majority of patients did not have macroscopic tumor. Taken together, the margins for grade II meningiomas cannot be transferred unreflectedly to meningiomas grade III. Regarding the existing studies a limited safety margin might be an option for some atypical meningiomas grade II, but for anaplastic meningiomas grade III a limited safety margin might be harmful and could lead to tumor relapse. The present data suggest that safety margins for anaplastic meningiomas have to be sufficiently large.

Over the last decades, modern radiotherapy techniques (IMRT or volumetric modulated arc therapy (VMAT)) were introduced in the treatment of meningiomas. Anvari et al. conclude that utilizing high-technology equipment and new techniques might improve the outcome after RT [37].

Madani et al. describe that dose-painting intensity-modulated proton therapy (IMPT) using a SIB is a feasible therapy option with excellent dose coverage with minimal or no dose to brain, brainstem or optical system [38]. A study by Simon concludes that carbon-ion RT could be the preferred therapy option [39]. Similarly, Harrabi et al. describe that proton beam RT shows dosimetric advantages over conventional radiotherapy that might be essential for neurologic function [40]. Therefore, future concepts might include carbon-ion or proton RT.

The present study is limited due to its retrospective nature and the small number of patients. Obviously the number of or presented patients is smaller than in most of the mentioned studies in Table 3. In contrast you have to consider that in the other mentioned studies, there are all kind of meningioma, not only high-grade meningiomas as presented here.

The follow-up time has a wide range, as some patients had to be censored very early. Furthermore, a multivariate analysis was not reasonably feasible due to the small number of cases, which is another weakness of the analysis.

Conclusion

Even following postoperative radiotherapy, high-grade meningiomas relapsed frequently “in-field” of the prior target volume.

We consider postoperative radiotherapy after resection of a high-grade meningioma essential to minimize the risk of local failure, even if a gross total resection of the tumor was performed. Especially patients with macroscopic tumor should receive postoperative RT immediately. Patients who cannot be treated with a resection of the tumor at all should receive a definitive radiotherapy.

Concerning target volume delineation, the present data show a good local control with the applied margins and confirm the common used safety margins. We suggest a CTV margin of 15 mm starting from the GTV and additional 3–5 mm for the PTV, depending on the available image-guidance.

It would be valuable to have future studies including a higher number of patients to evaluate which patients would benefit from a dose escalation, e.g. in form of a simultaneous integrated boost, or a CTV margin reduction.

Abbreviations

CI:

Confidence interval

CT:

Computer tomography

CTV:

Clinical target volume

GTV:

Gross tumor volume

IGRT:

Image-guided application of radiotherapy

IMRT:

Intensity modulated radiotherapy

LPFS:

Local progression-free survival

MRI:

Magnetic resonance imaging

OAR:

Organs at risk

OS:

Overall survival

PFS:

Progression-free survival

PTV:

Planning target volume

PTVext:

Planning target volume extended

PTVhom:

Planning target volume homogenous

RT:

Radiotherapy

SIB:

Simultaneous integrated boost

SUVmax:

Maximum standardized uptake value

VMAT:

Volumetric modulated arc therapy

WHO:

World Health Organisation

References

  1. Claus EB, Bondy ML, Schildkraut JM, Wiemels JL, Wrensch M, Black PM. Epidemiology of intracranial meningioma. Neurosurgery. 2005;57(6):1088–95. discussion 1088-1095

    Article  PubMed  Google Scholar 

  2. Rogers CL, Perry A, Pugh S, Vogelbaum MA, Brachman D, McMillan W, Jenrette J, Barani I, Shrieve D, Sloan A, Bovi J, Kwok Y, Burri SH, Chao ST, Spalding AC, Anscher MS, Bloom B, Mehta M. Pathology concordance levels for meningioma classification and grading in NRG oncology RTOG trial 0539. Neuro-Oncology. 2016;18(4):565–74. https://doi.org/10.1093/neuonc/nov247.

    Article  PubMed  Google Scholar 

  3. Rogers L, Mehta M. Role of radiation therapy in treating intracranial meningiomas. Neurosurg Focus. 2007;23(4):E4. https://doi.org/10.3171/FOC-07/10/E4.

    Article  PubMed  Google Scholar 

  4. Carlson ML, Glasgow AE, Jacob JT, Habermann EB, Link MJ. The short-term and intermediate-term risk of second neoplasms after diagnosis and treatment of unilateral vestibular schwannoma: analysis of 9460 cases. Int J Radiat Oncol Biol Phys. 2016;95(4):1149–57. https://doi.org/10.1016/j.ijrobp.2016.03.005.

