Next Article in Journal
Slip versus Slop: A Head-to-Head Comparison of UV-Protective Clothing to Sunscreen
Next Article in Special Issue
HSA-Binding Prodrugs-Based Nanoparticles Endowed with Chemo and Photo-Toxicity against Breast Cancer
Previous Article in Journal
Additive Intralesional Interleukin-2 Improves Progression-Free Survival in a Distinct Subgroup of Melanoma Patients with Prior Progression under Immunotherapy
Previous Article in Special Issue
Synergistic Action of Immunotherapy and Nanotherapy against Cancer Patients Infected with SARS-CoV-2 and the Use of Artificial Intelligence
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Combined Fluorescence-Guided Resection and Intracavitary Thermotherapy with Superparamagnetic Iron-Oxide Nanoparticles for Recurrent High-Grade Glioma: Case Series with Emphasis on Complication Management

1
Department of Neurosurgery, University Hospital Münster, 48149 Münster, Germany
2
European Institute for Molecular Imaging (EIMI), University of Münster, 48149 Münster, Germany
3
Biomarkers & Translational Technologies (BTT), Pharma Research & Early Development (pRED), F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
4
Department of Neurosurgery, Haukeland University Hospital, 5021 Bergen, Norway
5
Deparment of Neurosurgery, Hufeland Klinikum, 99974 Mühlhausen, Germany
6
Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149 Münster, Germany
*
Author to whom correspondence should be addressed.
The authors contributed equally to this work.
Cancers 2022, 14(3), 541; https://doi.org/10.3390/cancers14030541
Submission received: 21 December 2021 / Revised: 13 January 2022 / Accepted: 16 January 2022 / Published: 21 January 2022
(This article belongs to the Special Issue Nanoparticle-Based Combination Therapy and Diagnosis for Cancer)

Abstract

:

Simple Summary

Recurrent high-grade gliomas are difficult to treat. Here, we report on our single-center experience in combining fluorescence-guided tumor resection with 5-ALA and local thermotherapy with superparamagnetic iron nanoparticles. In total, 18 patients were operated on and received thermotherapy with or without additional radiotherapy. The median progression-free survival was 5.5 months and median overall survival was 9.5 months. Although no major side effects were observed during active treatment, 72% of the patients developed cerebral edema requiring steroid treatment or even surgical removal of the nanoparticles. In conclusion, the combination of fluorescence-guided resection and intracavitary thermotherapy provides a novel and promising treatment option for improving local tumor control in recurrent high-grade gliomas, but further refinements of the treatment protocol are needed to decrease major side effects.

Abstract

Background: Concepts improving local tumor control in high-grade glioma (HGG) are desperately needed. The aim of this study is to report an extended series of cases treated with a combination of 5-ALA-fluorescence-guided resection (FGR) and intracavitary thermotherapy with superparamagnetic iron oxide nanoparticles (SPION). Methods: We conducted a single-center retrospective review of all recurrent HGG treated with FGR and intracavitary thermotherapy (n = 18). Patients underwent six hyperthermia sessions in an alternating magnetic field and received additional adjuvant therapies on a case-by-case basis. Results: Nine patients were treated for first tumor recurrence; all other patients had suffered at least two recurrences. Nine patients received combined radiotherapy and thermotherapy. The median progression-free survival was 5.5 (95% CI: 4.67–6.13) months and median overall survival was 9.5 (95% CI: 7.12–11.79) months. No major side effects were observed during active treatment. Thirteen patients (72%) developed cerebral edema and more clinical symptoms during follow-up and were initially treated with dexamethasone. Six (33%) of these patients underwent surgical removal of nanoparticles due to refractory edema. Conclusions: The combination of FGR and intracavitary thermotherapy with SPION provides a new treatment option for improving local tumor control in recurrent HGG. The development of cerebral edema is a major issue requiring further refinements of the treatment protocol.

1. Introduction

In spite of guideline-adherent treatment, the prognosis of high-grade gliomas (HGG) is poor [1,2]. Over time, developments such as fluorescence-guided resection (FGR) [3,4], intraoperative neuro-monitoring (IOM) [5,6], intraoperative magnetic resonance imaging (ioMRI) [7,8] and novel treatment methods such as Tumor-Treating Fields (TTF) [9] have led to limited improvements of survival times.
Fluorescence-guided resection with 5-aminolevulinic acid (5-ALA) is currently the only approved and one of the most widely used procedures in adult glioma surgery. The efficacy of 5-ALA-assisted resection and its impact on the degree of resection, progression-free survival (PFS), and overall survival (OS) in primary cases have been demonstrated in numerous studies [3,4,5,10,11]. Stummer et al. identified a significantly higher OS when no residual tumor was visible after surgery (16.7 vs. 11.8 months, p < 0.0001) [12]. Moreover, this method is based on the intracellular accumulation of fluorescent protoporphyrin IX (PpIX). This selective accumulation is the reason for the high sensitivity and specificity of the fluorescence signal [13,14,15,16,17,18], also in the case of recurrent malignant brain tumors [19].
Therapies for tumor recurrence are not well defined, and the overall level of evidence for available treatments is low. Current guideline recommendations include re-resection, re-irradiation, chemotherapy and experimental treatment options [20]. In recent years, several clinical trials have yielded only limited results [21,22]. Even though HGG affect the whole brain, most surgical cases recur locally at the margins of resection cavities [23,24]. Therefore, improving local tumor control is the aim of all local therapy strategies, such as locally applied chemotherapy [25] and photodynamic therapy (PDT) [23,26].
Early clinical trials demonstrated the effectiveness and safety of stereotactic application of superparamagnetic iron oxide nanoparticles (SPION) subjected to an alternating magnetic field (AMF) in combination with irradiation in patients with recurrent HGG [27,28]. SPION with AMF is an approved treatment within the European Union. Hyperthermia generated by SPION has been shown to possibly elicit a potent antitumor immune response [29,30,31] with enhanced infiltration of natural killer (NK) cells, macrophages, dendritic cells and CD4+ and CD8+ T cells [32].
One major technical issue with stereotactic application is the inadequate distribution of SPION [28]. Additionally, with stereotactic procedures, immediate cytoreductive treatment is not possible. With this in mind, we developed a novel technique of administering the particles into the resection cavity immediately after FGR. The main advantage of this method is that the surgeon has optimal control over particle distribution, enabling him to apply larger particle volumes while preventing spillage into the ventricles.
The primary aim of this study is to present our results concerning effectiveness and the outcome, in term of overall survival (OS), with the secondary objective to evaluate the safety of combined FGR and local intracavitary thermotherapy with superparamagnetic iron oxide nanoparticles in an extended series of patients treated at our center.

