ϟ

Ghazaleh Tabatabai

Here are all the papers by Ghazaleh Tabatabai that you can download and read on OA.mg.
Ghazaleh Tabatabai’s last known institution is . Download Ghazaleh Tabatabai PDFs here.

Claim this Profile →
DOI: 10.1016/s1470-2045(12)70164-x
2012
Cited 975 times
Temozolomide chemotherapy alone versus radiotherapy alone for malignant astrocytoma in the elderly: the NOA-08 randomised, phase 3 trial
Background Radiotherapy is the standard care in elderly patients with malignant astrocytoma and the role of primary chemotherapy is poorly defined. We did a randomised trial to compare the efficacy and safety of dose-dense temozolomide alone versus radiotherapy alone in elderly patients with anaplastic astrocytoma or glioblastoma. Methods Between May 15, 2005, and Nov 2, 2009, we enrolled patients with confirmed anaplastic astrocytoma or glioblastoma, age older than 65 years, and a Karnofsky performance score of 60 or higher. Patients were randomly assigned 100 mg/m2 temozolomide, given on days 1–7 of 1 week on, 1 week off cycles, or radiotherapy of 60·0 Gy, administered over 6–7 weeks in 30 fractions of 1·8–2·0 Gy. The primary endpoint was overall survival. We assessed non-inferiority with a 25% margin, analysed for all patients who received at least one dose of assigned treatment. This trial is registered with ClinicalTrials.gov, number NCT01502241. Findings Of 584 patients screened, we enrolled 412. 373 patients (195 randomly allocated to the temozolomide group and 178 to the radiotherapy group) received at least one dose of treatment and were included in efficacy analyses. Median overall survival was 8·6 months (95% CI 7·3–10·2) in the temozolomide group versus 9·6 months (8·2–10·8) in the radiotherapy group (hazard ratio [HR] 1·09, 95% CI 0·84–1·42, pnon-inferiority=0·033). Median event-free survival (EFS) did not differ significantly between the temozolomide and radiotherapy groups (3·3 months [95% CI 3·2–4·1] vs 4·7 [4·2–5·2]; HR 1·15, 95% CI 0·92–1·43, pnon-inferiority=0·043). Tumour MGMT promoter methylation was seen in 73 (35%) of 209 patients tested. MGMT promoter methylation was associated with longer overall survival than was unmethylated status (11·9 months [95% CI 9·0 to not reached] vs 8·2 months [7·0–10·0]; HR 0·62, 95% CI 0·42–0·91, p=0·014). EFS was longer in patients with MGMT promoter methylation who received temozolomide than in those who underwent radiotherapy (8·4 months [95e% CI 5·5–11·7] vs 4·6 [4·2–5·0]), whereas the opposite was true for patients with no methylation of the MGMT promoter (3·3 months [3·0–3·5] vs 4·6 months [3·7–6·3]). The most frequent grade 3–4 intervention-related adverse events were neutropenia (16 patients in the temozolomide group vs two in the radiotherapy group), lymphocytopenia (46 vs one), thrombocytopenia (14 vs four), raised liver-enzyme concentrations (30 vs 16), infections (35 vs 23), and thromboembolic events (24 vs eight). Interpretation Temozolomide alone is non-inferior to radiotherapy alone in the treatment of elderly patients with malignant astrocytoma. MGMT promoter methylation seems to be a useful biomarker for outcomes by treatment and could aid decision-making. Funding Merck Sharp & Dohme.
DOI: 10.1038/s41586-018-0810-y
2018
Cited 652 times
Actively personalized vaccination trial for newly diagnosed glioblastoma
Patients with glioblastoma currently do not sufficiently benefit from recent breakthroughs in cancer treatment that use checkpoint inhibitors1,2. For treatments using checkpoint inhibitors to be successful, a high mutational load and responses to neoepitopes are thought to be essential3. There is limited intratumoural infiltration of immune cells4 in glioblastoma and these tumours contain only 30-50 non-synonymous mutations5. Exploitation of the full repertoire of tumour antigens-that is, both unmutated antigens and neoepitopes-may offer more effective immunotherapies, especially for tumours with a low mutational load. Here, in the phase I trial GAPVAC-101 of the Glioma Actively Personalized Vaccine Consortium (GAPVAC), we integrated highly individualized vaccinations with both types of tumour antigens into standard care to optimally exploit the limited target space for patients with newly diagnosed glioblastoma. Fifteen patients with glioblastomas positive for human leukocyte antigen (HLA)-A*02:01 or HLA-A*24:02 were treated with a vaccine (APVAC1) derived from a premanufactured library of unmutated antigens followed by treatment with APVAC2, which preferentially targeted neoepitopes. Personalization was based on mutations and analyses of the transcriptomes and immunopeptidomes of the individual tumours. The GAPVAC approach was feasible and vaccines that had poly-ICLC (polyriboinosinic-polyribocytidylic acid-poly-L-lysine carboxymethylcellulose) and granulocyte-macrophage colony-stimulating factor as adjuvants displayed favourable safety and strong immunogenicity. Unmutated APVAC1 antigens elicited sustained responses of central memory CD8+ T cells. APVAC2 induced predominantly CD4+ T cell responses of T helper 1 type against predicted neoepitopes.
DOI: 10.1016/s0140-6736(18)31791-4
2019
Cited 397 times
Lomustine-temozolomide combination therapy versus standard temozolomide therapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter (CeTeG/NOA–09): a randomised, open-label, phase 3 trial
There is an urgent need for more effective therapies for glioblastoma. Data from a previous unrandomised phase 2 trial suggested that lomustine-temozolomide plus radiotherapy might be superior to temozolomide chemoradiotherapy in newly diagnosed glioblastoma with methylation of the MGMT promoter. In the CeTeG/NOA-09 trial, we aimed to further investigate the effect of lomustine-temozolomide therapy in the setting of a randomised phase 3 trial.In this open-label, randomised, phase 3 trial, we enrolled patients from 17 German university hospitals who were aged 18-70 years, with newly diagnosed glioblastoma with methylated MGMT promoter, and a Karnofsky Performance Score of 70% and higher. Patients were randomly assigned (1:1) with a predefined SAS-generated randomisation list to standard temozolomide chemoradiotherapy (75 mg/m2 per day concomitant to radiotherapy [59-60 Gy] followed by six courses of temozolomide 150-200 mg/m2 per day on the first 5 days of the 4-week course) or to up to six courses of lomustine (100 mg/m2 on day 1) plus temozolomide (100-200 mg/m2 per day on days 2-6 of the 6-week course) in addition to radiotherapy (59-60 Gy). Because of the different schedules, patients and physicians were not masked to treatment groups. The primary endpoint was overall survival in the modified intention-to-treat population, comprising all randomly assigned patients who started their allocated chemotherapy. The prespecified test for overall survival differences was a log-rank test stratified for centre and recursive partitioning analysis class. The trial is registered with ClinicalTrials.gov, number NCT01149109.Between June 17, 2011, and April 8, 2014, 141 patients were randomly assigned to the treatment groups; 129 patients (63 in the temozolomide and 66 in the lomustine-temozolomide group) constituted the modified intention-to-treat population. Median overall survival was improved from 31·4 months (95% CI 27·7-47·1) with temozolomide to 48·1 months (32·6 months-not assessable) with lomustine-temozolomide (hazard ratio [HR] 0·60, 95% CI 0·35-1·03; p=0·0492 for log-rank analysis). A significant overall survival difference between groups was also found in a secondary analysis of the intention-to-treat population (n=141, HR 0·60, 95% CI 0·35-1·03; p=0·0432 for log-rank analysis). Adverse events of grade 3 or higher were observed in 32 (51%) of 63 patients in the temozolomide group and 39 (59%) of 66 patients in the lomustine-temozolomide group. There were no treatment-related deaths.Our results suggest that lomustine-temozolomide chemotherapy might improve survival compared with temozolomide standard therapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter. The findings should be interpreted with caution, owing to the small size of the trial.German Federal Ministry of Education and Research.
DOI: 10.1038/s41586-019-1775-1
2019
Cited 337 times
Longitudinal molecular trajectories of diffuse glioma in adults
The evolutionary processes that drive universal therapeutic resistance in adult patients with diffuse glioma remain unclear1,2. Here we analysed temporally separated DNA-sequencing data and matched clinical annotation from 222 adult patients with glioma. By analysing mutations and copy numbers across the three major subtypes of diffuse glioma, we found that driver genes detected at the initial stage of disease were retained at recurrence, whereas there was little evidence of recurrence-specific gene alterations. Treatment with alkylating agents resulted in a hypermutator phenotype at different rates across the glioma subtypes, and hypermutation was not associated with differences in overall survival. Acquired aneuploidy was frequently detected in recurrent gliomas and was characterized by IDH mutation but without co-deletion of chromosome arms 1p/19q, and further converged with acquired alterations in the cell cycle and poor outcomes. The clonal architecture of each tumour remained similar over time, but the presence of subclonal selection was associated with decreased survival. Finally, there were no differences in the levels of immunoediting between initial and recurrent gliomas. Collectively, our results suggest that the strongest selective pressures occur during early glioma development and that current therapies shape this evolution in a largely stochastic manner.
DOI: 10.1038/nm.3214
2013
Cited 310 times
Acid sphingomyelinase–ceramide system mediates effects of antidepressant drugs
Major depression is a highly prevalent severe mood disorder that is treated with antidepressants. The molecular targets of antidepressants require definition. We investigated the role of the acid sphingomyelinase (Asm)-ceramide system as a target for antidepressants. Therapeutic concentrations of the antidepressants amitriptyline and fluoxetine reduced Asm activity and ceramide concentrations in the hippocampus, increased neuronal proliferation, maturation and survival and improved behavior in mouse models of stress-induced depression. Genetic Asm deficiency abrogated these effects. Mice overexpressing Asm, heterozygous for acid ceramidase, treated with blockers of ceramide metabolism or directly injected with C16 ceramide in the hippocampus had higher ceramide concentrations and lower rates of neuronal proliferation, maturation and survival compared with controls and showed depression-like behavior even in the absence of stress. The decrease of ceramide abundance achieved by antidepressant-mediated inhibition of Asm normalized these effects. Lowering ceramide abundance may thus be a central goal for the future development of antidepressants.
DOI: 10.1093/neuonc/noab150
2021
Cited 271 times
EANO guideline on the diagnosis and management of meningiomas
Abstract Meningiomas are the most common intracranial tumors. Yet, only few controlled clinical trials have been conducted to guide clinical decision making, resulting in variations of management approaches across countries and centers. However, recent advances in molecular genetics and clinical trial results help to refine the diagnostic and therapeutic approach to meningioma. Accordingly, the European Association of Neuro-Oncology (EANO) updated its recommendations for the diagnosis and treatment of meningiomas. A provisional diagnosis of meningioma is typically made by neuroimaging, mostly magnetic resonance imaging. Such provisional diagnoses may be made incidentally. Accordingly, a significant proportion of meningiomas, notably in patients that are asymptomatic or elderly or both, may be managed by a watch-and-scan strategy. A surgical intervention with tissue, commonly with the goal of gross total resection, is required for the definitive diagnosis according to the WHO classification. A role for molecular profiling including gene panel sequencing and genomic methylation profiling is emerging. A gross total surgical resection including the involved dura is often curative. Inoperable or recurrent tumors requiring treatment can be treated with radiosurgery, if the size or the vicinity of critical structures allows that, or with fractionated radiotherapy (RT). Treatment concepts combining surgery and radiosurgery or fractionated RT are increasingly used, although there remain controversies regard timing, type, and dosing of the various RT approaches. Radionuclide therapy targeting somatostatin receptors is an experimental approach, as are all approaches of systemic pharmacotherapy. The best albeit modest results with pharmacotherapy have been obtained with bevacizumab or multikinase inhibitors targeting vascular endothelial growth factor receptor, but no standard of care systemic treatment has been yet defined.
DOI: 10.1158/1078-0432.ccr-14-2737
2015
Cited 261 times
<i>MGMT</i> Promoter Methylation Is a Strong Prognostic Biomarker for Benefit from Dose-Intensified Temozolomide Rechallenge in Progressive Glioblastoma: The DIRECTOR Trial
Abstract Purpose: Rechallenge with temozolomide (TMZ) at first progression of glioblastoma after temozolomide chemoradiotherapy (TMZ/RT→TMZ) has been studied in retrospective and single-arm prospective studies, applying temozolomide continuously or using 7/14 or 21/28 days schedules. The DIRECTOR trial sought to show superiority of the 7/14 regimen. Experimental Design: Patients with glioblastoma at first progression after TMZ/RT→TMZ and at least two maintenance temozolomide cycles were randomized to Arm A [one week on (120 mg/m2 per day)/one week off] or Arm B [3 weeks on (80 mg/m2 per day)/one week off]. The primary endpoint was median time-to-treatment failure (TTF) defined as progression, premature temozolomide discontinuation for toxicity, or death from any cause. O6-methylguanine DNA methyltransferase (MGMT) promoter methylation was prospectively assessed by methylation-specific PCR. Results: Because of withdrawal of support, the trial was prematurely closed to accrual after 105 patients. There was a similar outcome in both arms for median TTF [A: 1.8 months; 95% confidence intervals (CI), 1.8–3.2 vs. B: 2.0 months; 95% CI, 1.8–3.5] and overall survival [A: 9.8 months (95% CI, 6.7–13.0) vs. B: 10.6 months (95% CI, 8.1–11.6)]. Median TTF in patients with MGMT-methylated tumors was 3.2 months (95% CI, 1.8–7.4) versus 1.8 months (95% CI, 1.8–2) in MGMT-unmethylated glioblastoma. Progression-free survival rates at 6 months (PFS-6) were 39.7% with versus 6.9% without MGMT promoter methylation. Conclusions: Temozolomide rechallenge is a treatment option for MGMT promoter-methylated recurrent glioblastoma. Alternative strategies need to be considered for patients with progressive glioblastoma without MGMT promoter methylation. Clin Cancer Res; 21(9); 2057–64. ©2015 AACR.
DOI: 10.1038/s41586-021-03363-z
2021
Cited 241 times
A vaccine targeting mutant IDH1 in newly diagnosed glioma
Mutated isocitrate dehydrogenase 1 (IDH1) defines a molecularly distinct subtype of diffuse glioma1-3. The most common IDH1 mutation in gliomas affects codon 132 and encodes IDH1(R132H), which harbours a shared clonal neoepitope that is presented on major histocompatibility complex (MHC) class II4,5. An IDH1(R132H)-specific peptide vaccine (IDH1-vac) induces specific therapeutic T helper cell responses that are effective against IDH1(R132H)+ tumours in syngeneic MHC-humanized mice4,6-8. Here we describe a multicentre, single-arm, open-label, first-in-humans phase I trial that we carried out in 33 patients with newly diagnosed World Health Organization grade 3 and 4 IDH1(R132H)+ astrocytomas (Neurooncology Working Group of the German Cancer Society trial 16 (NOA16), ClinicalTrials.gov identifier NCT02454634). The trial met its primary safety endpoint, with vaccine-related adverse events restricted to grade 1. Vaccine-induced immune responses were observed in 93.3% of patients across multiple MHC alleles. Three-year progression-free and death-free rates were 0.63 and 0.84, respectively. Patients with immune responses showed a two-year progression-free rate of 0.82. Two patients without an immune response showed tumour progression within two years of first diagnosis. A mutation-specificity score that incorporates the duration and level of vaccine-induced IDH1(R132H)-specific T cell responses was associated with intratumoral presentation of the IDH1(R132H) neoantigen in pre-treatment tumour tissue. There was a high frequency of pseudoprogression, which indicates intratumoral inflammatory reactions. Pseudoprogression was associated with increased vaccine-induced peripheral T cell responses. Combined single-cell RNA and T cell receptor sequencing showed that tumour-infiltrating CD40LG+ and CXCL13+ T helper cell clusters in a patient with pseudoprogression were dominated by a single IDH1(R132H)-reactive T cell receptor.
DOI: 10.1038/s41586-021-03850-3
2021
Cited 200 times
A clinically applicable integrative molecular classification of meningiomas
DOI: 10.1212/wnl.0b013e3182a95680
2013
Cited 199 times
Prognostic or predictive value of <i>MGMT</i> promoter methylation in gliomas depends on <i>IDH1</i> mutation
To explore whether the isocitrate dehydrogenase 1 (IDH1) or 1p/19q status determines the prognostic vs predictive role of O(6)-methylguanine-DNA methyltransferase (MGMT) promoter methylation in the Neuro-Oncology Working Group of the German Cancer Society (NOA)-04 trial anaplastic glioma biomarker cohort.Patients (n = 183) of the NOA-04 trial with known MGMT and IDH1 status were analyzed for interdependency of the prognostic vs predictive role of MGMT promoter methylation from IDH1 or 1p/19q status and treatment, using progression-free survival (PFS) as an endpoint. An independent validation cohort of the German Glioma Network (n = 75) and the NOA-08 trial (n = 34) served as a confirmation cohort.In tumors with IDH1 mutation, MGMT promoter methylation was associated with prolonged PFS with chemotherapy ± radiotherapy (RT) or RT-only groups, and is thus prognostic. In tumors without IDH1 mutation, MGMT promoter methylation was associated with increased PFS in patients treated with chemotherapy, but not in those who received RT alone as the first-line treatment, and is thus chemotherapy-predictive. In contrast, 1p/19q codeletions showed no such association with the prognostic vs predictive value of MGMT.MGMT promoter methylation is a predictive biomarker for benefit from alkylating agent chemotherapy in patients with IDH1-wild-type, but not IDH1-mutant, malignant gliomas of World Health Organization grades III/IV. Combined IDH1/MGMT assessment may help to individualize clinical decision-making in neuro-oncology.
DOI: 10.1093/neuonc/nov326
2016
Cited 188 times
Complete resection of contrast-enhancing tumor volume is associated with improved survival in recurrent glioblastoma—results from the DIRECTOR trial
The role of reoperation for recurrent glioblastoma (GBM) remains unclear. Prospective studies are lacking. Here, we studied the association of clinical outcome with extent of resection upon surgery for recurrent GBM in the patient cohort of DIRECTOR, a prospective randomized multicenter trial comparing 2 dose-intensified temozolomide regimens at recurrence of GBM.We analyzed prospectively collected clinical and imaging data from the DIRECTOR cohort (N = 105). Volumetric analysis was performed on gadolinium contrast-enhanced MRI as well as fluid attenuated inversion recovery/T2 MRI and correlated with PFS after initial progression (PFS2) and post-recurrence survival (PRS). Quality of life was monitored by the EORTC QLQ-C30 and QLQ-BN20 questionnaires at 8-week intervals.Seventy-one patients received surgery at first recurrence. Prognostic factors, including age, MGMT promoter methylation, and Karnofsky performance score, were balanced between patients with and without reoperation. Outcome in patients with versus without surgery at recurrence was similar for PFS2 (2.0 mo vs 1.9 mo, P = .360) and PRS (11.4 mo vs 9.8 mo, P = .633). Among reoperated patients, post-surgery imaging was available in 59 cases. In these patients, complete resection of contrast-enhancing tumor (N = 40) versus residual detection of contrast enhancement (N = 19) was associated with improved PRS (12.9 mo [95% CI: 11.5-18.2] vs 6.5 mo [95% CI: 3.6-9.9], P < .001) and better quality of life. Incomplete tumor resection was associated with inferior PRS compared with patients who did not undergo surgery (6.5 vs 9.8 mo, P = .052). Quality of life was similar in these 2 groups.Surgery at first recurrence of GBM improves outcome if complete resection of contrast-enhancing tumor is achieved.
DOI: 10.1093/neuonc/noz061
2019
Cited 173 times
DNA methylation profiling to predict recurrence risk in meningioma: development and validation of a nomogram to optimize clinical management
Abstract Background Variability in standard-of-care classifications precludes accurate predictions of early tumor recurrence for individual patients with meningioma, limiting the appropriate selection of patients who would benefit from adjuvant radiotherapy to delay recurrence. We aimed to develop an individualized prediction model of early recurrence risk combining clinical and molecular factors in meningioma. Methods DNA methylation profiles of clinically annotated tumor samples across multiple institutions were used to develop a methylome model of 5-year recurrence-free survival (RFS). Subsequently, a 5-year meningioma recurrence score was generated using a nomogram that integrated the methylome model with established prognostic clinical factors. Performance of both models was evaluated and compared with standard-of-care models using multiple independent cohorts. Results The methylome-based predictor of 5-year RFS performed favorably compared with a grade-based predictor when tested using the 3 validation cohorts (ΔAUC = 0.10, 95% CI: 0.03–0.018) and was independently associated with RFS after adjusting for histopathologic grade, extent of resection, and burden of copy number alterations (hazard ratio 3.6, 95% CI: 1.8–7.2, P &lt; 0.001). A nomogram combining the methylome predictor with clinical factors demonstrated greater discrimination than a nomogram using clinical factors alone in 2 independent validation cohorts (ΔAUC = 0.25, 95% CI: 0.22–0.27) and resulted in 2 groups with distinct recurrence patterns (hazard ratio 7.7, 95% CI: 5.3–11.1, P &lt; 0.001) with clinical implications. Conclusions The models developed and validated in this study provide important prognostic information not captured by previously established clinical and molecular factors which could be used to individualize decisions regarding postoperative therapeutic interventions, in particular whether to treat patients with adjuvant radiotherapy versus observation alone.
DOI: 10.1093/neuonc/noac099
2022
Cited 164 times
Radiotherapy combined with nivolumab or temozolomide for newly diagnosed glioblastoma with unmethylated <i>MGMT</i> promoter: An international randomized phase III trial
Addition of temozolomide (TMZ) to radiotherapy (RT) improves overall survival (OS) in patients with glioblastoma (GBM), but previous studies suggest that patients with tumors harboring an unmethylated MGMT promoter derive minimal benefit. The aim of this open-label, phase III CheckMate 498 study was to evaluate the efficacy of nivolumab (NIVO) + RT compared with TMZ + RT in newly diagnosed GBM with unmethylated MGMT promoter.Patients were randomized 1:1 to standard RT (60 Gy) + NIVO (240 mg every 2 weeks for eight cycles, then 480 mg every 4 weeks) or RT + TMZ (75 mg/m2 daily during RT and 150-200 mg/m2/day 5/28 days during maintenance). The primary endpoint was OS.A total of 560 patients were randomized, 280 to each arm. Median OS (mOS) was 13.4 months (95% CI, 12.6 to 14.3) with NIVO + RT and 14.9 months (95% CI, 13.3 to 16.1) with TMZ + RT (hazard ratio [HR], 1.31; 95% CI, 1.09 to 1.58; P = .0037). Median progression-free survival was 6.0 months (95% CI, 5.7 to 6.2) with NIVO + RT and 6.2 months (95% CI, 5.9 to 6.7) with TMZ + RT (HR, 1.38; 95% CI, 1.15 to 1.65). Response rates were 7.8% (9/116) with NIVO + RT and 7.2% (8/111) with TMZ + RT; grade 3/4 treatment-related adverse event (TRAE) rates were 21.9% and 25.1%, and any-grade serious TRAE rates were 17.3% and 7.6%, respectively.The study did not meet the primary endpoint of improved OS; TMZ + RT demonstrated a longer mOS than NIVO + RT. No new safety signals were detected with NIVO in this study. The difference between the study treatment arms is consistent with the use of TMZ + RT as the standard of care for GBM.ClinicalTrials.gov NCT02617589.
DOI: 10.1093/neuonc/noad008
2023
Cited 27 times
EANO guideline on rational molecular testing of gliomas, glioneuronal, and neuronal tumors in adults for targeted therapy selection
The mainstay of treatment for adult patients with gliomas, glioneuronal and neuronal tumors consists of combinations of surgery, radiotherapy, and chemotherapy. For many systemic cancers, targeted treatments are a part of the standard of care, however, the predictive significance of most of these targets in central nervous system (CNS) tumors remains less well-studied. Despite that, there is increasing use of advanced molecular diagnostics that identify potential targets, and tumor-agnostic regulatory approvals on targets also present in CNS tumors have been granted. This raises the question of when and for which targets it is meaningful to test in adult patients with CNS tumors. This evidence-based guideline reviews the evidence available for targeted treatment for alterations in the RAS/MAPK pathway (BRAF, NF1), in growth factor receptors (EGFR, ALK, fibroblast growth factor receptor (FGFR), neurotrophic tyrosine receptor kinase (NTRK), platelet-derived growth factor receptor alpha, and ROS1), in cell cycle signaling (CDK4/6, MDM2/4, and TSC1/2) and altered genomic stability (mismatch repair, POLE, high tumor mutational burden (TMB), homologous recombination deficiency) in adult patients with gliomas, glioneuronal and neuronal tumors. At present, targeted treatment for BRAF p.V600E alterations is to be considered part of the standard of care for patients with recurrent gliomas, pending regulatory approval. For approved tumor agnostic treatments for NTRK fusions and high TMB, the evidence for efficacy in adult patients with CNS tumors is very limited, and treatment should preferably be given within prospective clinical registries and trials. For targeted treatment of CNS tumors with FGFR fusions or mutations, clinical trials are ongoing to confirm modest activity so far observed in basket trials. For all other reviewed targets, evidence of benefit in CNS tumors is currently lacking, and testing/treatment should be in the context of available clinical trials.
DOI: 10.1038/emboj.2011.301
2011
Cited 150 times
CAMTA1 is a novel tumour suppressor regulated by miR-9/9<sup>*</sup>in glioblastoma stem cells
Cancer stem cells or cancer initiating cells are believed to contribute to cancer recurrence after therapy. MicroRNAs (miRNAs) are short RNA molecules with fundamental roles in gene regulation. The role of miRNAs in cancer stem cells is only poorly understood. Here, we report miRNA expression profiles of glioblastoma stem cell-containing CD133(+) cell populations. We find that miR-9, miR-9(*) (referred to as miR-9/9(*)), miR-17 and miR-106b are highly abundant in CD133(+) cells. Furthermore, inhibition of miR-9/9(*) or miR-17 leads to reduced neurosphere formation and stimulates cell differentiation. Calmodulin-binding transcription activator 1 (CAMTA1) is a putative transcription factor, which induces the expression of the anti-proliferative cardiac hormone natriuretic peptide A (NPPA). We identify CAMTA1 as an miR-9/9(*) and miR-17 target. CAMTA1 expression leads to reduced neurosphere formation and tumour growth in nude mice, suggesting that CAMTA1 can function as tumour suppressor. Consistently, CAMTA1 and NPPA expression correlate with patient survival. Our findings could provide a basis for novel strategies of glioblastoma therapy.