    Article  PubMed  Google Scholar 

  5. Perry A, Scheithauer BW, Stafford SL, Lohse CM, Wollan PC. “Malignancy” in meningiomas. Cancer. 1999;85(9):2046–56. https://doi.org/10.1002/(SICI)1097-0142(19990501)85:9<2046::AID-CNCR23>3.0.CO;2-M.

    Article  PubMed  CAS  Google Scholar 

  6. Goldbrunner R, Minniti G, Preusser M, Jenkinson MD, Sallabanda K, Houdart E, von Deimling A, Stavrinou P, Lefranc F, Lund-Johansen M, Moyal EC, Brandsma D, Henriksson R, Soffietti R, Weller M. EANO guidelines for the diagnosis and treatment of meningiomas. Lancet Oncol. 2016;17(9):e383–91. https://doi.org/10.1016/S1470-2045(16)30321-7.

    Article  PubMed  Google Scholar 

  7. Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P, Ellison DW. The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol. 2016;131(6):803–20. https://doi.org/10.1007/s00401-016-1545-1.

    Article  PubMed  Google Scholar 

  8. Combs SE, Ganswindt U, Foote RL, Kondziolka D, Tonn JC. State-of-the-art treatment alternatives for base of skull meningiomas: complementing and controversial indications for neurosurgery, stereotactic and robotic based radiosurgery or modern fractionated radiation techniques. Radiat Oncol. 2012;7:226. https://doi.org/10.1186/1748-717X-7-226.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Maier H, Ofner D, Hittmair A, Kitz K, Budka H. Classic, atypical, and anaplastic meningioma: three histopathological subtypes of clinical relevance. J Neurosurg. 1992;77(4):616–23. https://doi.org/10.3171/jns.1992.77.4.0616.

    Article  PubMed  CAS  Google Scholar 

  10. Wiemels J, Wrensch M, Claus EB. Epidemiology and etiology of meningioma. J Neuro-Oncol. 2010;99(3):307–14. https://doi.org/10.1007/s11060-010-0386-3.

    Article  Google Scholar 

  11. Backer-Grondahl T, Moen BH, Torp SH. The histopathological spectrum of human meningiomas. Int J Clin Exp Pathol. 2012;5(3):231–42.

    PubMed  PubMed Central  Google Scholar 

  12. Kaur G, Sayegh ET, Larson A, Bloch O, Madden M, Sun MZ, Barani IJ, James CD, Parsa AT. Adjuvant radiotherapy for atypical and malignant meningiomas: a systematic review. Neuro-Oncology. 2014;16(5):628–36. https://doi.org/10.1093/neuonc/nou025.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Modha A, Gutin PH. Diagnosis and treatment of atypical and anaplastic meningiomas: a review. Neurosurgery. 2005;57(3):538–50. discussion 538-550

    Article  PubMed  Google Scholar 

  14. Kumar N, Kumar R, Khosla D, Salunke PS, Gupta SK, Radotra BD. Survival and failure patterns in atypical and anaplastic meningiomas: a single-center experience of surgery and postoperative radiotherapy. J Cancer Res Ther. 2015;11(4):735–9. https://doi.org/10.4103/0973-1482.151426.

    Article  PubMed  CAS  Google Scholar 

  15. De Monte F. Current management of meningiomas. Oncology (Williston Park). 1995;9(1):83–91. 96; discussion 96, 99-101

    CAS  Google Scholar 

  16. Wang C, Kaprealian TB, Suh JH, Kubicky CD, Ciporen JN, Chen Y, Jaboin JJ. Overall survival benefit associated with adjuvant radiotherapy in WHO grade II meningioma. Neuro-Oncology. 2017;19(9):1263–70. https://doi.org/10.1093/neuonc/nox007.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Condra KS, Buatti JM, Mendenhall WM, Friedman WA, Marcus RB Jr, Rhoton AL. Benign meningiomas: primary treatment selection affects survival. Int J Radiat Oncol Biol Phys. 1997;39(2):427–36.

    Article  PubMed  CAS  Google Scholar 

  18. Combs SE, Nusslin F, Wilkens JJ. Individualized radiotherapy by combining high-end irradiation and magnetic resonance imaging. Strahlenther Onkol. 2016;192(4):209–15. https://doi.org/10.1007/s00066-016-0944-5.