2. Methods

2.1. Patients

This case series includes all patients treated with a combination of FGR and intracavitary SPION thermotherapy in a single-center academic setting from 2015 to 2020 (Figure 1). All patients with recurrent HGG considered as candidates for surgical resection were offered additional intracavitary thermotherapy as an adjunctive treatment modality. Data analysis was performed retrospectively, with all procedures and analyses being approved by the local ethics committee according to the declaration of Helsinki (2020-531-f-S). Informed consent was obtained from all subjects involved in the study.

2.2. Nanoparticles

As previously reported [32], the semifluid solution MFL AS-1 (NanoTherm®, MagForce AG, Berlin, Germany) containing SPION with an iron concentration of 112 mg/mL was applied during surgery. The fluid is manufactured according to European medical device regulations and has been approved for the treatment of brain tumors since 2011.

2.3. Fluorescence-Guided Resection and Nanoparticle Application

Tumor resection was performed using a standard microsurgical technique. 5-aminolevulinic acid (5-ALA, Gliolan®, Medac GmbH, Wedel, Germany) was applied orally 4 h prior to anesthesia induction at a concentration of 20 mg/kg body weight. The surgical aim was to remove the fluorescent tumor. In cases with a motor-eloquent tumor location, dynamic MEP mapping was used for corticospinal tract mapping. Speech-eloquent tumors were resected under awake conditions for speech monitoring, as described previously [33]. After tumor resection, the cavity wall was coated with a hydroxycellulose mesh (Tabotamp®, Johnson & Johnson Medical GmbH, Ethicon, Norderstedt, Germany), and subsequently SPION were applied to the hydroxycellulose mesh using a 1 mL syringe. Up to three layers of hydroxycellulose and SPION were applied, if possible. Additionally, a closed-end catheter (TK-01, MagForce AG, Berlin, Germany) was led through the particle deposits to allow temperature measurement during treatment in the AMF device (Nano-Activator®, MagForce AG, Berlin, Germany). The bone flap was re-fixed using absorbable, non-metal plates (SonicWeld Rx®, KLS Martin, Tuttlingen, Germany). Metallic body implants up to 40 cm from the tumor, including dental implants and bone flap fixation plates, were removed during surgery or were removed before. Patients in which tumor resection led to opening of the ventricle were not eligible for the application of nanoparticles to decrease the risk of unintended particle distribution within the CSF spaces.
Due to artifacts caused by the SPION, we could not rely on postoperative MRI scans when estimating the extent of resection (EOR). Therefore, we rated EOR as gross total resection (GTR) if all fluorescing tumor tissue was removed, near total resection (NTR) whenever a weak and diffuse fluorescent signal was detected at the end of surgery, and subtotal resection (STR) if a strong and compact fluorescence signal had to be left over, e.g., in eloquent regions. Previous studies have proven a close association between the fluorescence signal and EOR on postoperative MRI [3,5,12].

2.4. Thermotherapy

After surgery, computed tomography (CT) was performed (Figure 2). These images were fused with preoperative MRI scans using the treatment simulation software NanoPlan® (MagForce AG), as published previously [32,34,35,36]. NanoPlan® simulates heat generation as a function of the nanoparticle density and intensity of AMF. However, due to the lack of technical possibilities for measuring local tissue perfusion over the course of a hyperthermia application, this simulation tends to be imprecise. Therefore, temperature measurements must be performed during the treatment session to readjust the simulation. During the first hyperthermia application, a fiber-optic temperature sensor (Optocon, Dresden, Germany) was inserted into the closed-end catheter that was placed during surgery. Thermotherapy was performed in the AMF applicator operating at a frequency of 100 kHz and with field intensities of 2.5–15 kA/m, as published previously [32]. The highest temperature along the thermocatheter course was used to fine-tune the field strength. Two one-hour treatments were scheduled per week. In cases with concomitant radiotherapy, the first thermotherapy session was scheduled 3 days before the start of radiotherapy, while another five sessions were conducted at days 1, 4, 8, 11, 15 ± 1 day. Radiotherapy was performed as previously described [32] and, when planned for the same day, took place within a time interval of 2 h to each thermotherapy session.

2.5. Follow Up

As noted above, the placement of iron nanoparticles does not allow for adequate postoperative imaging with MRI. Patients who live close to our center were followed up with 18F-Fluoro-ethyl-tyrosine (18F-FET)-PET-CT, including post-contrast studies, once every three months. With all other patients, post-contrast CT scans were used for follow-up. If the nanoparticles had to be removed over the course of disease, MRI was again chosen as the optimal imaging modality. In order to further visualize the immunological response during thermotherapy, we conducted a N,N-diethyl-2-[4-(2-fluoroethoxy)phenyl]-5,7-dimethylpyrazolo[1,5-a]pyrimidine-3-acetamide (18F-DPA-714) PET scan for translocator protein (TSPO) in individual cases, as previously described [37].

2.6. Data Analyses

The IBM SPSS Statistics 25.0 package (IBM, Armonk, New York, NY, USA) was used for statistical analyses. Data were analyzed by standard descriptive statistics, using absolute and relative frequencies for categorical variables, median and interquartile range (IQR) for continuous variables, and mean and standard deviation (SD) for metrical variables. The Mann–Whitney U-Test (MWU) was used for ordinal and Fisher’s exact test for categorical variables. Time-to-event analyses were performed using Kaplan–Meier curves and log-rank test. Progression-free survival (PFS) defines the time from the procedure until progression according to modified RANO criteria or death [38]. Overall survival (OS) was defined as the time from procedure to death. A probability value less than 0.05 was considered statistically significant.

3. Results

Eighteen patients with recurrent HGG were treated with FGR and subsequent intracavitary thermotherapy. Eight patients (44%) were female. The median age was 51 years (IQR: 43–61). According to the WHO classification of 2006 [39], sixteen patients (89%) were diagnosed with glioblastoma, and each of the remaining two were diagnosed with anaplastic astrocytoma and anaplastic oligodendroglioma, respectively. Fourteen cases showed IDH wildtype (78%). The MGMT promotor was non-methylated in 13 cases (76%). Nine patients (50%) were treated for first tumor recurrence; all other patients suffered at least two recurrences (Table 1). The median time between initial diagnosis and surgical treatment investigated in this study was 14 months (IQR: 7–45). All patients received thermotherapy twice a week at a median temperature of 47.0 °C (IQR: 44.5–53.3). Nine patients (50%) additionally received concurrent radiotherapy at a dose of 39.6 Gy (5 × 1.8 Gy/week); all other patients did not receive radiotherapy due to dose limitations (Figure 1). Initially, no salvage chemotherapy was administered. During follow-up, one patient received temozolomide and one patient received bevacizumab for palliative edema treatment (Table 2).

3.1. Survival Analysis

Tumor progression was defined according to the modified RANO criteria [38]. The median progression-free survival (PFS) for the study population was 5.5 months (95% CI: 4.67–6.13) after thermotherapy, and median overall survival (OS) was 9.5 months (95% CI: 7.12–11.79) (Figure 3). Survival differences could neither be observed between patients treated for first recurrence and patients treated for second recurrence or later (p = 0.283 for PFS; p = 0.608 for OS) nor in patients who received both thermotherapy and radiotherapy and those having received thermotherapy alone (p = 0.232 for PFS; p = 0.450 for OS).