DOI: 10.1158/1078-0432.ccr-10-0705
2010
Cited 129 times
GDF-15 Contributes to Proliferation and Immune Escape of Malignant Gliomas
Abstract Purpose: Growth and differentiation factor (GDF)-15 is a member of the transforming growth factor (TGF)-β family. GDF-15 is necessary for the maintenance of pregnancy but has also been linked to other physiologic and pathologic conditions. Experimental Design: The expression of GDF-15 in glioma cell lines was assessed by quantitative reverse transcriptase-PCR and immunoblot. GDF-15 levels in situ and in the peripheral blood of glioma patients were examined by immunohistochemistry and enzyme-linked immunosorbent assay, respectively. The effects of short hairpin RNA-mediated GDF-15 inhibition on proliferation and immunogenicity of SMA-560 glioma cells were investigated by [methyl-3H]thymidine incorporation and immune-mediated target cell lysis. The impact of GDF-15 on glioma growth in vivo was assessed in syngeneic mice. Results: GDF-15 is expressed by gliomas of different WHO grades as assessed by immunohistochemistry. The high expression of GDF-15 in tumor tissue translates into elevated GDF-15 serum levels in glioblastoma patients compared with healthy controls. GDF-15 mRNA and protein are also detectable in human and mouse glioma cells in vitro. Silencing of GDF-15 by RNA interference reduces the proliferation of malignant glioma cells. Immunologically, the depletion of glioma-derived GDF-15 enhances the susceptibility of mouse glioma cells towards syngeneic natural killer cells and splenocytes. This results in a reduced in vivo tumorigenicity and increased T-cell infiltration of GDF-15–deficient glioma cells in syngeneic mice. Conclusions: Although previous studies focusing on ectopic overexpression of GDF-15 have proposed unclear or antitumorigenic effects of GDF-15 in glioma cells, we here show that GDF-15 at endogenous levels contributes to proliferation and immune escape of malignant gliomas in an immunocompetent host. Clin Cancer Res; 16(15); 3851–9. ©2010 AACR.
DOI: 10.1007/s00401-010-0750-6
2010
Cited 128 times
Molecular diagnostics of gliomas: the clinical perspective
Significant progress has been made in the molecular diagnostic subtyping of brain tumors, in particular gliomas. In contrast to the classical molecular markers in this field, p53 and epidermal growth factor receptor (EGFR) status, the clinical significance of which has remained controversial, at least three important molecular markers with clinical implications have now been identified: 1p/19q codeletion, O 6-methylguanine methyltransferase (MGMT) promoter methylation and isocitrate dehydrogenase-1 (IDH1) mutations. All three are favorable prognostic markers. 1p/19q codeletion and IDH1 mutations are also useful to support and extend the histological classification of gliomas since they are strongly linked to oligodendroglial morphology and grade II/III gliomas, as opposed to glioblastoma, respectively. MGMT promoter methylation is the only potentially predictive marker, at least for alkylating agent chemotherapy in glioblastoma. Beyond these classical markers, the increasing repertoire of anti-angiogenic agents that are currently explored within registration trials for gliomas urgently calls for efforts to identify molecular markers that predict the benefit derived from these novel treatments, too.
DOI: 10.1002/cam4.50
2012
Cited 125 times
Targeting hyperactivation of the <scp>AKT</scp> survival pathway to overcome therapy resistance of melanoma brain metastases
Brain metastases are the most common cause of death in patients with metastatic melanoma, and the RAF-MEK-ERK and PI3K-AKT signaling pathways are key players in melanoma progression and drug resistance. The BRAF inhibitor vemurafenib significantly improved overall survival. However, brain metastases still limit the effectiveness of this therapy. In a series of patients, we observed that treatment with vemurafenib resulted in substantial regression of extracerebral metastases, but brain metastases developed. This study aimed to identify factors that contribute to treatment resistance in brain metastases. Matched brain and extracerebral metastases from melanoma patients had identical ERK, p-ERK, and AKT immunohistochemistry staining patterns, but there was hyperactivation of AKT (p-AKT) and loss of PTEN expression in the brain metastases. Mutation analysis revealed no differences in BRAF, NRAS, or KIT mutation status in matched brain and extracerebral metastases. In contrast, AKT, p-AKT, and PTEN expression was identical in monolayer cultures derived from melanoma brain and extracerebral metastases. Furthermore, melanoma cells stimulated by astrocyte-conditioned medium showed higher AKT activation and invasiveness than melanoma cells stimulated by fibroblast-conditioned medium. Inhibition of PI3K-AKT signaling resensitized melanoma cells isolated from a vemurafenib-resistant brain metastasis to vemurafenib. Brain-derived factors appear to induce hyperactivation of the AKT survival pathway and to promote the survival and drug resistance of melanoma cells in the brain. Thus, inhibition of PI3K-AKT signaling shows potential for enhancing and/or prolonging the antitumor effect of BRAF inhibitors or other anticancer agents in melanoma brain metastases.
DOI: 10.1093/neuonc/noy020
2018
Cited 114 times
Glioma through the looking GLASS: molecular evolution of diffuse gliomas and the Glioma Longitudinal Analysis Consortium
Adult diffuse gliomas are a diverse group of brain neoplasms that inflict a high emotional toll on patients and their families. The Cancer Genome Atlas and similar projects have provided a comprehensive understanding of the somatic alterations and molecular subtypes of glioma at diagnosis. However, gliomas undergo significant cellular and molecular evolution during disease progression. We review the current knowledge on the genomic and epigenetic abnormalities in primary tumors and after disease recurrence, highlight the gaps in the literature, and elaborate on the need for a new multi-institutional effort to bridge these knowledge gaps and how the Glioma Longitudinal Analysis Consortium (GLASS) aims to systemically catalog the longitudinal changes in gliomas. The GLASS initiative will provide essential insights into the evolution of glioma toward a lethal phenotype, with the potential to reveal targetable vulnerabilities and, ultimately, improved outcomes for a patient population in need.
DOI: 10.1093/neuonc/noy143
2019
Cited 107 times
Imaging and diagnostic advances for intracranial meningiomas
The archetypal imaging characteristics of meningiomas are among the most stereotypic of all central nervous system (CNS) tumors. In the era of plain film and ventriculography, imaging was only performed if a mass was suspected, and their results were more suggestive than definitive. Following more than a century of technological development, we can now rely on imaging to non-invasively diagnose meningioma with great confidence and precisely delineate the locations of these tumors relative to their surrounding structures to inform treatment planning. Asymptomatic meningiomas may be identified and their growth monitored over time; moreover, imaging routinely serves as an essential tool to survey tumor burden at various stages during the course of treatment, thereby providing guidance on their effectiveness or the need for further intervention. Modern radiological techniques are expanding the power of imaging from tumor detection and monitoring to include extraction of biologic information from advanced analysis of radiological parameters. These contemporary approaches have led to promising attempts to predict tumor grade and, in turn, contribute prognostic data. In this supplement article, we review important current and future aspects of imaging in the diagnosis and management of meningioma, including conventional and advanced imaging techniques using CT, MRI, and nuclear medicine.
DOI: 10.1093/neuonc/noy136
2019
Cited 105 times
Advances in multidisciplinary therapy for meningiomas
Surgery has long been established as the first-line treatment for the majority of symptomatic and enlarging meningiomas, and evidence for its success is derived from retrospective case series. Despite surgical resection, a subset of meningiomas display aggressive behavior with early recurrences that are difficult to treat. The decision to radically resect meningiomas and involved structures is balanced against the risk for neurological injury in patients. Radiation therapy has largely been used as a complementary and safe therapeutic strategy in meningiomas with evidence primarily stemming from retrospective, single-institution reports. Two of the first cooperative group studies (RTOG 0539 and EORTC 22042) evaluating the outcomes of adjuvant radiation therapy in higher-risk meningiomas have shown promising preliminary results. Historically, systemic therapy has resulted in disappointing results in meningiomas. However, several clinical trials are under way evaluating the efficacy of chemotherapies, such as trabectedin, and novel molecular agents targeting Smoothened, AKT1, and focal adhesion kinase in patients with recurrent meningiomas.
DOI: 10.1186/s40478-016-0331-6
2016
Cited 102 times
ATRX immunostaining predicts IDH and H3F3A status in gliomas
Gliomas are the most frequent intraaxial CNS neoplasms with a heterogeneous molecular background. Recent studies on diffuse gliomas have shown frequent alterations in the genes involved in chromatin remodelling pathways such as α-thalassemia/mental-retardation-syndrome-X-linked gene (ATRX). Yet, the reliability of ATRX in predicting isocitrate dehydrogenase (IDH) and H3 histone, family 3A (H3F3A) mutations in gliomas, is unclear.We analysed the ATRX expression status by immunohistochemistry, in a large series of 1064 gliomas and analysed the results in correlation to IDH, H3F3A and loss of heterozygosity (LOH) 1p/19q status in these tumors. We also investigated the prognostic potential of ATRX concerning the clinical outcome of patients with diffuse gliomas.According to our results, loss of nuclear ATRX expression was accompanied with an astrocytic tumor lineage and a younger age of onset. ATRX loss in astrocytomas was also strongly associated with IDH1/2 and H3F3A mutation (p < 0.0001). Among 196 glial tumors with nuclear ATRX loss, 173 (89 %) had an IDH1 or IDH2 mutation. Among the remaining 23 cases (11 %) with ATRX loss and IDH wild type status, 7 cases had a H3F3A G34R mutation (3 %) and 2 cases had a H3F3A K27M mutation (1 %). ATRX retention in IDH1/2 mutant tumors was strongly associated with LOH 1p/19q and oligodendroglioma histology (p < 0.0001). We also confirmed the significant prognostic role of ATRX. Diffuse gliomas with ATRX loss (n = 137, median 1413 days, 95 % CI: 1065-1860 days) revealed a significantly better clinical outcome compared with tumors with ATRX retention (n = 335, median: 609, 95 % CI: 539-760 days, HR = 1.81, p < 0.0001).In conclusion, ATRX is a potential marker for prediction of IDH/H3F3A mutations and substratification of diffuse gliomas into survival relevant tumor groups. Such classification is of great importance for further clinical decision making especially concerning the therapeutic options available for diffuse gliomas.
DOI: 10.1093/brain/aws351
2013
Cited 98 times
Integrin control of the transforming growth factor-β pathway in glioblastoma
Transforming growth factor-b is a central mediator of the malignant phenotype of glioblastoma, the most common and malignant form of intrinsic brain tumours.Transforming growth factor-b promotes invasiveness and angiogenesis, maintains cancer cell stemness and induces profound immunosuppression in the host.Integrins regulate cellular adhesion and transmit signals important for cell survival, proliferation, differentiation and motility, and may be involved in the activation of transforming growth factor-b.We report that avb3, avb5 and avb8 integrins are broadly expressed not only in glioblastoma blood vessels but also in tumour cells.Exposure to av, b3 or b5 neutralizing antibodies, RNA interference-mediated integrin gene silencing or pharmacological integrin inhibition using the cyclic RGD peptide EMD 121974 (cilengitide) results in reduced phosphorylation of Smad2 in most glioma cell lines, including glioma-initiating cell lines and reduced transforming growth factor-b-mediated reporter gene activity, coinciding with reduced transforming growth factor-b protein levels in the supernatant.Time course experiments indicated that the loss of transforming growth factor-b bioactivity due to integrin inhibition likely results from two distinct mechanisms: an early effect on activation of preformed inactive protein, and second, major effect on transforming growth factor-b gene transcription as confirmed by decreased activity of the transforming growth factor-b gene promoter and decreased transforming growth factor-b 1 and transforming growth factor-b 2 messenger RNA expression levels.In vivo, EMD 121974 (cilengitide), which is currently in late clinical development as an antiangiogenic agent in newly diagnosed glioblastoma, was a weak antagonist of pSmad2 phosphorylation.These results validate integrin inhibition as a promising strategy not only to inhibit angiogenesis, but also to block transforming growth factor-b-controlled features of malignancy including invasiveness, stemness and immunosuppression in human glioblastoma.
DOI: 10.1093/neuonc/noy178
2019
Cited 91 times
Molecular and translational advances in meningiomas
Meningiomas are the most common primary intracranial neoplasm. The current World Health Organization (WHO) classification categorizes meningiomas based on histopathological features, but emerging molecular data demonstrate the importance of genomic and epigenomic factors in the clinical behavior of these tumors. Treatment options for symptomatic meningiomas are limited to surgical resection where possible and adjuvant radiation therapy for tumors with concerning histopathological features or recurrent disease. At present, alternative adjuvant treatment options are not available in part due to limited historical biological analysis and clinical trial investigation on meningiomas. With advances in molecular and genomic techniques in the last decade, we have witnessed a surge of interest in understanding the genomic and epigenomic landscape of meningiomas. The field is now at the stage to adopt this molecular knowledge to refine meningioma classification and introduce molecular algorithms that can guide prediction and therapeutics for this tumor type. Animal models that recapitulate meningiomas faithfully are in critical need to test new therapeutics to facilitate rapid-cycle translation to clinical trials. Here we review the most up-to-date knowledge of molecular alterations that provide insight into meningioma behavior and are ready for application to clinical trial investigation, and highlight the landscape of available preclinical models in meningiomas.
DOI: 10.1158/2326-6066.cir-18-0865
2019
Cited 77 times
Tumor Vessel Normalization, Immunostimulatory Reprogramming, and Improved Survival in Glioblastoma with Combined Inhibition of PD-1, Angiopoietin-2, and VEGF
Abstract Glioblastoma (GBM) is a non-T-cell–inflamed cancer characterized by an immunosuppressive microenvironment that impedes dendritic cell maturation and T-cell cytotoxicity. Proangiogenic cytokines such as VEGF and angiopoietin-2 (Ang-2) have high expression in glioblastoma in a cell-specific manner and not only drive tumor angiogenesis and vascular permeability but also negatively regulate T-lymphocyte and innate immune cell responses. Consequently, the alleviation of immunosuppression might be a prerequisite for successful immune checkpoint therapy in GBM. We here combined antiangiogenic and immune checkpoint therapy and demonstrated improved therapeutic efficacy in syngeneic, orthotopic GBM models. We observed that blockade of VEGF, Ang-2, and programmed cell death protein-1 (PD-1) significantly extended survival compared with vascular targeting alone. In the GBM microenvironment, triple therapy increased the numbers of CTLs, which inversely correlated with myeloid-derived suppressor cells and regulatory T cells. Transcriptome analysis of GBM microvessels indicated a global vascular normalization that was highest after triple therapy. Our results propose a rationale to overcome tumor immunosuppression and the current limitations of VEGF monotherapy by integrating the synergistic effects of VEGF/Ang-2 and PD-1 blockade to reinforce antitumor immunity through a normalized vasculature.
DOI: 10.1093/neuonc/noy152
2019
Cited 63 times
Life after surgical resection of a meningioma: a prospective cross-sectional study evaluating health-related quality of life
Few studies have evaluated the health-related quality of life (HRQoL) of patients with meningiomas. Here, we report the largest prospective, longitudinal cross-sectional cohort study of HRQoL in meningiomas to date, in order to identify possible actionable determinants of global HRQoL.Adults who had undergone resection of a grade I intracranial meningioma and were in routine follow-up at a single large tertiary center underwent HRQoL assessment using the QLQ-C30 questionnaire administered opportunistically at follow-up visits. Averaged transformed QLQ-C30 scores at 12-month intervals were compared with scores from a normative reference population, with reference to known minimal clinically meaningful difference (CMD) in scores. To evaluate for possible determinants of changes in global HRQoL, global HRQoL scores were correlated (Spearman's Rho) with subdomain and symptom scores and with interval time from surgical resection.A total of 291 postoperative patients with histologically confirmed and surgically treated grade I meningiomas consented to participation and a total of 455 questionnaires were included for analysis. Patients with meningiomas reported reduced global HRQoL at nearly every 12-month interval with clinically and statistically significant impairments at 12, 48, 108, and 120 months postoperative compared with the normative population (P < 0.05). Meningioma patients at the 12-month interval also reported a reduction of each subdomain of HRQoL assessment (P < 0.05); however, a CMD was only seen in cognitive functioning. Physical, emotional, cognitive, and social subdomains, as well as fatigue and sleep/insomnia, were significantly associated with global HRQoL at the first 12-month interval. Overall, there was no significant correlation between time from surgery and global HRQoL or the subdomain functional or symptom sections of the QLQ-C30.Meningioma patients report considerable limitations in HRQoL for more than 120 months after surgery, particularly in cognitive, emotional, and social function, as well as suffering significant fatigue and sleep impairment compared with a normative reference population. The majority of these reported functional impairments and symptoms are strongly associated with global HRQoL and thus can be considered determinants of global HRQoL that if treated, have the potential to improve HRQoL for our meningioma patients. This hypothesis requires future study of targeted interventions to determine their efficacy.
DOI: 10.1093/neuonc/noab036
2021
Cited 50 times
Loss of H3K27me3 in meningiomas
Abstract Background There is a critical need for objective and reliable biomarkers of outcome in meningiomas beyond WHO classification. Loss of H3K27me3 has been reported as a prognostically unfavorable alteration in meningiomas. We sought to independently evaluate the reproducibility and prognostic value of H3K27me3 loss by immunohistochemistry (IHC) in a multicenter study. Methods IHC staining for H3K27me3 and analyses of whole slides from 181 meningiomas across three centers was performed. Staining was analyzed by dichotomization into loss and retained immunoreactivity, and using a 3-tiered scoring system in 151 cases with clear staining. Associations of grouping with outcome were performed using Kaplan-Meier survival estimates. Results A total of 21 of 151 tumors (13.9%) demonstrated complete loss of H3K27me3 staining in tumor with retained endothelial staining. Overall, loss of H3K27me3 portended a worse outcome with shorter times to recurrence in our cohort, particularly for WHO grade 2 tumors which were enriched in our study. There were no differences in recurrence-free survival (RFS) for WHO grade 3 patients with retained vs loss of H3K27me3. Scoring by a 3-tiered system did not add further insights into the prognostic value of this H3K27me3 loss. Overall, loss of H3K27me3 was not independently associated with RFS after controlling for WHO grade, extent of resection, sex, age, and recurrence status of tumor on multivariable Cox regression analysis. Conclusions Loss of H3K27me3 identifies a subset of WHO grade 2 and possibly WHO grade 1 meningiomas with increased recurrence risk. Pooled analyses of a larger cohort of samples with standardized reporting of clinical definitions and staining patterns are warranted.
DOI: 10.1093/neuonc/noac188
2022
Cited 28 times
EANO - EURACAN - SNO Guidelines on circumscribed astrocytic gliomas, glioneuronal, and neuronal tumors
In the new WHO 2021 Classification of CNS Tumors the chapter "Circumscribed astrocytic gliomas, glioneuronal and neuronal tumors" encompasses several different rare tumor entities, which occur more frequently in children, adolescents, and young adults. The Task Force has reviewed the evidence of diagnostic and therapeutic interventions, which is low particularly for adult patients, and draw recommendations accordingly. Tumor diagnosis, based on WHO 2021, is primarily performed using conventional histological techniques; however, a molecular workup is important for differential diagnosis, in particular, DNA methylation profiling for the definitive classification of histologically unresolved cases. Molecular factors are increasing of prognostic and predictive importance. MRI finding are non-specific, but for some tumors are characteristic and suggestive. Gross total resection, when feasible, is the most important treatment in terms of prolonging survival and achieving long-term seizure control. Conformal radiotherapy should be considered in grade 3 and incompletely resected grade 2 tumors. In recurrent tumors reoperation and radiotherapy, including stereotactic radiotherapy, can be useful. Targeted therapies may be used in selected patients: BRAF and MEK inhibitors in pilocytic astrocytomas, pleomorphic xanthoastrocytomas, and gangliogliomas when BRAF altered, and mTOR inhibitor everolimus in subependymal giant cells astrocytomas. Sequencing to identify molecular targets is advocated for diagnostic clarification and to direct potential targeted therapies.
DOI: 10.1038/s41591-023-02354-z
2023
Cited 17 times
The German Network for Personalized Medicine to enhance patient care and translational research
DOI: 10.1007/s00401-023-02571-3
2023
Cited 10 times
Increased mRNA expression of CDKN2A is a transcriptomic marker of clinically aggressive meningiomas
Abstract Homozygous deletion of CDKN2A/B was recently incorporated into the World Health Organization classification for grade 3 meningiomas. While this marker is overall rare in meningiomas, its relationship to other CDKN2A alterations on a transcriptomic, epigenomic, and copy number level has not yet been determined. We therefore utilized multidimensional molecular data of 1577 meningioma samples from 6 independent cohorts enriched for clinically aggressive meningiomas to comprehensively interrogate the spectrum of CDKN2A alterations through DNA methylation, copy number variation, transcriptomics, and proteomics using an integrated molecular approach. Homozygous CDKN2A/B deletions were identified in only 7.1% of cases but were associated with significantly poorer outcomes compared to tumors without these deletions. Heterozygous CDKN2A/B deletions were identified in 2.6% of cases and had similarly poor outcomes as those with homozygous deletions. Among tumors with intact CDKN2A/B (without a homozygous or heterozygous deletion), we found a distinct difference in outcome based on mRNA expression of CDKN2A, with meningiomas that had elevated mRNA expression (CDKN2A high ) having a significantly shorter time to recurrence. The expression of CDKN2A was independently prognostic after accounting for copy number loss and consistently increased with WHO grade and more aggressive molecular and methylation groups irrespective of cohort. Despite the discordant and mutually exclusive status of the CDKN2A gene in these groups, both CDKN2A high meningiomas and meningiomas with CDKN2A deletions were enriched for similar cell cycle pathways but at different checkpoints. High mRNA expression of CDKN2A was also associated with gene hypermethylation, Rb-deficiency, and lack of response to CDK inhibition. p16 immunohistochemistry could not reliably differentiate between meningiomas with and without CDKN2A deletions but appeared to correlate better with mRNA expression. These findings support the role of CDKN2A mRNA expression as a biomarker of clinically aggressive meningiomas with potential therapeutic implications.
DOI: 10.1016/j.ctro.2023.100706
2024
Dosimetric feasibility analysis and presentation of an isotoxic dose-escalated radiation therapy concept for glioblastoma used in the PRIDE trial (NOA-28; ARO-2022-12)
Background and purposeThe PRIDE trial (NOA-28; ARO-2022-12; NCT05871021) is scheduled to start recruitment in October 2023. Its primary objective is to enhance median overall survival (OS), compared to historical median OS rates, in patients with methylguanine methlyltransferase (MGMT) promotor unmethylated glioblastoma by incorporating isotoxic dose escalation to 75 Gy in 30 fractions. To achieve isotoxicity and counteract the elevated risk of radiation necrosis (RN) associated with dose-escalated regimens, the addition of protective concurrent bevacizumab (BEV) serves as an innovative approach. The current study aims to assess the dosimetric feasibility of the proposed concept.Materials and methodsA total of ten patients diagnosed with glioblastoma were included in this dosimetric analysis. Delineation of target volumes for the reference plans adhered to the ESTRO-EANO 2023 guideline. The experimental plans included an additional volume for the integrated boost. Additionally, the 60 Gy-volume was reduced by using a margin of 1.0 cm instead of 1.5 cm. To assess the risk of symptomatic RN, the Normal Tissue Complication Probability (NTCP) was calculated and compared between the reference and experimental plans.ResultsMedian NTCP of the reference plan (NTCPref) and of the experimental plan (NTCPex) were 0.24 (range 0.11–0.29) and 0.42 (range 0.18–0.54), respectively. NTCPex was a median of 1.77 (range 1.60–1.99) times as high as the NTXPref. In a logarithmic comparison, the risk of RN is enhanced by a factor of median 2.00 (range 1.66–2.35). The defined constraints for the organs at risk were feasible.ConclusionWhen considering the potential protective effect of BEV, which we hypothesized might reduce the risk of RN by approximately two-fold, achieving isotoxicity with the proposed dose-escalated experimental plan for the PRIDE trial seems feasible.
DOI: 10.1002/jnr.21224
2007
Cited 118 times
Expression pattern of the water channel aquaporin‐4 in human gliomas is associated with blood–brain barrier disturbance but not with patient survival
Abstract Aquaporin‐4 (AQP4), the most prominent CNS water channel, is restricted to the glia limitans and astrocytic endfeet. We previously showed the loss of spatial AQP4 expression in glioblastomas and a redistribution across the cell surface. However, opposing AQP4 functions have been described: protective in vasogenic but detrimental in cytotoxic brain edema. Thus, specific AQP4 induction to prevent or reduce vasogenic edema is suggested. To elucidate the AQP4 role in brain tumors, we investigated 189 WHO grade I–IV gliomas by immunohistochemistry and the prognostic significance for patients' survival. In gliomas, a remarkable de novo AQP4 redistribution was observed in comparison with normal CNS tissue. Surprisingly, the highest membraneous staining levels were seen in pilocytic astrocytomas WHO grade I and grade IV glioblastomas, both significantly higher than in WHO grade II astrocytomas. AQP4 up‐regulation was associated with brain edema formation; however, no association between survival and WHO grade‐dependent AQP4 expression was seen. Hence, AQP4 redistribution may go along with other tumor properties, such as vascular proliferation and resulting blood–brain barrier disturbance, features usually prominent in pilocytic astrocytomas WHO I and glioblastomas WHO grade IV. In summary, our findings question the protective role of AQP4 in vasogenic brain edema. Although AQP4 was associated with brain edema formation, one has to question the suitability of AQP4 induction as a promising approach in vasogenic brain edema prevention and treatment. In addition, our results provide unexpectedly high AQP4 levels in pilocytic astrocytomas and present AQP4 as tumor progression marker in WHO grade II–IV astrocytomas. © 2007 Wiley‐Liss, Inc.