    Article  PubMed  Google Scholar 

  19. Kantz S, Sohn M, Troeller A, Reiner M, Weingandt H, Alber M, Belka C, Ganswindt U. Impact of MLC properties and IMRT technique in meningioma and head-and-neck treatments. Radiat Oncol. 2015;10:184. https://doi.org/10.1186/s13014-015-0447-z.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Marks LB, Yorke ED, Jackson A, Ten Haken RK, Constine LS, Eisbruch A, Bentzen SM, Nam J, Deasy JO. Use of normal tissue complication probability models in the clinic. Int J Radiat Oncol Biol Phys. 2010;76(3 Suppl):S10–9. https://doi.org/10.1016/j.ijrobp.2009.07.1754.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Mayo C, Martel MK, Marks LB, Flickinger J, Nam J, Kirkpatrick J. Radiation dose-volume effects of optic nerves and chiasm. Int J Radiat Oncol Biol Phys. 2010;76(3 Suppl):S28–35. https://doi.org/10.1016/j.ijrobp.2009.07.1753.

    Article  PubMed  Google Scholar 

  22. Mayo C, Yorke E, Merchant TE. Radiation associated brainstem injury. Int J Radiat Oncol Biol Phys. 2010;76(3 Suppl):S36–41. https://doi.org/10.1016/j.ijrobp.2009.08.078.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Lee SW, Fraass BA, Marsh LH, Herbort K, Gebarski SS, Martel MK, Radany EH, Lichter AS, Sandler HM. Patterns of failure following high-dose 3-D conformal radiotherapy for high-grade astrocytomas: a quantitative dosimetric study. Int J Radiat Oncol Biol Phys. 1999;43(1):79–88.

    Article  PubMed  CAS  Google Scholar 

  24. Simpson D. The recurrence of intracranial meningiomas after surgical treatment. J Neurol Neurosurg Psychiatry. 1957;20(1):22–39.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Guo L, Wang G, Feng Y, Yu T, Guo Y, Bai X, Ye Z. Diffusion and perfusion weighted magnetic resonance imaging for tumor volume definition in radiotherapy of brain tumors. Radiat Oncol. 2016;11(1):123. https://doi.org/10.1186/s13014-016-0702-y.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Martin F, Magnier F, Berger L, Miroir J, Chautard E, Verrelle P, Lapeyre M, Biau J. Fractionated stereotactic radiotherapy of benign skull-base tumors: a dosimetric comparison of volumetric modulated arc therapy with Rapidarc (R) versus non-coplanar dynamic arcs. Radiat Oncol. 2016;11:58. https://doi.org/10.1186/s13014-016-0632-8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Rachinger W, Stoecklein VM, Terpolilli NA, Haug AR, Ertl L, Poschl J, Schuller U, Schichor C, Thon N, Tonn JC. Increased 68Ga-DOTATATE uptake in PET imaging discriminates meningioma and tumor-free tissue. J Nucl Med. 2015;56(3):347–53. https://doi.org/10.2967/jnumed.114.149120.

    Article  PubMed  CAS  Google Scholar 

  28. Goyal LK, Suh JH, Mohan DS, Prayson RA, Lee J, Barnett GH. Local control and overall survival in atypical meningioma: a retrospective study. Int J Radiat Oncol Biol Phys. 2000;46(1):57–61.

    Article  PubMed  CAS  Google Scholar 

  29. Milosevic MF, Frost PJ, Laperriere NJ, Wong CS, Simpson WJ. Radiotherapy for atypical or malignant intracranial meningioma. Int J Radiat Oncol Biol Phys. 1996;34(4):817–22.

    Article  PubMed  CAS  Google Scholar 

  30. Choi Y, Lim DH, Jo K, Nam DH, Seol HJ, Lee JI. Efficacy of postoperative radiotherapy for high grade meningiomas. J Neuro-Oncol. 2014;119(2):405–12. https://doi.org/10.1007/s11060-014-1507-1.

    Article  Google Scholar 

  31. Champeaux C, Wilson E, Shieff C, Khan AA, Thorne L. WHO grade II meningioma: a retrospective study for outcome and prognostic factor assessment. J Neuro-Oncol. 2016;129(2):337–45. https://doi.org/10.1007/s11060-016-2181-2.