3.2. Safety and Complications

No major side effects were observed in the immediate perioperative phase or during AMF treatment. However, thirteen patients (72%) developed cerebral edema with clinical symptoms during treatment follow-up (median: 70 days, IQR: 50–105) and were treated with dexamethasone. In six of these thirteen cases (46%), nanoparticles had to be removed surgically due to refractory edema. Four (31%) of these developed impaired surgical site infections after prolonged treatment with steroids (median: 72 days, IQR: 59–116). One patient (No. 4) developed transient mild myelopathy, with cervical MRI suggesting a T2 hyperintense medullary heat injury caused by a small spinal deposit of dislocated nanoparticles (Figure 4).
The mean peak temperature during treatments was 47 °C (IQR: 44.5–53.3). We found no significant association between peak temperature and edema development (p > 0.05). Moreover, no significant correlations between peak temperature and steroid treatment, reoperation, OS, PFS, or time interval to edema development (all p > 0.05) (all p > 0.05) were found. Further analyses did not show any association between additional radiotherapy and the development of edema (p > 0.05).

4. Discussion

In this analysis of an extended case series, we evaluated the effectiveness, safety and technical issues of combined 5-ALA FGR and intracavitary thermotherapy. We found a median PFS of 5.5 months (95% CI: 4.67–6.13) and median OS of 9.5 months (95% CI: 7.12–11.79) after surgery and thermotherapy. In contrast to earlier studies, we did not observe a significant value of re-irradiation in combination with thermotherapy on OS and PFS [32,40,41,42]. Moreover, we found no significant difference between the survival outcomes of patients treated for their first recurrence vs. second recurrence or later.
As for now, there is no standard treatment for patients with recurrent HGG. Recent non-randomized trials show that patients may benefit from repeated resection with a median PFS of 1.9 months and OS of 6.5–12.9 months [43,44,45,46]. Re-irradiation was also associated with a modest survival benefit compared to best supportive care alone. Survival times could be improved when re-irradiation was combined with other treatment modalities (median OS of 8.2 months vs. 12.2 months) [47]. Regarding systemic treatments in recurrent GBM, therapeutic options include salvage chemotherapy with temozolomide, lomustine, bevacizumab or combinations, and regorafenib. A median PFS of 1.5–4.2 months and median OS of 6.0 to 10.6 months were reported [48,49,50,51,52]. Decision making is even more complex in cases of second or later recurrence. In general, thermotherapy is not a novel concept in neuro-oncology. Laser interstitial therapy (LITT), a stereotactic procedure which is limited to smaller lesions, showed comparable survival outcomes for recurrent glioblastoma [53,54]. Recently, our group also published a series of recurrent glioblastoma patients treated with intraoperative open photodynamic therapy after FGR with comparable survival curves [23]. Importantly, in comparison to other modalities, NanoTherm® nanoparticles have been certified according to the European medical device regulations for the treatment of brain tumors since 2011. Moreover, intracavitary nanoparticle application allows one to address more complex tumor architecture and can be directly combined with microsurgical resection.

4.1. Combining FGR with Application of Nanoparticles

In comparison to the classical stereotactic application of nanoparticles [27], its addition to fluorescence-guided resection has several advantages. Cytoreduction using FGR and IOM shows good oncological and functional results [55]. This allows us to provide thermotherapy in addition to the best standard of care. As opposed to stereotactic application of nanoparticles, higher volumes of nanoparticles can be applied without causing elevated intracranial pressure. Moreover, as an open procedure enables the neurosurgeon to distribute the particles more accurately under direct visual control, common problems connected to the stereotactic method including leakage and backwash of the nanoparticles, which can be avoided. Finally, with one surgical corridor, there is no need for multiple trajectories, which are required in cases of larger tumors with stereotactic application alone [27,28].

4.2. Complication Management

After a short period of time (median: 70 days, IQR: 50–105) more than half of the patients developed perifocal edema with additional neurological deficits, requiring treatment with higher steroid doses. In six of these cases, nanoparticles had to be removed surgically due to refractory edema (n = 6; 33%). In our previous publication we focused on the immunological aspect of this reaction that must be considered as part of the treatment. In histopathology, brain tissue revealed large amounts of aggregated nanoparticles located in necrotic tissue without evidence of tumor activity. At the borders, nanoparticles were found to be incorporated by phagocytes. The surrounding tissue exhibited a strongly proinflammatory state with increased T cell, NK cell and myeloid cell infiltration [32]. These reactions were also found in this case series (patients 9 and 13). As well as edema formation, four patients also developed surgical site infection, probably due the prolonged treatment with steroids (n = 4; 22%). Therefore, the administration of high doses of corticosteroids (dexamethasone more than 4 mg/day) for a longer period (>6 weeks) should be avoided. As an alternative, bevacizumab could be used for edema control, as has been demonstrated for the treatment of radionecrosis [56].
Otherwise, the surgical removal of the nanoparticles and of necrotic tissue bulks from the tumor core must be discussed. In addition, thermotherapy should be limited to patients who are not in need of corticosteroids before treatment starts. Moreover, patients with compromised clinical performance status who may not tolerate temporary increases in edema secondary to necrosis, or are in risk of other major side effects, will not be eligible for this therapy—as is also the case for all other therapies.
In comparison to stereotactic applications [27,28], nanoparticle application after tumor resection allows for a much better distribution and higher concentration of nanoparticles at the borders of the tumor cavity. Yet, elution into the ventricles and basal cisterns must be avoided to prevent complications such as in patient 4. Therefore, any contacts between the resection cavity and ventricles or cisterns should be sealed, e.g., using fibrinogen-coated collagen pads, if possible.