DOI: 10.1093/brain/awl173
2006
Cited 118 times
Irradiation and hypoxia promote homing of haematopoietic progenitor cells towards gliomas by TGF- -dependent HIF-1 -mediated induction of CXCL12
Previously we defined a pathway of transforming growth factor beta (TGF-β) and stromal cell-derived factor-1/CXC chemokine ligand 12 (SDF-1α/CXCL12) dependent migration of adult haematopoietic stem and progenitor cells (HPC) towards glioma cells in vitro and their homing to experimental gliomas in vivo. Hypoxia is a critical aspect of the microenvironment of gliomas and irradiation is an essential part of the standard therapy. To evaluate the therapeutic potential of HPC as vectors for a cell-based therapy of gliomas, we investigated the impact of hypoxia and irradiation on the attraction of HPC by glioma cells. Temozolomide (TMZ) treatment and hyperthermia served as controls. Supernatants of irradiated or hypoxic LNT-229 glioma cells promote HPC migration in vitro. Reporter assays reveal that the CXCL12 promoter activity is enhanced in LNT-229 cells at 24 h after irradiation at 8 Gy or after exposure to 1% oxygen for 12 h. The irradiation- and hypoxia-induced release of CXCL12 depends on hypoxia inducible factor-1 alpha (HIF-1α), but not on p53. Induction of transcriptional activity of HIF-1α by hypoxia or irradiation requires an intact TGF-β signalling cascade. This delineates a novel stress signalling cascade in glioma cells involving TGF-β, HIF-1α and CXCL12. Stress stimuli can be irradiation, hypoxia or TMZ, but not hyperthermia. Cerebral irradiation of nude mice at 21 days after intracerebral implantation of LNT-229 glioma induces tumour satellite formation and enhances the glioma tropism of HPC to the tumour bulk and even to these satellites in vivo. These data suggest that the use of HPC as cellular vectors in the treatment of glioblastoma may well be combined with irradiation or other anti-angiogenic therapies that induce tumour hypoxia.
1998
Cited 116 times
Mutation of the PTEN (MMAC1) tumor suppressor gene in a subset of glioblastomas but not in meningiomas with loss of chromosome arm 10q.
The PTEN (MMAC1) gene, which has been identified as a tumor suppressor gene at 10q23.3, is mutated in multiple malignant tumors, including glioblastomas [J. Li et al., Science (Washington DC), 275: 1943-1947, 1997; P. A. Steck et al., Nat. Genet., 15: 356-362, 1997]. Among tumors of the central nervous system, loss of 10q is not restricted to glioblastomas but is also common in atypical and anaplastic meningiomas. Therefore, we have investigated 36 glioblastomas and 34 meningiomas (2 benign, 17 atypical, and 15 anaplastic meningiomas) for loss on 10q, as well as deletion, mutation, and expression of PTEN. Analysis of eight microsatellites from 10q revealed loss of heterozygosity (LOH) in 25 of 36 glioblastomas (69%). Twenty-three of these tumors demonstrated LOH at all informative loci. Two glioblastomas showed LOH restricted to markers located distally to PTEN, with breakpoints mapping telomeric to D10S541 and D10S185. One glioblastoma demonstrated evidence of homozygous deletion of PTEN by differential PCR analysis. PTEN mutations were detected in 9 of 36 glioblastomas (25%). Seven of these tumors showed LOH at all informative loci from 10q, indicating complete loss of wild-type PTEN. Although loss of 10q was detected by comparative genomic hybridization and/or LOH analysis in 14 of the 34 meningiomas investigated (41%), none of these tumors showed evidence of PTEN mutations or homozygous gene deletions. Our findings corroborate that PTEN is inactivated in a subset of glioblastomas. However, the lack of detectable PTEN alterations in a considerable fraction of glioblastomas and all meningiomas with 10q loss strongly supports the hypothesis that at least one additional tumor suppressor gene is located on 10q.
DOI: 10.1016/s1044-7431(03)00232-x
2003
Cited 110 times
Wnt signalling inhibits neural differentiation of embryonic stem cells by controlling bone morphogenetic protein expression
Wnt signalling plays an important role in both embryonic development and in tumourigenesis. Activation of the signalling cascade by wnt, but also mutations of the adenomatous polyposis coli (APC) protein and of the phosphorylation domain of beta-catenin, result in accumulation of active beta-catenin in the nucleus, where it binds to TCF/LEF transcription factors. We studied the effect of wnt signalling in embryonic stem cells by either inactivating APC or by introducing a dominant active form of beta-catenin. Both resulted in inhibition of neural differentiation in vitro and after brain grafting and in activation of downstream targets of wnt signalling, such as cyclins, c-myc, and bone morphogenetic proteins (BMP). Neural differentiation could be partially restored by the addition of the BMP antagonist noggin. This suggests a mechanism regulating the fate of differentiating embryonic stem cells.
DOI: 10.1038/onc.2009.104
2009
Cited 109 times
Aryl hydrocarbon receptor inhibition downregulates the TGF-β/Smad pathway in human glioblastoma cells
DOI: 10.1007/s11523-010-0156-3
2010
Cited 86 times
Targeting integrins in malignant glioma
The integrin family of cell adhesion receptors is emerging as a promising target of anticancer therapy. AlphaVbeta3 and alphaVbeta5 integrins are overexpressed on both glioma cells and tumor vasculature. Cilengitide, the most advanced specific integrin inhibitor in oncology, has shown antitumor activity against glioma in early clinical trials. Durable remissions have been observed in phase I and phase II trials for recurrent glioblastoma (GBM) with both lower and higher doses of cilengitide. Pilot trials in newly diagnosed glioblastoma in conjunction with standard chemoradiotherapy have been encouraging. Preclinical data suggest synergy with concomitant chemo- and radiation therapy. A pivotal phase III study (CENTRIC) in newly diagnosed GBM patients is currently recruiting. This paper summarizes the current understanding of the role of integrins and their inhibition in gliomagenesis. The background and design of ongoing trials are outlined.
DOI: 10.1215/15228517-2009-012
2009
Cited 83 times
Cilengitide modulates attachment and viability of human glioma cells, but not sensitivity to irradiation or temozolomide in vitro
Cilengitide is a cyclic peptide antagonist of integrins alphavbeta3 and alphavbeta5 that is currently being evaluated as a novel therapeutic agent for recurrent and newly diagnosed glioblastoma. Its mode of action is thought to be mainly antiangiogenic but may include direct effects on tumor cells, notably on attachment, migration, invasion, and viability. In this study we found that, at clinically relevant concentrations, cilengitide (1-100 microM) induces detachment in some but not all glioma cell lines, while the effect on cell viability is modest. Detachment induced by cilengitide could not be predicted by the level of expression of the cilengitide target molecules, alphavbeta3 and alphavbeta5, at the cell surface. Glioma cell death induced by cilengitide was associated with the generation of caspase activity, but caspase activity was not required for cell death since ectopic expression of cytokine response modifier (crm)-A or coexposure to the broad-spectrum caspase inhibitor zVAD-fmk was not protective. Moreover, forced expression of the antiapoptotic protein marker Bcl-X(L) or altering the p53 status did not modulate cilengitide-induced cell death. No consistent effects of cilengitide on glioma cell migration or invasiveness were observed in vitro. Preliminary clinical results indicate a preferential benefit from cilengitide added to temozolomide-based radiochemotherapy in patients with O(6)-methylguanine DNA methyltransferase (MGMT) gene promoter methylation. Accordingly, we also examined whether the MGMT status determines glioma cell responses to cilengitide alone or in combination with temozolomide. Neither ectopic expression of MGMT in MGMT-negative cells nor silencing the MGMT gene in MGMT-positive cells altered glioma cell responses to cilengitide alone or to cilengitide in combination with temozolomide. These data suggest that the beneficial clinical effects derived from cilengitide in vivo may arise from altered perfusion, which promotes temozolomide delivery to glioma cells.
DOI: 10.1007/s00441-010-1123-0
2011
Cited 80 times
Glioblastoma stem cells
Glioblastomas are highly malignant primary brain tumors with one of the worst survival rates among all human cancers. With a more profound understanding of the cellular and molecular mechanisms of tumor initiation and acquired resistance to conventional radio- and chemotherapy, novel therapeutic targets might be discovered to optimize therapeutic approaches. In this regard, the identification of a small cellular subpopulation, called glioblastoma stem cell or stem-like cells or glioma-initiating cells or brain tumor propagating cells, has gained attention. In this article, we briefly summarize the current state of knowledge about this tumor cell population and discuss future directions for basic and clinical research.
DOI: 10.1007/s00259-014-2921-5
2014
Cited 80 times
In vivo visualization of prostate-specific membrane antigen in glioblastoma
DOI: 10.1093/eurheartj/ehs331
2012
Cited 74 times
Deletion of the ageing gene p66Shc reduces early stroke size following ischaemia/reperfusion brain injury
Stroke is a leading cause of morbidity and mortality, and its incidence increases with age. Both in animals and in humans, oxidative stress appears to play an important role in ischaemic stroke, with or without reperfusion. The adaptor protein p66(Shc) is a key regulator of reactive oxygen species (ROS) production and a mediator of ischaemia/reperfusion damage in ex vivo hearts. Hence, we hypothesized that p66(Shc) may be involved in ischaemia/reperfusion brain damage. To this end, we investigated whether genetic deletion of p66(Shc) protects from ischaemia/reperfusion brain injury.Transient middle cerebral artery occlusion (MCAO) was performed to induce ischaemia/reperfusion brain injury in wild-type (Wt) and p66(Shc) knockout mice (p66(Shc-/-)), followed by 24 h of reperfusion. Cerebral blood flow and blood pressure measurements revealed comparable haemodynamics in both experimental groups. Neuronal nuclear antigen immunohistochemical staining showed a significantly reduced stroke size in p66(Shc-/-) when compared with Wt mice (P < 0.05, n = 7-8). In line with this, p66(Shc-/-) mice exhibited a less impaired neurological function and a decreased production of free radicals locally and systemically (P < 0.05, n = 4-5). Following MCAO, protein levels of gp91phox nicotinamide adenine dinucleotide phosphate oxidase subunit were increased in brain homogenates of Wt (P < 0.05, n = 4), but not of p66(Shc-/-) mice. Further, reperfusion injury in Wt mice induced p66(Shc) protein in the basilar and middle cerebral artery, but not in brain tissue, suggesting a predominant involvement of vascular p66(Shc).In the present study, we show that the deletion of the ageing gene p66(Shc) protects mice from ischaemia/reperfusion brain injury through a blunted production of free radicals. The ROS mediator p66(Shc) may represent a novel therapeutical target for the treatment of ischaemic stroke.
DOI: 10.1158/1078-0432.ccr-16-0064
2016
Cited 69 times
PI3K Pathway Inhibition Achieves Potent Antitumor Activity in Melanoma Brain Metastases<i>In Vitro</i>and<i>In Vivo</i>
Abstract Purpose: Great advances have recently been made in treating patients with metastatic melanoma. However, existing therapies are less effective on cerebral than extracerebral metastases. This highlights the potential role of the brain environment on tumor progression and drug resistance and underlines the need for “brain-specific” therapies. We previously showed that the PI3K-AKT survival pathway is hyperactivated in brain but not extracerebral melanoma metastases and that astrocyte-conditioned medium activates AKT in melanoma cells in vitro. We therefore tested the PI3K inhibitor buparlisib as an antitumor agent for melanoma brain metastases. Experimental Design and Results: Buparlisib inhibited AKT activity, decreased proliferation, and induced apoptosis in metastatic melanoma cell lines and short-term brain melanoma cells, irrespective of their BRAF and NRAS mutation status. In addition, buparlisib inhibited hyperactivated AKT and induced apoptosis in melanoma cells that were stimulated with astrocyte-conditioned medium. The growth of tumors induced by injecting human BRAF- and NRAS-mutant metastatic melanoma cells into the brain of mice was significantly inhibited by buparlisib. Conclusions: These results emphasize the value of targeting the PI3K pathway as a strategy to develop drugs for melanoma brain metastases. Clin Cancer Res; 22(23); 5818–28. ©2016 AACR.
DOI: 10.1093/annonc/mdy120
2018
Cited 67 times
Bevacizumab plus hypofractionated radiotherapy versus radiotherapy alone in elderly patients with glioblastoma: the randomized, open-label, phase II ARTE trial
The addition of bevacizumab to temozolomide-based chemoradiotherapy (TMZ/RT → TMZ) did not prolong overall survival (OS) in patients with newly diagnosed glioblastoma in phase III trials. Elderly and frail patients are underrepresented in clinical trials, but early reports suggested preferential benefit in this population.ARTE was a 2 : 1 randomized, multi-center, open-label, non-comparative phase II trial of hypofractionated RT (40 Gy in 15 fractions) with bevacizumab (10 mg/kg×14 days) (arm A, N = 50) or without bevacizumab (arm B, N = 25) in patients with newly diagnosed glioblastoma aged ≥65 years. The primary objective was to obtain evidence for prolongation of median OS by the addition of bevacizumab to RT. Response was assessed by RANO criteria. Quality of life (QoL) was monitored by the EORTC QLQ-C30/BN20 modules. Exploratory studies included molecular subtyping by 450k whole methylome and gene expression analyses.Median PFS was longer in arm A than in arm B (7.6 and 4.8 months, P = 0.003), but OS was similar (12.1 and 12.2 months, P = 0.77). Clinical deterioration was delayed and more patients came off steroids in arm A. Prolonged PFS in arm A was confined to tumors with the receptor tyrosine kinase (RTK) I methylation subtype (HR 0.25, P = 0.014) and proneural gene expression (HR 0.29, P = 0.025). In a Cox model of OS controlling for established prognostic factors, associations with more favorable outcome were identified for age <70 years (HR 0.52, P = 0.018) and Karnofsky performance score 90%-100% (HR 0.51, P = 0.026). Including molecular subtypes into that model identified an association of the RTK II gene methylation subtype with inferior OS (HR 1.73, P = 0.076).Efficacy outcomes and exploratory analyses of ARTE do not support the hypothesis that the addition of bevacizumab to RT generally prolongs survival in elderly glioblastoma patients. Molecular biomarkers may identify patients with preferential benefit from bevacizumab.NCT01443676.
DOI: 10.1093/neuonc/nou218
2014
Cited 66 times
Differential regulation of TGF-β–induced, ALK-5–mediated VEGF release by SMAD2/3 versus SMAD1/5/8 signaling in glioblastoma
BackgroundThe transforming growth factor (TGF)-β and vascular endothelial growth factor (VEGF) pathways have a major role in the pathogenesis of glioblastoma, notably immunosuppression, migration, and angiogenesis, but their interactions have remained poorly understood.
DOI: 10.1002/ana.24522
2015
Cited 65 times
Predictors of preoperative and early postoperative seizures in patients with intra‐axial primary and metastatic brain tumors: A retrospective observational single center study
Objective Antiepileptic treatment of brain tumor patients mainly depends on the individual physician's choice rather than on well‐defined predictive factors. We investigated the predictive value of defined clinical parameters to formulate a model of risk estimations for subpopulations of brain tumor patients. Methods We enclosed 650 patients &gt; 18 years of age who underwent brain tumor surgery and included a number of clinical data. Logistic regressions were performed to determine the effect sizes of seizure‐related risk factors and to develop prognostic scores for the occurrence of preoperative and early postoperative seizures. Results A total of 492 patients (334 gliomas) were eligible for logistic regression for preoperative seizures, and 338 patients for early postoperative seizures. Age ≤ 60 years (odds ratio [OR] = 1.66, p = 0.020), grades I and II glioma (OR = 4.00, p = 0.0002), total tumor/edema volume ≤ 64cm 3 (OR = 2.18, p = 0.0003), and frontal location (OR = 2.28, p = 0.034) demonstrated an increased risk for preoperative seizures. Isocitrate–dehydrogenase mutations (OR = 2.52, p = 0.026) were an independent risk factor in the glioma subgroup. Age ≥ 60 years (OR = 3.32, p = 0.041), total tumor/edema volume ≤ 64cm 3 (OR = 3.17, p = 0.034), complete resection (OR = 15.50, p = 0.0009), diencephalic location (OR = 12.2, p = 0.013), and high‐grade tumors (OR = 5.67, p = 0.013) were significant risk factors for surgery‐related seizures. Antiepileptics (OR = 1.20, p = 0.60) did not affect seizure occurrence. For seizure occurrence, patients could be stratified into 3 prognostic preoperative and into 2 prognostic early postoperative groups. Interpretation Based on the developed prognostic scores, seizure prophylaxis should be considered in high‐risk patients and patient stratification for prospective studies may be feasible in the future. Ann Neurol 2015;78:917–928
DOI: 10.1007/s11060-016-2272-0
2016
Cited 57 times
In vivo molecular profiling of human glioma using diffusion kurtosis imaging
DOI: 10.18632/oncotarget.4310
2015
Cited 56 times
Modulation of cerebral endothelial cell function by TGF-β in glioblastoma: VEGF-dependent angiogenesis versus endothelial mesenchymal transition
Glioblastoma are among the most angiogenic tumors.The molecular mechanisms that control blood vessel formation by endothelial cells (EC) in glioblastoma remain incompletely understood.Transforming growth factor-β (TGF-β) is a key regulatory cytokine that has proinvasive and stemness-maintaining autocrine properties in glioblastoma and confers immunosuppression to the tumor microenvironment.Here we characterize potential pro-and anti-angiogenic activities of TGF-β in the context of glioblastoma in vitro, using human brain-derived microvascular endothelial cells (hCMEC/D3) and glioblastoma-derived endothelial cells (GMEC) as model systems.We find that TGF-β induces vascular endothelial growth factor (VEGF) and placental growth factor (PlGF) mRNA expression and protein release in a TGF-β receptor (TβR) II / activin-like kinase (ALK)-5-dependent manner under normoxia and hypoxia, defining potential indirect proangiogenic activity of TGF-β in glioblastoma.In parallel, exogenous TGF-β has also inhibitory effects on EC properties and induces endothelialmesenchymal transition (EndMT) in hCMEC and GMEC.Accordingly, direct inhibition of endogenous TGF-β/ALK-5 signalling increases EC properties such as tube formation, von-Willebrand factor (vWF) and claudin (CLDN) 5 expression.Yet, the supernatant of TGF-β-stimulated hCMEC and GMEC strongly promotes EC-related gene expression and tube formation in a cediranib-sensitive manner.These observations shed light on the complex pro-and anti-angiogenic pathways involving the cross-talk between TGF-β and VEGF/PLGF signalling in glioblastoma which may involve parallel stimulation of angiogenesis and EndMT in distinct target cell populations.