    Article  Google Scholar 

  32. Goldsmith BJ, Wara WM, Wilson CB, Larson DA. Postoperative irradiation for subtotally resected meningiomas. A retrospective analysis of 140 patients treated from 1967 to 1990. J Neurosurg. 1994;80(2):195–201. https://doi.org/10.3171/jns.1994.80.2.0195.

    Article  PubMed  CAS  Google Scholar 

  33. Katz TS, Amdur RJ, Yachnis AT, Mendenhall WM, Morris CG. Pushing the limits of radiotherapy for atypical and malignant meningioma. Am J Clin Oncol. 2005;28(1):70–4.

    Article  PubMed  Google Scholar 

  34. Maranzano E, Draghini L, Casale M, Arcidiacono F, Anselmo P, Trippa F, Giorgi C. Long-term outcome of moderate hypofractionated stereotactic radiotherapy for meningiomas. Strahlenther Onkol. 2015;191(12):953–60. https://doi.org/10.1007/s00066-015-0915-2.

    Article  PubMed  Google Scholar 

  35. Bostrom JP, Seifert M, Greschus S, Schafer N, Glas M, Lammering G, Herrlinger U. Bevacizumab treatment in malignant meningioma with additional radiation necrosis. An MRI diffusion and perfusion case study. Strahlenther Onkol. 2014;190(4):416–21. https://doi.org/10.1007/s00066-013-0505-0.

    Article  PubMed  CAS  Google Scholar 

  36. Press RH, Prabhu RS, Appin CL, Brat DJ, Shu HK, Hadjipanayis C, Olson JJ, Oyesiku NM, Curran WJ, Crocker I. Outcomes and patterns of failure for grade 2 meningioma treated with reduced-margin intensity modulated radiation therapy. Int J Radiat Oncol Biol Phys. 2014;88(5):1004–10. https://doi.org/10.1016/j.ijrobp.2013.12.037.

    Article  PubMed  Google Scholar 

  37. Anvari K, Hosseini S, Rahighi S, Toussi MS, Roshani N, Torabi-Nami M. Intracranial meningiomas: prognostic factors and treatment outcome in patients undergoing postoperative radiation therapy. Adv Biomed Res. 2016;5:83. https://doi.org/10.4103/2277-9175.182214.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Madani I, Lomax AJ, Albertini F, Trnkova P, Weber DC. Dose-painting intensity-modulated proton therapy for intermediate- and high-risk meningioma. Radiat Oncol. 2015;10:72. https://doi.org/10.1186/s13014-015-0384-x.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Simon F, Dittmar JO, Brons S, Orschiedt L, Urbschat S, Weber KJ, Debus J, Combs SE, Rieken S. Integrin-based meningioma cell migration is promoted by photon but not by carbon-ion irradiation. Strahlenther Onkol. 2015;191(4):347–55. https://doi.org/10.1007/s00066-014-0778-y.

    Article  PubMed  Google Scholar 

  40. Harrabi SB, Bougatf N, Mohr A, Haberer T, Herfarth K, Combs SE, Debus J, Adeberg S. Dosimetric advantages of proton therapy over conventional radiotherapy with photons in young patients and adults with low-grade glioma. Strahlenther Onkol. 2016;192(11):759–69. https://doi.org/10.1007/s00066-016-1005-9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank Mrs. Sabine Sirges-Szasz and Ms. Vera Muehlberger for their support in contacting patients and/or the patients’ general practitioners for information about progression and survival. There was no writing assistance.

Availability of data and materials

Please contact author for data requests.

Author information

Authors and Affiliations

Authors

Contributions

BZ and MN planned, coordinated and conducted the retrospective analysis. NA helped with data acquisition concerning the nuclear medical data. BZ, UG and MN analyzed the diagnostic imaging data as well as the treatment planning data. BZ provided follow-up data. CM helped providing patient data. BZ, UG, SC, CM, NA, CS, CB and MN drafted the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Maximilian Niyazi.

Ethics declarations

Ethics approval

Approval by the ethics committee of Ludwig-Maximilians-Universität is on record no. 545–16.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

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

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zollner, B., Ganswindt, U., Maihöfer, C. et al. Recurrence pattern analysis after [68Ga]-DOTATATE-PET/CT -planned radiotherapy of high-grade meningiomas. Radiat Oncol 13, 110 (2018). https://doi.org/10.1186/s13014-018-1056-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13014-018-1056-4

Keywords