4.3. Outlook

As hyperthermia must be assumed to be the major cause for edema formation, special attention must be directed to temperature simulation and adjustment. Currently, the maximum temperature registered along the course of the closed-end catheter is used to correct the simulation and to tune the AMF. It might be more reasonable to adjust the AMF according to the temperature at the tumor–SPION border, for this is the region in which biological heat effects are expected to be the most pronounced.
We are working on solutions to measure temperatures simultaneously at multiple sites along the thermometry catheter, which should preferably be positioned along the rim of the tumor cavity. These measurements will be used for real-time corrections of the temperature simulation to create a more precise temperature chart. Another option might be to place a second catheter to obtain more reference points for the simulation. It is not easy to envision, but most the desirable process for accurate therapy steering would be a non-invasive method able to create a three-dimensional temperature chart under the conditions of a high-energy alternating magnetic field.
The main determinants of tissue heat generation are field strength, nanoparticle density and tissue perfusion. A standard perfusion value averaged over the whole brain is currently used for temperature simulation. Because of postoperative scarring, necrosis, varying cell densities or preexisting edema, etc., local perfusion will almost always differ from this generalized assumption. As brain perfusion mapping can be easily and routinely accomplished using dynamic contrast CT techniques, the NanoPlan® software is currently modified to include this modality for a voxel-based correction of the simulation.
A higher number of nanoparticle sheets allows for lower AMF energy, which facilitates field strength adjustments during treatment sessions. However, too large volumes may foster space-occupying necrotic reactions with post-treatment edema, as seen in case number 13 (Table 2). More highly concentrated SPION solutions might be engineered to address this problem.
One novel option to demonstrate and monitor post-treatment inflammatory reactions and the immunologic tumor microenvironment (TME) is dual-tracer PET imaging with 18F-FET and 18F-DPA-714 (a ligand of microglial translocator protein (TSPO)). In the case of patient 8, this imaging was conducted six months after thermotherapy (Figure 5). TSPO signaling clearly exceeded the FET signal, which might indicate the activation of glioma-associated myeloid cells beyond the tumor borders delineated by FET-PET. Combining these imaging biomarkers supports the characterization of the TME, as discussed in an earlier study [37]. Regarding thermotherapy, TSPO imaging shows perifocal immunologic reactions that clearly exceed solid tumor volumes. Therefore, TSPO PET may contribute to a more elaborate classification of recurrence that helps the clinician to perfectly tailor the thermotherapy settings [57].
At present, we assume that FGR and intracavitary thermotherapy are a good alternative treatment option for patients with recurrent and resectable HGG in non-eloquent regions with moderate edema. This particularly includes HGG patients at first recurrence with a non-methylated MGMT promotor and tumor progression during the treatment with alkylating agents, but also HGG patients with second or later recurrence. However, our results are based on a retrospective review of a heterogenous series of recurrent HGG. Further investigations are necessary to refine several technical aspects, such as the applied SPION volume, or the calculation and monitoring of therapy temperature. Recently, a patient registry has been established to collect treatment data prospectively.

5. Conclusions

FGR in combination with intracavitary thermotherapy is an interesting treatment modality for patients with malignant glioma. In addition to cytoreductive treatment, non-ablative hyperthermia can induce an inflammatory reaction; however, the frequent development of cerebral edema requires the refinement of the treatment protocol.

Author Contributions

Conceptualization, M.S., M.M., J.W., O.G.; Methodology, M.S. and M.M.; Software, M.S. and M.M.; Validation, S.S., J.W., O.G., W.S. and B.Z.; Formal Analysis, M.S., M.M., B.Z. and O.G.; Investigation, M.S. and J.W.; Resources, M.S., A.-K.B. and J.W.; Data Curation, M.S., A.-K.B. and B.Z.; Writing—Original Draft Preparation, M.S. and M.M.; Writing—Review and Editing, M.S., M.M., S.S., M.H., B.B., N.W., J.W., W.S. and O.G.; Visualization, M.S. and M.M.; Supervision, W.S.; Project Administration, M.S., J.W. and W.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Institutional Review Board (or Ethics Committee) of University of Münster, Germany (2020-531-f-S and date of approval: 18 August 2020).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

All data generated or analysed during this study are included in this published article.

Conflicts of Interest

Walter Stummer reports lecture activities for Medac (Wedel, Germany), Carl Zeiss Meditech (Oberkochen, Germany) and MagForce AG (Berlin, Germany). Oliver Grauer has received financial support from MagForce AG (Berlin, Germany) for attending symposia. Michael Schwake and Stephanie Schipmann report consultant activities for MagForce AG (Berlin, Germany). All other authors have no personal, financial, or institutional interest in any of the drugs, materials, or devices described in this article.