DOI: 10.1007/s00062-017-0590-z
2017
Cited 53 times
Glioma Grading and Determination of IDH Mutation Status and ATRX loss by DCE and ASL Perfusion
DOI: 10.1007/s00432-018-02836-5
2019
Cited 53 times
High frequency of H3 K27M mutations in adult midline gliomas
DOI: 10.15252/emmm.201708492
2019
Cited 53 times
Efficacy of systemic temozolomide‐activated phage‐targeted gene therapy in human glioblastoma
Research Article27 February 2019Open Access Source DataTransparent process Efficacy of systemic temozolomide-activated phage-targeted gene therapy in human glioblastoma Justyna Magdalena Przystal Justyna Magdalena Przystal Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK Search for more papers by this author Sajee Waramit Sajee Waramit Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK Search for more papers by this author Md Zahidul Islam Pranjol Md Zahidul Islam Pranjol Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK Search for more papers by this author Wenqing Yan Wenqing Yan Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK Search for more papers by this author Grace Chu Grace Chu Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK Search for more papers by this author Aitthiphon Chongchai Aitthiphon Chongchai Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand Search for more papers by this author Gargi Samarth Gargi Samarth Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK Search for more papers by this author Nagore Gene Olaciregui Nagore Gene Olaciregui Institute de Recerca Sant Joan de Deu, Barcelona, Spain Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona, Spain Search for more papers by this author Ghazaleh Tabatabai Ghazaleh Tabatabai Interdisciplinary Division of Neuro-Oncology, Hertie Institute for Clinical Brain Research, Center for CNS Tumors, Comprehensive Cancer Center, University Hospital Tübingen, Eberhard Karls University, Tübingen, Germany German Cancer Consortium (DKTK), DKFZ Partner Site Tübingen, Tübingen, Germany Search for more papers by this author Angel Montero Carcaboso Angel Montero Carcaboso Institute de Recerca Sant Joan de Deu, Barcelona, Spain Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona, Spain Search for more papers by this author Eric Ofori Aboagye Eric Ofori Aboagye Comprehensive Cancer Imaging Centre, Imperial College London, Faculty of Medicine, London, UK Search for more papers by this author Keittisak Suwan Keittisak Suwan Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK Search for more papers by this author Amin Hajitou Corresponding Author Amin Hajitou [email protected] orcid.org/0000-0003-1119-5686 Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK Search for more papers by this author Justyna Magdalena Przystal Justyna Magdalena Przystal Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK Search for more papers by this author Sajee Waramit Sajee Waramit Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK Search for more papers by this author Md Zahidul Islam Pranjol Md Zahidul Islam Pranjol Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK Search for more papers by this author Wenqing Yan Wenqing Yan Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK Search for more papers by this author Grace Chu Grace Chu Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK Search for more papers by this author Aitthiphon Chongchai Aitthiphon Chongchai Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand Search for more papers by this author Gargi Samarth Gargi Samarth Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK Search for more papers by this author Nagore Gene Olaciregui Nagore Gene Olaciregui Institute de Recerca Sant Joan de Deu, Barcelona, Spain Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona, Spain Search for more papers by this author Ghazaleh Tabatabai Ghazaleh Tabatabai Interdisciplinary Division of Neuro-Oncology, Hertie Institute for Clinical Brain Research, Center for CNS Tumors, Comprehensive Cancer Center, University Hospital Tübingen, Eberhard Karls University, Tübingen, Germany German Cancer Consortium (DKTK), DKFZ Partner Site Tübingen, Tübingen, Germany Search for more papers by this author Angel Montero Carcaboso Angel Montero Carcaboso Institute de Recerca Sant Joan de Deu, Barcelona, Spain Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona, Spain Search for more papers by this author Eric Ofori Aboagye Eric Ofori Aboagye Comprehensive Cancer Imaging Centre, Imperial College London, Faculty of Medicine, London, UK Search for more papers by this author Keittisak Suwan Keittisak Suwan Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK Search for more papers by this author Amin Hajitou Corresponding Author Amin Hajitou [email protected] orcid.org/0000-0003-1119-5686 Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK Search for more papers by this author Author Information Justyna Magdalena Przystal1,8,9, Sajee Waramit1, Md Zahidul Islam Pranjol1,10, Wenqing Yan1, Grace Chu1, Aitthiphon Chongchai2, Gargi Samarth1, Nagore Gene Olaciregui3,4, Ghazaleh Tabatabai5,6, Angel Montero Carcaboso3,4, Eric Ofori Aboagye7, Keittisak Suwan1 and Amin Hajitou *,1 1Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK 2Thailand Excellence Centre for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand 3Institute de Recerca Sant Joan de Deu, Barcelona, Spain 4Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona, Spain 5Interdisciplinary Division of Neuro-Oncology, Hertie Institute for Clinical Brain Research, Center for CNS Tumors, Comprehensive Cancer Center, University Hospital Tübingen, Eberhard Karls University, Tübingen, Germany 6German Cancer Consortium (DKTK), DKFZ Partner Site Tübingen, Tübingen, Germany 7Comprehensive Cancer Imaging Centre, Imperial College London, Faculty of Medicine, London, UK 8Present address: Interdisciplinary Division of Neuro-Oncology, Hertie Institute for Clinical Brain Research, University Hospital Tübingen, Eberhard Karls University, Tübingen, Germany 9Present address: German Cancer Consortium (DKTK), DKFZ Partner Site Tübingen, Tübingen, Germany 10Present address: William Harvey Research Institute, Queen Mary University of London, London, UK *Corresponding author. Tel: +44 207 594 6546; E-mail: [email protected] EMBO Mol Med (2019)11:e8492https://doi.org/10.15252/emmm.201708492 PDFDownload PDF of article text and main figures. Peer ReviewDownload a summary of the editorial decision process including editorial decision letters, reviewer comments and author responses to feedback. ToolsAdd to favoritesDownload CitationsTrack CitationsPermissions ShareFacebookTwitterLinked InMendeleyWechatReddit Figures & Info Abstract Glioblastoma multiforme (GBM) is the most lethal primary intracranial malignant neoplasm in adults and most resistant to treatment. Integration of gene therapy and chemotherapy, chemovirotherapy, has the potential to improve treatment. We have introduced an intravenous bacteriophage (phage) vector for dual targeting of therapeutic genes to glioblastoma. It is a hybrid AAV/phage, AAVP, designed to deliver a recombinant adeno-associated virus genome (rAAV) by the capsid of M13 phage. In this vector, dual tumor targeting is first achieved by phage capsid display of the RGD4C ligand that binds the αvβ3 integrin receptor. Second, genes are expressed from a tumor-activated and temozolomide (TMZ)-induced promoter of the glucose-regulated protein, Grp78. Here, we investigated systemic combination therapy using TMZ and targeted suicide gene therapy by the RGD4C/AAVP-Grp78. Firstly, in vitro we showed that TMZ increases endogenous Grp78 gene expression and boosts transgene expression from the RGD4C/AAVP-Grp78 in human GBM cells. Next, RGD4C/AAVP-Grp78 targets intracranial tumors in mice following intravenous administration. Finally, combination of TMZ and RGD4C/AAVP-Grp78 targeted gene therapy exerts a synergistic effect to suppress growth of orthotopic glioblastoma. Synopsis Following intravenous administration, the RGD4C/AAVP-Grp78 vector delivers the therapeutic AAV genome into GBM via binding to integrins. Treatment with temozolomide (TMZ) induces the Grp78 promoter activity and subsequently the therapeutic gene expression resulting in synergistic anti-tumor action. RGD4C/AAVP-Grp78-HSVtk binds to αvβ3 integrin in GBM, leading to vector internalisation and delivery of the recombinant rAAV genome in the nucleus to generate expression of the HSVtk. Expression of HSVtk results in phosphorylation of ganciclovir (GCV) within GBM and subsequent tumor destruction. Administration of TMZ induces the Grp78 promoter activity through the UPR pathway, resulting in further expression of the HSVtk and subsequent phosphorylation of GCV. Consequently combination of TMZ with targeted intravenous RGD4C/AAVP-Grp78-HSVtk/GCV gene therapy results in synergistic destruction of GBM. Introduction Glioblastoma, also known as glioblastoma multiforme (GBM) or grade IV astrocytoma, is the most common and deadliest of primary brain tumors in adults with 14.6-month median survival time and a 2% 5-year survival rate (Kwiatkowska et al, 2013). This depressing prognosis occurs with current, aggressive gold standard treatment regimens, such as chemotherapy. Even more recent trials with anti-angiogenic and molecularly targeted therapies have shown only slight improvements in GBM outcome (Gilbert et al, 2014). Therefore, novel remedies that effectively treat or improve/complement existing treatments are pressingly needed. Temozolomide (TMZ), an alkylating agent able to cross the blood–brain barrier (BBB), is well-tolerated and widely used as part of standard chemotherapy for newly diagnosed and recurrent GBM, but has limited efficacy (Thon et al, 2013; Messaoudi et al, 2015; Taal et al, 2015). We hypothesized that combining TMZ chemotherapy with gene therapy should yield a synergistic effect against GBM. Gene therapy for GBM has been attempted for the past 24 years, but progress has been hindered mostly by lack of tumor selectivity and inefficiency of eukaryotic viral vectors (Natsume & Yoshida, 2008; Kwiatkowska et al, 2013). Gene therapy of brain tumors is further hampered by the BBB (Maguire et al, 2014). Although this could be circumvented by delivery of vectors locally, the invasiveness of repeated intracerebral injections and the diffuse/not discrete nature of GBM place great limitations. We have introduced a unique prokaryotic viral-based approach of systemic intravenous gene delivery to target tumors specifically by using the harmless and non-pathogenic filamentous M13 bacteriophage, a virus that infects only bacterial cells and lacks native tropism to normal tissues in human and eukaryotes in general (Asavarut & Hajitou, 2014). A key strategy of our systemic delivery platform involves the construction of a hybrid bacteriophage vector termed AAV-phage, or AAVP (Fig EV1A). In this vector, the single-stranded genome of human adeno-associated virus (AAV) was inserted within the M13 phage single-stranded genome, resulting in a phage capsid which incorporates AAV genomes(s) (Hajitou et al, 2006, 2007). Importantly, the phage capsid was engineered to display the CDCRGDCFC (RGD4C) ligand (Fig EV1A) that binds the heterodimer αvβ3 integrin cell surface receptor, which is overexpressed on tumor cells and supporting angiogenic vasculature in most tumor types including human GBM (Hajitou et al, 2006; Schnell et al, 2008). Upon binding to αvβ3 integrin receptor and subsequent entry of the RGD4C/AAVP viral particles into cells, the AAV genome is released to express genes in tumors from a cytomegalovirus, CMV, promoter (Hajitou et al, 2006). To date, we and collaborators have established that these vectors target various preclinical models of human cancer including soft tissue sarcoma, melanoma, breast, prostate, and pancreatic cancers (Hajitou et al, 2006, 2008; Tandle et al, 2009; Yuan et al, 2013; Dobroff et al, 2016; Smith et al, 2016). Additionally, a study by the National Cancer Institute, USA, using our vector in pet dogs with natural cancers demonstrated that RGD4C/AAVP delivered the cytokine, tumor necrosis factor-alpha (TNFα), selectively, to spontaneous cancers (Paoloni et al, 2009). Remarkably, repeated vector dosing proved safe and resulted in complete tumor eradication in several dogs with aggressive cancers (Paoloni et al, 2009). To improve the vector platform for use in targeted gene therapy against GBM, we have refined the technology by replacing the CMV promoter with the tumor-specific Grp78 promoter and designed the dual tumor targeting RGD4C/AAVP-Grp78 vector (Kia et al, 2012, 2013). This novel vector effectively ensured further tumor selectivity, through transcriptional targeting as previously reported (Kia et al, 2012). We also demonstrated that the RGD4C/AAVP-Grp78 vector provides much longer lasting transgene expression than the RGD4C/AAVP-CMV vector carrying a CMV promoter, in vitro and in vivo in subcutaneous GBM following intravenous administration (Kia et al, 2012). The Grp78 promoter is marginally active in healthy tissues; however, potent activation has been observed in aggressive tumors, including GBM (Dong et al, 2004; Pyrko et al, 2007; Virrey et al, 2008), and is induced by stress and conditions of tumor microenvironment such as glucose deprivation, chronic anoxia, and acidic pH. It is further worth considering that the induction of the Grp78 gene expression and activation confers drug resistance in a variety of human tumors, including gliomas (Li & Lee, 2006; Lee, 2007; Pyrko et al, 2007). Grp78 can also be induced by TMZ in GBM (Pyrko et al, 2007), and its activation has been associated with GBM resistance to TMZ (Pyrko et al, 2007; Virrey et al, 2008). Thus, further increase in gene expression from the RGD4C/AAVP-Grp78 can be ensured through TMZ activation of the Grp78 promoter. Consequently, we postulated that RGD4C/AAVP-Grp78 is a suitable candidate for use in combination with TMZ against GBM. Herein, we investigated the effects of combining TMZ chemotherapy and targeted gene therapy with RGD4C/AAVP-Grp78, termed chemovirotherapy, against intracranial orthotopic models of human glioblastoma. Firstly, we demonstrated that a list of various human glioblastoma cell lines as well as primary GBM and primary GBM stem cells express αvβ3 integrin, a receptor of RGD4C, resulting in ligand-directed gene delivery by RGD4C/AAVP in vitro. Then, we showed that TMZ treatment of human glioblastoma cell lines increased expression of the endogenous Grp78, through the UPR (unfolded protein response) pathway, subsequently stimulating RGD4C/AAVP-Grp78-mediated gene expression. We also found that TMZ amplified the destruction of GBM cells in combination with RGD4C/AAVP-Grp78-HSVtk encoding the Herpes simplex virus type I thymidine kinase in the presence of ganciclovir (GCV); we used the HSVtk mutant SR39 (Black et al, 2001). Next, we confirmed that RGD4C/AAVP-Grp78 targets orthotopic glioblastoma in mice after intravenous administration selectively binding to tumor cells and tumor vasculature without accumulation in the healthy brains. Additionally, the combination of TMZ and RGD4C/AAVP-Grp78-HSVtk/GCV administered systemically elicited strong anti-tumor activity against intracranial glioblastoma established in vivo from GBM cell lines and primary GBM, and in both immunodeficient and immunocompetent mice. Unless technically, the in vivo effect was measured synergistic, compared to TMZ or RGD4C/AAVP-Grp78-HSVtk/GCV alone. The HSVtk enzyme phosphorylates prodrug nucleoside analogues such as GCV and converts them into nucleoside analogue triphosphates. These triphosphate GCV compounds are then incorporated into the cellular genome, inhibit DNA polymerase, and subsequently induce cell death by apoptosis (Hamel et al, 1996; Natsume & Yoshida, 2008). It is important to note that the HSVtk/GCV approach also elicits a bystander effect, which means that cells containing the HSVtk kill neighboring non-transduced cells through the gap junctional intercellular communications (GJIC) that allow the transfer of the converted cytotoxic drug and/or toxic metabolites between these cells as we have previously reported (Trepel et al, 2009; Duarte et al, 2012). After a few days, “this bystander effect” results in increased cell killing by the RGD4C/AAVP-HSVtk vector and may potentially overcome the requirement for all malignant cells to be transduced in order to achieve meaningful tumor regression. Altogether, these findings indicate that this combination therapy strategy offers significant translational potential in the treatment regime for GBM patients. Click here to expand this figure. Figure EV1. The targeted RGD4C/AAVP viral particle A. The vector bears the αvβ3 integrin-targeting double-cyclic RGD4C ligand on the pIII minor coat protein. The virus structure consists of 2,700–3,000 copies of the major coat protein pVIII with approximately five copies of the four minor capsid proteins pIII, pVI, pVII, and pIX, which are located at the ends of the filamentous particle. The AAV transgene cassette flanked by the inverted terminal repeats (ITR) from AAV2 is inserted in an intergenomic region of the bacteriophage genome. Expression of the HSVtk or Luc transgenes is under the control of either CMV or Grp78 promoters. pA: polyadenylation signal. B. Induction of RGD4C/AAVP-Grp78 by curcumin in primary glioma. Pediatric human primary glioma cells transduced with RGD4C/AAVP-Grp78-Luc or non-targeted/AAVP-Grp78-Luc control vector were treated with curcumin at day 3 post-transduction. Results represent the RLU measured at day 6 post-transduction and normalized to untreated and non-transduced control cells. Data shown are representative of three independent experiments, n = 3. Data analysis was done by two-way ANOVA and Bonferroni's multiple comparison test. Data information: Data are expressed as mean ± SEM. Source data are available online for this figure. Download figure Download PowerPoint Results Human glioblastoma cell lines express integrin receptors of RGD4C and are subsequently transduced by the RGD4C/AAVP vector As initial experiments, we conducted in vitro studies on cell lines by using three models of human glioblastoma cells, namely LN229, U87, and SNB19, considered as common cellular models of this disease. First, we investigated expression of the integrins αvβ3 and αvβ5, receptors for RGD4C/AAVP, by immunofluorescent staining of αV, β3, and β5 integrin subunits. As shown in Fig 1A, all tumor cells tested were positive for expression of αv, β3, and β5 integrins, with varying expression of each integrin. Next, we investigated RGD4C/AAVP-mediated gene delivery to these tumor cells and used vectors carrying the reporter Luciferase (Luc) gene, which allows quantitative analysis of gene expression, and analyzed Luc expression over time. Cells were incubated with targeted RGD4C/AAVP-Luc or control non-targeted/AAVP-Luc vector (lacking the RGD4C). RGD4C/AAVP-mediated gene expression was demonstrated in all the human glioblastoma cells tested, in an efficient way and which increased over time (Fig 1B). Importantly, gene expression mediated by RGD4C/AAVP was selective, targeted, and dependent on RGD4C ligand binding to integrin receptors as no Luc expression was detected in cells treated with the control non-targeted/AAVP-Luc (Fig 1B). Figure 1. Targeted transduction of human glioblastoma cell lines and induction of RGD4C/AAVP-Grp78 by TMZ A. Immunofluorescence staining of LN229, U87, and SNB19 tumor cells with primary antibodies against αv, β3 or β5 integrin subunits. Scale bars, 20 μm for LN229 and SNB19, and 50 μm for U87. B. Targeted gene delivery into LN229, U87, and SNB19 cells by RGD4C/AAVP-Luc carrying the Luc reporter gene. Cells were seeded into 48-well plates before transduction. Non-targeted/AAVP-Luc was used as negative control for targeted transduction. Results represent the average relative luminescence units (RLU)/1 μg protein. Data shown are representative of two experiments, n = 3. C. Western blot analysis of the endogenous Grp78 protein expression levels in LN229, U87, and SNB19 cells following treatment with increasing concentrations of TMZ. Grp78 level was normalized to GAPDH. Data shown in right panel are representative of two experiments, n = 3. D. Induction of Grp78 promoter activity by TMZ in RGD4C/AAVP-Grp78-Luc cells. LN229, U87, and SNB19 cells stably transduced with RGD4C/AAVP-Grp78-Luc or RGD4C/AAVP-CMV-Luc were grown in the presence of TMZ for the indicated times. Results represent the average RLU/1 μg protein. Data shown are representative of two experiments, n = 3. Two-way ANOVA with Bonferroni correction (GraphPad Prism 6) was used for data analysis. Data information: Data are expressed as mean ± SEM. Source data are available online for this figure. Source Data for Figure 1 [emmm201708492-sup-0002-SDataFig1.pdf] Download figure Download PowerPoint Temozolomide treatment of human glioblastoma cells results in induction of the endogenous Grp78 gene and boosts gene expression from the RGD4C/AAVP-Grp78 Numerous studies have reported that treatment with chemotherapeutic drugs stimulates endogenous Grp78 gene expression in tumors (Pyrko et al, 2007). It has also been reported that treatment with TMZ induces expression of endogenous Grp78 in glioblastoma patients, which is associated with glioblastoma resistance to TMZ (Virrey et al, 2008). Thus, we examined whether TMZ can increase the Grp78 promoter activity in the context of RGD4C/AAVP-Grp78 vector in vitro and subsequently enhance gene expression. First, we sought to confirm that TMZ activates expression of the endogenous Grp78 gene in glioblastoma cell lines. The LN229, U87, and SNB19 cells were treated with a range of TMZ concentrations (25–250 μM) for 12 h, followed by Western blot analysis and band intensity quantification. As shown in Fig 1C, the expression of endogenous Grp78 increased in a dose-dependent manner in LN229, U87, and SNB19 cells. We also noted that Grp78 expression in SNB19 reached a plateau between 100 and 250 μM of TMZ. We next set out to determine the effect of TMZ on transgene expression mediated by the RGD4C/AAVP-Grp78 in human glioblastoma cells. Vectors carrying the Luc reporter gene were utilized, and stably transduced LN229, U87, and SNB19 cells were generated by using puro® vectors that confer resistance to puromycin. Vectors expressing the Luc gene from the CMV promoter (RGD4C/AAVP-CMV-Luc) were also included in this experiment. Based on our data in Fig 1C, we used TMZ concentrations of 100 μM for LN229 and SNB19, and 60 μM for U87. Remarkably, TMZ induced a sharp increase in Luc expression in all LN229, U87, and SNB19 glioblastoma cells in which the Luc transgene was mediated by the RGD4C/AAVP-Grp78-Luc (Fig 1D). In contrast, Luc expression driven by the RGD4C/AAVP-CMV-Luc remained unaltered by TMZ in all tumor cells (Fig 1D). In LN229 and U87 cells, the rise in Luc expression occurred at 9 h post-TMZ treatment and continued gradually to reach 20- and 15-fold higher than RGD4C/AAVP-CMV-Luc at 12 h, respectively (Fig 1D). In the SNB19 cell line, the boost in Luc expression occurred at 24 h post-TMZ treatment and reached ~ 4.3-fold increase (Fig 1D). To further investigate induction of the RGD4C/AAVP-Grp78 vector in human glioma by other anti-cancer agents, known for their ability to cross the BBB, we treated human glioma cells with increasing concentrations of curcumin (0–40 μM). Curcumin, a natural and non-toxic dietary plant-based product, has been highlighted for its potential as an anti-cancer agent, due to its ability to induce tumor cell death with no systemic side effects. Studies have shown that curcumin can prevent proliferation of cancer cells including GBM (Klinger & Mittal, 2016). Addition of curcumin to tumor cells at day 3 post-transduction with RGD4C/AAVP-Grp78-Luc vector resulted in a dose-dependent increase of Luc gene expression that climbed markedly in the presence of 40 μM of curcumin (Fig EV1B). Temozolomide induces Grp78 promoter activation through the UPR signaling pathway To investigate whether the boost of RGD4C/AAVP-Grp78 gene expression by TMZ is mediated through the conserved signaling cascade termed UPR (unfolded protein response) pathway that regulates the endogenous Grp78 gene, we examined three transmembrane proteins representing the three arms of the UPR signaling pathway (Rutkowski & Kaufman, 2004; Pfaffenbach & Lee, 2011; Luo & Lee, 2013; Nagelkerke et al, 2014). These experiments were performed following TMZ treatment of LN229, U87, and SNB19 tumor cells stably transduced with the RGD4C/AAVP-Grp78-Luc vector, consistent with the experiments in Fig 1. We first focused on protein kinase RNA-like ER kinase (PERK); after homo-dimerization and trans-autophosphorylation, PERK phosphorylates Ser51 of the α-subunit of the eukaryotic translation initiation factor-2 (eIF2α; Luo & Lee, 2013; Ron & Walter, 2007) that selectively promotes translation of the transcription factor ATF4 which induces the Grp78 promoter. Western blot analyses of tumor cells showed that TMZ induces expression of the phospho-eIF2α that is detectable at 2, 1, and 3 h post-TMZ treatment in LN229, U87, and SNB19 cells, respectively, and induction of expression persisted for up to 24 h post-TMZ treatment (Fig 2A). Next, we evaluated TMZ induction of expression of the activating transcription factor 6 (ATF6). We found that TMZ induces the ATF6-p90 expression detectable at 1 h in LN229 and 2 h in U87 cells, and then increased over time for 24 h post-treatment (Fig 2A). The induction of ATF6-p90 in SNB19 cells was noticed 24 h after TMZ treatment (Fig 2A). Previous studies have shown that an increase in the 90-kDa ATF6 level indicates initiation of the endoplasmic reticulum (ER) stress pathway and subsequently activation of UPR (Teske et al, 2011; Kia et al, 2012), which is a cellular stress response related to the ER stress. Finally, during ER stress, activated inositol-requiring enzyme 1 (IRE1) cleaves a 26-base intron from mRNA of X-box binding protein 1 (XBP1) resulting in a product that acts as a transcription factor for the Grp78 promoter (Teske et al, 2011). Semi-quantitative RT–PCR analyses showed a slight increase in spliced XBP1 mRNA at 24 h post-TMZ treatment of LN229 (Fig 2B), while a greater and gradual increase in the level of spliced XBP1 mRNA was found in the U87 cell line upon TMZ treatment (Fig 2B). In contrast, no spliced XBP1 mRNA was detected in SNB19 glioblastoma cells (Fig 2B). Figure 2. TMZ activation of the UPR pathway and subsequent enhancement of glioblastoma cell killing in vitro by the RGD4C/AAVP-Grp78-HSVtk and GCV A. TMZ induction of phospho-eIF2α and ATF6-p90 expression. Human glioblastoma cells transduced with RGD4C/AAVP-Grp78 were analyzed by Western blot following treatment with TMZ (100 μM for LN229 and SNB19, 60 μM for U87). GAPDH was used as a control. B. RT–PCR analysis of constitutive expression and splicing of XBP1 in glioblastoma cells transduced with RGD4C/AAVP-Grp78, in the presence of TMZ. Sizes of the PCR products were 289 bp for unspliced XBP1 and 263 bp for spliced XBP1, and the lower size band is not specific. Time points were selected among those tested in the Western blot in (A), subsequently the image containing 0, 1, and 2 h was juxtaposed to images of 6, 12, and 24 h. A white line has been added between the gel pieces that have been juxtaposed. C. Tumor cell killing in vitro by the HSVtk/GCV approach. Glioblastoma cells stably transduced with RGD4C/AAVP-Grp78-HSVtk or RGD4C/AAVP-CMV-HSVtk were treated with either GCV (10 μM) or TMZ (100 μM for LN229 and SNB19, 60 μM for U87) or combination of both GCV and TMZ. Cells were stained with MitoSOX and analyzed by FACS at day 4 post-treatment. Data shown are representative of three experiments, n = 3. Two-way ANOVA with Tukey's multiple comparison test (GraphPad Prism 6) was used for data analysis. Data information: Data are expressed as mean ± SEM. Source data are available online for this figure. Source Data for Figure 2 [emmm201708492-sup-0003-SDataFig2.pdf] Download figure Download PowerPoint Combination of TMZ with RGD4C/AAVP-Grp78-HSVtk and GCV produces strong tumor cell killing in vitro Our previous studies showed that the RGD4C/AAVP-Grp78-HSVtk is superior to the conventional RGD4C/AAVP-CMV-HSVtk vector in inducing glioblastoma cell killing in vitro as well as GBM growth inhibition in vivo following intravenous administration of vectors carrying the HSVtk, and intraperitoneal injection of GCV (Kia et al, 2012). Herein, we compared tumor cell killin
DOI: 10.1074/mcp.ra118.000792
2018
Cited 49 times
Identification of Tumor Antigens Among the HLA Peptidomes of Glioblastoma Tumors and Plasma
Glioblastoma multiforme (GBM) is the most aggressive brain tumor with poor prognosis to most patients. Immunotherapy of GBM is a potentially beneficial treatment option, whose optimal implementation may depend on familiarity with tumor specific antigens, presented as HLA peptides by the GBM cells. Furthermore, early detection of GBM, such as by a routine blood test, may improve survival, even with the current treatment modalities. This study includes large-scale analyses of the HLA peptidome (immunopeptidome) of the plasma-soluble HLA molecules (sHLA) of 142 plasma samples, and the membranal HLA of GBM tumors of 10 of these patients' tumor samples. Tumor samples were fresh-frozen immediately after surgery and the plasma samples were collected before, and at multiple visits after surgery. In total, this HLA peptidome analysis involved 52 different HLA allotypes and resulted in the identification of more than 35,000 different HLA peptides. Strong correlations were observed in the signal intensities and in the repertoires of identified peptides between the tumors and plasma-soluble HLA peptidomes of the individual patients, whereas low correlations were observed between these HLA peptidomes and the tumors' proteomes. HLA peptides derived from Cancer/Testis Antigens (CTAs) were selected based on their presence among the HLA peptidomes of the patients and absence of expression of their source genes from any healthy and essential human tissues, except from immune-privileged sites. Additionally, peptides were selected as potential biomarkers if their levels in the plasma-sHLA peptidome were significantly reduced after the removal of tumor mass. The CTAs identified among the analyzed HLA peptidomes provide new opportunities for personalized immunotherapy and for early diagnosis of GBM.