References

  1. Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.B.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef] [PubMed]
  2. Goldbrunner, R.; Minniti, G.; Preusser, M.; Jenkinson, M.D.; Sallabanda, K.; Houdart, E.; von Deimling, A.; Stavrinou, P.; Lefranc, F.; Lund-Johansen, M.; et al. EANO guidelines for the diagnosis and treatment of meningiomas. Lancet Oncol. 2016, 17, e383–e391. [Google Scholar] [CrossRef] [Green Version]
  3. Stummer, W.; Pichlmeier, U.; Meinel, T.; Wiestler, O.D.; Zanella, F.; Reulen, H.-J.; ALA-Glioma Study Group. Fluorescence-guided surgery with 5-aminolevulinic acid for resection of malignant glioma: A randomised controlled multicentre phase III trial. Lancet Oncol. 2006, 7, 392–401. [Google Scholar] [CrossRef]
  4. Stummer, W.; Novotny, A.; Stepp, H.; Goetz, C.; Bise, K.; Reulen, H.J. Fluorescence-guided resection of glioblastoma multiforme by using 5-aminolevulinic acid-induced porphyrins: A prospective study in 52 consecutive patients. J. Neurosurg. 2000, 93, 1003–1013. [Google Scholar] [CrossRef]
  5. Schucht, P.; Beck, J.; Abu-Isa, J.; Andereggen, L.; Murek, M.; Seidel, K.; Stieglitz, L.; Raabe, A. Gross Total Resection Rates in Contemporary Glioblastoma Surgery. Neurosurgery 2012, 71, 927–936. [Google Scholar] [CrossRef] [Green Version]
  6. Seidel, K.; Beck, J.; Stieglitz, L.; Schucht, P.; Raabe, A. The warning-sign hierarchy between quantitative subcortical motor mapping and continuous motor evoked potential monitoring during resection of supratentorial brain tumors; Clinical article. J. Neurosurg. 2013, 118, 287–296. [Google Scholar] [CrossRef] [Green Version]
  7. Hadani, M.; Spiegelman, R.; Feldman, Z.; Berkenstadt, H.; Ram, Z. Novel, compact, intraoperative magnetic resonance imaging-guided system for conventional neurosurgical operating rooms. Neurosurgery 2001, 48, 799–807; discussion 807–809. [Google Scholar]
  8. Eyüpoglu, I.Y.; Hore, N.; Savaskan, N.E.; Grummich, P.; Roessler, K.; Buchfelder, M.; Ganslandt, O. Improving the Extent of Malignant Glioma Resection by Dual Intraoperative Visualization Approach. PLoS ONE 2012, 7, e44885. [Google Scholar] [CrossRef]
  9. Stupp, R.; Taillibert, S.; Kanner, A.; Read, W.; Steinberg, D.M.; Lhermitte, B.; Toms, S.; Idbaih, A.; Ahluwalia, M.S.; Fink, K.; et al. Effect of tumor-treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma a randomized clinical trial. JAMA J. Am. Med. Assoc. 2017, 318, 2306–2316. [Google Scholar] [CrossRef] [Green Version]
  10. Díez Valle, R.; Slof, J.; Galván, J.; Arza, C.; Romariz, C.; Vidal, C. Observational, retrospective study of the effectiveness of 5-aminolevulinic acid in malignant glioma surgery in Spain (The VISIONA study). Neurologia 2014, 29, 131–138. [Google Scholar] [CrossRef]
  11. Aldave, G.; Tejada, S.; Pay, E.; Marigil, M.; Bejarano, B.; Idoate, M.A.; Díez-Valle, R. Prognostic value of residual fluorescent tissue in glioblastoma patients after gross total resection in 5-aminolevulinic Acid-guided surgery. Neurosurgery 2013, 72, 915–920; discussion 920–921. [Google Scholar] [CrossRef]
  12. Stummer, W.; Reulen, H.-J.J.; Meinel, T.; Pichlmeier, U.; Schumacher, W.; Tonn, J.-C.C.; Rohde, V.; Oppel, F.; Turowski, B.; Woiciechowsky, C.; et al. Extent of resection and survival in glioblastoma multiforme: Identification of and adjustment for bias. Neurosurgery 2008, 62, 564–576; discussion 564–576. [Google Scholar] [CrossRef]
  13. Valdés, P.A.; Samkoe, K.; O’Hara, J.A.; Roberts, D.W.; Paulsen, K.D.; Pogue, B.W. Deferoxamine iron chelation increases delta-aminolevulinic acid induced protoporphyrin IX in xenograft glioma model. Photochem. Photobiol. 2010, 86, 471–475. [Google Scholar] [CrossRef] [Green Version]
  14. Valdés, P.A.; Leblond, F.; Kim, A.; Harris, B.T.; Wilson, B.C.; Fan, X.; Tosteson, T.D.; Hartov, A.; Ji, S.; Erkmen, K.; et al. Quantitative fluorescence in intracranial tumor: Implications for ALA-induced PpIX as an intraoperative biomarker. J. Neurosurg. 2011, 115, 11–17. [Google Scholar] [CrossRef] [Green Version]
  15. Stummer, W.; Tonn, J.-C.; Goetz, C.; Ullrich, W.; Stepp, H.; Bink, A.; Pietsch, T.; Pichlmeier, U. 5-Aminolevulinic Acid-derived Tumor Fluorescence. Neurosurgery 2014, 74, 310–320. [Google Scholar] [CrossRef] [Green Version]
  16. Hefti, M.; von Campe, G.; Moschopulos, M.; Siegner, A.; Looser, H.; Landolt, H. 5-Aminolevulinic acid induced protoporphyrin IX fluorescence in high-grade glioma surgery: A one-year experience at a single institutuion. Swiss Med. Wkly. 2008, 138, 180–185. [Google Scholar] [CrossRef]
  17. Cage, T.A.; Pekmezci, M.; Prados, M.; Berger, M.S. Subependymal spread of recurrent glioblastoma detected with the intraoperative use of 5-aminolevulinic acid: Case report. J. Neurosurg. 2013, 118, 1220–1223. [Google Scholar] [CrossRef]
  18. Müther, M.; Koch, R.; Weckesser, M.; Sporns, P.; Schwindt, W.; Stummer, W. 5-Aminolevulinic Acid Fluorescence-Guided Resection of 18F-FET-PET Positive Tumor Beyond Gadolinium Enhancing Tumor Improves Survival in Glioblastoma. Neurosurgery 2019, 85, E1020–E1029. [Google Scholar] [CrossRef] [Green Version]
  19. Nabavi, A.; Thurm, H.; Zountsas, B.; Pietsch, T.; Lanfermann, H.; Pichlmeier, U.; Mehdorn, M.; 5-ALA Recurrent Glioma Study Group. Five-aminolevulinic acid for fluorescence-guided resection of recurrent malignant gliomas: A phase ii study. Neurosurgery 2009, 65, 1070–1076; discussion 1076–1077. [Google Scholar] [CrossRef]
  20. Weller, M.; van den Bent, M.; Tonn, J.C.; Stupp, R.; Preusser, M.; Cohen-Jonathan-Moyal, E.; Henriksson, R.; Le Rhun, E.; Balana, C.; Chinot, O.; et al. European Association for Neuro-Oncology (EANO) guideline on the diagnosis and treatment of adult astrocytic and oligodendroglial gliomas. Lancet Oncol. 2017, 18, e315–e329. [Google Scholar] [CrossRef] [Green Version]
  21. Desjardins, A.; Gromeier, M.; Herndon, J.E.; Beaubier, N.; Bolognesi, D.P.; Friedman, A.H.; Friedman, H.S.; McSherry, F.; Muscat, A.M.; Nair, S.; et al. Recurrent Glioblastoma Treated with Recombinant Poliovirus. N. Engl. J. Med. 2018, 379, 150–161. [Google Scholar] [CrossRef] [PubMed]