DOI: 10.1074/mcp.ra119.001524
2019
Cited 45 times
Identification of Tumor Antigens Among the HLA Peptidomes of Glioblastoma Tumors and Plasma
Glioblastoma multiforme (GBM) is the most aggressive brain tumor with poor prognosis to most patients. Immunotherapy of GBM is a potentially beneficial treatment option, whose optimal implementation may depend on familiarity with tumor specific antigens, presented as HLA peptides by the GBM cells. Further, early detection of GBM, such as by a routine blood test, may improve survival, even with the current treatment modalities. This study includes large-scale analyses of the HLA peptidome (immunopeptidome) of the plasma-soluble HLA molecules (sHLA) of 142 plasma samples, and the membranal HLA of GBM tumors of 10 of these patients' tumor samples. Tumor samples were fresh-frozen immediately after surgery and the plasma samples were collected before, and at multiple visits after surgery. In total, this HLA peptidome analysis involved 52 different HLA allotypes and resulted in the identification of more than 35,000 different HLA peptides. Strong correlations were observed in the signal intensities and in the repertoires of identified peptides between the tumors and plasma-soluble HLA peptidomes of the individual patients, whereas low correlations were observed between these HLA peptidomes and the tumors' proteomes. HLA peptides derived from Cancer/Testis Antigens (CTAs) were selected based on their presence among the HLA peptidomes of the patients and absence of expression of their source genes from any healthy and essential human tissues, except from immune-privileged sites. Additionally, peptides were selected as potential biomarkers if their levels in the plasma-sHLA peptidome were significantly reduced after the removal of tumor mass. The CTAs identified among the analyzed HLA peptidomes provide new opportunities for personalized immunotherapy and for early diagnosis of GBM. Glioblastoma multiforme (GBM) is the most aggressive brain tumor with poor prognosis to most patients. Immunotherapy of GBM is a potentially beneficial treatment option, whose optimal implementation may depend on familiarity with tumor specific antigens, presented as HLA peptides by the GBM cells. Further, early detection of GBM, such as by a routine blood test, may improve survival, even with the current treatment modalities. This study includes large-scale analyses of the HLA peptidome (immunopeptidome) of the plasma-soluble HLA molecules (sHLA) of 142 plasma samples, and the membranal HLA of GBM tumors of 10 of these patients' tumor samples. Tumor samples were fresh-frozen immediately after surgery and the plasma samples were collected before, and at multiple visits after surgery. In total, this HLA peptidome analysis involved 52 different HLA allotypes and resulted in the identification of more than 35,000 different HLA peptides. Strong correlations were observed in the signal intensities and in the repertoires of identified peptides between the tumors and plasma-soluble HLA peptidomes of the individual patients, whereas low correlations were observed between these HLA peptidomes and the tumors' proteomes. HLA peptides derived from Cancer/Testis Antigens (CTAs) were selected based on their presence among the HLA peptidomes of the patients and absence of expression of their source genes from any healthy and essential human tissues, except from immune-privileged sites. Additionally, peptides were selected as potential biomarkers if their levels in the plasma-sHLA peptidome were significantly reduced after the removal of tumor mass. The CTAs identified among the analyzed HLA peptidomes provide new opportunities for personalized immunotherapy and for early diagnosis of GBM. Glioblastoma multiforme (GBM) 1The abbreviations used are: GBMglioblastomaMHCmajor histocompatibility complexHLAhuman leukocytes antigenCTAcancer/testis antigenTSAtumor specific antigensTAAtumor associated antigenssHLAsoluble human leukocytes antigenmHLAmembranal human leukocytes antigenFDRfalse discovery rateBBBblood brain barrier. 1The abbreviations used are: GBMglioblastomaMHCmajor histocompatibility complexHLAhuman leukocytes antigenCTAcancer/testis antigenTSAtumor specific antigensTAAtumor associated antigenssHLAsoluble human leukocytes antigenmHLAmembranal human leukocytes antigenFDRfalse discovery rateBBBblood brain barrier. is the most common and aggressive primary brain tumor, with a median survival of about 15 months (1Alifieris C. Trafalis D.T. Glioblastoma multiforme: Pathogenesis and treatment.Pharmacol. Ther. 2015; 152: 63-82Crossref PubMed Scopus (456) Google Scholar, 2Thakkar J.P. Dolecek T.A. Horbinski C. Ostrom Q.T. Lightner D.D. Barnholtz-Sloan J.S. Villano J.L. Epidemiologic and molecular prognostic review of glioblastoma.Cancer Epidemiol. Biomarkers Prev. 2014; 23: 1985-1996Crossref PubMed Scopus (674) Google Scholar). The grim prognosis facing most GBM patients calls for improvements in targeted therapeutic treatments, as well as in discovery of proper biomarkers for early detection of the disease. Recently, immunotherapy of different cancers, including GBM, has drawn significant attention (3Terasaki M. Shibui S. Narita Y. Fujimaki T. Aoki T. Kajiwara K. Sawamura Y. Kurisu K. Mineta T. Yamada A. Itoh K. Phase I trial of a personalized peptide vaccine for patients positive for human leukocyte antigen–A24 with recurrent or progressive glioblastoma multiforme.J. Clin. Oncol. 2011; 29: 337-344Crossref PubMed Scopus (96) Google Scholar, 4Neidert M.C. Schoor O. Trautwein C. Trautwein N. Christ L. Melms A. Honegger J. Rammensee H.-G. Herold-Mende C. Dietrich P.-Y. Stevanović S. Natural HLA class I ligands from glioblastoma: extending the options for immunotherapy.J. Neurooncol. 2013; 111: 285-294Crossref PubMed Scopus (18) Google Scholar), reviewed in (5Patel M.A. Pardoll D.M. Concepts of immunotherapy for glioma.J. Neurooncol. 2015; 123: 323-330Crossref PubMed Scopus (11) Google Scholar, 6Cohen-Inbar O. Zaaroor M. Immunological aspects of malignant gliomas.Can. J. Neurol. Sci. 2016; 43: 494-502Crossref PubMed Scopus (10) Google Scholar, 7Polivka J. Polivka J. Holubec L. Kubikova T. Priban V. Hes O. Pivovarcikova K. Treskova I. Advances in experimental targeted therapy and immunotherapy for patients with glioblastoma multiforme.Anticancer Res. 2017; 37: 21-33Crossref PubMed Scopus (89) Google Scholar, 8Swartz A.M. Batich K.A. Fecci P.E. Sampson J.H. Peptide vaccines for the treatment of glioblastoma.J. Neurooncol. 2015; 123: 433-440Crossref PubMed Scopus (26) Google Scholar, 9Ampie L. Woolf E.C. Dardis C. Immunotherapeutic advancements for glioblastoma.Front. Oncol. 2015; 5: 12Crossref PubMed Scopus (31) Google Scholar, 10Oh T. Sayegh E.T. Fakurnejad S. Oyon D. Lamano J.B. DiDomenico J.D. Bloch O. Parsa A.T. Vaccine therapies in malignant glioma.Curr. Neurol. Neurosci. Rep. 2015; 15: 508Crossref PubMed Scopus (16) Google Scholar, 11Srinivasan V.M. Ferguson S.D. Lee S. Weathers S.-P. Kerrigan B.C.P. Heimberger A.B. Tumor vaccines for malignant gliomas.Neurotherapeutics. 2017; 14: 345-357Crossref PubMed Scopus (37) Google Scholar, 12Kamran N. Calinescu A. Candolfi M. Chandran M. Mineharu Y. Asad A.S. Koschmann C. Nunez F.J. Lowenstein P.R. Castro M.G. Recent advances and future of immunotherapy for glioblastoma.Expert Opin. Biol. Ther. 2016; 16: 1245-1264Crossref PubMed Scopus (56) Google Scholar, 13Ott P.A. Hu Z. Keskin D.B. Shukla S.A. Sun J. Bozym D.J. Zhang W. Luoma A. Giobbie-Hurder A. Peter L. Chen C. Olive O. Carter T.A. Li S. Lieb D.J. Eisenhaure T. Gjini E. Stevens J. Lane W.J. Javeri I. Nellaiappan K. Salazar A.M. Daley H. Seaman M. Buchbinder E.I. Yoon C.H. Harden M. Lennon N. Gabriel S. Rodig S.J. Barouch D.H. Aster J.C. Getz G. Wucherpfennig K. Neuberg D. Ritz J. Lander E.S. Fritsch E.F. Hacohen N. Wu C.J. An immunogenic personal neoantigen vaccine for patients with melanoma.Nature. 2017; 547: 1-25Crossref Scopus (1533) Google Scholar). Much of the recent excitement about cancer immunotherapy stems from the success in treating patients with immune checkpoint modulators, which induce anti-cancer T cell immune reactions that can break tolerance and bring about complete responses in increasingly larger percentages of patients (14Sharma P. Allison J.P.P. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential.Cell. 2015; 161: 205-214Abstract Full Text Full Text PDF PubMed Scopus (1506) Google Scholar, 15Palucka A.K. Coussens L.M. The basis of oncoimmunology.Cell. 2016; 164: 1233-1247Abstract Full Text Full Text PDF PubMed Scopus (531) Google Scholar). Identification of tumor antigens is needed for development of effective cancer immunotherapy, including GBM, and a good source for such antigens are the pools of HLA-bound peptides presented preferentially, or even exclusively, by the tumor cells (16Dutoit V. Herold-Mende C. Hilf N. Schoor O. Beckhove P. Bucher J. Dorsch K. Flohr S. Fritsche J. Lewandrowski P. Lohr J. Rammensee H.-G. Stevanovic S. Trautwein C. Vass V. Walter S. Walker P.R. Weinschenk T. Singh-Jasuja H. Dietrich P.-Y. Exploiting the glioblastoma peptidome to discover novel tumour-associated antigens for immunotherapy.Brain. 2012; 135: 1042-1054Crossref PubMed Scopus (137) Google Scholar), reviewed in (17Bassani-Sternberg M. Coukos G. Mass spectrometry-based antigen discovery for cancer immunotherapy.Curr. Opin. Immunol. 2016; 41: 9-17Crossref PubMed Scopus (92) Google Scholar, 18Rammensee H.-G. Singh-Jasuja H. HLA ligandome tumor antigen discovery for personalized vaccine approach.Expert Rev. Vaccines. 2013; 12: 1211-1217Crossref PubMed Scopus (72) Google Scholar, 19Pol J. Bloy N. Buqué A. Eggermont A. Sautès-fridman C. Galon J. Tartour E. Kroemer G. Galluzzi L. Pol J. Bloy N. Buqué A. Eggermont A. Cremer I. Sautès-fridman C. Galon J. Tartour E. Zitvogel L. Kroemer G. Galluzzi L. Watch T. Pol J. Bloy N. Buqué A. Eggermont A. Cremer I. Zitvogel L. Pol J. Bloy N. Buqu A. Buque A. Eggermont A. Cremer I. Sautes-Fridman C. Galon J. Tartour E. Zitvogel L. Kroemer G. Galluzzi L. Trial Watch : Peptide-based anticancer vaccines Trial Watch : Peptide-based anticancer vaccines.Oncoimmunology. 2015; 4: e974411Crossref PubMed Scopus (93) Google Scholar, 20Comber J.D. Philip R. MHC class I antigen presentation and implications for developing a new generation of therapeutic vaccines.Ther. Adv. vaccines. 2014; 2: 77-89Crossref PubMed Scopus (81) Google Scholar). Tumor-specific antigens (TSA) and tumor-associated antigens (TAA) that can serve as candidates for cancer immunotherapeutics have been searched for extensively. Indeed, many such antigens were already identified, yet none has induced sufficiently strong anti-cancer immune reaction to eradicate the tumors in large cohorts of patients (18Rammensee H.-G. Singh-Jasuja H. HLA ligandome tumor antigen discovery for personalized vaccine approach.Expert Rev. Vaccines. 2013; 12: 1211-1217Crossref PubMed Scopus (72) Google Scholar, 20Comber J.D. Philip R. MHC class I antigen presentation and implications for developing a new generation of therapeutic vaccines.Ther. Adv. vaccines. 2014; 2: 77-89Crossref PubMed Scopus (81) Google Scholar, 21Heemskerk B. Kvistborg P. Schumacher T.N.M. The cancer antigenome.EMBO J. 2012; 32: 194-203Crossref PubMed Scopus (185) Google Scholar). One subset of preferred TAAs are Cancer/Testis Antigens (CTA), which are abnormally expressed in the malignant cells, and are normally expressed only in fetal tissues and in immune privileged sites, such as the male germ cells, placenta and ovary, but are absent from the normal somatic cells of any healthy tissue (22Chen Y.T. Scanlan M.J. Sahin U. Türeci O. Gure A.O. Tsang S. Williamson B. Stockert E. Pfreundschuh M. Old L.J. A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening.Proc. Natl. Acad. Sci. U.S.A. 1997; 94: 1914-1918Crossref PubMed Scopus (1093) Google Scholar), reviewed in (23Whitehurst A.W. Cause and consequence of cancer/testis antigen activation in cancer.Annu. Rev. Pharmacol. Toxicol. 2014; 54: 251-272Crossref PubMed Scopus (170) Google Scholar). Another special groups of tumor antigens are neoantigens, which are attracting significant attention as potential targets for cancer immunotherapy (24Schumacher T.N. Schreiber R.D. Neoantigens in cancer immunotherapy.Science. 2015; 348: 69-74Crossref PubMed Scopus (2981) Google Scholar). These are more likely common in tumors with higher mutational load and therefore less frequently found in GBM. glioblastoma major histocompatibility complex human leukocytes antigen cancer/testis antigen tumor specific antigens tumor associated antigens soluble human leukocytes antigen membranal human leukocytes antigen false discovery rate blood brain barrier. glioblastoma major histocompatibility complex human leukocytes antigen cancer/testis antigen tumor specific antigens tumor associated antigens soluble human leukocytes antigen membranal human leukocytes antigen false discovery rate blood brain barrier. HLA class I molecules are predominantly expressed as membrane anchored, cell-surface proteins (mHLA). Yet, the HLA molecules are also released from cells (25Charlton R.K. Zmijewski C.M. Soluble HL-A7 antigen: localization in the beta-lipoprotein fraction of human serum.Science. 1970; 170: 636-637Crossref PubMed Scopus (148) Google Scholar, 26van Rood J.J. van Leeuwen A. van Santen M.C. Anti HL-A2 inhibitor in normal human serum.Nature. 1970; 226: 366-367Crossref PubMed Scopus (200) Google Scholar), reviewed in (27Tabayoyong W.B. Zavazava N. Soluble HLA revisited.Leuk. Res. 2007; 31: 121-125Crossref PubMed Scopus (36) Google Scholar), and can be recovered from the soluble HLA molecules in human plasma (sHLA) for analysis of their bound peptidome (28Bassani-Sternberg M. Barnea E. Beer I. Avivi I. Katz T. Admon A. Soluble plasma HLA peptidome as a potential source for cancer biomarkers.Proc. Natl. Acad. Sci. U.S.A. 2010; 107: 18769-18776Crossref PubMed Scopus (101) Google Scholar, 29Ritz D. Gloger A. Weide B. Garbe C. Neri D. Fugmann T. High-sensitivity HLA class I peptidome analysis enables a precise definition of peptide motifs and the identification of peptides from cell lines and patients' sera.Proteomics. 2016; 16: 1570-1580Crossref PubMed Scopus (44) Google Scholar, 30Ritz D. Gloger A. Neri D. Fugmann T. Purification of soluble HLA class I complexes from human serum or plasma deliver high quality immuno peptidomes required for biomarker discovery.Proteomics. 2017; 17: 1-6Crossref Scopus (17) Google Scholar). Confounding factors that limit the use of the sHLA peptidome as a source for biomarkers are the presence of small amounts of sHLA molecules in the plasma of all healthy individuals (27Tabayoyong W.B. Zavazava N. Soluble HLA revisited.Leuk. Res. 2007; 31: 121-125Crossref PubMed Scopus (36) Google Scholar, 31Puppo F. Scudeletti M. Indiveri F. Ferrone S. Serum HLA class I antigens: markers and modulators of an immune response?.Immunol. Today. 1995; 16: 124-127Abstract Full Text PDF PubMed Scopus (112) Google Scholar), and in addition, some of the sHLA alleles are released to the circulation in larger amounts than others (32Adamashvili I.M. Fraser P.A. McDonald J.C. Association of serum concentration of soluble class I HLA with HLA allotypes.Transplantation. 1996; 61: 984-987Crossref PubMed Scopus (28) Google Scholar, 33Campoli M. Ferrone S. Tumor escape mechanisms: potential role of soluble HLA antigens and NK cells activating ligands.Tissue Antigens. 2008; 72: 321-334Crossref PubMed Scopus (59) Google Scholar). It has been suggested that shedding of the sHLA molecules represents a mechanism for evasion of immune recognition of tumor cells, which release larger amounts of sHLA molecules relative to the healthy cells (33Campoli M. Ferrone S. Tumor escape mechanisms: potential role of soluble HLA antigens and NK cells activating ligands.Tissue Antigens. 2008; 72: 321-334Crossref PubMed Scopus (59) Google Scholar). Because the sHLA molecules carry with them their original load of bound peptides, the immunopeptidomes of the sHLA molecules of cancer patients include disproportionally large fractions of peptides originating from the tumor cells, even though the tumors constitute just small fraction of the body mass. It is therefore intuitive to assume that biomarkers of disease can be found among those sHLA peptides. Such plasma-sHLA molecules could then be a useful source of cancer biomarkers for early detection of recurrence during follow-up (28Bassani-Sternberg M. Barnea E. Beer I. Avivi I. Katz T. Admon A. Soluble plasma HLA peptidome as a potential source for cancer biomarkers.Proc. Natl. Acad. Sci. U.S.A. 2010; 107: 18769-18776Crossref PubMed Scopus (101) Google Scholar). Analyses of the HLA peptidomes, based on immunoaffinity purification of the mHLA molecules from cell lines, tumor tissues or patients' plasma, followed by analysis of the bound peptides by chromatography and tandem mass spectrometry (34Hunt D.F. Henderson R.A. Shabanowitz J. Sakaguchi K. Michel H. Sevilir N. Cox a. L. Appella E. Engelhard V.H. Characterization of peptides bound to the class I MHC molecule HLA-A2.1 by mass spectrometry.Science. 1992; 255: 1261-1263Crossref PubMed Scopus (1051) Google Scholar) currently results in identification of thousands of HLA bound peptides (35Granados D.P. Laumont C.M. Thibault P. Perreault C. The nature of self for T cells-a systems-level perspective.Curr. Opin. Immunol. 2015; 34: 1-8Crossref PubMed Scopus (55) Google Scholar, 36Schumacher F.-R. Delamarre L. Jhunjhunwala S. Modrusan Z. Phung Q.T. Elias J.E. Lill J.R. Building proteomic tool boxes to monitor MHC class I and class II peptides.Proteomics. 2017; 17: 1600061Crossref Scopus (24) Google Scholar, 37de Verteuil D. Granados D.P. Thibault P. Perreault C. Origin and plasticity of MHC I-associated self peptides.Autoimmun. Rev. 2012; 11: 627-635Crossref PubMed Scopus (40) Google Scholar, 38Fritsche J. Rakitsch B. Hoffgaard F. Römer M. Schuster H. Kowalewski D.J. Priemer M. Stos-Zweifel V. Hoerzer H. Satelli A. Sonntag A. Goldfinger V. Song C. Mahr A. Ott M. Schoor O. Weinschenk T. Hörzer H. Satelli A. Sonntag A. Goldfinger V. Song C. Mahr A. Ott M. Schoor O. Weinschenk T. Translating immunopeptidomics to immunotherapy-decision-making for patient and personalized target selection.Proteomics. 2018; : 1700284Crossref Scopus (22) Google Scholar). HLA peptides were previously isolated from GBM cell lines (39Shraibman B. Kadosh D.M. Barnea E. Admon A. Human Leukocyte Antigen (HLA) Peptides derived from tumor antigens induced by inhibition of DNA methylation for development of drug-facilitated immunotherapy.Mol. Cell. Proteomics. 2016; 15: 3058-3070Abstract Full Text Full Text PDF PubMed Scopus (34) Google Scholar) and fresh tumors, including GBM (4Neidert M.C. Schoor O. Trautwein C. Trautwein N. Christ L. Melms A. Honegger J. Rammensee H.-G. Herold-Mende C. Dietrich P.-Y. Stevanović S. Natural HLA class I ligands from glioblastoma: extending the options for immunotherapy.J. Neurooncol. 2013; 111: 285-294Crossref PubMed Scopus (18) Google Scholar, 16Dutoit V. Herold-Mende C. Hilf N. Schoor O. Beckhove P. Bucher J. Dorsch K. Flohr S. Fritsche J. Lewandrowski P. Lohr J. Rammensee H.-G. Stevanovic S. Trautwein C. Vass V. Walter S. Walker P.R. Weinschenk T. Singh-Jasuja H. Dietrich P.-Y. Exploiting the glioblastoma peptidome to discover novel tumour-associated antigens for immunotherapy.Brain. 2012; 135: 1042-1054Crossref PubMed Scopus (137) Google Scholar). Similarly, the HLA peptidomes of tumors, such as melanoma (40Weinschenk T. Gouttefangeas C. Schirle M. Obermayr F. Walter S. Schoor O. Kurek R. Loeser W. Bichler K. Wernet D. Stevanović S. Rammensee H.-G. Integrated functional genomics approach for the design of patient-individual antitumor vaccines.Cancer Res. 2002; 62: 5818-5827PubMed Google Scholar, 41Bassani-Sternberg M. Bräunlein E. Klar R. Engleitner T. Sinitcyn P. Audehm S. Straub M. Weber J. Slotta-Huspenina J. Specht K. Martignoni M.E. Werner A. Hein R. Busch H D Peschel C. Rad R. Cox J. Mann M. Krackhardt A.M. Direct identification of clinically relevant neoepitopes presented on native human melanoma tissue by mass spectrometry.Nat. Commun. 2016; 7: 13404Crossref PubMed Scopus (398) Google Scholar), renal cell carcinoma (42Seliger B. Dressler S.P. Massa C. Recktenwald C.V. Altenberend F. Bukur J. Marincola F.M. Wang E. Stevanovic S. Lichtenfels R. Identification and characterization of human leukocyte antigen class I ligands in renal cell carcinoma cells.Proteomics. 2011; 11: 2528-2541Crossref PubMed Scopus (16) Google Scholar, 43Klatt M.G. Kowalewski D.J. Schuster H. Di Marco M. Hennenlotter J. Stenzl A. Rammensee H.-G. Stevanović S. Carcinogenesis of renal cell carcinoma reflected in HLA ligands: A novel approach for synergistic peptide vaccination design.Oncoimmunology. 2016; 5: e1204504Crossref PubMed Scopus (14) Google Scholar), leukemia (44Kowalewski D.J. Schuster H. Backert L. Berlin C. Kahn S. Kanz L. Salih H.R. Rammensee H.-G. Stevanovic S. Stickel J.S. HLA ligandome analysis identifies the underlying specificities of spontaneous antileukemia immune responses in chronic lymphocytic leukemia (CLL).Proc. Natl. Acad. Sci. U.S.A. 2015; 112: E166-E175Crossref PubMed Scopus (124) Google Scholar) and multiple myeloma (45Walz S. Stickel J.S. Kowalewski D.J. Schuster H. Weisel K. Backert L. Kahn S. Nelde A. Stroh T. Handel M. Kohlbacher O. Kanz L. Salih H.R. Rammensee H. Stevanović S. The antigenic landscape of multiple myeloma: mass spectrometry (re)defines targets for T-cell-based immunotherapy.Blood. 2015; 126: 1203-1213Crossref PubMed Scopus (78) Google Scholar) have been characterized. Further, several thousands of peptides have successfully been isolated from cancer patients' plasma-sHLA molecules (28Bassani-Sternberg M. Barnea E. Beer I. Avivi I. Katz T. Admon A. Soluble plasma HLA peptidome as a potential source for cancer biomarkers.Proc. Natl. Acad. Sci. U.S.A. 2010; 107: 18769-18776Crossref PubMed Scopus (101) Google Scholar, 30Ritz D. Gloger A. Neri D. Fugmann T. Purification of soluble HLA class I complexes from human serum or plasma deliver high quality immuno peptidomes required for biomarker discovery.Proteomics. 2017; 17: 1-6Crossref Scopus (17) Google Scholar). In this research, more than 35,000 plasma-sHLA and tumor-mHLA peptides from GBM patients were identified. These large HLA peptidomes included several hundred peptides, derived from both known TAAs and newly defined potential CTAs. We propose that these may serve as useful biomarkers and candidates for immunotherapeutics for GBM, as well as for other cancers. One hundred six plasma samples were obtained from 52 different GBM patients, collected prior to tumor removal and at different time points after the surgery (supplemental Table S1). Additionally, control plasma of healthy donors (n = 6) and patients diagnosed with ankylosing spondylitis (n = 30), as an exemplar inflammatory process, were used for this study. In addition, tumor samples were obtained from 10 of the 52 GBM patients for mHLA peptidome analysis, as well as for tumor proteome analysis. Peripheral blood (PB) and tumor tissues samples from GBM patients (University Hospital Heidelberg, Leiden University Medical Centre, Vall d'Hebron University Hospital, Université de Genève, Southampton Universities Hospital, Heidelberg University Medical Center, Herlev Hospital, and Rigshospitalet) and noncancerous controls (Universitario Central de Asturias and Bnei-Zion Hospital). The HLA tissue typing of the GBM patients and noncancerous controls, healthy, and Ankylosing Spondylitis patients are listed only with gender, age, and stage of disease in supplemental Table S1. All human bio-specimens were obtained with informed consent, and after approval of the relevant ethics committees. Peripheral blood samples were collected into EDTA tubes and cleared of the cells by centrifugation for 10 min at 1200 × g, at room temperature. The cleared plasma was stored frozen at −80 °C until use for sHLA purification. Tumor samples were frozen in liquid nitrogen immediately after dissection and stored at −80 °C until use, for both mHLA peptidome and proteome analyses of the same tissue extracts. Membranal HLA class I molecules were purified from GBM tumor tissues by immunoaffinity from the cells after lysis with 0.25% sodium deoxycholate, 0.2 mm iodoacetamide, 1 mm EDTA, 1:200 Protease Inhibitors Mixture (Sigma), 1 mm PMSF, and 1% octyl-d-glucopyranoside (Sigma) in PBS at 4 °C for 1 h. The lysate was cleared by centrifugation for 45 min at 18,000 rpm, at 4 °C. HLA class I molecules from the cleared lysate or from the fresh human plasma were immunoaffinity purified using the W6/32 mAb bound to Amino-Link beads (Thermo-Fisher Scientific, Waltham, MA) as in (28Bassani-Sternberg M. Barnea E. Beer I. Avivi I. Katz T. Admon A. Soluble plasma HLA peptidome as a potential source for cancer biomarkers.Proc. Natl. Acad. Sci. U.S.A. 2010; 107: 18769-18776Crossref PubMed Scopus (101) Google Scholar, 46Milner E. Gutter-Kapon L. Bassani-Strenberg M. Barnea E. Beer I. Admon A. The effect of proteasome inhibition on the generation of the human leukocyte antigen (HLA) peptidome.Mol. Cell. Proteomics. 2013; 12: 1853-1864Abstract Full Text Full Text PDF PubMed Scopus (58) Google Scholar). The HLA molecules with their bound peptides were eluted from the affinity column with five column volumes of 1% TFA. The eluted HLA class I proteins, and the released peptides were loaded on disposable C18 micro-columns (Harvard Apparatus, Holliston, MA), and the peptides fraction was recovered with 30% acetonitrile in 0.1% TFA, whereas the protein fraction was recovered with 80% acetonitrile in 0.1% TFA, as in (46Milner E. Gutter-Kapon L. Bassani-Strenberg M. Barnea E. Beer I. Admon A. The effect of proteasome inhibition on the generation of the human leukocyte antigen (HLA) peptidome.Mol. Cell. Proteomics. 2013; 12: 1853-1864Abstract Full Text Full Text PDF PubMed Scopus (58) Google Scholar). The peptide fractions were dried using vacuum centrifugation, reconstituted in 100 μl of 0.1% TFA, reloaded on C18 Stage-Tips (47Ishihama Y. Rappsilber J. Mann M. Modular stop and go extraction tips with stacked disks for parallel and multidimensional peptide fractionation in proteomics.J. Proteome Res. 2006; 5: 988-994Crossref PubMed Scopus (224) Google Scholar), eluted with 80% acetonitrile, dried, and reconstituted with 0.1% formic acid for LC-MS/MS analysis. Proteomics analysis was performed after trypsin digestion of the proteins in gel slices. Briefly, 40 μg protein sample from each tissue was ran in 10% acrylamide gel, stained with Coomassie, and each lane of the gel was sliced into five slices. The proteins in the gel slices were reduced with 2.8 mm DTT at 60 °C for 30 min and carbamidomethylated with 8.8 mm iodoacetamide in 100 mm ammonium bicarbonate at room temperature for 30 min and digested overnight at 37 °C in 10% acetonitrile and 10 mm ammonium bicarbonate with modified trypsin (Promega) at a 1:10 (wt/wt) enzyme-to-substrate ratio. This was repeated for another 4 h with a similar amount of trypsin, followed by 150 min gradients of LC-MS/MS of the tryptic peptides from each gel slice. Both the HLA and tryptic peptides were resolved by capillary chromatography and electrospray tandem mass spectrometry using a Q-Exactive-Plus mass spectrometer fitted with a capillary Ultimate 3000 RSLC nano-capillary UHPLC (Thermo-Fisher Scientific). The reversed phase chromatographs were with about 30 cm long, 75-micron inner diameter capillary columns, home-packed with 3.5 m silica ReproSil-Pur C18-AQ resin (Dr. Maisch GmbH, Ammerbuch-Entringen, Germany) as in (48Ishihama Y. Rappsilber J. Andersen J.S. Mann M. Microcolumns with self-assembled particle frits for proteomics.J. Chromatogr. A. 2002; 979: 233-239Crossref PubMed Scopus (260) Google Scholar). HLA and tryptic peptides were eluted with a linear gradient of 5–28% of acetonitrile in 0.1% formic acid. For both the HLA peptides and for the in-gel digested fractions, the capillary HPLC gradients were run at flow rates of 0.15 μl/minutes during 120 min. Data was acquired using a data-dependent “top 10” method, fragmenting the peptides by higher-energy collisional dissociation (HCD). Full scan MS spectra were acquired at a resolution of 70,000 at 200 m/z with a target value of 3 ×106 ions. MS/MS ions were accumulated to an AGC target value of 105 with a maximum injection time of 100 msec. For the HLA peptides with unassigned precursor ion charge states, or charge states of four and above, no fragmentation was performed. For tryptic peptides, the fragmentation was performed on charge states between 2 to 7. The peptide match option was set to Preferred. Normalized collision energy was set to 25% and MS/MS resolution was 17,500 at 200 m/z. Fragmented m/z values were dynamically excluded from further selection for 20 s. The MS data was analyzed by the MaxQuant computational proteomics platform (49Cox J. Mann M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification.Nat. Biotechnol. 2008; 26: 1367-1372Crossref PubMed Scopus (9154) Google Scholar) version 1.5.3.8 and searched with the Andromeda search engine (50Cox J. Neuhauser N. Michalski A. Scheltema R.A. Olsen J.V. Mann M. Andromeda: a peptide search engine integrated into the MaxQuant environment.J. Proteome Res. 2011; 10: 1794-1805Crossref PubMed Scopus (3450) Google Scholar). Peptide identifications were based on the human section of the UniProt database (http://www.uniprot.org/) of July 2015 containing 69,693 proteins from 91,459 entries, including one sequence of mutated EGFR-VIII. An additional FASTA file, containing 638 entries of mutated peptides (sequences in supplemental Table S10) was used during the same MaxQuant analysis (however, none of these was identified in the HLA peptidome anal
DOI: 10.1093/neuonc/noaa303
2020
Cited 41 times
H3K27me3 loss indicates an increased risk of recurrence in the Tübingen meningioma cohort
Abstract Background A loss of the trimethylation of lysine 27 of histone H3 (H3K27me3) in meningioma has been recently suggested as an adjunct to identify subsets of higher risk of recurrence. The aim of the present study was to assess the prognostic value of H3K27 histone trimethylation and its potential clinical utility in the “Tübingen meningioma cohort.” Methods Patients who underwent meningioma resection between October 2003 and December 2015 at the University Hospital Tübingen were included. Immunohistochemical stainings for H3K27me3 and the proliferation marker MIB1 were assessed and correlated with clinical parameters using univariate and multivariate Cox regressions as well as Pearson's chi-squared and log-rank test. Results Overall, 1268 meningiomas were analyzed with a female to male ratio of 2.6 and a mean age of 58.7 years (range 8.3–91.0). With 163 cases lost to follow up, 1103 cases were available for further analysis with a mean follow-up of 40.3 months (range 1.1–186.3). Male gender, younger age, intracranial tumor localization, progressive tumor, subtotal resection, higher WHO grade, increased MIB1 rate, and loss of H3K27me3 were significant negative prognostic factors in the univariate analysis. H3K27me3 status and all other prognostic factors, except age and tumor location, remained significant in the multivariate model. Furthermore, adjuvant radiotherapy was an independent positive prognostic factor. Conclusions Loss of H3K27me3 combined with MIB1 labeling index are independent prognostic factors in meningioma. These data from the Tübingen meningioma cohort support the clinical utility of H3K27me3 immunohistochemical staining in meningioma and its integration into the routine histopathological workup.