  22. Wick, W.; Gorlia, T.; Bendszus, M.; Taphoorn, M.; Sahm, F.; Harting, I.; Brandes, A.A.; Taal, W.; Domont, J.; Idbaih, A.; et al. Lomustine and bevacizumab in progressive glioblastoma. N. Engl. J. Med. 2017, 377, 1954–1963. [Google Scholar] [CrossRef] [PubMed]
  23. Schipmann, S.; Müther, M.; Stögbauer, L.; Zimmer, S.; Brokinkel, B.; Holling, M.; Grauer, O.; Suero Molina, E.; Warneke, N.; Stummer, W. Combination of ALA-induced fluorescence-guided resection and intraoperative open photodynamic therapy for recurrent glioblastoma: Case series on a promising dual strategy for local tumor control. J. Neurosurg. 2020, 134, 426–436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Wallner, K.E.; Galicich, J.H.; Krol, G.; Arbit, E.; Malkin, M.G. Patterns of failure following treatment for glioblastoma multiforme and anaplastic astrocytoma. Int. J. Radiat. Oncol. Biol. Phys. 1989, 16, 1405–1409. [Google Scholar] [CrossRef]
  25. Westphal, M.; Hilt, D.C.; Bortey, E.; Delavault, P.; Olivares, R.; Warnke, P.C.; Whittle, I.R.; Jääskeläinen, J.; Ram, Z. A phase 3 trial of local chemotherapy with biodegradable carmustine (BCNU) wafers (Gliadel wafers) in patients with primary malignant glioma. Neuro. Oncol. 2003, 5, 79–88. [Google Scholar] [CrossRef]
  26. Stepp, H.; Beck, T.; Pongratz, T.; Meinel, T.; Kreth, F.-W.; Tonn, J.C.; Stummer, W. ALA and malignant glioma: Fluorescence-guided resection and photodynamic treatment. J. Environ. Pathol. Toxicol. Oncol. 2007, 26, 157–164. [Google Scholar] [CrossRef]
  27. Maier-Hauff, K.; Ulrich, F.; Nestler, D.; Niehoff, H.; Wust, P.; Thiesen, B.; Orawa, H.; Budach, V.; Jordan, A. Efficacy and safety of intratumoral thermotherapy using magnetic iron-oxide nanoparticles combined with external beam radiotherapy on patients with recurrent glioblastoma multiforme. J. Neurooncol. 2011, 103, 317–324. [Google Scholar] [CrossRef] [Green Version]
  28. Maier-Hauff, K.; Rothe, R.; Scholz, R.; Gneveckow, U.; Wust, P.; Thiesen, B.; Feussner, A.; Deimling, A.; Waldoefner, N.; Felix, R.; et al. Intracranial thermotherapy using magnetic nanoparticles combined with external beam radiotherapy: Results of a feasibility study on patients with glioblastoma multiforme. J. Neurooncol. 2007, 81, 53–60. [Google Scholar] [CrossRef]
  29. Yanase, M.; Shinkai, M.; Honda, H.; Wakabayashi, T.; Yoshida, J.; Kobayashi, T. Antitumor Immunity Induction by Intracellular Hyperthermia Using Magnetite Cationic Liposomes. Jpn. J. Cancer Res. 1998, 89, 775–782. [Google Scholar] [CrossRef]
  30. Ito, A.; Shinkai, M.; Honda, H.; Yoshikawa, K.; Saga, S.; Wakabayashi, T.; Yoshida, J.; Kobayashi, T. Heat shock protein 70 expression induces antitumor immunity during intracellular hyperthermia using magnetite nanoparticles. Cancer Immunol. Immunother. 2003, 52, 80–88. [Google Scholar] [CrossRef]
  31. Tanaka, K.; Ito, A.; Kobayashi, T.; Kawamura, T.; Shimada, S.; Matsumoto, K.; Saida, T.; Honda, H. Intratumoral injection of immature dendritic cells enhances antitumor effect of hyperthermia using magnetic nanoparticles. Int. J. Cancer 2005, 116, 624–633. [Google Scholar] [CrossRef]
  32. Grauer, O.; Jaber, M.; Hess, K.; Weckesser, M.; Schwindt, W.; Maring, S.; Wölfer, J.; Stummer, W. Combined intracavitary thermotherapy with iron oxide nanoparticles and radiotherapy as local treatment modality in recurrent glioblastoma patients. J. Neurooncol. 2019, 141, 83–94. [Google Scholar] [CrossRef] [Green Version]
  33. Molina, E.S.; Schipmann, S.; Mueller, I.; Wölfer, J.; Ewelt, C.; Maas, M.; Brokinkel, B.; Stummer, W. Conscious sedation with dexmedetomidine compared with asleep-awake-asleep craniotomies in glioma surgery: An analysis of 180 patients. J. Neurosurg. 2018, 129, 1223–1230. [Google Scholar] [CrossRef] [Green Version]
  34. Plotkin, M.; Gneveckow, U.; Meier-Hauff, K.; Amthauer, H.; Feußner, A.; Denecke, T.; Gutberlet, M.; Jordan, A.; Felix, R.; Wust, P. 18F-FET PET for planning of thermotherapy using magnetic nanoparticles in recurrent glioblastoma. Int. J. Hyperth. 2006, 22, 319–325. [Google Scholar] [CrossRef]
  35. Yoo, T.S.; Ackerman, M.J.; Lorensen, W.E.; Schroeder, W.; Chalana, V.; Aylward, S.; Metaxas, D.; Whitaker, R. Engineering and algorithm design for an image processing API: A technical report on ITK—The Insight Toolkit. Stud. Health Technol. Inform. 2002, 85, 586–592. [Google Scholar] [CrossRef]
  36. Gneveckow, U.; Jordan, A.; Scholz, R.; Brüss, V.; Waldöfner, N.; Ricke, J.; Feussner, A.; Hildebrandt, B.; Rau, B.; Wust, P. Description and characterization of the novel hyperthermia- and thermoablation-system MFH 300F for clinical magnetic fluid hyperthermia. Med. Phys. 2004, 31, 1444–1451. [Google Scholar] [CrossRef]
  37. Zinnhardt, B.; Müther, M.; Roll, W.; Backhaus, P.; Jeibmann, A.; Foray, C.; Barca, C.; Döring, C.; Tavitian, B.; Dollé, F.; et al. TSPO imaging-guided characterization of the immunosuppressive myeloid tumor microenvironment in patients with malignant glioma. Neuro. Oncol. 2020, 22, 1030–1043. [Google Scholar] [CrossRef]
  38. Wen, P.Y.; Macdonald, D.R.; Reardon, D.A.; Cloughesy, T.F.; Sorensen, A.G.; Galanis, E.; DeGroot, J.; Wick, W.; Gilbert, M.R.; Lassman, A.B.; et al. Updated response assessment criteria for high-grade gliomas: Response assessment in neuro-oncology working group. J. Clin. Oncol. 2010, 28, 1963–1972. [Google Scholar] [CrossRef]
  39. Louis, D.N.; Perry, A.; Reifenberger, G.; von Deimling, A.; Figarella-Branger, D.; Cavenee, W.K.; Ohgaki, H.; Wiestler, O.D.; Kleihues, P.; Ellison, D.W. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A summary. Acta Neuropathol. 2016, 131, 803–820. [Google Scholar] [CrossRef] [Green Version]
  40. Issels, R.D.; Lindner, L.H.; Verweij, J.; Wust, P.; Reichardt, P.; Schem, B.C.; Abdel-Rahman, S.; Daugaard, S.; Salat, C.; Wendtner, C.M.; et al. Neo-adjuvant chemotherapy alone or with regional hyperthermia for localised high-risk soft-tissue sarcoma: A randomised phase 3 multicentre study. Lancet Oncol. 2010, 11, 561–570. [Google Scholar] [CrossRef] [Green Version]
  41. Thiesen, B.; Jordan, A. Clinical applications of magnetic nanoparticles for hyperthermia. Int. J. Hyperth. 2008, 24, 467–474. [Google Scholar] [CrossRef] [PubMed]
  42. Van Der Zee, J.; González González, D.; Van Rhoon, G.C.; Van Dijk, J.D.P.; Van Putten, W.L.J.; Hart, A.A.M. Comparison of radiotherapy alone with radiotherapy plus hyperthermia in locally advanced pelvic tumours: A prospective, randomised, multicentre trial. Lancet 2000, 355, 1119–1125. [Google Scholar] [CrossRef]
  43. Ringel, F.; Pape, H.; Sabel, M.; Krex, D.; Bock, H.C.; Misch, M.; Weyerbrock, A.; Westermaier, T.; Senft, C.; Schucht, P.; et al. Clinical benefit from resection of recurrent glioblastomas: Results of a multicenter study including 503 patients with recurrent glioblastomas undergoing surgical resection. Neuro. Oncol. 2016, 18, 96–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Chaichana, K.L.; Zadnik, P.; Weingart, J.D.; Olivi, A.; Gallia, G.L.; Blakeley, J.; Lim, M.; Brem, H.; Quiñones-Hinojosa, A. Multiple resections for patients with glioblastoma: Prolonging survival. J. Neurosurg. 2013, 118, 812–820. [Google Scholar] [CrossRef] [Green Version]
  45. Helseth, R.; Helseth, E.; Johannesen, T.B.; Langberg, C.W.; Lote, K.; Rønning, P.; Scheie, D.; Vik, A.; Meling, T.R. Overall survival, prognostic factors, and repeated surgery in a consecutive series of 516 patients with glioblastoma multiforme. Acta Neurol. Scand. 2010, 122, 159–167. [Google Scholar] [CrossRef]
  46. Suchorska, B.; Weller, M.; Tabatabai, G.; Senft, C.; Hau, P.; Sabel, M.C.; Herrlinger, U.; Ketter, R.; Schlegel, U.; Marosi, C.; et al. Complete resection of contrast-enhancing tumor volume is associated with improved survival in recurrent glioblastoma-results from the DIRECTOR trial. Neuro. Oncol. 2016, 18, 549–556. [Google Scholar] [CrossRef]
  47. Shi, W.; Scannell Bryan, M.; Gilbert, M.R.; Mehta, M.P.; Blumenthal, D.T.; Brown, P.D.; Valeinis, E.; Hopkins, K.; Souhami, L.; Andrews, D.W.; et al. Investigating the Effect of Reirradiation or Systemic Therapy in Patients With Glioblastoma After Tumor Progression: A Secondary Analysis of NRG Oncology/Radiation Therapy Oncology Group Trial 0525. Int. J. Radiat. Oncol. Biol. Phys. 2018, 100, 38–44. [Google Scholar] [CrossRef]
  48. Perry, J.R.; Bélanger, K.; Mason, W.P.; Fulton, D.; Kavan, P.; Easaw, J.; Shields, C.; Kirby, S.; Macdonald, D.R.; Eisenstat, D.D.; et al. Phase II trial of continuous dose-intense temozolomide in recurrent malignant glioma: RESCUE study. J. Clin. Oncol. 2010, 28, 2051–2057. [Google Scholar] [CrossRef]
  49. Weller, M.; Tabatabai, G.; Kästner, B.; Felsberg, J.; Steinbach, J.P.; Wick, A.; Schnell, O.; Hau, P.; Herrlinger, U.; Sabel, M.C.; et al. MGMT Promoter Methylation Is a Strong Prognostic Biomarker for Benefit from Dose-Intensified Temozolomide Rechallenge in Progressive Glioblastoma: The DIRECTOR Trial. Clin. Cancer Res. 2015, 21, 2057–2064. [Google Scholar] [CrossRef] [Green Version]
  50. Batchelor, T.T.; Mulholland, P.; Neyns, B.; Nabors, L.B.; Campone, M.; Wick, A.; Mason, W.; Mikkelsen, T.; Phuphanich, S.; Ashby, L.S.; et al. Phase III randomized trial comparing the efficacy of cediranib as monotherapy, and in combination with lomustine, versus lomustine alone in patients with recurrent glioblastoma. J. Clin. Oncol. 2013, 31, 3212–3218. [Google Scholar] [CrossRef] [Green Version]
  51. Friedman, H.S.; Prados, M.D.; Wen, P.Y.; Mikkelsen, T.; Schiff, D.; Abrey, L.E.; Yung, W.K.A.; Paleologos, N.; Nicholas, M.K.; Jensen, R.; et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J. Clin. Oncol. 2009, 27, 4733–4740. [Google Scholar] [CrossRef] [Green Version]
  52. Lombardi, G.; De Salvo, G.L.; Brandes, A.A.; Eoli, M.; Rudà, R.; Faedi, M.; Lolli, I.; Pace, A.; Daniele, B.; Pasqualetti, F.; et al. Regorafenib compared with lomustine in patients with relapsed glioblastoma (REGOMA): A multicentre, open-label, randomised, controlled, phase 2 trial. Lancet. Oncol. 2019, 20, 110–119. [Google Scholar] [CrossRef]
  53. Diaz, R.; Ivan, M.E.; Hanft, S.; Vanni, S.; Manzano, G.; Jagid, J.; Komotar, R.J. Laser Interstitial Thermal Therapy. Neurosurgery 2016, 79, S3–S7. [Google Scholar] [CrossRef] [Green Version]
  54. Munoz-Casabella, A.; Alvi, M.A.; Rahman, M.; Burns, T.C.; Brown, D.A. Laser Interstitial Thermal Therapy for Recurrent Glioblastoma: Pooled Analyses of Available Literature. World Neurosurg. 2021, 153, 91–97.e1. [Google Scholar] [CrossRef]
  55. Chohan, M.O.; Berger, M.S. 5-Aminolevulinic acid fluorescence guided surgery for recurrent high-grade gliomas. J. Neurooncol. 2019, 141, 517–522. [Google Scholar] [CrossRef]
  56. Levin, V.A.; Bidaut, L.; Hou, P.; Kumar, A.J.; Wefel, J.S.; Bekele, B.N.; Prabhu, S.; Loghin, M.; Gilbert, M.R.; Jackson, E.F. Randomized double-blind placebo-controlled trial of bevacizumab therapy for radiation necrosis of the central nervous system. Int. J. Radiat. Oncol. Biol. Phys. 2011, 79, 1487–1495. [Google Scholar] [CrossRef] [Green Version]
  57. Haider, A.S.; van den Bent, M.; Wen, P.Y.; Vogelbaum, M.A.; Chang, S.; Canoll, P.D.; Horbinski, C.M.; Huse, J.T. Toward a standard pathological and molecular characterization of recurrent glioma in adults: A Response Assessment in Neuro-Oncology effort. Neuro. Oncol. 2020, 22, 450–456. [Google Scholar] [CrossRef]
Figure 1. Flow chart of treatment.
Figure 1. Flow chart of treatment.
Cancers 14 00541 g001
Figure 2. (A) Magnetic resonance imaging performed preoperatively showing contrast-enhanced tumor in the right parietal lobe. (B) Contrast-enhanced computed tomography performed at the first postoperative day. (C) Computed tomography in patient 13 demonstrating significant edema two months after surgery.
Figure 2. (A) Magnetic resonance imaging performed preoperatively showing contrast-enhanced tumor in the right parietal lobe. (B) Contrast-enhanced computed tomography performed at the first postoperative day. (C) Computed tomography in patient 13 demonstrating significant edema two months after surgery.