DOI: 10.1158/1078-0432.ccr-20-4256
2021
Cited 34 times
Phase I Assessment of Safety and Therapeutic Activity of BAY1436032 in Patients with IDH1-Mutant Solid Tumors
BAY1436032, an inhibitor of mutant isocitrate dehydrogenase 1 (mIDH1), was active against multiple IDH1-R132X solid tumors in preclinical models. This first-in-human study was designed to determine the safety and pharmacokinetics of BAY1436032, and to evaluate its potential pharmacodynamics and antitumor effects.The study comprised of dose escalation and dose expansion cohorts. BAY1436032 tablets were orally administered twice daily on a continuous basis in subjects with mIDH1 solid tumors.In dose escalation, 29 subjects with various tumor types were administered BAY1436032 across five doses (150-1,500 mg twice daily). BAY1432032 exhibited a relatively short half-life. Most evaluable subjects experienced target inhibition as indicated by a median maximal reduction of plasma R-2-hydroxyglutarate levels of 76%. BAY1436032 was well tolerated and an MTD was not identified. A dose of 1,500 mg twice daily was selected for dose expansion, where 52 subjects were treated in cohorts representing four different tumor types [lower grade glioma (LGG), glioblastoma, intrahepatic cholangiocarcinoma, and a basket cohort of other tumor types]. The best clinical outcomes were in subjects with LGG (n = 35), with an objective response rate of 11% (one complete response and three partial responses) and stable disease in 43%. As of August 2020, four of these subjects were in treatment for >2 years and still ongoing. Objective responses were observed only in LGG.BAY1436032 was well tolerated and showed evidence of target inhibition and durable objective responses in a small subset of subjects with LGG.
DOI: 10.3390/cancers13102400
2021
Cited 29 times
Targeting CSF1R Alone or in Combination with PD1 in Experimental Glioma
Glioblastoma is an aggressive primary tumor of the central nervous system. Targeting the immunosuppressive glioblastoma-associated microenvironment is an interesting therapeutic approach. Tumor-associated macrophages represent an abundant population of tumor-infiltrating host cells with tumor-promoting features. The colony stimulating factor-1/ colony stimulating factor-1 receptor (CSF-1/CSF1R) axis plays an important role for macrophage differentiation and survival. We thus aimed at investigating the antiglioma activity of CSF1R inhibition alone or in combination with blockade of programmed death (PD) 1. We investigated combination treatments of anti-CSF1R alone or in combination with anti-PD1 antibodies in an orthotopic syngeneic glioma mouse model, evaluated post-treatment effects and assessed treatment-induced cytotoxicity in a coculture model of patient-derived microtumors (PDM) and autologous tumor-infiltrating lymphocytes (TILs) ex vivo. Anti-CSF1R monotherapy increased the latency until the onset of neurological symptoms. Combinations of anti-CSF1R and anti-PD1 antibodies led to longterm survivors in vivo. Furthermore, we observed treatment-induced cytotoxicity of combined anti-CSF1R and anti-PD1 treatment in the PDM/TILs cocultures ex vivo. Our results identify CSF1R as a promising therapeutic target for glioblastoma, potentially in combination with PD1 inhibition.
DOI: 10.1200/jco.22.01862
2023
Cited 9 times
Intraoperative MRI-Guided Resection Is Not Superior to 5-Aminolevulinic Acid Guidance in Newly Diagnosed Glioblastoma: A Prospective Controlled Multicenter Clinical Trial
Prospective data suggested a superiority of intraoperative MRI (iMRI) over 5-aminolevulinic acid (5-ALA) for achieving complete resections of contrast enhancement in glioblastoma surgery. We investigated this hypothesis in a prospective clinical trial and correlated residual disease volumes with clinical outcome in newly diagnosed glioblastoma.This is a prospective controlled multicenter parallel-group trial with two center-specific treatment arms (5-ALA and iMRI) and blinded evaluation. The primary end point was complete resection of contrast enhancement on early postoperative MRI. We assessed resectability and extent of resection by an independent blinded centralized review of preoperative and postoperative MRI with 1-mm slices. Secondary end points included progression-free survival (PFS) and overall survival (OS), patient-reported quality of life, and clinical parameters.We recruited 314 patients with newly diagnosed glioblastomas at 11 German centers. A total of 127 patients in the 5-ALA and 150 in the iMRI arm were analyzed in the as-treated analysis. Complete resections, defined as a residual tumor ≤0.175 cm³, were achieved in 90 patients (78%) in the 5-ALA and 115 (81%) in the iMRI arm (P = .79). Incision-suture times (P < .001) were significantly longer in the iMRI arm (316 v 215 [5-ALA] minutes). Median PFS and OS were comparable in both arms. The lack of any residual contrast enhancing tumor (0 cm³) was a significant favorable prognostic factor for PFS (P < .001) and OS (P = .048), especially in methylguanine-DNA-methyltransferase unmethylated tumors (P = .006).We could not confirm superiority of iMRI over 5-ALA for achieving complete resections. Neurosurgical interventions in newly diagnosed glioblastoma shall aim for safe complete resections with 0 cm³ contrast-enhancing residual disease, as any other residual tumor volume is a negative predictor for PFS and OS.
DOI: 10.1093/noajnl/vdad012
2023
Cited 8 times
Clinical outcome of biomarker-guided therapies in adult patients with tumors of the nervous system
The clinical utility of molecular profiling and targeted therapies for neuro-oncology patients outside of clinical trials is not established. We aimed at investigating feasibility and clinical utility of molecular profiling and targeted therapy in adult patients with advanced tumors in the nervous system within a prospective observational study.molecular tumor board (MTB)@ZPM (NCT03503149) is a prospective observational precision medicine study for patients with advanced tumors. After inclusion of patients, we performed comprehensive molecular profiling, formulated ranked biomarker-guided therapy recommendations based on consensus by the MTB, and collected prospective clinical outcome data.Here, we present initial data of 661 adult patients with tumors of the nervous system enrolled by December 31, 2021. Of these, 408 patients were presented at the MTB. Molecular-instructed therapy recommendations could be made in 380/408 (93.1%) cases and were prioritized by evidence levels. Therapies were initiated in 86/380 (22.6%) cases until data cutoff. We observed a progression-free survival ratio >1.3 in 31.3% of patients.Our study supports the clinical utility of biomarker-guided therapies for neuro-oncology patients and indicates clinical benefit in a subset of patients. Our data might inform future clinical trials, translational studies, and even clinical care.
DOI: 10.1002/ana.21057
2007
Cited 71 times
Synergistic antiglioma activity of radiotherapy and enzastaurin
Abstract Objective Radiotherapy is an essential treatment modality for malignant gliomas, but it exerts adverse effects via promotion of glioma cell invasion in experimental glioma. Furthermore, irradiation induces vascular endothelial growth factor (VEGF) levels in gliomas, which is associated with poor prognosis. Here, we investigate the combination of the protein kinase C‐β inhibitor enzastaurin (ENZA) and radiotherapy in vitro and in vivo in comparison with either treatment alone. Methods We analyzed the effects of ENZA and irradiation on migration, apoptosis, and proliferation of glioma cells, as well as VEGF secretion in vitro. Neurotoxicity of ENZA was assessed in cerebellar granule neurons. After orthotopic intracerebral implantation of LNT‐229 glioma cells in nude mice, the effects of in situ cerebral irradiation and oral application of ENZA on survival, tumor size, VEGF expression, apoptosis, and microvessel density in vivo were analyzed. Results Combining cerebral irradiation with ENZA leads to longer survival in vivo. ENZA diminishes tumor volume, irradiation‐induced tumor satellite formation, upregulation of VEGF expression in vitro and in vivo, as well as enhanced microvessel density in vivo. Importantly, ENZA is not neurotoxic in vitro or in vivo. Interpretation Long‐term administration of ENZA after radiotherapy is feasible and leads to long‐term survival without neurotoxicity. Ann Neurol 2007
DOI: 10.1007/s11060-014-1365-x
2014
Cited 53 times
Cilengitide treatment of newly diagnosed glioblastoma patients does not alter patterns of progression
The integrin antagonist cilengitide has been explored as an adjunct with anti-angiogenic properties to standard of care temozolomide chemoradiotherapy (TMZ/RT → TMZ) in newly diagnosed glioblastoma. Preclinical data as well as anecdotal clinical observations indicate that anti-angiogenic treatment may result in altered patterns of tumor progression. Using a standardized approach, we analyzed patterns of progression on MRI in 21 patients enrolled onto a phase 2 trial of cilengitide added to TMZ/RT → TMZ in newly diagnosed glioblastoma. Thirty patients from the experimental treatment arm of the EORTC/NCIC pivotal TMZ trial served as a reference. MRIcro software was used to map location and extent of initial preoperative and recurrent tumors on MRI of both groups into the same stereotaxic space which were then analyzed using an automated tool of image analysis. Clinical and outcome data of the cilengitide-treated patients were similar to those of the EORTC/NCIC trial except for a higher proportion of patients with a methylated O6-methylguanyl-DNA-methyltransferase gene promoter. Analysis of recurrence pattern revealed neither a difference in the size of the recurrent tumor nor in the distance of the recurrences from the preoperative tumor location between groups. Overall frequencies of distant recurrences were 20 % in the reference group and 19 % (4/21 patients) in the cilengitide group. Compared with TMZ/RT → TMZ alone, the addition of cilengitide does not alter patterns of progression. This analysis does not support concerns that integrin antagonism by cilengitide may induce a more aggressive phenotype at progression, but also provides no evidence for an anti-invasive activity of cilengitide in patients with newly diagnosed glioblastoma.
DOI: 10.1093/neuonc/nox160
2017
Cited 49 times
Molecular differences in IDH wildtype glioblastoma according to MGMT promoter methylation
O6-methylguanine-DNA-methyltransferase (MGMT) promoter methylation status is a predictive biomarker in glioblastoma. We investigated whether this marker furthermore defines a molecularly distinct tumor subtype with clinically different outcome. We analyzed copy number variation (CNV) and methylation profiles of 1095 primary and 92 progressive isocitrate dehydrogenase wildtype glioblastomas, including paired samples from 49 patients. DNA mutation data from 182 glioblastoma samples of The Cancer Genome Atlas (TCGA) and RNA expression from 107 TCGA and 55 Chinese Glioma Genome Atlas samples were analyzed. Among untreated glioblastomas, MGMT promoter methylated (mMGMT) and unmethylated (uMGMT) tumors did not show different CNV or specific gene mutations, but a higher mutation count in mMGMT tumors. We identified 3 methylation clusters. Cluster 1 showed the highest average methylation and was enriched for mMGMT tumors. Seventeen genes including gastrulation brain homeobox 2 (GBX2) were found to be hypermethylated and downregulated on the mRNA level in mMGMT tumors. In progressive glioblastomas, platelet derived growth factor receptor alpha (PDGFRA) and GLI2 amplifications were enriched in mMGMT tumors. Methylated MGMT tumors gain PDGFRA amplification of PDGFRA, whereas uMGMT tumors with amplified PDGFRA frequently lose this amplification upon progression. Glioblastoma patients surviving <6 months and with mMGMT harbored less frequent epidermal growth factor receptor (EGFR) amplifications, more frequent TP53 mutations, and a higher tumor necrosis factor–nuclear factor-kappaB (TNF-NFκB) pathway activation compared with patients surviving >12 months. MGMT promoter methylation status does not define a molecularly distinct glioblastoma subpopulation among untreated tumors. Progressive mMGMT glioblastomas and mMGMT tumors of patients with short survival tend to have more unfavorable molecular profiles.
DOI: 10.1159/000480643
2017
Cited 41 times
Targeting the Potassium Channel Kv1.3 Kills Glioblastoma Cells
Glioblastoma (GBM) is one of the most aggressive cancers, counting for a high number of the newly diagnosed patients with central nervous system (CNS) cancers in the United States and Europe. Major features of GBM include aggressive and invasive growth as well as a high resistance to treatment. Kv1.3, a potassium channel of the shaker family, is expressed in the inner mitochondrial membrane of many cancer cells. Inhibition of mitochondrial Kv1.3 was shown to induce apoptosis in several tumor cells at doses that were not lethal for normal cells.We investigated the expression of Kv1.3 in different glioma cell lines by immunocytochemistry, western blotting and electron microscopy and analyzed the effect of newly synthesized, mitochondria-targeted, Kv1.3 inhibitors on the induction of cell death in these cells. Finally, we performed in vivo studies on glioma bearing mice.Here, we report that Kv1.3 is expressed in mitochondria of human and murine GL261, A172 and LN308 glioma cells. Treatment with the novel Kv1.3 inhibitors PAPTP or PCARBTP as well as with clofazimine induced massive cell death in glioma cells, while Psora-4 and PAP-1 were almost without effect. However, in vivo experiments revealed that the drugs had no effect on orthotopic brain tumors in vivo.These data serve as proof of principle that Kv1.3 inhibitors kills GBM cells, but drugs that act in vivo against glioblastoma must be developed to translate these findings in vivo.
DOI: 10.1016/j.omto.2018.12.010
2019
Cited 39 times
Measles Virus-Based Treatments Trigger a Pro-inflammatory Cascade and a Distinctive Immunopeptidome in Glioblastoma
Glioblastoma is an aggressive primary brain tumor with bad prognosis. On the other hand, oncolytic measles virus (MeV) therapy is an experimental glioma treatment strategy with clinical safety and first evidence of anti-tumoral efficacy. Therefore, we investigated the combination of MeV with conventional therapies by cytotoxic survival assays in long-term glioma cell lines LN229, LNZ308, and glioma stem-like GS8 cells, as well as the basal viral infectivity in primary glioblastoma cultures T81/16, T1094/17, and T708/16. We employed Chou-Talalay analysis to identify the synergistic treatment sequence chemotherapy, virotherapy, and finally radiotherapy (CT-VT-RT). RNA sequencing and immunopeptidome analyses were used to delineate treatment-induced molecular and immunological profiles. CT-VT-RT displayed synergistic anti-glioma activity and initiated a type 1 interferon response, along with canonical Janus kinase-signal transducers and activators of transcription (JAK-STAT) signaling, and downstream interferon-stimulated genes were induced, resulting in apoptotic cascades. Furthermore, antigen presentation along with immunostimulatory chemokines was increased in CT-VT-RT-treated glioma cells, indicating a treatment-induced pro-inflammatory phenotype. We identified novel treatment-induced viral and tumor-associated peptides through HLA ligandome analysis. Our data delineate an actionable treatment-induced molecular and immunological signature of CT-VT-RT, and they could be exploited for the design of novel tailored treatment strategies involving virotherapy and immunotherapy.
DOI: 10.3390/vaccines9050428
2021
Cited 25 times
Designing a SARS-CoV-2 T-Cell-Inducing Vaccine for High-Risk Patient Groups
We describe the results of two vaccinations of a self-experimenting healthy volunteer with SARS-CoV-2-derived peptides performed in March and April 2020, respectively. The first set of peptides contained eight peptides predicted to bind to the individual’s HLA molecules. The second set consisted of ten peptides predicted to bind promiscuously to several HLA-DR allotypes. The vaccine formulation contained the new TLR 1/2 agonist XS15 and was administered as an emulsion in Montanide as a single subcutaneous injection. Peripheral blood mononuclear cells isolated from blood drawn before and after vaccinations were assessed using Interferon-γ ELISpot assays and intracellular cytokine staining. We detected vaccine-induced CD4 T cell responses against six out of 11 peptides predicted to bind to HLA-DR after 19 days, following vaccination, for one peptide already at day 12. We used these results to support the design of a T-cell-inducing vaccine for application in high-risk patients, with weakened lymphocyte performance. Meanwhile, an according vaccine, incorporating T cell epitopes predominant in convalescents, is undergoing clinical trial testing.
DOI: 10.1093/neuonc/noad038
2023
Cited 5 times
Diagnosis and management of complications from the treatment of primary central nervous system tumors in adults
Abstract Central nervous system (CNS) tumor patients commonly undergo multimodality treatment in the course of their disease. Adverse effects and complications from these interventions have not been systematically studied, but pose significant challenges in clinical practice and impact function and quality of life, especially in the management of long-term brain tumor survivors. Here, the European Association of Neuro-Oncology (EANO) has developed recommendations to prevent, diagnose, and manage adverse effects and complications in the adult primary brain CNS tumor (except lymphomas) patient population with a specific focus on surgery, radiotherapy, and pharmacotherapy. Specifically, we also provide recommendations for dose adaptations, interruptions, and reexposure for pharmacotherapy that may serve as a reference for the management of standard of care in clinical trials. We also summarize which interventions are unnecessary, inactive or contraindicated. This consensus paper should serve as a reference for the conduct of standard therapy within and outside of clinical trials.
DOI: 10.1093/brain/awh563
2005
Cited 73 times
Lessons from the bone marrow: how malignant glioma cells attract adult haematopoietic progenitor cells
Stem and progenitor cells (PCs) of various lineages have become attractive vehicles to improve therapeutic gene delivery to cancers, notably glioblastoma.Here we report that adult human and murine haematopoietic PCs display a tropism for intracerebral gliomas but not for normal brain tissue in mice.Organotypic hippocampal slice culture and spheroid confrontation assays confirm a directed PC migration towards glioma cells ex vivo and in vitro.RNA interference-mediated disruption of transforming growth factor beta (TGF-b) synthesis by the glioma cells strongly inhibits PC migration.We delineate a CXC chemokine ligand (CXCL) 12-dependent pathway of TGF-b-induced PC migration that is facilitated by MMP-9-mediated stem cell factor cleavage in vitro.Moreover, neutralizing antibodies to CXCL12 strongly reduce PC homing to experimental gliomas in vivo.Thus, we define here the molecular mechanism underlying the glioma tropism of the probably most easily accessible PC population suitable for cancer therapy, that is, adult haematopoietic PC.
DOI: 10.1016/j.jneuroim.2007.07.002
2007
Cited 58 times
Elevated HLA-E levels in human glioblastomas but not in grade I to III astrocytomas correlate with infiltrating CD8+ cells
HLA-E is a ligand for the immune-inhibitory NKG2A receptor expressed on NK and T cells. To investigate HLA-E expression and immune cell infiltration in human astrocytic tumors in vivo, we analyzed normal CNS controls and astrocytomas of all WHO grades by immunohistochemistry. Both, CD8(+) immune cell infiltration and HLA-E expression were significantly higher in astrocytic tumors than in normal brain. Further, HLA-E expression levels and immune cell infiltration were significantly correlated in WHO grade IV glioblastomas. Thus, HLA-E overexpression in glioblastomas may be triggered by T and NK cell infiltration.
DOI: 10.1093/brain/awt333
2013
Cited 43 times
Thymosin beta 4 gene silencing decreases stemness and invasiveness in glioblastoma
Thymosin beta 4 is a pleiotropic actin-sequestering polypeptide that is involved in wound healing and developmental processes. Thymosin beta 4 gene silencing promotes differentiation of neural stem cells whereas thymosin beta 4 overexpression initiates cortical folding of developing brain hemispheres. A role of thymosin beta 4 in malignant gliomas has not yet been investigated. We analysed thymosin beta 4 staining on tissue microarrays and performed interrogations of the REMBRANDT and the Cancer Genome Atlas databases. We investigated thymosin beta 4 expression in seven established glioma cell lines and seven glioma-initiating cell lines and induced or silenced thymosin beta 4 expression by lentiviral transduction in LNT-229, U87MG and GS-2 cells to study the effects of altered thymosin beta 4 expression on gene expression, growth, clonogenicity, migration, invasion, self-renewal and differentiation capacity in vitro, and tumorigenicity in vivo. Thymosin beta 4 expression increased with grade of malignancy in gliomas. Thymosin beta 4 gene silencing in LNT-229 and U87MG glioma cells inhibited migration and invasion, promoted starvation-induced cell death in vitro and enhanced survival of glioma-bearing mice. Thymosin beta 4 gene silencing in GS-2 cells inhibited self-renewal and promoted differentiation in vitro and decreased tumorigenicity in vivo. Gene expression analysis suggested a thymosin beta 4-dependent regulation of mesenchymal signature genes and modulation of TGFβ and p53 signalling networks. We conclude that thymosin beta 4 should be explored as a novel molecular target for anti-glioma therapy.
DOI: 10.1016/j.wneu.2016.10.062
2017
Cited 39 times
Risk Factors of Preoperative and Early Postoperative Seizures in Patients with Meningioma: A Retrospective Single-Center Cohort Study
Well-defined risk factors for the identification of patients with meningioma who might benefit from preoperative or early postoperative seizure prophylaxis are unknown. We investigated and quantified risk factors to determine individual risks of seizure occurrence in patients with meningioma.A total of 634 adult patients with meningioma were included in this retrospective cohort study. Patient gender and age, tumor location, grade and volume, usage of antiepileptic drugs (AEDs) and extent of resection were determined.Preoperative and early postoperative seizures occurred in 15% (n = 97) and 5% (n = 21) of the patients, respectively. Overall, 502 and 418 patients were eligible for multivariate logistic regression analyses of preoperative and early postoperative seizures, respectively. Male gender (odds ratio [OR], 2.06; P = 0.009), a non-skull base location (OR, 4.43; P < 0.001), and a tumor volume of >8 cm3 (OR, 3.05; P = 0.002) were associated with a higher risk of preoperative seizures and were used to stratify the patients into 3 prognostic groups. The high-risk subgroup of patients with meningioma showed a seizure rate of >40% (OR, 9.8; P < 0.001). Only a non-skull base tumor location (OR, 2.61; P = 0.046) was identified as a significant risk factor for early postoperative seizures. AEDs did not reduce early postoperative seizure occurrence.Seizure prophylaxis might be considered for patients at high risk of developing seizures who are for other reasons being considered for watchful waiting instead of resection. In contrast, our data do not provide any evidence of the efficacy of perioperative AEDs in patients with meningioma.
DOI: 10.1002/jmri.25221
2016
Cited 35 times
MR spectroscopy for in vivo assessment of the oncometabolite 2‐hydroxyglutarate and its effects on cellular metabolism in human brain gliomas at 9.4T
To examine in vivo metabolic alterations in the isocitrate dehydrogenase (IDH) mutated gliomas using magnetic resonance spectroscopy (MRS) at magnetic field 9.4T.Spectra were acquired with a 9.4T whole-body scanner with the use of a custom-built head coil (16 channel transmit and 31 channel receive). A modified stimulated echo acquisition mode (STEAM) sequence was used for localization. Eighteen patients with brain tumors of probable glial origin participated in this study. The study was performed in accordance with the guidelines of the local Ethics Committee.The increased spectral resolution allowed us to directly address metabolic alterations caused by the specific pathophysiology of IDH mutations including the presence of the oncometabolite 2-hydroxglutarate (2HG) and a significant decrease of the pooled glutamate and glutamine (20%, P = 0.024), which probably reflects an attempt to replenish α-ketoglutarate lost by conversion to 2HG. We also observed significantly reduced glutathione (GSH) levels (39%, P = 0.019), which could be similarly caused by depletion of dihydronicotinamide-adenine dinucleotide phosphate (NADPH) during this conversion in IDH mutant gliomas.We demonstrate that MRS at 9.4T provides a noninvasive measure of 2HG in vivo, which may be used for therapy planning and prognostication, and may provide insights into related pathophysiologic metabolic alterations associated with IDH mutations. J. MAGN. RESON. IMAGING 2016;44:823-833.
DOI: 10.1016/j.neurad.2017.07.005
2018
Cited 35 times
In vivo assessment of tumor heterogeneity in WHO 2016 glioma grades using diffusion kurtosis imaging: Diagnostic performance and improvement of feasibility in routine clinical practice
To assess the diagnostic performance of normalized and non-normalized diffusion kurtosis imaging (DKI) metrics extracted from different tumor volume data for grading glioma according to the integrated approach of the revised 2016 WHO classification. Sixty patients with histopathologically confirmed glioma, who provided written informed consent, were retrospectively assessed between 01/2013 and 08/2016 from a prospective trial approved by the local institutional review board. Mean kurtosis (MK) and mean diffusivity (MD) metrics from DKI were assessed by two blinded physicians from four different volumes of interest (VOI): whole solid tumor including (VOItu-ed) and excluding perifocal edema (VOItu), infiltrative zone (VOIed), and single slice of solid tumor core (VOIslice). Intra-class correlation coefficient (ICC) was calculated to assess inter-rater agreement. One-way ANOVA was used to compare MK between 2016 CNS WHO tumor grades. Friedman's test compared MK and MD of each VOI. Spearman's correlation coefficient was used to correlate MK with 2016 CNS WHO tumor grades. ROC analysis was performed on MK for significant results. The MK assessment showed excellent inter-rater agreement for each VOI (ICC, 0.906–0.955). MK was significantly lower in IDHmutant astrocytoma (0.40 ± 0.07), than in 1p/19q-confirmed oligodendroglioma (0.54 ± 0.10, P = 0.001) or IDHwild-type glioblastoma (0.68 ± 0.13, P < 0.001). MK and 2016 WHO tumor grades were strongly and positively correlated (VOItu-ed, r = 0.684; VOItu, r = 0.734; VOIed, r = 0.625; VOIslice, r = 0.698; P < 0.001). Non-normalized MK values obtained from VOItu and VOIslice showed the best reproducibility and highest diagnostic performance for stratifying glioma according to the integrated approach of the recent 2016 WHO classification.
DOI: 10.1016/s1470-2045(19)30502-9
2019
Cited 28 times
Health-related quality of life and neurocognitive functioning with lomustine–temozolomide versus temozolomide in patients with newly diagnosed, MGMT-methylated glioblastoma (CeTeG/NOA-09): a randomised, multicentre, open-label, phase 3 trial
Background The CeTeG/NOA-09 trial showed significantly longer overall survival with combined lomustine–temozolomide therapy compared with standard temozolomide for patients with glioblastoma with methylated MGMT promoter. The trial also aimed to investigate the effect of lomustine–temozolomide therapy on health-related quality of life (HRQOL) and neurocognitive function, which we report here. Methods In this randomised, multicentre, open-label, phase 3 trial, newly diagnosed, chemoradiotherapy-naive patients with MGMT-methylated glioblastoma, aged 18–70 years, with a Karnofsky performance score of 70% or higher, were recruited and enrolled at 17 university hospitals in Germany. Patients received standard radiotherapy (60 Gy) and were randomly assigned (1:1, stratified by centre by allocating complete blocks of six to a centre, without masking) to either six 6-week courses of oral combined lomustine (100 mg/m2 on day 1) plus temozolomide (100–200 mg/m2 on days 2–6) or standard oral temozolomide (75 mg/m2 daily during radiotherapy plus six 4-week courses of temozolomide [150–200 mg/m2] on days 1–5, every 4 weeks). The primary endpoint was overall survival. HRQOL, assessed using the European Organisation for Research and Treatment of Cancer (EORTC) quality of life questionnaire core-30 and the EORTC brain cancer module (BN20); and neurocognitive function, assessed using the Mini Mental State Examination (MMSE), plus a neurocognitive test battery (NOA-07), including Trail Making Test A and B (TMT-A and B), working memory tests, and tests for lexical (Controlled Oral Word Association [COWA]) and semantic verbal fluency, were secondary endpoints analysed in the modified intention-to-treat population (mITT; all randomly assigned patients who received at least one dose of study chemotherapy). We used linear mixed-model analyses to investigate differences between treatment groups regarding HRQOL (clinically relevant ≥10 points) and MMSE scores (clinically relevant ≥3 points). The trial is registered with ClinicalTrials.gov, NCT01149109. Findings Between June 17, 2011 and April 8, 2014, 141 patients were randomly assigned and 129 patients began treatment and were included in the mITT population (63 in the temozolomide and 66 in the lomustine–temozolomide group). Median follow-up for HRQOL (the item global health) was 19·4 months (IQR 7·8–38·6), for MMSE was 15·3 months (4·1–29·6), and for COWA was 11·0 months (0–27·5). We found no significant impairment regarding any item of HRQOL in the lomustine–temozolomide group (difference between the groups for global health 0·30 [95% CI −0·23 to 0·83]; p=0·26). Differences in MMSE were in favour of the temozolomide group (difference −0·11 [95% CI −0·19 to −0·03]; p=0·0058) but were not clinically relevant (1·76/30 points over 4 years). We found no significant difference between the groups in any subtest of the neurocognitive test battery (difference for COWA 0·04 [95% CI −0·01 to 0·09]; p=0·14). Interpretation The absence of systematic and clinically relevant changes in HRQOL and neurocognitive function combined with the survival benefit of lomustine–temozolomide versus temozolomide alone suggests that a long-term net clinical benefit exists for patients with newly diagnosed glioblastoma with methylation of the MGMT promoter and supports the use of lomustine–temozolomide as a treatment option for these patients. Funding German Federal Ministry of Education and Research.
DOI: 10.1016/j.ejca.2020.04.019
2020
Cited 27 times
Conceptual framework for precision cancer medicine in Germany: Consensus statement of the Deutsche Krebshilfe working group ‘Molecular Diagnostics and Therapy’
Precision cancer medicine (PCM) holds great promises to offer more effective therapies to patients based on molecular profiling of their individual tumours. Although the PCM approach seems intuitive, multiple conceptional and structural challenges interfere with the broad implementation of PCM into clinical practice. Accordingly, concerted national and international efforts are needed to guide the further development and broad adoption of PCM in Germany. With support of the 'German Cancer Aid' (Deutsche Krebshilfe [DKH]) a task force 'Molecular Diagnostics and Therapy' was implemented. In two workshops supported by the DKH, delegates from the fourteen comprehensive cancer centresidentified key topics essential to implement quality-guided, harmonized and adaptable PCM. Based on an online questionnaire and using a modified Delphi approach, nine statements were drafted and evaluated within the group. These statements could serve as a basis to define a collaborative strategy for PCM in the future with the aim to sustain and further improve its quality.
DOI: 10.2967/jnumed.121.262051
2021
Cited 20 times
Impact of <sup>18</sup>F-FET PET/MRI on Clinical Management of Brain Tumor Patients
Multiparametric PET/MRI with the amino-acid analog O-(2-18F-fluoroethyl)-l-tyrosine (18F-FET) enables the simultaneous assessment of molecular, morphologic, and functional brain tumor characteristics. Although it is considered the most accurate noninvasive approach in brain tumors, its relevance for patient management is still under debate. Here, we report the diagnostic performance of 18F-FET PET/MRI and its impact on clinical management in a retrospective patient cohort. Methods: We retrospectively analyzed brain tumor patients who underwent 18F-FET PET/MRI between 2017 and 2018. 18F-FET PET/MRI examinations were indicated clinically because of equivocal standard imaging results or the clinical course. Histologic confirmation or clinical and standard imaging follow-up served as the reference standard. We evaluated 18F-FET PET/MRI accuracy in identifying malignancy in untreated suspected lesions (category, new diagnosis) and true progression during adjuvant treatment (category, detection of progression) in a clinical setting. Using multiple regression, we also estimated the contribution of single modalities to produce an optimal PET/MRI outcome. We assessed the recommended and applied therapies before and after 18F-FET PET/MRI and noted whether the treatment changed on the basis of the 18F-FET PET/MRI outcome. Results: We included 189 patients in the study. 18F-FET PET/MRI allowed the identification of malignancy at new diagnosis with an accuracy of 85% and identified true progression with an accuracy of 93%. Contrast enhancement, 18F-FET PET uptake, and tracer kinetics were the major contributors to an optimal PET/MRI outcome. In the previously equivocal patients, 18F-FET PET/MRI changed the clinical management in 33% of the untreated lesions and 53% of the cases of tumor progression. Conclusion: Our results suggest that 18F-FET PET/MRI helps clarify equivocal conditions and profoundly supports the clinical management of brain tumor patients. The optimal modality setting for 18F-FET PET/MRI and the clinical value of a simultaneous examination need further exploration. At a new diagnosis, multiparametric 18F-FET PET/MRI might help prevent unnecessary invasive procedures by ruling out malignancy; however, adding static 18F-FET PET to an already existing MRI examination seems to be of equal value. At detection of progression, multiparametric 18F-FET PET/MRI may increase therapy effectiveness by distinguishing between tumor progression and therapy-related imaging alterations.
DOI: 10.1186/s42466-022-00184-x
2022
Cited 13 times
AMPLIFY-NEOVAC: a randomized, 3-arm multicenter phase I trial to assess safety, tolerability and immunogenicity of IDH1-vac combined with an immune checkpoint inhibitor targeting programmed death-ligand 1 in isocitrate dehydrogenase 1 mutant gliomas
Isocitrate dehydrogenase (IDH) mutations are disease-defining mutations in IDH-mutant astrocytomas and IDH-mutant and 1p/19q-codeleted oligodendrogliomas. In more than 80% of these tumors, point mutations in IDH type 1 (IDH1) lead to expression of the tumor-specific protein IDH1R132H. IDH1R132H harbors a major histocompatibility complex class II (MHCII)-restricted neoantigen that was safely and successfully targeted in a first-in human clinical phase 1 trial evaluating an IDH1R132H 20-mer peptide vaccine (IDH1-vac) in newly diagnosed astrocytomas concomitant to standard of care (SOC).AMPLIFY-NEOVAC is a randomized, 3-arm, window-of-opportunity, multicenter national phase 1 trial to assess safety, tolerability and immunogenicity of IDH1-vac combined with avelumab (AVE), an immune checkpoint inhibitor (ICI) targeting programmed death-ligand 1 (PD-L1). The target population includes patients with resectable IDH1R132H-mutant recurrent astrocytoma or oligodendroglioma after SOC. Neoadjuvant and adjuvant immunotherapy will be administered to 48 evaluable patients. In arm 1, 12 patients will receive IDH1-vac; in arm 2, 12 patients will receive the combination of IDH1-vac and AVE, and in arm 3, 24 patients will receive AVE only. Until disease progression according to immunotherapy response assessment for neuro-oncology (iRANO) criteria, treatment will be administered over a period of maximum 43 weeks (primary treatment phase) followed by facultative maintenance treatment.IDH1R132H 20-mer peptide is a shared clonal driver mutation-derived neoepitope in diffuse gliomas. IDH1-vac safely targets IDH1R132H in newly diagnosed astrocytomas. AMPLIFY-NEOVAC aims at (1) demonstrating safety of enhanced peripheral IDH1-vac-induced T cell responses by combined therapy with AVE compared to IDH1-vac only and (2) investigating intra-glioma abundance and phenotypes of IDH1-vac induced T cells in exploratory post-treatment tissue analyses. In an exploratory analysis, both will be correlated with clinical outcome.NCT03893903.
DOI: 10.1093/neuonc/noq176
2010
Cited 45 times
APO010, a synthetic hexameric CD95 ligand, induces human glioma cell death in vitro and in vivo
Death receptor targeting has emerged as one of the promising novel approaches of cancer therapy. The activation of one such prototypic death receptor, CD95 (Fas/APO-1), has remained controversial because CD95 agonistic molecules have exhibited either too strong toxicity or too little activity. The natural CD95 ligand (CD95L) is a cytokine, which needs to trimerize to mediate a cell death signal. Mega-Fas-Ligand, now referred to as APO010, is a synthetic hexameric CD95 agonist that exhibits strong antitumor activity in various tumor models. Here, we studied the effects of APO010 in human glioma models in vitro and in vivo. Compared with a cross-linked soluble CD95L or a CD95-agonistic antibody, APO010 exhibited superior activity in glioma cell lines expressing CD95 and triggered caspase-dependent cell death. APO010 reduced glioma cell viability in synergy when combined with temozolomide. The locoregional administration of APO010 induced glioma cell death in vivo and prolonged the survival of tumor-bearing mice. A further exploration of APO010 as a novel antiglioma agent is warranted.
DOI: 10.1016/j.jneuroim.2012.05.010
2012
Cited 39 times
HLA-E contributes to an immune-inhibitory phenotype of glioblastoma stem-like cells
Cancer stem cells are an attractive target for immunotherapeutic approaches to glioblastoma. However, an immune inhibitory phenotype of cells currently classified as “glioma-initiating cells” (GIC) might counteract recognition by immune effector cells. Here, we investigate the contribution of the non-classical MHC molecule HLA-E to the immunosuppressive phenotype of GIC. HLA-E is expressed in GIC lines and its expression is reduced upon differentiation of GIC in serum-containing culture conditions. Constitutive HLA-E inhibits natural killer (NK) cell-mediated lysis of GIC since small-interfering RNA-mediated HLA-E gene silencing enhances the immunogenicity of GIC. Increased GIC lysis was observed both in the CD133+ and in the CD133− compartment. Furthermore, the use of interferon-γ as a possible agent to boost an immune response against glioblastoma cells might be limited by the concurrent upregulation of HLA-E.
DOI: 10.2174/138161211797249189
2011
Cited 35 times
The Role of Integrins in Glioma Biology and Anti-Glioma Therapies
The tumor environment is critical for tumor maintenance and progression. Integrins are a large family of cell surface receptors mediating the interaction of tumor cells with their microenvironment and play important roles in glioma biology, including migration, invasion, angiogenesis and tumor stem cell anchorage. Here, we review preclinical and clinical data on integrin inhibition in malignant gliomas. Various pharmacological approaches to the modulation of integrin signaling have been explored including antibodies and peptide-based agents. Cilengitide, a cyclic RGD-mimetic peptide of αvβ3 and αvβ5 integrins is in advanced clinical development in glioblastoma. Cilengitide had only limited activity as a single agent in glioblastoma, but, when added to standard radiochemotherapy, appeared to prolong progression-free and overall survival in patients with newly diagnosed glioblastomas and methylation of the promoter of the O⁶ methylguanine methyltransferase (MGMT) gene. MGMT gene promoter methylation in turn predicts benefit from alkylating chemotherapy. A phase III randomized clinical trial in conjunction with standard radiochemotherapy in newly diagnosed glioblastoma patients with MGMT gene promoter methylation has recently completed accrual (EORTC 26071-22072). A companion trial explores a dose-escalated regimen of cilengitide added to radiotherapy plus temozolomide in patients without MGMT gene promoter methylation. Promising results in these trials would probably result in a broader interest in integrins as targets for glioma therapy and hopefully the development of a broader panel of anti-integrin agents.
DOI: 10.1158/0008-5472.can-12-2532
2013
Cited 32 times
Multimodal Elucidation of Choline Metabolism in a Murine Glioma Model Using Magnetic Resonance Spectroscopy and 11C-Choline Positron Emission Tomography
The metabolites, transporters, and enzymes involved in choline metabolism are regarded as biomarkers for disease progression in a variety of cancers, but their in vivo detection is not ideal. Both magnetic resonance spectroscopy [MRS using chemical shift imaging (CSI) total choline (tCho)] and C-choline positron emission tomography (PET) can probe this pathway, but they have not been compared side by side. In this study, we used the spontaneous murine astrocytoma model SMA560 injected intracranially into syngeneic VM/Dk mice, analyzing animals at various postimplantation time points using dynamic microPET imaging and CSI MRS. We observed an increase in tumor volume and C-choline uptake between days 5 and 18. Similarly, tCho levels decreased at days 5 to 18. We found a negative correlation between the tCho and PET results in the tumor and a positive correlation between the tCho tumor-to-brain ratio and choline uptake in the tumor. PCR results confirmed expected increases in expression levels for most of the transporters and enzymes. Using MRS quantification, a good agreement was found between CSI and C-choline PET data, whereas a negative correlation occurred when CSI was not referenced. Thus, C-choline PET and MRS methods seemed to be complementary in strengths. While advancing tumor proliferation caused an increasing C-choline uptake, gliosis and inflammation potentially accounted for a high peritumoral tCho signal in CSI, as supported by histology and secondary ion mass spectrometry imaging. Our findings provide definitive evidence of the use of MRS, CSI, and PET for imaging tumors in vivo.
DOI: 10.1634/theoncologist.2016-0347
2017
Cited 30 times
Prolonged Temozolomide Maintenance Therapy in Newly Diagnosed Glioblastoma
Abstract Background The impact of prolonging temozolomide (TMZ) maintenance beyond six cycles in newly diagnosed glioblastoma (GBM) remains a topic of discussion. We investigated the effects of prolonged TMZ maintenance on progression-free survival (PFS) and overall survival (OS). Patients and Methods In this retrospective single-center cohort study, we included patients with GBM who were treated with radiation therapy with concomitant and adjuvant TMZ. For analysis, patients were considered who either completed six TMZ maintenance cycles (group B), continued with TMZ therapy beyond six cycles (group C), or stopped TMZ maintenance therapy within the first six cycles (group A). Patients with progression during the first six TMZ maintenance cycles were excluded. Results Clinical data from 107 patients were included for Kaplan-Meier analyses and 102 for Cox regressions. Median PFS times were 8.1 months (95% confidence interval [CI] 6.1–12.4) in group A, 13.7 months (95% CI 10.6–17.5) in group B, and 20.9 months (95% CI 15.2–43.5) in group C. At first progression, response rates of TMZ/lomustine rechallenge were 47% in group B and 13% in group C. Median OS times were 12.7 months (95% CI 10.3–16.8) in group A, 25.2 months (95% CI 17.7–55.5) in group B, and 28.6 months (95% CI 24.4–open) in group C. Nevertheless, multivariate Cox regression for patients in group C compared with group B that accounted for imbalances of other risk factors showed no different relative risk (RR) for OS (RR 0.77, p = .46). Conclusion Our data do not support a general extension of TMZ maintenance therapy beyond six cycles.
DOI: 10.1093/neuonc/now112
2016
Cited 27 times
Limited role for transforming growth factor–β pathway activation-mediated escape from VEGF inhibition in murine glioma models
The vascular endothelial growth factor (VEGF) and transforming growth factor (TGF)-β pathways regulate key biological features of glioblastoma. Here we explore whether the TGF-β pathway, which promotes angiogenesis, invasiveness, and immunosuppression, acts as an escape pathway from VEGF inhibition.The role of the TGF-β pathway in escape from VEGF inhibition was assessed in vitro and in vivo and by gene expression profiling in syngeneic mouse glioma models.We found that TGF-β is an upstream regulator of VEGF, whereas VEGF pathway activity does not alter the TGF-β pathway in vitro. In vivo, single-agent activity was observed for the VEGF antibody B20-4.1.1 in 3 and for the TGF-β receptor 1 antagonist LY2157299 in 2 of 4 models. Reduction of tumor volume and blood vessel density, but not induction of hypoxia, correlated with benefit from B20-4.1.1. Reduction of phosphorylated (p)SMAD2 by LY2157299 was seen in all models but did not predict survival. Resistance to B20 was associated with anti-angiogenesis escape pathway gene expression, whereas resistance to LY2157299 was associated with different immune response gene signatures in SMA-497 and GL-261 on transcriptomic profiling. The combination of B20 with LY2157299 was ineffective in SMA-497 but provided prolongation of survival in GL-261, associated with early suppression of pSMAD2 in tumor and host immune cells, prolonged suppression of angiogenesis, and delayed accumulation of tumor infiltrating microglia/macrophages.Our study highlights the biological heterogeneity of murine glioma models and illustrates that cotargeting of the VEGF and TGF-β pathways might lead to improved tumor control only in subsets of glioblastoma.
DOI: 10.1007/s11060-019-03337-2
2019
Cited 27 times
A Phase Ib/II, open-label, multicenter study of INC280 (capmatinib) alone and in combination with buparlisib (BKM120) in adult patients with recurrent glioblastoma
To estimate the maximum tolerated dose (MTD) and/or identify the recommended Phase II dose (RP2D) for combined INC280 and buparlisib in patients with recurrent glioblastoma with homozygous phosphatase and tensin homolog (PTEN) deletion, mutation or protein loss.This multicenter, open-label, Phase Ib/II study included adult patients with glioblastoma with mesenchymal-epithelial transcription factor (c-Met) amplification. In Phase Ib, patients received INC280 as capsules or tablets in combination with buparlisib. In Phase II, patients received INC280 only. Response was assessed centrally using Response Assessment in Neuro-Oncology response criteria for high-grade gliomas. All adverse events (AEs) were recorded and graded.33 patients entered Phase Ib, 32 with altered PTEN. RP2D was not declared due to potential drug-drug interactions, which may have resulted in lack of efficacy; thus, Phase II, including 10 patients, was continued with INC280 monotherapy only. Best response was stable disease in 30% of patients. In the selected patient population, enrollment was halted due to limited activity with INC280 monotherapy. In Phase Ib, the most common treatment-related AEs were fatigue (36.4%), nausea (30.3%) and increased alanine aminotransferase (30.3%). MTD was identified at INC280 Tab 300 mg twice daily + buparlisib 80 mg once daily. In Phase II, the most common AEs were headache (40.0%), constipation (30.0%), fatigue (30.0%) and increased lipase (30.0%).The combination of INC280/buparlisib resulted in no clear activity in patients with recurrent PTEN-deficient glioblastoma. More stringent molecular selection strategies might produce better outcomes.NCT01870726.
DOI: 10.1186/s13014-019-1438-2
2019
Cited 25 times
Retrospective analysis of fractionated intensity-modulated radiotherapy (IMRT) in the interdisciplinary management of primary optic nerve sheath meningiomas
Abstract Background As optic nerve sheath meningiomas (ONSM) are rare, there are no prospective studies. Our retrospective analysis focusses on a cohort of patients with uniform disease characteristics all treated with the same radiotherapy regimen. We describe treatment decision making, radiotherapy planning and detailed neuro-ophthalmological outcome of the patients. Methods 26 patients with unilateral ONSM extending only to the orbit and the optic canal were evaluated for neuro-ophthalmological outcome. Radiation treatment was planned in a simultaneous integrated boost approach to gross tumor volume (GTV) + 2 mm / 5 mm to 54 Gy / 51 Gy in 1.8 Gy / 1.7 Gy fractions. Follow-up was done by specialized neuro-ophthalmologists. Visual acuity and visual field defects were evaluated after therapy as well as during follow-up. Results Interdisciplinary treatment decision for patients with ONSM follows a rather complex decision tree. Radiation treatment planning (equivalent uniform dose (EUD), maximum dose to the optic nerve) improved with experience over time. With this patient selection visual acuity as well as visual field improved significantly at first follow-up after treatment. For visual acuity this also applied to patients with severe defects before treatment. Long term evaluation showed 16 patients with improved visual function, 6 were stable, in 4 patients visual function declined. Interdisciplinary case discussion rated the visual decline as radiation-associated in two patients. Conclusions With stringent patient selection radiotherapy for unilateral primary ONSM to 51 Gy / 54 Gy is safe and leads to significantly improved visual function. Interdisciplinary treatment decision and experience of the radiation oncology team play a major role.
DOI: 10.1007/s10143-020-01428-7
2020
Cited 22 times
The role of Simpson grading in meningiomas after integration of the updated WHO classification and adjuvant radiotherapy
Abstract Since the introduction of the Simpson grading for the extent of resection in meningiomas in 1957, its usefulness in modern neurosurgery has been challenged. Especially, the updated WHO classification regarding brain invasion and the efficacy of radiation therapy has not been taken into account when evaluating the prognostic role of the Simpson grading in this era. We analyzed the clinical and histopathological data of 1571 meningiomas that were surgically resected in the authors’ institution between July 2003 and March 2017. Operative reports were reviewed regarding the extent of resection according to Simpson grading. Meningioma subtype according to the updated WHO classification of 2016 and clinical characteristics and time to tumor progression were analyzed. The mean follow-up was 38.4 months (range 1.2 to 195.6). A higher rate of tumor recurrence was observed for male gender, younger age, recurrent tumors, non-spinal tumor localization, higher WHO, and Simpson grades in the univariate analysis. In the multivariate analysis older age, recurrent tumors and higher WHO grades remained negative prognostic factors. Among the different Simpson grades, the relative risk for recurrence was highest for grade IV compared to all other grades (each p &lt; 0.0001), while there was no difference between Simpson grades I and II. Adjuvant radiotherapy showed lower rates of tumor recurrence. Subtotal microsurgical resection remains an independent prognostic factor with a higher rate of tumor recurrence. The prognostic benefit of radical treatment of the dural attachment is questionable and needs to be considered when weighing the intraoperative risks of radicality.
DOI: 10.1158/1078-0432.ccr-20-2096
2021
Cited 17 times
MRI and 18FET-PET Predict Survival Benefit from Bevacizumab Plus Radiotherapy in Patients with Isocitrate Dehydrogenase Wild-type Glioblastoma: Results from the Randomized ARTE Trial
To explore a prognostic or predictive role of MRI and O-(2-18F-fluoroethyl)-L-tyrosine (18FET) PET parameters for outcome in the randomized multicenter trial ARTE that compared bevacizumab plus radiotherapy with radiotherpay alone in elderly patients with glioblastoma.Patients with isocitrate dehydrogenase wild-type glioblastoma ages 65 years or older were included in this post hoc analysis. Tumor volumetric and apparent diffusion coefficient (ADC) analyses of serial MRI scans from 67 patients and serial 18FET-PET tumor-to-brain intensity ratios (TBRs) from 31 patients were analyzed blinded for treatment arm and outcome. Multivariate Cox regression analysis was done to account for established prognostic factors and treatment arm.Overall survival benefit from bevacizumab plus radiotherapy compared with radiotherapy alone was observed for larger pretreatment MRI contrast-enhancing tumor [HR per cm3 0.94; 95% confidence interval (CI), 0.89-0.99] and for higher ADC (HR 0.18; CI, 0.05-0.66). Higher 18FET-TBR on pretreatment PET scans was associated with inferior overall survival in both arms. Response assessed by standard MRI-based Response Assessment in Neuro-Oncology criteria was associated with overall survival in the bevacizumab plus radiotherapy arm by trend only (P = 0.09). High 18FET-TBR of noncontrast-enhancing tumor portions during bevacizumab therapy was associated with inferior overall survival on multivariate analysis (HR 5.97; CI, 1.16-30.8).Large pretreatment contrast-enhancing tumor mass and higher ADCs identify patients who may experience a survival benefit from bevacizumab plus radiotherapy. Persistent 18FET-PET signal of no longer contrast-enhancing tumor after concomitant bevacizumab plus radiotherapy suggests pseudoresponse and predicts poor outcome.
DOI: 10.1186/s13063-021-05977-0
2022
Cited 11 times
Phase I/II trial of meclofenamate in progressive MGMT-methylated glioblastoma under temozolomide second-line therapy—the MecMeth/NOA-24 trial
Glioblastoma is the most frequent and malignant primary brain tumor. Even in the subgroup with O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation and favorable response to first-line therapy, survival after relapse is short (12 months). Standard therapy for recurrent MGMT-methylated glioblastoma is not standardized and may consist of re-resection, re-irradiation, and chemotherapy with temozolomide (TMZ), lomustine (CCNU), or a combination thereof. Preclinical results show that meclofenamate (MFA), originally developed as a nonsteroidal anti-inflammatory drug (NSAID) and registered in the USA, sensitizes glioblastoma cells to temozolomide-induced toxicity via inhibition of gap junction-mediated intercellular cytosolic traffic and demolishment of tumor microtube (TM)-based network morphology.In this study, combined MFA/TMZ therapy will be administered (orally) in patients with first relapse of MGMT-methylated glioblastoma. A phase I component (6-12 patients, 2 dose levels of MFA + standard dose TMZ) evaluates safety and feasibility and determines the dose for the randomized phase II component (2 × 30 patients) with progression-free survival as the primary endpoint.This study is set up to assess toxicity and first indications of efficacy of MFA repurposed in the setting of a very difficult-to-treat recurrent tumor. The trial is a logical next step after the identification of the role of resistance-providing TMs in glioblastoma, and results will be crucial for further trials targeting TMs. In case of favorable results, MFA may constitute the first clinically feasible TM-targeted drug and therefore might bridge the idea of a TM-targeted therapeutic approach from basic insights into clinical reality.EudraCT 2021-000708-39 . Registered on 08 February 2021.
DOI: 10.1007/s00066-022-01994-3
2022
Cited 11 times
DEGRO practical guideline for central nervous system radiation necrosis part 1: classification and a multistep approach for diagnosis
The Working Group for Neuro-Oncology of the German Society for Radiation Oncology in cooperation with members of the Neuro-Oncology Working Group of the German Cancer Society aimed to define a practical guideline for the diagnosis and treatment of radiation-induced necrosis (RN) of the central nervous system (CNS).Panel members of the DEGRO working group invited experts, participated in a series of conferences, supplemented their clinical experience, performed a literature review, and formulated recommendations for medical treatment of RN including bevacizumab in clinical routine.Diagnosis and treatment of RN requires multidisciplinary structures of care and defined processes. Diagnosis has to be made on an interdisciplinary level with the joint knowledge of a neuroradiologist, radiation oncologist, neurosurgeon, neuropathologist, and neuro-oncologist. A multistep approach as an opportunity to review as many characteristics as possible to improve diagnostic confidence is recommended. Additional information about radiotherapy (RT) techniques is crucial for the diagnosis of RN. Misdiagnosis of untreated and progressive RN can lead to severe neurological deficits. In this practice guideline, we propose a detailed nomenclature of treatment-related changes and a multistep approach for their diagnosis.
DOI: 10.1172/jci.insight.153526
2022
Cited 10 times
Tissue metabolites in diffuse glioma and their modulations by IDH1 mutation, histology, and treatment
The discovery of the oncometabolite 2-hydroxyglutarate in isocitrate dehydrogenase 1-mutated (IDH1-mutated) tumor entities affirmed the role of metabolism in cancer. However, large databases with tissue metabolites that are modulated by IDH1 mutation remain an area of development. Here, we present an unprecedented and valuable resource for tissue metabolites in diffuse glioma and their modulations by IDH1 mutation, histology, and tumor treatments in 101 tissue samples from 73 diffuse glioma patients (24 astrocytoma, 17 oligodendroglioma, 32 glioblastoma), investigated by NMR-based metabolomics and supported by RNA-Seq. We discovered comparison-specific metabolites and pathways modulated by IDH1 (IDH1 mutation status cohort) and tumor entity. The Longitudinal investigation cohort provides metabolic profiles of untreated and corresponding treated glioma samples at first progression. Most interestingly, univariate and multivariate cox regressions and Kaplan-Meier analyses revealed that tissue metabolites correlate with progression-free and overall survival. Thus, this study introduces potentially novel candidate prognostic and surrogate metabolite biomarkers for future prospective clinical studies, aiming at further refining patient stratification in diffuse glioma. Furthermore, our data will facilitate the generation of so-far-unanticipated hypotheses for experimental studies to advance our molecular understanding of glioma biology.
DOI: 10.1136/jitc-2021-003368
2022
Cited 10 times
Systemic high-dose dexamethasone treatment may modulate the efficacy of intratumoral viral oncolytic immunotherapy in glioblastoma models
Intratumoral viral oncolytic immunotherapy is a promising new approach for the treatment of a variety of solid cancers. CAN-2409 is a replication-deficient adenovirus that delivers herpes simplex virus thymidine kinase to cancer cells, resulting in local conversion of ganciclovir or valacyclovir into a toxic metabolite. This leads to highly immunogenic cell death, followed by a local immune response against a variety of cancer neoantigens and, next, a systemic immune response against the injected tumor and uninjected distant metastases. CAN-2409 treatment has shown promising results in clinical studies in glioblastoma (GBM). Patients with GBM are usually given the corticosteroid dexamethasone to manage edema. Previous work has suggested that concurrent dexamethasone therapy may have a negative effect in patients treated with immune checkpoint inhibitors in patients with GBM. However, the effects of dexamethasone on the efficacy of CAN-2409 treatment have not been explored.In vitro experiments included cell viability and neurosphere T-cell killing assays. Effects of dexamethasone on CAN-2409 in vivo were examined using a syngeneic murine GBM model; survival was assessed according to Kaplan-Meier; analyses of tumor-infiltrating lymphocytes were performed with mass cytometry (CyTOF - cytometry by time-of-flight). Data were analyzed using a general linear model, with one-way analysis of variance followed by Dunnett's multiple comparison test, Kruskal-Wallis test, Dunn's multiple comparison test or statistical significance analysis of microarrays.In a mouse model of GBM, we found that high doses of dexamethasone combined with CAN-2409 led to significantly reduced median survival (29.0 days) compared with CAN-2409 treatment alone (39.5 days). CyTOF analyses of tumor-infiltrating immune cells demonstrated potent immune stimulation induced by CAN-2409 treatment. These effects were diminished when high-dose dexamethasone was used. Functional immune cell characterization suggested increased immune cell exhaustion and tumor promoting profiles after dexamethasone treatment.Our data suggest that concurrent high-dose dexamethasone treatment may impair the efficacy of oncolytic viral immunotherapy of GBM, supporting the notion that dexamethasone use should be balanced between symptom control and impact on the therapeutic outcome.
DOI: 10.1038/sj.cdd.4401786
2005
Cited 43 times
BCL-xL: time-dependent dissociation between modulation of apoptosis and invasiveness in human malignant glioma cells
Conditionally BCL-xL-overexpressing LNT-229 Tet-On glioma cell clones were generated to investigate whether the ‘antiapoptosis phenotype’ and the ‘motility phenotype’ mediated by BCL-2 family proteins in glioma cells could be separated. BCL-xL induction led to an immediate and concentration-dependent protection of LNT-229 cells from apoptosis. BCL-xL induction for up to 3 days did not result in altered invasiveness. In contrast, long-term BCL-xL induction for 21 days resulted in increased transforming growth factor-β2 expression, and in metalloproteinase-2 and -14 dependent, but integrin independent, increased invasiveness. Withdrawal of doxycycline (Dox) abolished the protection from apoptosis whereas the ‘invasion phenotype’ remained stable. Dox stimulation of BCL-xL-inducible LNT-229 cells conferred infiltrative growth to BCL-xL-positive glioma cells in vivo and reduced the survival of tumor-bearing mice. These data allow to dissect a direct antiapoptotic action of BCL-xL from an indirect effect, presumably mediated by altered gene expression, which modifies tumor cell invasiveness in vitro and in vivo.
DOI: 10.1016/j.ejrad.2017.08.008
2017
Cited 25 times
Histogram analysis of diffusion kurtosis imaging estimates for in vivo assessment of 2016 WHO glioma grades: A cross-sectional observational study
To assess the diagnostic performance of histogram analysis of diffusion kurtosis imaging (DKI) maps for in vivo assessment of the 2016 World Health Organization Classification of Tumors of the Central Nervous System (2016 CNS WHO) integrated glioma grades.Seventy-seven patients with histopathologically-confirmed glioma who provided written informed consent were retrospectively assessed between 01/2014 and 03/2017 from a prospective trial approved by the local institutional review board. Ten histogram parameters of mean kurtosis (MK) and mean diffusivity (MD) metrics from DKI were independently assessed by two blinded physicians from a volume of interest around the entire solid tumor. One-way ANOVA was used to compare MK and MD histogram parameter values between 2016 CNS WHO-based tumor grades. Receiver operating characteristic analysis was performed on MK and MD histogram parameters for significant results.The 25th, 50th, 75th, and 90th percentiles of MK and average MK showed significant differences between IDH1/2wild-type gliomas, IDH1/2mutated gliomas, and oligodendrogliomas with chromosome 1p/19q loss of heterozygosity and IDH1/2mutation (p<0.001). The 50th, 75th, and 90th percentiles showed a slightly higher diagnostic performance (area under the curve (AUC) range; 0.868-0.991) than average MK (AUC range; 0.855-0.988) in classifying glioma according to the integrated approach of 2016 CNS WHO.Histogram analysis of DKI can stratify gliomas according to the integrated approach of 2016 CNS WHO. The 50th (median), 75th, and the 90th percentiles showed the highest diagnostic performance. However, the average MK is also robust and feasible in routine clinical practice.
DOI: 10.2217/imt-2018-0149
2019
Cited 23 times
Immunotherapy plus surgery/radiosurgery is associated with favorable survival in patients with melanoma brain metastasis
Melanoma brain metastases (MBM) are associated with a dismal prognosis. Few clinical trials evaluated the impact of immunotherapy (IT) and targeted therapy (TT) alone or in combination with surgery and radiotherapy in this population.Retrospective analysis of data from 163 patients diagnosed with MBM between January 2014 and December 2016. Prognostic factors of overall survival were analyzed using Kaplan-Meier survival curves, classification and regression tree and multivariate Cox regression analysis.The median follow-up was 25 months; median overall survival (mOS) for all patients was 7 months. For patients receiving IT, the mOS was 13 months and 7 months for patients receiving TT or chemotherapy (CT). The mOS for patients treated with surgery/radiosurgery in combination with IT, TT and CT was 25, 14 and 11 months, respectively.New systemic therapies, especially IT, improve mOS in patients with MBM, particularly when combined with surgery/radiosurgery upfront.
DOI: 10.1200/jco.2018.36.15_suppl.2001
2018
Cited 22 times
A mutation-specific peptide vaccine targeting IDH1R132H in patients with newly diagnosed malignant astrocytomas: A first-in-man multicenter phase I clinical trial of the German Neurooncology Working Group (NOA-16).
2001 Background: Hot-spot point mutations in the gene for isocitrate dehydrogenase type 1 (IDH1R132H) are a frequent founder event in gliomas and other tumors. Preclinical studies have defined IDH1R132H as a clonal neoantigen presented on MHC class II to induce tumor-specific therapeutic T helper cell responses. Methods: NOA-16 (NCT02454634) is a first-in-man, multicenter, phase I trial, which enrolled 33 patients with newly diagnosed WHO °III and °IV astrocytomas with IDH1R132H mutations. After completion of radiochemotherapy a total of eight vaccinations with an IDH1R132H peptide in incomplete Freund’s adjuvant produced at a central GMP site was to be administered subcutaneously with topical imiquimod over a period of 32 weeks together with maintenance temozolomide. The primary end points were safety and immunogenicity. Results: The safety dataset comprised 249 vaccines administered to 32 patients. One patient withdrew after screening. 29 patients received all eight vaccines. Vaccine-related adverse events (AE) were restricted to grade 1 reactions, according to common toxicity criteria for AE(CTCAE v4.0). Two serious AE were observed in two patients; one probably related to the peptide vaccine. 28/30 patients (93.3%) evaluable for immunogenicity displayed IDH1R132H-specific T cellular (detected by ELISPOT assays in 24/30 (80%)) or humoral (detected by ELISA in 26/30 patients (87%)) immune responses not detectable before vaccination. Until end of study (EOS, week 32), 4/32 (12.5 %) patients had progressive disease (PD) according to RANO criteria, all other patients (N = 28, 87.5%) had stable disease (SD). 12/32 (37.5%) patients displayed pseudoprogressions. Single-cell T cell receptor (TCR) sequencing allowed for the identification of IDH1R132H-specific TCRs. Conclusions: NOA-16 met its primary endpoints by demonstrating safety and immunogenicity of a mutation-specific IDH1R132H peptide vaccine. Pseudoprogressions observed after the initiation of the vaccine may indicate intratumoral immune reactions warranting further development, including TCR cell therapy. Clinical trial information: NCT02454634.
DOI: 10.1200/po.19.00359
2020
Cited 18 times
Next-Generation Sequencing of Advanced GI Tumors Reveals Individual Treatment Options
Precision oncology connects highly complex diagnostic procedures with patient histories to identify individualized treatment options in interdisciplinary molecular tumor boards (MTBs). Detailed data on MTB-guided treatments and outcome with a focus on advanced GI cancers have not been reported yet.Next-generation sequencing of tumor and normal tissue pairs was performed between April 2016 and February 2018. After identification of relevant molecular alterations, available clinical studies or in-label, off-label, or matched experimental treatment options were recommended. Follow-up data and a response assessment that was based on radiologic imaging were recorded.Ninety-six patients were presented to the MTB of Tuebingen University Hospital. Sixteen (17%) showed "pathogenic" or "likely pathogenic" germline variants. Recommendations on the basis of molecular alterations or tumor mutational burden were given for 41 patients (43%). Twenty-five received the suggested drug, and 20 were evaluable for best response assessment. Three patients (15%) reached a partial response (PR), and 6 (30%), stable disease (SD), whereas 11 (55%) had tumor progression (progressive disease). Median progression-free survival (PFS) for all treated and evaluable patients was 2.8 months (range, 1.0-9.0 months), and median overall survival (OS) of all treated patients was 5.2 months (range, 0.1 months to not reached). Patients with SD for ≥ 3 months or PR compared with progressive disease showed both a statistically significant longer median PFS (7.8 months [95% CI, 4.2 to 11.4 months] v 2.2 months [95% CI, 1.5 to 2.8 months], P < .0001) and median OS (18.0 months [95% CI, 10.4 to 25.6 months] v 3.8 months [95% CI, 2.3 to 5.4 months], P < .0001).Next-generation sequencing diagnostics of advanced GI cancers identified a substantial number of pathogenic or likely pathogenic germline variants and unique individual treatment options. Patients with PR or SD in the course of MTB-recommended treatments seemed to benefit with respect to PFS and OS.
DOI: 10.1007/s00432-021-03906-x
2022
Cited 9 times
Frequent FGFR1 hotspot alterations in driver-unknown low-grade glioma and mixed neuronal-glial tumors
Low-grade gliomas (LGG) and mixed neuronal-glial tumors (MNGT) show frequent MAPK pathway alterations. Oncogenic fibroblast growth factor receptor 1 (FGFR1) tyrosinase kinase domain has been reported in brain tumors of various histologies. We sought to determine the frequency of FGFR1 hotspot mutations N546 and K656 in driver-unknown LGG/MNGT and examined FGFR1 immunohistochemistry as a potential tool to detect those alterations.We analyzed 476 LGG/MNGT tumors for KIAA-1549-BRAF fusion, IDH1/2, TERT promotor, NF1, H3F3A and the remaining cases for FGFR1 mutation frequency and correlated FGFR1 immunohistochemistry in 106 cases.368 of 476 LGG/MNGT tumors contained non-FGFR1 alterations. We identified 9 FGFR1 p.N546K and 4 FGFR1 p.K656E mutations among the 108 remaining driver-unknown samples. Five tumors were classified as dysembryoplastic neuroepithelial tumor (DNT), 4 as pilocytic astrocytoma (PA) and 3 as rosette-forming glioneuronal tumor (RGNT). FGFR1 mutations were associated with oligodendroglia-like cells, but not with age or tumor location. FGFR1 immunohistochemical expression was observed in 92 cases. FGFR1 immunoreactivity score was higher in PA and DNT compared to diffuse astrocytoma, but no correlation between FGFR1 mutation in tumors and FGFR1 expression level was observed.FGFR1 hotspot mutations are the fifth most prevailing alteration in LGG/MNGT. Performing FGFR1 sequencing analysis in driver-unknown low-grade brain tumors could yield up to 12% FGFR1 N546/K656 mutant cases.
DOI: 10.1007/s00066-022-01973-8
2022
Cited 9 times
DEGRO practical guideline for central nervous system radiation necrosis part 2: treatment
The Working Group for Neurooncology of the German Society for Radiation Oncology (DEGRO; AG NRO) in cooperation with members of the Neurooncological Working Group of the German Cancer Society (DKG-NOA) aimed to define a practical guideline for the diagnosis and treatment of radiation-induced necrosis (RN) of the central nervous system (CNS).Panel members of the DEGRO working group invited experts, participated in a series of conferences, supplemented their clinical experience, performed a literature review, and formulated recommendations for medical treatment of RN, including bevacizumab, in clinical routine.Diagnosis and treatment of RN requires multidisciplinary structures of care and defined processes. Diagnosis has to be made on an interdisciplinary level with the joint knowledge of a neuroradiologist, radiation oncologist, neurosurgeon, neuropathologist, and neurooncologist. If the diagnosis of blood-brain barrier disruptions (BBD) or RN is likely, treatment should be initiated depending on the symptoms, location, and dynamic of the lesion. Multiple treatment options are available (such as observation, surgery, steroids, and bevacizumab) and the optimal approach should be discussed in an interdisciplinary setting. In this practice guideline, we offer detailed treatment strategies for various scenarios.
DOI: 10.3171/2009.11.jns09901
2010
Cited 31 times
Soluble CD70: a novel immunotherapeutic agent for experimental glioblastoma
Object Given the overall poor outcome with current treatment strategies in malignant gliomas, immunotherapy has been considered a promising experimental approach to glioblastoma for more than 2 decades. A cell surface molecule, CD70, may induce potent antitumor immune responses via activation of the costimulatory receptor CD27 expressed on immune effector cells. There is evidence that a soluble form of CD70 (sCD70) may exhibit biological activity, too. A soluble costimulatory ligand is attractive because it may facilitate immune activation and may achieve a superior tissue distribution. Methods To test the antiglioma effect of sCD70, the authors genetically modified SMA-560 mouse glioma cells to secrete the extracellular domain of CD70. They assessed the immunogenicity of the transfected cells in cocultures with immune effector cells by the determination of immune cell proliferation and the release of interferon-γ. Syngeneic VM/Dk mice were implanted orthotopically with control or sCD70-releasing glioma cells to determine a survival benefit mediated by sCD70. Depletion studies were performed to identify the cellular mediators of prolonged survival of sCD70-releasing glioma-bearing mice. Results The authors found that ectopic expression of sCD70 enhanced the proliferation and interferon-γ release of syngeneic splenocytes in vitro. More importantly, sCD70 prolonged the survival of syngeneic VM/Dk mice bearing intracranial SMA-560 gliomas. The survival rate at 60 days increased from 5 to 45%. Antibody-mediated depletion of CD8-positive T cells abrogates the survival advantage conferred by sCD70. Conclusions These data suggest that sCD70 is a potent stimulator of antiglioma immune responses that depend critically on CD8-positive T cells. Soluble CD70 could be a powerful adjuvant for future immunotherapy trials for glioblastoma.
DOI: 10.1016/j.wneu.2017.07.034
2017
Cited 22 times
The Prognostic Impact of Ventricular Opening in Glioblastoma Surgery: A Retrospective Single Center Analysis
Ventricular opening during glioblastoma (GBM) resection is controversial. Sufficient evidence regarding its prognostic role is missing. We investigated the impact of ventricular opening on overall survival (OS), hydrocephalus development, and postoperative morbidity in patients with GBM. Patients who underwent primary GBM resection between 2006 and 2013 were assessed retrospectively. Established predictors for overall survival (age, Karnofsky Performance Status, extent of resection, O-6-methylguanine-DNA methyltransferase promoter methylation status, isocitrate dehydrogenase mutation status) and further clinical data (postoperative status, further treatment, preoperative tumor volume, proximity to the ventricle) were included in univariate and multivariate analyses. Thirteen (5.7%) of 229 patients developed a hydrocephalus. Multivariate logistic regression showed that neither ventricular opening, tumor size, proximity to the ventricle, nor extent of resection were significant risk factors for hydrocephalus. Ventricular opening did not delay postoperative therapy and was not associated with neurological morbidity. Kaplan-Meier analysis demonstrated that patients who underwent ventricular opening (n = 114) exhibited a median OS of 14.3 months (12.9–16.5), whereas patients who did not undergo ventricular opening (n = 115) exhibited a median OS of 18.6 months (16.1–20.8). However, multivariate Cox regression (n = 134) did not confirm ventricular opening as an independent negative predictor of OS (risk ratio 1.09, P = 0.77). Instead, it showed that a greater preoperative tumor volume >22.8 cm3 was a negative predictor of OS (risk ratio 1.76, P = 0.02). Because extent of resection is a strong independent predictor of OS and ventricular opening is safe, neurosurgeons should consider ventricular opening to achieve maximal tumor resection.
DOI: 10.1007/s00062-018-0676-2
2018
Cited 21 times
In Vivo Molecular Profiling of Human Glioma
DOI: 10.1093/neuonc/noy148.028
2018
Cited 19 times
ATIM-33. NOA-16: A FIRST-IN-MAN MULTICENTER PHASE I CLINICAL TRIAL OF THE GERMAN NEUROONCOLOGY WORKING GROUP EVALUATING A MUTATION-SPECIFIC PEPTIDE VACCINE TARGETING IDH1R132H IN PATIENTS WITH NEWLY DIAGNOSED MALIGNANT ASTROCYTOMAS
In preclinical studies we have defined IDH1R132H as a clonal neoantigen presented on MHC class II. A peptide vaccine encompassing IDH1R132H induces tumor-specific T helper cell responses effective in controlling syngeneic IDH1R132H-mutant tumors in humanized mouse models. NOA-16 (NCT02454634) is a first-in-man, multicenter, phase I trial testing the safety and immunogenicity of an IDH1R132H peptide in incomplete Freunds adjuvant in patients with newly diagnosed, IDH1R132H mutant WHO °III and °IV astrocytomas. Between September 2015 and October 2016, 32 patients were enrolled in seven German sites. 23 patients (71.9%) received radiochemotherapy with temozolomide, six patients (18.8%) received radiotherapy alone and three patients (9.4%) received temozolomide alone. 249 vaccines were administered, 29 (90.6%) of the patients of the safety set (N=32) and 27 (90.0%) patients of the immunogenicity set (N=30) received all eight vaccines. No regime-limiting toxicity was observed. The majority of the patients (N=29, 90.6%) experienced treatment related adverse events (trAE), 1 (3.1%) of them had treatment related SAE. None of the reported AEs were severe. 28/30 (93.3%) patients, who were evaluable for immunogenicity, displayed mutation-specific T cellular (24/30 (80%)) or humoral (26/30 patients (87%)) immune responses not detectable before vaccination. Until end of study no deaths were observed. 4/32 (12.5 %) patients had PD according to RANO criteria, all other patients (N=28, 87.5%) had SD. 12/32 (37.5%) patients displayed pseudoprogressions after the initiation of the vaccine. Single-cell T cell receptor (TCR) sequencing allowed for the identification of IDH1R132H-specific TCRs. In conclusion, NOA-16 met its primary endpoints by demonstrating safety and immunogenicity of a mutation-specific IDH1R132H peptide vaccine given with standard of care in patients with newly diagnosed IDH1R132H mutant malignant astrocytoma.
DOI: 10.1111/j.1365-2990.2008.00957.x
2009
Cited 26 times
Diagnostic value of WT1 in neuroepithelial tumours
Currently, clinical trials using WT1 (Wilms tumour gene) peptide vaccines are conducted in haematopoietic malignancies and solid cancers. Single reports showed that the Wilms tumour gene product WT1 is also expressed in astrocytic neoplasms. Our aim was to investigate WT1 expression in a large cohort of various neuroepithelial tumours of different World Health Organization (WHO) grades and in normal central nervous system (CNS) tissue specimens to test its potential value as a diagnostic marker.Specimens were assessed by RT-PCR, Western blotting and immunohistochemistry. The samples investigated in our study consisted of 334 human neuroepithelial tumours, among those 33 oligodendrogliomas, 219 astrocytomas (including 105 glioblastomas) and 47 ependymomas.Our results showed a de novo WT1 expression in neuroepithelial tumours. In diffuse astrocytomas and ependymomas, WT1 expression increased significantly with the grade of malignancy. In contrast, no significant difference was seen between WHO grade-II and -III oligodendrogliomas. Controlling for WHO grade, the comparison of oligodendrogliomas with ependymal and astrocytic tumours showed higher expression values for the latter.Our study shows that WT1 is expressed de novo in numerous neuroepithelial tumours and increases with the grade of malignancy. These results suggest an important role of WT1 in tumourigenesis and progression in human brain tumours.