Cancers 14 00541 g002
Figure 3. Kaplan–Meier plots demonstrating progression-free survival (PFS) and overall survival (OS) in months, after fluorescence-guided resection (FGR) and thermotherapy. Strokes mark censored data points. Median progression-free survival (PFS) for the study population was 5.5 months (95% CI: 4.67–6.13) after thermotherapy and median overall survival (OS) was 9.5 months (95% CI: 7.21–11.79).
Figure 3. Kaplan–Meier plots demonstrating progression-free survival (PFS) and overall survival (OS) in months, after fluorescence-guided resection (FGR) and thermotherapy. Strokes mark censored data points. Median progression-free survival (PFS) for the study population was 5.5 months (95% CI: 4.67–6.13) after thermotherapy and median overall survival (OS) was 9.5 months (95% CI: 7.21–11.79).
Cancers 14 00541 g003
Figure 4. T2-weighted magnetic resonance imaging of the cervical spine of patient 4 in sagittal (left) and axial (right) planes. Note the intramedullary hyperintense lesion of the spinal cord at the level of C2-3 adjacent to metal artefacts (arrow).
Figure 4. T2-weighted magnetic resonance imaging of the cervical spine of patient 4 in sagittal (left) and axial (right) planes. Note the intramedullary hyperintense lesion of the spinal cord at the level of C2-3 adjacent to metal artefacts (arrow).
Cancers 14 00541 g004
Figure 5. Patient 8, six months after completion of thermotherapy. Combination of CT post contrast (A), 18F-FET- (B) and 18F-DPA-714-PET (C) imaging. Note the increasing volumes of contrast enhancement, FET positivity and DPA positivity, respectively; the latter is referring to the infiltration of myeloid cells within the tumor microenvironment.
Figure 5. Patient 8, six months after completion of thermotherapy. Combination of CT post contrast (A), 18F-FET- (B) and 18F-DPA-714-PET (C) imaging. Note the increasing volumes of contrast enhancement, FET positivity and DPA positivity, respectively; the latter is referring to the infiltration of myeloid cells within the tumor microenvironment.
Cancers 14 00541 g005
Table 1. Baseline patient characteristics.
Table 1. Baseline patient characteristics.
IDAge (Years)SexInitial DiagnosisTime Since Initial Diagnosis (Months)Number of RecurrencesCurrent DiagnosisMGMT PromotorLocation
142FGlioblastoma, IDH mutated61Glioblastoma, IDH mutatedmethylatedLP
260MGlioblastoma, IDH wild type132Glioblastoma, IDH wild typeunmethylatedRP
359MGlioblastoma, IDH wild type151Glioblastoma, IDH wild typeunmethylatedLP
465MGlioblastoma, IDH wild type82Glioblastoma, IDH wild typeunmethylatedRP
575FGlioblastoma, IDH wild type41Glioblastoma, IDH wild typeunmethylatedRF
642MGlioblastoma, IDH wild type34Glioblastoma, IDH wild typemethylatedLF
763FGlioblastoma, IDH wild type131Glioblastoma, IDH wildtypeunmethylatedLP
836FAnaplastic astrocytoma, IDH mutated572Anaplastic astrocytoma, IDH mutatedunmethylatedRF
938MGlioblastoma, IDH wildtype422Glioblastoma, IDH wildtypeunmethylatedLO
1062MGlioblastoma, IDH wildtype71Glioblastoma, IDH wildtypeunmethylatedLO
1142FGlioblastoma, IDH wildtype331Glioblastoma, IDH wildtypeunmethylatedLO
1240FDiffuse Astrocytoma, IDH mutated2483Anaplastic Astrocytoma, IDH mutatedmethylatedRF
1358MGlioblastoma, IDH wildtype61Glioblastoma, IDH wildtypeunmethylatedRP
1444FGlioblastoma, IDH wildtype81Glioblastoma, IDH wildtypeunmethylatedLF
1546MGlioblastoma, IDH wildtype101Glioblastoma, IDH wildtypeunmethylatedRP
1657MDiffuse Astrocytoma, IDH mutated1673Anaplastic Oligodendroglioma, IDH mutatednot availableLT
1758FGlioblastoma, IDH wildtype163Glioblastoma, IDH wildtypeunmethylatedLT
1845MGlioblastoma, NOS1333Glioblastoma, IDH mutatedmethylatedLT
R, right. L, left. F, frontal. P, parietal. T, temporal. O, occipital.
Table 2. Data on thermotherapy and follow up.
Table 2. Data on thermotherapy and follow up.
Patient IDEORVolume of Nanoparticles (mL)Peak Temperature (°C)Re-Irradiation (Gy)Salvage ChemotherapyTime to Edema (Months)Revision Due to EdemaPFS (Months)OS (Months)
1STR2.85939.6-3.67Yes72 *72
2GTR3.26039.6-0.33No77
3NTR25639.6-1.97Yes1124
4STR2.15339.6-NonYes69
5GTR553--4.43No57
6NTR354--NonNo34
7NRT3.249--NonNo35
8GTR4.344-Temozolomide5.07No3535
9GTR3.450--2.73No410
10GTR3.84839.6Bevacizumab3.33No88
11NTR54539.6-3.33No510
12NTR342- NonNo717
13NTR74639.6-1.5Yes67
14GTR33939.6-2.0No56
15GTR1.943.339.6-2.33Yes315
16NTR545--NonNo712
17STR544.6--0.77No35
18STR545--1.83Yes211
EOR, extent of resection. GTR, gross total resection, NTR, near total resection. STR, subtotal resection. PFS, progression-free survival. OS, overall survival. * Patients without evidence of progression. † patients still alive.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Schwake, M.; Müther, M.; Bruns, A.-K.; Zinnhardt, B.; Warneke, N.; Holling, M.; Schipmann, S.; Brokinkel, B.; Wölfer, J.; Stummer, W.; et al. Combined Fluorescence-Guided Resection and Intracavitary Thermotherapy with Superparamagnetic Iron-Oxide Nanoparticles for Recurrent High-Grade Glioma: Case Series with Emphasis on Complication Management. Cancers 2022, 14, 541. https://doi.org/10.3390/cancers14030541

AMA Style

Schwake M, Müther M, Bruns A-K, Zinnhardt B, Warneke N, Holling M, Schipmann S, Brokinkel B, Wölfer J, Stummer W, et al. Combined Fluorescence-Guided Resection and Intracavitary Thermotherapy with Superparamagnetic Iron-Oxide Nanoparticles for Recurrent High-Grade Glioma: Case Series with Emphasis on Complication Management. Cancers. 2022; 14(3):541. https://doi.org/10.3390/cancers14030541

Chicago/Turabian Style

Schwake, Michael, Michael Müther, Ann-Katrin Bruns, Bastian Zinnhardt, Nils Warneke, Markus Holling, Stephanie Schipmann, Benjamin Brokinkel, Johannes Wölfer, Walter Stummer, and et al. 2022. "Combined Fluorescence-Guided Resection and Intracavitary Thermotherapy with Superparamagnetic Iron-Oxide Nanoparticles for Recurrent High-Grade Glioma: Case Series with Emphasis on Complication Management" Cancers 14, no. 3: 541. https://doi.org/10.3390/cancers14030541

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop