58 results on '"Hervey-Jumper, S"'
Search Results
2. High interobserver agreement in the semiquantitative classification of 5-ALA fluorescence levels in newly diagnosed glioblastomas
- Author
-
Mischkulnig, M, Kiesel, B, Borkovec, M, Wadiura, L, Benner, D, Hosmann, A, Hervey-Jumper, S, Knosp, E, Rössler, K, Berger, MS, and Widhalm, G
- Subjects
ddc: 610 ,610 Medical sciences ,Medicine - Abstract
Objective: Fluorescence-guided resection of glioblastomas (GBM) using 5-aminolevulinic acid (5-ALA) improves intraoperative tumor visualization and is thus widely used nowadays. During resection, different fluorescence levels can usually be distinguished within the same tumor. Recently, we demonstrated[for full text, please go to the a.m. URL], 71. Jahrestagung der Deutschen Gesellschaft für Neurochirurgie (DGNC), 9. Joint Meeting mit der Japanischen Gesellschaft für Neurochirurgie
- Published
- 2020
- Full Text
- View/download PDF
3. Mutational Analysis and Single Cell Sequencing of Melanoma Brain Metastases.
- Author
-
Vasudevan, H., Delley, C., Aabedi, A., Nguyen, M.P., Morshed, R.A., Young, J.S., Demaree, B., Diwanji, D., Hervey-Jumper, S., Boreta, L., Fogh, S.E., Nakamura, J.L., Theodosopoulos, P.V., Phillips, J., Daras, M., Tsai, K., Sneed, P.K., Aghi, M.K., Raleigh, D., and Braunstein, S.E.
- Subjects
- *
BRAIN metastasis , *CELL analysis , *PROGRESSION-free survival , *GENOMICS , *CEREBRAL arteriovenous malformations , *PROGNOSIS , *MELANOMA - Abstract
Melanoma brain metastases are a significant source of morbidity and mortality. While radiosurgery and (CNS) penetrant therapies have improved clinical outcomes, understanding the molecular landscape and microenvironment of brain metastases is critical to devise improved treatments for patients. Here, we perform bulk and single cell genomic analysis of melanoma brain metastases to identify molecular mechanisms underlying therapeutic response. 50 consecutive patients who underwent surgical resection with histo-pathologically confirmed melanoma brain metastases and known BRAFV600E status treated at a single institution were retrospectively identified. In 25 patients (50%) with sufficient brain metastasis tissue for targeted next generation sequencing, DNA mutations were assessed with a CLIA certified sequencing assay spanning over 500 cancer genes. Single cell RNA sequencing was performed on n=5 samples with sufficient frozen tissue. Overall survival (OS) and CNS progression free survival (CNS PFS) were estimated using the Kaplan-Meier method from time of brain metastasis diagnosis. Single cell analysis was performed using the 10x platform and Seurat R package. The median patient age was 64 years old (27 – 78 years), and the median clinical follow up was 17 months. 21 patients (42%) had BRAFV600E melanoma brain metastases. BRAFV600E mutation was associated with worse CNS PFS (p=0.04, log-rank test) and OS (p=0.03, log-rank test). Multivariate analysis incorporating age, Karnofsky performance score, and presence of extracranial disease supports BRAF status as an independent prognostic factor for OS (p<0.05). In patients undergoing targeted next generation sequencing, the most common pathogenic variant was TERT promoter mutation (n=19; 76%) followed by TP53 mutation (n=9; 36%). The four TCGA molecular melanoma subgroups were equally represented: NRAS mutant (n=8; 32%), NF1 mutant (n=6; 24%), BRAF mutant (n=6; 24%) or triple wildtype (n=5; 20%) Evaluation of clinical outcomes in the context of next generation sequencing results revealed no differences by TERT status but demonstrated worse overall survival in BRAF mutant tumors (median OS: 11 months; p=0.01, log-rank test). Single cell sequencing of 31,219 nuclei across 5 samples revealed distinct subpopulations between BRAF mutant and BRAF wildtype tumors in both the tumor and the brain microenvironment, with increased immune infiltration in BRAF wildtype tumors and greater neuronal subpopulations in BRAF mutant lesions. Our data suggest BRAF may be useful as a prognostic biomarker and TERT promoter mutations as a recurrent intracranial molecular alteration in melanoma. Single cell analysis shows brain metastases exhibit distinct neuronal and immune populations. Future work will require integration of molecular status with treatment to help guide therapy and interrogation of larger, prospective, multi-institutional cohorts to validate these observations. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
4. Timing of Adjuvant Radiotherapy and Survival Analysis for Molecularly Defined Low Grade Glioma.
- Author
-
Liu, S.J., Chen, W.C., Zhang, Y., Young, J.S., Morshed, R.A., Villanueva-Meyer, J., Phillips, J., Oberheim, N.A., Aghi, M.K., Sneed, P.K., Braunstein, S.E., Berger, M.S., Molinaro, A.M., Hervey-Jumper, S., and Raleigh, D.
- Subjects
- *
SURVIVAL analysis (Biometry) , *GLIOMAS , *SURVIVAL rate , *CHI-squared test , *OVERALL survival , *BRAIN tumors - Abstract
The optimal treatment for patients with diffuse low-grade glioma (LGG) is controversial. Level I evidence support the use of radiotherapy (RT) for histologic LGG, but incorporation of molecular biomarkers in the WHO classification compels additional investigation to define the optimal treatment and timing of RT. We hypothesized molecularly defined LGG (IDH-mutant astrocytoma [astro] and IDH-mutant, 1p/19q-codeleted oligodendrogliomas [oligo]) have different survival outcomes following early vs delayed RT, knowledge that may guide RT in the context of resection and chemotherapy. We performed a retrospective analysis of adult patients who were newly diagnosed with a WHO Grade 2 IDH-mutant astro or IDH-mutant, 1p/19q-codel oligo and had initial resection at a single institution from Jan 1998 to Nov 2017. Wilcoxon rank sum and Chi-squared tests were used to compare continuous and categorical variables between cohorts, respectively. Survival analysis was performed using the Kaplan-Meier method. Patients without clinical progression or death were censored at last follow-up date. Pre-operative and post-operative tumor volumes were quantified using 3D Slicer and defined as the T2 FLAIR hyperintense lesion to calculate extent of resection (EOR). 392 patients (202 astro, 190 oligo) were analyzed with a median follow up of 9.3 yr (95% CI: 8.19–10.16). Median overall survival (OS) was 13.0 yr for astro (95% CI: 11.4 – 18.6) and not reached for oligo (95% CI: 19.9 – NA). 168 patients (42.8%) received RT during their treatment course, with the majority (84%) treated at time of first disease progression. Chemotherapy was used in 81.7% of patients who received RT (TMZ in 90.3% of cases). The median RT dose was 54 Gy (range: 40 – 60 Gy). RT was more likely for astro (51.0%) than for oligo (34.2%) (p < 0.001), and a lower median EOR was associated with RT (88% vs 95%; p = 0.017). RT was not associated with increased OS for astro (p = 0.12) or oligo (p = 0.31) for all comers. For patients < 40 yr of age who had subtotal resections of astros (< 85% EOR), RT increased median OS with a trend toward significance when compared to no RT (13.1 yr [95% CI: 7.3 – NA] vs 9.1 yr [95% CI: 5.4 – NA], p = 0.1). RT within 1 yr of diagnosis was associated with improved median PFS compared to patients who received RT after 1 yr for astro (7.0 yr [95% CI: 5.5 – NA] vs 3.6 yr [95% CI: 2.9 – 4.8], p < 0.001), but not for oligo (p = 0.4). RT within 1 yr of diagnosis increased subsequent survival following RT for astro (median 12.9 yr [95% CI: 7.9 – NA] vs 4.4 yr [95% CI: 3.4 – 8.5], p < 0.001), with a trend toward improvement for oligo (p = 0.08). Patients with astro < 40 yr of age exhibited the greatest improvement in subsequent survival following RT, when RT was delivered within 1 yr of diagnosis (0 recorded deaths out of n = 12, p = 0.001). Retrospective data from a single institution support the use of early RT for patients < 40 yr of age with molecularly-defined astrocytomas. RT may also improve OS in this population after subtotal resection. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
5. Salvage Resection and Intracranial Cesium-131 Brachytherapy for Brain Tumors.
- Author
-
Chen, W.C., Lafreniere, M., Phuong, C., Lometti, M., Morin, O., Ziemer, B.P., Vasudevan, H., Hervey-Jumper, S., Theodosopoulos, P.V., Magill, S., Fogh, S.E., Nakamura, J.L., Boreta, L., Sneed, P.K., McDermott, M.W., Raleigh, D., and Braunstein, S.E.
- Subjects
- *
BRAIN tumors , *RADIOISOTOPE brachytherapy , *SURGICAL excision , *SURGICAL complications , *LOW dose rate brachytherapy , *LICENSE fees , *BRAIN metastasis , *RADIOSURGERY - Abstract
Purpose/objective(s): Management of progressive brain tumors following high dose radiotherapy is controversial. In select cases, surgical resection can provide rapid relief of regional mass effect and edema, and highly focal adjuvant re-irradiation can be administered in the form of brachytherapy. Permanent low-dose rate cesium-131 brachytherapy may offer dosimetric and radiobiologic advantages over other isotopes. The purpose of this study was to examine the safety and efficacy of intracranial cesium-131 brachytherapy after surgical resection in a heavily pre-treated patient cohort.Materials/methods: A retrospective chart review was conducted of all patients treated with intraoperative intracranial cesium-131 brachytherapy from an institutional database. Treatment consisted of maximal safe resection followed by implantation of suture-stranded cesium-131 seeds, maintaining approximately 10mm spacing between seeds, with a planned target dose of 80 Gy to 5mm depth. Local failure was defined as recurrence in or contacting the resection cavity after salvage resection and brachytherapy. Regional failure was defined as recurrence within 10mm of the resection cavity. Recurrence was defined as a new lesion or growth of ≥25% in any axial dimension on interval MRI. Toxicity was collected based on CTCAE 4.0.Results: A total of 35 patients with a median imaging follow up of 17.0 months (interquartile range [IQR] 9.7-25.9) underwent 41 consecutive cesium-131 implants between 2016 and 2020. 20 patients (59%; N = 22 implants) were treated for recurrent brain metastasis, 12 patients (34%; N = 16 implants) were treated for recurrent atypical (N = 8) or anaplastic meningioma (N = 4), and 3 patients (9%) were treated for recurrent glioma (N = 2) or hemangiopericytoma (N = 1). A majority of lesions had received prior surgery (N = 28, 66.7%, range 0-3 prior surgeries) and 34 patients had received prior high dose radiation. 34 of 41 (82.9%) tumors had received prior radiosurgery, and 12 (29.2%) had undergone prior external beam radiation (range 50-60 Gy). 17 of 41 lesions (41.5%) had radiologic evidence of adverse radiation/treatment effect (ARE) prior to salvage therapy. Median tumor size was 3.0cm (IQR 2.3-3.7). Overall actuarial local/loco-regional control at 1-year was 91.6%/84.7% (89.8%/88.9% for metastases and 100%/82.5% for meningiomas). Crude loco-regional control was 86.3% for metastases, 75% for meningiomas, and 66.7% for other lesions. Symptomatic ARE following treatment was observed in 9.8% (N = 4) resection cavities. The surgical complication rate was 14.6% (N = 6 total, N = 3 [7.3%] grade 3-5) including 1 peri-operative death.Conclusion: Cesium-131 brachytherapy in this heavily pre-treated cohort resulted in good local control and an acceptable rate of symptomatic adverse radiation effect and toxicity and may be a reasonable treatment option for this select population.Author Disclosure: W.C. Chen: None. M. Lafreniere: None. C. Phuong: None. M. Lometti: None. O. Morin: None. B.P. Ziemer: None. H. Vasudevan: Research Grant; Children's Tumor Foundation. Patent/License Fees/Copyright; Genentech, Eli Lilly. S. Hervey-Jumper: None. P.V. Theodosopoulos: None. S. Magill: None. S.E. Fogh: Independent Contractor; Accuray. J.L. Nakamura: None. L. Boreta: None. P.K. Sneed: None. M.W. McDermott: Chair; Miami Baptist. D. Raleigh: None. S.E. Braunstein: Advisory Board; Radiation Oncology Questions, LLC. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
6. A New Benchmark of the Sharpest Dose Fall-Off for Hypo-Fractionated Radiosurgery of Large Single Target Brain Lesions.
- Author
-
Nano, T., Morin, O., Ziemer, B.P., Raleigh, D., Boreta, L., Nakamura, J.L., Fogh, S.E., Sneed, P.K., Hervey-Jumper, S., Theodosopoulos, P.V., Braunstein, S.E., and Ma, L.
- Subjects
- *
BRAIN damage , *BRAIN tumors , *RADIOSURGERY , *LICENSE fees , *INDEPENDENT contractors ,PLANNING techniques - Abstract
Purpose/objective(s): Previous studies have characterized dose gradient patterns for various hypo-fractionated brain radiosurgery (hSRS) treatment platforms. The Gamma Knife Icon (GKI) has consistently exhibited either superior or non-inferior peripheral dose fall-off and normal brain sparing characteristics when compared to other Linac-based hSRS treatment platforms. Given the prevalence of Linac-based treatments, identifying planning techniques to optimize treatment parameters is desirable. In this study, we investigated a novel Linac-based treatment approach that aimed to create the sharpest dose fall-off for hSRS of large brain lesions.Materials/methods: A cohort of patient cases (n = 10) with single brain lesions (volume 27.6+/-8.1 mL, range 20.-42.1 mL) treated with GKI at our institution were selected as sample test cases for our study. A non-coplanar unconstrained VMAT (NCU-VMAT) treatment planning approach was developed, and its script was implemented on a commercial treatment planning system for clinical Linac treatment equipped with the latest high-definition multi-leaf collimators (MLCs). Treatment plans produced via the NCU-VMAT approach were then compared against further optimized treatment plans from GKI, as well as conventional coplanar and non-coplanar VMAT treatment planning approaches. The comparison was carried out using DVH-derived parameters including target volume coverage, target dose conformity, modified dose fall-off index defined as the volume of 50% prescribed target dose divided by the target volume (PIV50).Results: For each case studied, NCU-VMAT achieved practically identical target coverage (0.98 ± 0.01) and Paddick dose conformity index (0.87 ± 0.02) compared to optimized GKI treatments. The mean PIV50 values were 2.99 ± 0.14, 3.77 ± 0.22, and 3.36 ± 0.19 for optimized GKI, conventional coplanar, and non-coplanar VMAT treatment plans, respectively. These results were in excellent agreement with previously published studies. However, the NCU-VMAT technique yielded a mean PIV50 of 2.41 ± 0.07. This represents an improvement of approximately 40% (P < 0.001, paired two-tailed Student t-test) over all existing approaches including GKI. Further analyses revealed that PIV50 of NCU-VMAT approached the theoretical minimum for all the studied cases.Conclusion: Our results demonstrate that in treatment of large brain tumors, a treatment planning approach using Bremsstrahlung x-ray based Linacs can achieve superior dose fall-off for intracranial hSRS compared to GKI. We dispel the myth that physical characteristics of Cobalt γ-rays are needed to create the sharpest dose fall-off. Given the prevalence of Linac-based treatments compared to GKI, we present a novel optimized planning technique that achieves a new benchmark of sharpest dose fall-off.Author Disclosure: T. Nano: None. O. Morin: None. B.P. Ziemer: None. D. Raleigh: None. L. Boreta: None. J.L. Nakamura: None. S.E. Fogh: Independent Contractor; Accuray. P.K. Sneed: None. S. Hervey-Jumper: None. P.V. Theodosopoulos: None. S.E. Braunstein: Advisory Board; Radiation Oncology Questions, LLC.L. Ma: Patent/License Fees/Copyright; University of California Regents. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
7. Endothelial cells create a stem cell niche in glioblastoma by providing NOTCH ligands that nurture self-renewal of cancer stem-like cells
- Author
-
Xing Fan, Jessica G. Crowley, Mark A. Costello, Shawn L. Hervey-Jumper, Francesco DiMeco, Caroline E. Talsma, Karin M. Muraszko, Xiaobing He, Jason Heth, Thant S. Zhu, Callie G. Flack, Angelo L. Vescovi, Lisa L. Hamm, Zhu, T, Costello, M, Talsma, C, Flack, C, Crowley, J, Hamm, L, He, X, Hervey Jumper, S, Heth, J, Muraszko, K, Dimeco, F, Vescovi, A, and Fan, X
- Subjects
Cancer Research ,Notch signaling pathway ,stem cell, glioblastoma, NOTCH ligands ,Nerve Tissue Proteins ,Cell Growth Processes ,Biology ,Article ,Nestin ,Mice ,Serrate-Jagged Proteins ,Intermediate Filament Proteins ,Antigens, CD ,Neurosphere ,Animals ,Humans ,AC133 Antigen ,RNA, Small Interfering ,Stem Cell Niche ,Adaptor Proteins, Signal Transducing ,Glycoproteins ,Tumor microenvironment ,Receptors, Notch ,Brain Neoplasms ,Calcium-Binding Proteins ,Endothelial Cells ,Membrane Proteins ,Juxtacrine signalling ,Xenograft Model Antitumor Assays ,Cell biology ,nervous system diseases ,Oncology ,Gene Knockdown Techniques ,Cancer cell ,Neoplastic Stem Cells ,Intercellular Signaling Peptides and Proteins ,Stem cell ,Glioblastoma ,Peptides - Abstract
One important function of endothelial cells in glioblastoma multiforme (GBM) is to create a niche that helps promote self-renewal of cancer stem-like cells (CSLC). However, the underlying molecular mechanism for this endothelial function is not known. Since activation of NOTCH signaling has been found to be required for propagation of GBM CSLCs, we hypothesized that the GBM endothelium may provide the source of NOTCH ligands. Here, we report a corroboration of this concept with a demonstration that NOTCH ligands are expressed in endothelial cells adjacent to NESTIN and NOTCH receptor-positive cancer cells in primary GBMs. Coculturing human brain microvascular endothelial cells (hBMEC) or NOTCH ligand with GBM neurospheres promoted GBM cell growth and increased CSLC self-renewal. Notably, RNAi-mediated knockdown of NOTCH ligands in hBMECs abrogated their ability to induce CSLC self-renewal and GBM tumor growth, both in vitro and in vivo. Thus, our findings establish that NOTCH activation in GBM CSLCs is driven by juxtacrine signaling between tumor cells and their surrounding endothelial cells in the tumor microenvironment, suggesting that targeting both CSLCs and their niche may provide a novel strategy to deplete CSLCs and improve GBM treatment. Cancer Res; 71(18); 6061–72. ©2011 AACR.
- Published
- 2011
8. Standard Dose and Dose-Escalated Radiation Therapy Are Associated with Favorable Survival in Select Elderly Patients with Newly Diagnosed Glioblastoma.
- Author
-
Jackson, W.C., Tsien, C., Junck, L., Leung, D., Hervey-Jumper, S., Orringer, D., Heth, J., Wahl, D.R., Spratt, D.E., Cao, Y., Lawrence, T.S., and Kim, M.M.
- Subjects
- *
RADIOTHERAPY , *GLIOBLASTOMA multiforme , *OLDER patients , *DIAGNOSIS - Published
- 2017
- Full Text
- View/download PDF
9. The role of vorasidenib in the treatment of isocitrate dehydrogenase-mutant glioma.
- Author
-
de la Fuente MI, Touat M, van den Bent MJ, Preusser M, Peters KB, Young RJ, Huang RY, Ellingson BM, Capper D, Phillips JJ, Halasz LM, Shih HA, Rudà R, Lim-Fat MJ, Blumenthal DT, Weller M, Arakawa Y, Whittle JR, Ducray F, Reardon DA, Bi WL, Minniti G, Rahman R, Hervey-Jumper S, Chang SM, and Wen PY
- Abstract
Isocitrate dehydrogenase (IDH)-mutant gliomas are the most common malignant primary brain tumors in young adults. This condition imposes a substantial burden on patients and their caregivers, marked by neurocognitive deficits and high mortality rates due to tumor progression, coupled with significant morbidity from current treatment modalities. Although surgery, radiation therapy, and chemotherapy improve survival, these treatments can adversely affect cognitive function, quality of life, finances, employment status, and overall independence. Consequently, there is an urgent need for innovative strategies that delay progression and the use of radiation therapy and chemotherapy. The recent Federal Drug Administration (FDA) approval of vorasidenib, a brain-penetrant small molecule targeting mutant IDH1/2 proteins, heralds a shift in the therapeutic landscape for IDH-mutant gliomas. In this review, we address the role of vorasidenib in the treatment of IDH-mutant gliomas, providing a roadmap for its incorporation into daily practice. We discuss ongoing clinical trials with vorasidenib and other IDH inhibitors, as single-agent or in combination with other therapies, as well as current challenges and future directions., (© The Author(s) 2024. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For commercial re-use, please contact reprints@oup.com for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
10. Practical and statistical aspects of subgroup analyses in surgical neuro-oncology: A comprehensive review from the PIONEER Consortium.
- Author
-
Gerritsen JKW, Karschnia P, Young JS, van den Bent MJ, Chang SM, Smith TR, Nahed BV, Rincon-Torroella J, Bettegowda C, Sanai N, Krieg SM, Maruyama T, Schucht P, Broekman MLD, Tonn JC, Wen PY, De Vleeschouwer S, Vincent AJPE, Hervey-Jumper S, Berger MS, Mekary RA, and Molinaro AM
- Abstract
Subgroup analyses are essential to generate new hypotheses or to estimate treatment effects in clinically meaningful subgroups of patients. They play an important role in taking the next step towards personalized surgical treatment for brain tumor patients. However, subgroup analyses must be used with consideration and care because they have significant potential risks. Although some recommendations are available on the pearls and pitfalls of these analyses, a comprehensive guide is lacking, especially one focused on surgical neuro-oncology patients. This paper, therefore, reviews and summarizes for the first time comprehensively the practical and statistical considerations that are critical to this field. First, we evaluate the considerations when choosing a study design for surgical neuro-oncology studies and examine those unique to this field. Second, we give an overview of the relevant aspects to interpret subgroup analyses adequately. Third, we discuss the practical and statistical elements necessary to appropriately design and use subgroup analyses. The paper aims to provide an in-depth and complete guide to better understand risk modeling and assist the reader with practical examples of designing, using, and interpreting subgroup analyses., (© The Author(s) 2024. Published by Oxford University Press on behalf of the Society for Neuro-Oncology.)
- Published
- 2024
- Full Text
- View/download PDF
11. Foundation models for fast, label-free detection of glioma infiltration.
- Author
-
Kondepudi A, Pekmezci M, Hou X, Scotford K, Jiang C, Rao A, Harake ES, Chowdury A, Al-Holou W, Wang L, Pandey A, Lowenstein PR, Castro MG, Koerner LI, Roetzer-Pejrimovsky T, Widhalm G, Camelo-Piragua S, Movahed-Ezazi M, Orringer DA, Lee H, Freudiger C, Berger M, Hervey-Jumper S, and Hollon T
- Abstract
A critical challenge in glioma treatment is detecting tumour infiltration during surgery to achieve safe maximal resection
1-3 . Unfortunately, safely resectable residual tumour is found in the majority of patients with glioma after surgery, causing early recurrence and decreased survival4-6 . Here we present FastGlioma, a visual foundation model for fast (<10 s) and accurate detection of glioma infiltration in fresh, unprocessed surgical tissue. FastGlioma was pretrained using large-scale self-supervision (around 4 million images) on rapid, label-free optical microscopy, and fine-tuned to output a normalized score that indicates the degree of tumour infiltration within whole-slide optical images. In a prospective, multicentre, international testing cohort of patients with diffuse glioma (n = 220), FastGlioma was able to detect and quantify the degree of tumour infiltration with an average area under the receiver operating characteristic curve of 92.1 ± 0.9%. FastGlioma outperformed image-guided and fluorescence-guided adjuncts for detecting tumour infiltration during surgery by a wide margin in a head-to-head, prospective study (n = 129). The performance of FastGlioma remained high across diverse patient demographics, medical centres and diffuse glioma molecular subtypes as defined by the World Health Organization. FastGlioma shows zero-shot generalization to other adult and paediatric brain tumour diagnoses, demonstrating the potential for our foundation model to be used as a general-purpose adjunct for guiding brain tumour surgeries. These findings represent the transformative potential of medical foundation models to unlock the role of artificial intelligence in the care of patients with cancer., Competing Interests: Competing interests: C.F., D.A.O. and T.H. are shareholders in Invenio Imaging. The other authors declare no competing interests., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
12. Development and validation of a clinical risk model for postoperative outcome in newly diagnosed glioblastoma: a report of the RANO resect group.
- Author
-
Karschnia P, Young JS, Youssef GC, Dono A, Häni L, Sciortino T, Bruno F, Juenger ST, Teske N, Dietrich J, Weller M, Vogelbaum MA, van den Bent M, Beck J, Thon N, Gerritsen JKW, Hervey-Jumper S, Cahill DP, Chang SM, Rudà R, Bello L, Schnell O, Esquenazi Y, Ruge MI, Grau SJ, Huang RY, Wen PY, Berger MS, Molinaro AM, and Tonn JC
- Abstract
Background: Following surgery, patients with newly diagnosed glioblastoma frequently enter clinical trials. Nuanced risk assessment is warranted to reduce imbalances between study arms. Here, we aimed (I) to analyze the interactive effects of residual tumor with clinical and molecular factors on outcome and (II) to define a postoperative risk assessment tool., Methods: The RANO resect group retrospectively compiled an international, seven-center training cohort of patients with newly diagnosed glioblastoma. The combined associations of residual tumor with molecular or clinical factors and survival were analyzed, and recursive partitioning analysis was performed for risk modeling. The resulting model was prognostically verified in a separate external validation cohort., Results: Our training cohort compromised 1003 patients with newly diagnosed isocitrate dehydrogenase-wildtype glioblastoma. Residual tumor, O6-methylguanine DNA methyltransferase (MGMT) promotor methylation status, age, and postoperative KPS were prognostic for survival and incorporated into regression tree analysis. By individually weighting the prognostic factors, an additive score (range, 0-9 points) integrating these four variables distinguished patients with low (0-2 points), intermediate (3-5 points), and high risk (6-9 points) for inferior survival. The prognostic value of our risk model was retained in treatment-based subgroups and confirmed in an external validation cohort of 258 patients with glioblastoma. Compared to previously postulated models, goodness-of-fit measurements were superior for our model., Conclusions: The novel RANO risk model serves as an easy-to-use, yet highly prognostic tool for postoperative patient stratification prior to further therapy. The model may serve to guide patient management and reduce imbalances between study arms in prospective trials., (© The Author(s) 2024. Published by Oxford University Press on behalf of the Society for Neuro-Oncology.)
- Published
- 2024
- Full Text
- View/download PDF
13. A brave new framework for glioma drug development.
- Author
-
Hotchkiss KM, Karschnia P, Schreck KC, Geurts M, Cloughesy TF, Huse J, Duke ES, Lathia J, Ashley DM, Nduom EK, Long G, Singh K, Chalmers A, Ahluwalia MS, Heimberger A, Bagley S, Todo T, Verhaak R, Kelly PD, Hervey-Jumper S, de Groot J, Patel A, Fecci P, Parney I, Wykes V, Watts C, Burns TC, Sanai N, Preusser M, Tonn JC, Drummond KJ, Platten M, Das S, Tanner K, Vogelbaum MA, Weller M, Whittle JR, Berger MS, and Khasraw M
- Subjects
- Humans, Antineoplastic Agents therapeutic use, Brain Neoplasms drug therapy, Brain Neoplasms pathology, Glioma drug therapy, Glioma pathology, Drug Development
- Abstract
Patients with brain tumours are motivated to participate in clinical trials involving repeat tissue sampling. Normalising the use of neoadjuvant and staged surgical trials necessitates collaboration among patients, regulatory agencies, and researchers. Initial and repetitive tissue sampling plays a crucial role in enhancing our understanding of resistance mechanisms and vulnerabilities in brain tumour therapy. Standardising biopsy techniques and ensuring technical uniformity across institutions are vital for effective interinstitutional collaboration. Although liquid biopsy technologies hold promise, they are not yet ready to replace tissue analysis. Clear communication about the risks and benefits of biopsies is essential, particularly regarding potential postoperative deficits. Changes in mindset and neurosurgical culture are imperative to achieve much needed breakthroughs in the development of new, effective therapies for brain tumours., Competing Interests: Declaration of interests MG declares research grant support from Evgen Pharm. KCS declares honoraria from Springworks Therapeutics and Novartis; declares research funding to her institution from Springworks Therapeutics; and serves on a data and safety monitoring board for Advarra. TFC is cofounder, major stock holder, consultant, and board member of Katmai Pharmaceuticals; holds stock for Erasca; is a member of the board and paid consultant for the 501c3 Global Coalition for Adaptive Research; holds stock in Chimerix and receives milestone payments and possible future royalties; is a member of the scientific advisory board for Break Through Cancer foundation and Cure Brain Cancer Foundation; has provided paid consulting services to Symbio, Mundipharma, Tango BlueRock, Vida Ventures, Lista Therapeutics, Stemline, Novartis, Roche, Sonalasense, Sagimet, Clinical Care Options, Ideology Health, Servier, Jubilant, Immvira, Gan & Lee, BrainStorm, Katmai, Sapience, Inovio, Vigeo Therapeutics, DNATrix, Tyme, SDP, Kintara, Bayer, Merck, Boehringer Ingelheim, VBL, Amgen, Kiyatec, AbbVie, VBI, Deciphera, Agios, Novocure, and Medscape; and has contracts with UCLA for the Brain Tumour Programme with Roche, VBI, Merck, Novartis, and Bristol Myers Squibb. The Regents of the University of California, TFC's employer, has licensed intellectual property co-invented by TFC to Katmai Pharmaceuticals. GL is consultant advisor for Agenus, Amgen, Array Biopharma, AstraZeneca, Bayer, BioNTech, Boehringer Ingelheim, Bristol Myers Squibb, Evaxion, Hexal AG (Sandoz Company), Highlight Therapeutics, IOBiotech, Immunocore, Innovent Biologics USA, Merck Sharpe & Dohme, Novartis, PHMR, Pierre Fabre, and Regeneron. AC declares research funding and honoraria from AstraZeneca, Benevolent AI, Duke Street Bio, and Evgen Pharmaceuticals; and has received honoraria from Storm Therapeutics. MSA declares a grant from Seagen; declares consulting fees from Bayer, Kiyatec, Insightec, GSK, Xoft, Nuvation, SDP Oncology, Apollomics, Prelude, Janssen, Voyager Therapeutics, Viewray, Caris Lifesciences, Pyramid Biosciences, Varian Medical Systems, Cairn Therapeutics, Anheart Therapeutics, Theraguix, Menarini Ricerche, Sumitomo Pharma Oncology, Autem Therapeutics, GT Medical Technologies, Allovir, and Equillium Bio; is on the data and safety monitoring board for VBI Vaccines; is on the scientific advisory board of Modifi biosciences and Bugworks; and is a shareholder for Mimivax, Cytodyn, MedInnovate Advisors, and Trisalus Lifesciences. SB reports consulting fees from and is a member of the advisory boards of Telix, Servier, Bayer, and Novocure; and research grants from Incyte, GSK, Lilly, Kite, and Novocure. JdG reports being an advisory board member for Kintara Pharmaceuticals, Kazia, MundiPharma, Insightec, Monteris, Carthera, Samus, Sapience, DSP Pharma, Telix, Servier, Alpha Pharmaceuticals, Nervianos, and CapitalOne; is a data safety monitoring board member for Chimerix and VBI; and has consulted for MundiPharma, Insightec, Carthera, Kintara, Deciphera, Kazia, and Nervianos. IP reports research funding from Merck. TCB reports consulting roles for Predicine; has received financial support from AbbVie and Predicine, with consulting fees paid to Mayo Clinic; and has been a member of the advisory board for Neurametrix. MPr reports honoraria from the following for-profit companies: Bayer, Bristol Myers Squibb, Novartis, Gerson Lehrman Group, CMC Contrast, GSK, Mundipharma, Roche, BMJ Journals, MedMedia, AstraZeneca, AbbVie, Lilly, Medahead, Daiichi Sankyo, Sanofi, Merck Sharp & Dohme, Tocagen, Adastra, Gan & Lee Pharmaceuticals, Janssen, Servier, Miltenyi, Boehringer Ingelheim, Telix, and Medscape. JCT reports research grants from Novocure and Munich Surgical Imaging; being on the advisory board of Advanced Accelerator Applications, Novartis; and Servier; and has received royalties from Springer Publishing. MPl reports being a founder of Tcelltech; has received research support from Bayer, Roche, Pfizer, and Merck; and is an advisory board member of Bayer and Servier. SD is speaker for the Congress of Neurological Surgeons (CNS) and the American Association of Neurological Surgeons; receives research funding from Alkermes; is a member of the Subcortical Surgery Group advisory board; has received royalties from Oxford University Press; and is provincial lead for CNS Oncology at Ontario Health. KT reports consulting fees from Oncohereos Biosciences; and advisory board roles for Advanced Accelerator Applications, Novartis; Sage Therapeutics; FYR Diagnostics; Cordance Medical; Telo Therapeutics; and Modifi Bio. MAV reports clinical trial funding to their institution from DeNovo Pharma, Infuseon, and Oncosynergy; and received honoraria fees from Biodexa and Servier Pharma. MW reports research grants from Novartis, Quercis, and Versameb; honoraria for advisory boards from Roche and Servier; and honoraria for consultation from Bayer, Curevac, Medac, Neurosense, Novartis, Novocure, Orbus, Philogen, and Servier. JRW reports research funding from AnHeart Therapeutics to their institution; received consulting fees from AnHeart Therapeutics and Servier; being on advisory boards for Roche and Merck; is a data safety monitoring member for Telix Pharmaceuticals; and is an employee of The Walter and Eliza Hall Institute and could be eligible for milestone and royalty payments related to Venetoclax. MK reports research grants from Bristol Myers Squibb, AbbVie, BioNTech, CNS Pharmaceuticals, Daiichi Sankyo, Immorna Therapeutics, Immvira Therapeutics, and Personalis; received consulting fees from The Jackson Lab for Genomic Research, AnHeart Therapeutics, Berg Pharma, George Clinical, Manarini Stemline, and Servier; received honoraria from GSK; and is on a data safety monitoring board for BPGbio. All other authors declare no competing interests., (Copyright © 2024 Elsevier Ltd. All rights reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
14. Central nervous system regulation of diffuse glioma growth and invasion: from single unit physiology to circuit remodeling.
- Author
-
Picart T and Hervey-Jumper S
- Subjects
- Humans, Neoplasm Invasiveness, Animals, Nerve Net physiopathology, Nerve Net pathology, Neurons pathology, Neurons physiology, Neurons metabolism, Glioma pathology, Glioma metabolism, Glioma physiopathology, Brain Neoplasms pathology, Brain Neoplasms metabolism, Brain Neoplasms physiopathology
- Abstract
Purpose: Understanding the complex bidirectional interactions between neurons and glioma cells could help to identify new therapeutic targets. Herein, the techniques and application of novel neuroscience tools implemented to study the complex interactions between brain and malignant gliomas, their results, and the potential therapeutic opportunities were reviewed., Methods: Literature search was performed on PubMed between 2001 and 2023 using the keywords "glioma", "glioblastoma", "circuit remodeling", "plasticity", "neuron networks" and "cortical networks". Studies including grade 2 to 4 gliomas, diffuse midline gliomas, and diffuse intrinsic pontine gliomas were considered., Results: Glioma cells are connected through tumour microtubes and form a highly connected network within which pacemaker cells drive tumorigenesis. Unconnected cells have increased invasion capabilities. Glioma cells are also synaptically integrated within neural circuitry. Neurons promote tumour growth via paracrine and direct electrochemical mechanisms, including glutamatergic AMPA-receptors. Increased glutamate release in the tumor microenvironment and loss of peritumoral GABAergic inhibitory interneurons result in network hyperexcitability and secondary epilepsy. Functional imaging, local field potentials and subcortical mapping, performed in awake patients, have defined patterns of malignant circuit remodeling. Glioma-induced remodeling is frequent in language and even motor cortical networks, depending on tumour biological parameters, and influences functional outcomes., Conclusion: These data offer new insights into glioma tumorigenesis. Future work will be needed to understand how tumor intrinsic molecular drivers influence neuron-glioma interactions but also to integrate these results to design new therapeutic options for patients., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
15. Associations between recurrence patterns and outcome in glioblastoma patients undergoing re-resection: A complementary report of the RANO resect group.
- Author
-
Karschnia P, Dono A, Young JS, Juenger ST, Teske N, Häni L, Sciortino T, Mau CY, Bruno F, Nunez L, Morshed RA, Haddad AF, Weller M, van den Bent M, Thon N, Beck J, Hervey-Jumper S, Molinaro AM, Tandon N, Rudà R, Vogelbaum MA, Bello L, Schnell O, Grau SJ, Chang SM, Berger MS, Esquenazi Y, and Tonn JC
- Subjects
- Humans, Combined Modality Therapy, Neoplasm Recurrence, Local surgery, Retrospective Studies, Glioblastoma surgery, Brain Neoplasms surgery
- Published
- 2024
- Full Text
- View/download PDF
16. Surgical management and outcome of newly diagnosed glioblastoma without contrast enhancement (low-grade appearance): a report of the RANO resect group.
- Author
-
Karschnia P, Dietrich J, Bruno F, Dono A, Juenger ST, Teske N, Young JS, Sciortino T, Häni L, van den Bent M, Weller M, Vogelbaum MA, Morshed RA, Haddad AF, Molinaro AM, Tandon N, Beck J, Schnell O, Bello L, Hervey-Jumper S, Thon N, Grau SJ, Esquenazi Y, Rudà R, Chang SM, Berger MS, Cahill DP, and Tonn JC
- Subjects
- Humans, Retrospective Studies, Prognosis, Magnetic Resonance Imaging methods, Glioblastoma diagnostic imaging, Glioblastoma surgery, Glioblastoma pathology, Brain Neoplasms diagnostic imaging, Brain Neoplasms surgery, Brain Neoplasms pathology
- Abstract
Background: Resection of the contrast-enhancing (CE) tumor represents the standard of care in newly diagnosed glioblastoma. However, some tumors ultimately diagnosed as glioblastoma lack contrast enhancement and have a 'low-grade appearance' on imaging (non-CE glioblastoma). We aimed to (a) volumetrically define the value of non-CE tumor resection in the absence of contrast enhancement, and to (b) delineate outcome differences between glioblastoma patients with and without contrast enhancement., Methods: The RANO resect group retrospectively compiled a global, eight-center cohort of patients with newly diagnosed glioblastoma per WHO 2021 classification. The associations between postoperative tumor volumes and outcome were analyzed. Propensity score-matched analyses were constructed to compare glioblastomas with and without contrast enhancement., Results: Among 1323 newly diagnosed IDH-wildtype glioblastomas, we identified 98 patients (7.4%) without contrast enhancement. In such patients, smaller postoperative tumor volumes were associated with more favorable outcome. There was an exponential increase in risk for death with larger residual non-CE tumor. Accordingly, extensive resection was associated with improved survival compared to lesion biopsy. These findings were retained on a multivariable analysis adjusting for demographic and clinical markers. Compared to CE glioblastoma, patients with non-CE glioblastoma had a more favorable clinical profile and superior outcome as confirmed in propensity score analyses by matching the patients with non-CE glioblastoma to patients with CE glioblastoma using a large set of clinical variables., Conclusions: The absence of contrast enhancement characterizes a less aggressive clinical phenotype of IDH-wildtype glioblastomas. Maximal resection of non-CE tumors has prognostic implications and translates into favorable outcome., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Society for Neuro-Oncology.)
- Published
- 2024
- Full Text
- View/download PDF
17. Segmentation of glioblastomas in early post-operative multi-modal MRI with deep neural networks.
- Author
-
Helland RH, Ferles A, Pedersen A, Kommers I, Ardon H, Barkhof F, Bello L, Berger MS, Dunås T, Nibali MC, Furtner J, Hervey-Jumper S, Idema AJS, Kiesel B, Tewari RN, Mandonnet E, Müller DMJ, Robe PA, Rossi M, Sagberg LM, Sciortino T, Aalders T, Wagemakers M, Widhalm G, Witte MG, Zwinderman AH, Majewska PL, Jakola AS, Solheim O, Hamer PCW, Reinertsen I, Eijgelaar RS, and Bouget D
- Subjects
- Humans, Europe, Image Processing, Computer-Assisted methods, Magnetic Resonance Imaging methods, Neoplasm, Residual diagnostic imaging, Neural Networks, Computer, Multicenter Studies as Topic, Datasets as Topic, Glioblastoma diagnostic imaging, Glioblastoma surgery, Glioblastoma pathology
- Abstract
Extent of resection after surgery is one of the main prognostic factors for patients diagnosed with glioblastoma. To achieve this, accurate segmentation and classification of residual tumor from post-operative MR images is essential. The current standard method for estimating it is subject to high inter- and intra-rater variability, and an automated method for segmentation of residual tumor in early post-operative MRI could lead to a more accurate estimation of extent of resection. In this study, two state-of-the-art neural network architectures for pre-operative segmentation were trained for the task. The models were extensively validated on a multicenter dataset with nearly 1000 patients, from 12 hospitals in Europe and the United States. The best performance achieved was a 61% Dice score, and the best classification performance was about 80% balanced accuracy, with a demonstrated ability to generalize across hospitals. In addition, the segmentation performance of the best models was on par with human expert raters. The predicted segmentations can be used to accurately classify the patients into those with residual tumor, and those with gross total resection., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
18. Metabolomic Profiles of Human Glioma Inform Patient Survival.
- Author
-
Scott AJ, Correa LO, Edwards DM, Sun Y, Ravikumar V, Andren AC, Zhang L, Srinivasan S, Jairath N, Verbal K, Muraszko K, Sagher O, Carty SA, Hervey-Jumper S, Orringer D, Kim MM, Junck L, Umemura Y, Leung D, Venneti S, Camelo-Piragua S, Lawrence TS, Ippolito JE, Al-Holou WN, Chinnaiyan P, Heth J, Rao A, Lyssiotis CA, and Wahl DR
- Subjects
- Humans, Mutation, Isocitrate Dehydrogenase genetics, Isocitrate Dehydrogenase metabolism, Glioma genetics, Glioma metabolism, Astrocytoma genetics, Astrocytoma metabolism, Astrocytoma pathology, Glioblastoma genetics, Glioblastoma metabolism, Glioblastoma pathology, Brain Neoplasms genetics, Brain Neoplasms metabolism
- Abstract
Aims: Targeting tumor metabolism may improve the outcomes for patients with glioblastoma (GBM). To further preclinical efforts targeting metabolism in GBM, we tested the hypothesis that brain tumors can be stratified into distinct metabolic groups with different patient outcomes. Therefore, to determine if tumor metabolites relate to patient survival, we profiled the metabolomes of human gliomas and correlated metabolic information with clinical data. Results: We found that isocitrate dehydrogenase-wildtype (IDHwt) GBMs are metabolically distinguishable from IDH mutated (IDHmut) astrocytomas and oligodendrogliomas. Survival of patients with IDHmut gliomas was expectedly more favorable than those with IDHwt GBM, and metabolic signatures can stratify IDHwt GBMs subtypes with varying prognoses. Patients whose GBMs were enriched in amino acids had improved survival, while those whose tumors were enriched for nucleotides, redox molecules, and lipid metabolites fared more poorly. These findings were recapitulated in validation cohorts using both metabolomic and transcriptomic data. Innovation: Our results suggest the existence of metabolic subtypes of GBM with differing prognoses, and further support the concept that metabolism may drive the aggressiveness of human gliomas. Conclusions: Our data show that metabolic signatures of human gliomas can inform patient survival. These findings may be used clinically to tailor novel metabolically targeted agents for GBM patients with different metabolic phenotypes. Antioxid. Redox Signal . 39, 942-956.
- Published
- 2023
- Full Text
- View/download PDF
19. Prognostic evaluation of re-resection for recurrent glioblastoma using the novel RANO classification for extent of resection: A report of the RANO resect group.
- Author
-
Karschnia P, Dono A, Young JS, Juenger ST, Teske N, Häni L, Sciortino T, Mau CY, Bruno F, Nunez L, Morshed RA, Haddad AF, Weller M, van den Bent M, Beck J, Hervey-Jumper S, Molinaro AM, Tandon N, Rudà R, Vogelbaum MA, Bello L, Schnell O, Grau SJ, Chang SM, Berger MS, Esquenazi Y, and Tonn JC
- Subjects
- Humans, Prognosis, Retrospective Studies, Glioblastoma drug therapy, Brain Neoplasms surgery, Brain Neoplasms pathology
- Abstract
Background: The value of re-resection in recurrent glioblastoma remains controversial as a randomized trial that specifies intentional incomplete resection cannot be justified ethically. Here, we aimed to (1) explore the prognostic role of extent of re-resection using the previously proposed Response Assessment in Neuro-Oncology (RANO) classification (based upon residual contrast-enhancing (CE) and non-CE tumor), and to (2) define factors consolidating the surgical effects on outcome., Methods: The RANO resect group retrospectively compiled an 8-center cohort of patients with first recurrence from previously resected glioblastomas. The associations of re-resection and other clinical factors with outcome were analyzed. Propensity score-matched analyses were constructed to minimize confounding effects when comparing the different RANO classes., Results: We studied 681 patients with first recurrence of Isocitrate Dehydrogenase (IDH) wild-type glioblastomas, including 310 patients who underwent re-resection. Re-resection was associated with prolonged survival even when stratifying for molecular and clinical confounders on multivariate analysis; ≤1 cm3 residual CE tumor was associated with longer survival than non-surgical management. Accordingly, "maximal resection" (class 2) had superior survival compared to "submaximal resection" (class 3). Administration of (radio-)chemotherapy in the absence of postoperative deficits augmented the survival associations of smaller residual CE tumors. Conversely, "supramaximal resection" of non-CE tumor (class 1) was not associated with prolonged survival but was frequently accompanied by postoperative deficits. The prognostic role of residual CE tumor was confirmed in propensity score analyses., Conclusions: The RANO resect classification serves to stratify patients with re-resection of glioblastoma. Complete resection according to RANO resect classes 1 and 2 is prognostic., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Society for Neuro-Oncology.)
- Published
- 2023
- Full Text
- View/download PDF
20. Combined cytotoxic and immune-stimulatory gene therapy for primary adult high-grade glioma: a phase 1, first-in-human trial.
- Author
-
Umemura Y, Orringer D, Junck L, Varela ML, West MEJ, Faisal SM, Comba A, Heth J, Sagher O, Leung D, Mammoser A, Hervey-Jumper S, Zamler D, Yadav VN, Dunn P, Al-Holou W, Hollon T, Kim MM, Wahl DR, Camelo-Piragua S, Lieberman AP, Venneti S, McKeever P, Lawrence T, Kurokawa R, Sagher K, Altshuler D, Zhao L, Muraszko K, Castro MG, and Lowenstein PR
- Subjects
- Adult, Female, Humans, Male, Chemoradiotherapy, Genetic Therapy, Adolescent, Middle Aged, Aged, Antineoplastic Agents, Glioblastoma genetics, Glioblastoma therapy, Glioma genetics, Glioma therapy
- Abstract
Background: High-grade gliomas have a poor prognosis and do not respond well to treatment. Effective cancer immune responses depend on functional immune cells, which are typically absent from the brain. This study aimed to evaluate the safety and activity of two adenoviral vectors expressing HSV1-TK (Ad-hCMV-TK) and Flt3L (Ad-hCMV-Flt3L) in patients with high-grade glioma., Methods: In this dose-finding, first-in-human trial, treatment-naive adults aged 18-75 years with newly identified high-grade glioma that was evaluated per immunotherapy response assessment in neuro-oncology criteria, and a Karnofsky Performance Status score of 70 or more, underwent maximal safe resection followed by injections of adenoviral vectors expressing HSV1-TK and Flt3L into the tumour bed. The study was conducted at the University of Michigan Medical School, Michigan Medicine (Ann Arbor, MI, USA). The study included six escalating doses of viral particles with starting doses of 1×10
10 Ad-hCMV-TK viral particles and 1×109 Ad-hCMV-Flt3L viral particles (cohort A), and then 1×1011 Ad-hCMV-TK viral particles and 1×109 Ad-hCMV-Flt3L viral particles (cohort B), 1×1010 Ad-hCMV-TK viral particles and 1×1010 Ad-hCMV-Flt3L viral particles (cohort C), 1×1011 Ad-hCMV-TK viral particles and 1×1010 Ad-hCMV-Flt3L viral particles (cohort D), 1×1010 Ad-hCMV-TK viral particles and 1×1011 Ad-hCMV-Flt3L viral particles (cohort E), and 1×1011 Ad-hCMV-TK viral particles and 1×1011 Ad-hCMV-Flt3L viral particles (cohort F) following a 3+3 design. Two 1 mL tuberculin syringes were used to deliver freehand a mix of Ad-hCMV-TK and Ad-hCMV-Flt3L vectors into the walls of the resection cavity with a total injection of 2 mL distributed as 0·1 mL per site across 20 locations. Subsequently, patients received two 14-day courses of valacyclovir (2 g orally, three times per day) at 1-3 days and 10-12 weeks after vector administration and standad upfront chemoradiotherapy. The primary endpoint was the maximum tolerated dose of Ad-hCMV-Flt3L and Ad-hCMV-TK. Overall survival was a secondary endpoint. Recruitment is complete and the trial is finished. The trial is registered with ClinicalTrials.gov, NCT01811992., Findings: Between April 8, 2014, and March 13, 2019, 21 patients were assessed for eligibility and 18 patients with high-grade glioma were enrolled and included in the analysis (three patients in each of the six dose cohorts); eight patients were female and ten were male. Neuropathological examination identified 14 (78%) patients with glioblastoma, three (17%) with gliosarcoma, and one (6%) with anaplastic ependymoma. The treatment was well-tolerated, and no dose-limiting toxicity was observed. The maximum tolerated dose was not reached. The most common serious grade 3-4 adverse events across all treatment groups were wound infection (four events in two patients) and thromboembolic events (five events in four patients). One death due to an adverse event (respiratory failure) occurred but was not related to study treatment. No treatment-related deaths occurred during the study. Median overall survival was 21·3 months (95% CI 11·1-26·1)., Interpretation: The combination of two adenoviral vectors demonstrated safety and feasibility in patients with high-grade glioma and warrants further investigation in a phase 1b/2 clinical trial., Funding: Funded in part by Phase One Foundation, Los Angeles, CA, The Board of Governors at Cedars-Sinai Medical Center, Los Angeles, CA, and The Rogel Cancer Center at The University of Michigan., Competing Interests: Declaration of interests We declare no competing interests., (Copyright © 2023 Elsevier Ltd. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
21. Prognostic validation of a new classification system for extent of resection in glioblastoma: A report of the RANO resect group.
- Author
-
Karschnia P, Young JS, Dono A, Häni L, Sciortino T, Bruno F, Juenger ST, Teske N, Morshed RA, Haddad AF, Zhang Y, Stoecklein S, Weller M, Vogelbaum MA, Beck J, Tandon N, Hervey-Jumper S, Molinaro AM, Rudà R, Bello L, Schnell O, Esquenazi Y, Ruge MI, Grau SJ, Berger MS, Chang SM, van den Bent M, and Tonn JC
- Subjects
- Humans, Prognosis, Retrospective Studies, Neurosurgical Procedures, Treatment Outcome, Glioblastoma surgery, Glioblastoma drug therapy, Brain Neoplasms surgery, Brain Neoplasms pathology
- Abstract
Background: Terminology to describe extent of resection in glioblastoma is inconsistent across clinical trials. A surgical classification system was previously proposed based upon residual contrast-enhancing (CE) tumor. We aimed to (1) explore the prognostic utility of the classification system and (2) define how much removed non-CE tumor translates into a survival benefit., Methods: The international RANO resect group retrospectively searched previously compiled databases from 7 neuro-oncological centers in the USA and Europe for patients with newly diagnosed glioblastoma per WHO 2021 classification. Clinical and volumetric information from pre- and postoperative MRI were collected., Results: We collected 1,008 patients with newly diagnosed IDHwt glioblastoma. 744 IDHwt glioblastomas were treated with radiochemotherapy per EORTC-26981/22981 (TMZ/RT→TMZ) following surgery. Among these homogenously treated patients, lower absolute residual tumor volumes (in cm3) were favorably associated with outcome: patients with "maximal CE resection" (class 2) had superior outcome compared to patients with "submaximal CE resection" (class 3) or "biopsy" (class 4). Extensive resection of non-CE tumor (≤5 cm3 residual non-CE tumor) was associated with better survival among patients with complete CE resection, thus defining class 1 ("supramaximal CE resection"). The prognostic value of the resection classes was retained on multivariate analysis when adjusting for molecular and clinical markers., Conclusions: The proposed "RANO categories for extent of resection in glioblastoma" are highly prognostic and may serve for stratification within clinical trials. Removal of non-CE tumor beyond the CE tumor borders may translate into additional survival benefit, providing a rationale to explicitly denominate such "supramaximal CE resection.", (© The Author(s) 2022. Published by Oxford University Press on behalf of the Society for Neuro-Oncology.)
- Published
- 2023
- Full Text
- View/download PDF
22. Cancer neuroscience: State of the field, emerging directions.
- Author
-
Winkler F, Venkatesh HS, Amit M, Batchelor T, Demir IE, Deneen B, Gutmann DH, Hervey-Jumper S, Kuner T, Mabbott D, Platten M, Rolls A, Sloan EK, Wang TC, Wick W, Venkataramani V, and Monje M
- Subjects
- Humans, Immune System, Neurons pathology, Tumor Microenvironment, Neoplasms pathology, Neurosciences
- Abstract
The nervous system governs both ontogeny and oncology. Regulating organogenesis during development, maintaining homeostasis, and promoting plasticity throughout life, the nervous system plays parallel roles in the regulation of cancers. Foundational discoveries have elucidated direct paracrine and electrochemical communication between neurons and cancer cells, as well as indirect interactions through neural effects on the immune system and stromal cells in the tumor microenvironment in a wide range of malignancies. Nervous system-cancer interactions can regulate oncogenesis, growth, invasion and metastatic spread, treatment resistance, stimulation of tumor-promoting inflammation, and impairment of anti-cancer immunity. Progress in cancer neuroscience may create an important new pillar of cancer therapy., Competing Interests: Declaration of interests M.M. holds equity in MapLight Therapeutics. M.M. and H.S.V. report the patent (US Patent #10,377,818) “Method for treating glioma.” F.W. and W.W. report the patent (WO2017020982A1) “Agents for use in the treatment of glioma.” F.W. is a co-founder of DC Europa Ltd (a company trading under the name Divide & Conquer) that is developing new medicines for the treatment of glioma. Divide & Conquer also provides research funding to F.W.’s lab under a research collaboration agreement., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
23. Molecularly determining cognition in glioma: New insights as the plot thickens.
- Author
-
Taylor JW, Weyer-Jamora C, and Hervey-Jumper S
- Subjects
- Cognition, Humans, Glioma genetics
- Published
- 2022
- Full Text
- View/download PDF
24. TERT promotor status does not add prognostic information in IDH -wildtype glioblastomas fulfilling other diagnostic WHO criteria: A report of the RANO resect group.
- Author
-
Karschnia P, Young JS, Dono A, Häni L, Juenger ST, Sciortino T, Bruno F, Teske N, Morshed RA, Haddad AF, Zhang Y, Stoecklein S, Vogelbaum MA, Beck J, Tandon N, Hervey-Jumper S, Molinaro AM, Rudà R, Bello L, Schnell O, Esquenazi Y, Ruge MI, Grau SJ, van den Bent M, Weller M, Berger MS, Chang SM, and Tonn JC
- Published
- 2022
- Full Text
- View/download PDF
25. CD34 microvascularity in low-grade glioma: correlation with 5-aminolevulinic acid fluorescence and patient prognosis in a multicenter study at three specialized centers.
- Author
-
Hosmann A, Jaber M, Roetzer-Pejrimovsky T, Timelthaler G, Borkovec M, Kiesel B, Wadiura LI, Millesi M, Mercea PA, Phillips J, Hervey-Jumper S, Berghoff AS, Hainfellner JA, Berger MS, Stummer W, and Widhalm G
- Subjects
- Humans, Aminolevulinic Acid, Prognosis, Retrospective Studies, Antigens, CD34 metabolism, Brain Neoplasms surgery, Glioma surgery
- Abstract
Objective: Early markers are urgently needed in low-grade glioma (LGG) evaluation to rapidly estimate the individual patient's prognosis and to determine the optimal postoperative management. Generally, visible 5-aminolevulinic acid (5-ALA) fluorescence is present in only a few LGGs. Recently, the authors identified visible 5-ALA fluorescence as a powerful intraoperative marker for unfavorable outcome in LGG treatment. However, its precise histopathological correlate is unclear. Neoangiogenesis represents a crucial event in tumor evolution, and CD34 is an established marker for vascular endothelial progenitors potentially indicating tumor progression. The aim of this study was thus to correlate 5-ALA fluorescence and CD34 microvascularity as well as to investigate the prognostic value of CD34 in a large series of LGGs., Methods: In this retrospective study including 3 specialized centers, patients with histopathologically confirmed isocitrate dehydrogenase-mutated LGGs (WHO grade II) receiving 5-ALA prior to resection were included. During surgery, the presence of visible fluorescence was analyzed and one representative tumor sample from the area with the maximum fluorescence effect (tumor with focal fluorescence or nonfluorescing tumor) was selected for each LGG. All fluorescing or nonfluorescing tumor samples were stained for CD34 and semiquantitatively analyzed for microvascular proliferation patterns (physiological vessels, branching capillaries, or microvessel clusters) as well as automatically quantified for CD34 microvessel density (MVD) by standardized histomorphometry software. These semiquantitative/quantitative CD34 data were correlated to the fluorescence status and patient outcome including progression-free survival (PFS), malignant transformation-free survival (MTFS), and overall survival (OS)., Results: In a total of 86 LGGs, visible fluorescence was found during surgery in 13 (15%) cases. First, the semiquantitative CD34 score significantly correlated with intraoperative fluorescence (p = 0.049). Accordingly, the quantitative CD34 MVD was significantly higher in tumors showing fluorescence (p = 0.03). Altogether, the semiquantitative CD34 score showed a strong correlation with quantitative CD34 MVD (p < 0.001). At a mean follow-up of 5.4 ± 2.6 years, microvessel clusters in semiquantitative analysis were a prognostic marker for poor PFS (p = 0.01) and MTFS (p = 0.006), but not OS (p = 0.28). Finally, quantitative CD34 MVD > 10 vessels/mm2 was a prognostic marker for poor PFS (p = 0.01), MTFS (p = 0.008), and OS (p = 0.049)., Conclusions: The data indicate that CD34 microvascularity is associated with intraoperative 5-ALA fluorescence and outcomes in patients with LGG. Thus, visible fluorescence in LGGs might indicate increased CD34 microvascularity, serving as an early prognostic marker for unfavorable patient outcome that is already available during surgery.
- Published
- 2022
- Full Text
- View/download PDF
26. The impact of heme biosynthesis regulation on glioma aggressiveness: Correlations with diagnostic molecular markers.
- Author
-
Mischkulnig M, Kiesel B, Rötzer-Pejrimovsky T, Borkovec M, Lang A, Millesi M, Wadiura LI, Hervey-Jumper S, Penninger JM, Berger MS, Widhalm G, and Erhart F
- Abstract
Background: The prognosis of diffusely infiltrating glioma patients is dismal but varies greatly between individuals. While characterization of gliomas primarily relied on histopathological features, molecular markers increasingly gained importance and play a key role in the recently published 5
th edition of the World Health Organization (WHO) classification. Heme biosynthesis represents a crucial pathway due to its paramount importance in oxygen transport, energy production and drug metabolism. Recently, we described a "heme biosynthesis mRNA expression signature" that correlates with histopathological glioma grade and survival. The aim of the current study was to correlate this heme biosynthesis mRNA expression signature with diagnostic molecular markers and investigate its continued prognostic relevance., Materials and Methods: In this study, patient data were derived from the "The Cancer Genome Atlas" (TCGA) lower-grade glioma and glioblastoma cohorts. We identified diffusely infiltrating gliomas correlating molecular tumor diagnosis according to the most recent WHO classification with heme biosynthesis mRNA expression. The following molecular markers were analyzed: EGFR amplification, TERT promoter mutation, CDKN2A/B homozygous loss, chromosome 7 + /10- aneuploidy, MGMT methylation, IDH mutation, ATRX loss, p53 mutation and 1p19q codeletion. Subsequently, we calculated the heme biosynthesis mRNA expression signature for correlation with distinct molecular glioma markers/molecular subgroups and performed survival analyses., Results: A total of 649 patients with available data on up-to-date molecular markers and heme biosynthesis mRNA expression were included. According to analysis of individual molecular markers, we found a significantly higher heme biosynthesis mRNA expression signature in gliomas with IDH wildtype ( p < 0.0005), without 1p19q codeletion ( p < 0.0005), with homozygous CDKN2A/B loss ( p < 0.0005) and with EGFR amplification ( p = 0.001 ). Furthermore, we observed that the heme biosynthesis mRNA expression signature increased with molecular subgroup aggressiveness ( p < 0.0005 ), being lowest in WHO grade 2 oligodendrogliomas and highest in WHO grade 4 glioblastomas. Finally, the heme biosynthesis mRNA expression signature was a statistically significant survival predictor after multivariate correction for all molecular markers ( p < 0.0005)., Conclusion: Our data demonstrate a significant correlation between heme biosynthesis regulation and diagnostic molecular markers and a prognostic relevance independent of these established markers. Consequently, heme biosynthesis expression is a promising biomarker for glioma aggressiveness and might constitute a potential target for novel therapeutic approaches., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Mischkulnig, Kiesel, Rötzer-Pejrimovsky, Borkovec, Lang, Millesi, Wadiura, Hervey-Jumper, Penninger, Berger, Widhalm and Erhart.)- Published
- 2022
- Full Text
- View/download PDF
27. Preoperative Brain Tumor Imaging: Models and Software for Segmentation and Standardized Reporting.
- Author
-
Bouget D, Pedersen A, Jakola AS, Kavouridis V, Emblem KE, Eijgelaar RS, Kommers I, Ardon H, Barkhof F, Bello L, Berger MS, Conti Nibali M, Furtner J, Hervey-Jumper S, Idema AJS, Kiesel B, Kloet A, Mandonnet E, Müller DMJ, Robe PA, Rossi M, Sciortino T, Van den Brink WA, Wagemakers M, Widhalm G, Witte MG, Zwinderman AH, De Witt Hamer PC, Solheim O, and Reinertsen I
- Abstract
For patients suffering from brain tumor, prognosis estimation and treatment decisions are made by a multidisciplinary team based on a set of preoperative MR scans. Currently, the lack of standardized and automatic methods for tumor detection and generation of clinical reports, incorporating a wide range of tumor characteristics, represents a major hurdle. In this study, we investigate the most occurring brain tumor types: glioblastomas, lower grade gliomas, meningiomas, and metastases, through four cohorts of up to 4,000 patients. Tumor segmentation models were trained using the AGU-Net architecture with different preprocessing steps and protocols. Segmentation performances were assessed in-depth using a wide-range of voxel and patient-wise metrics covering volume, distance, and probabilistic aspects. Finally, two software solutions have been developed, enabling an easy use of the trained models and standardized generation of clinical reports: Raidionics and Raidionics-Slicer. Segmentation performances were quite homogeneous across the four different brain tumor types, with an average true positive Dice ranging between 80 and 90%, patient-wise recall between 88 and 98%, and patient-wise precision around 95%. In conjunction to Dice, the identified most relevant other metrics were the relative absolute volume difference, the variation of information, and the Hausdorff, Mahalanobis, and object average symmetric surface distances. With our Raidionics software, running on a desktop computer with CPU support, tumor segmentation can be performed in 16-54 s depending on the dimensions of the MRI volume. For the generation of a standardized clinical report, including the tumor segmentation and features computation, 5-15 min are necessary. All trained models have been made open-access together with the source code for both software solutions and validation metrics computation. In the future, a method to convert results from a set of metrics into a final single score would be highly desirable for easier ranking across trained models. In addition, an automatic classification of the brain tumor type would be necessary to replace manual user input. Finally, the inclusion of post-operative segmentation in both software solutions will be key for generating complete post-operative standardized clinical reports., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Bouget, Pedersen, Jakola, Kavouridis, Emblem, Eijgelaar, Kommers, Ardon, Barkhof, Bello, Berger, Conti Nibali, Furtner, Hervey-Jumper, Idema, Kiesel, Kloet, Mandonnet, Müller, Robe, Rossi, Sciortino, Van den Brink, Wagemakers, Widhalm, Witte, Zwinderman, De Witt Hamer, Solheim and Reinertsen.)
- Published
- 2022
- Full Text
- View/download PDF
28. Proceedings of the Survivorship Care in Neuro-Oncology Workshop sponsored by the Comprehensive Oncology Network Evaluating Rare CNS Tumors (NCI-CONNECT).
- Author
-
Leeper HE, Tonorezos E, Mayer D, Bakitas M, Chang S, Cooley ME, Hervey-Jumper S, Miaskowski C, Sherwood P, Tsien C, Wallgren K, Willmarth N, Arons D, Acquaye A, King AL, Penas-Prado M, Vera E, Gilbert MR, and Armstrong TS
- Abstract
Background: Survivorship for those living with primary CNS cancers begins at diagnosis, continues throughout a person's life, and includes caregivers. Opportunities and challenges exist to advance survivorship care for those living with primary CNS cancers that necessitate stakeholder involvement., Methods: In June 2021, NCI-CONNECT convened a two-day virtual workshop about survivorship care in neuro-oncology. Two expert panels provided key recommendations and five working groups considered critical questions to identify strengths, weaknesses, opportunities, and threats to the advancement of survivorship care and developed recommendations and action items., Results: The following action items emanated from the workshop: seek endorsement of meeting report from stakeholder organizations; address barriers in access to survivorship care and provider reimbursement; advance survivorship research through NIH and private grant support; develop a survivorship tool kit for providers, people living with primary CNS cancers and their caregivers; provide accessible educational content for neuro-oncology, neurology, and oncology community providers about survivorship care in neuro-oncology; and establish core competencies for survivorship care for neuro-oncology providers to be included in training and standardized exams., Conclusions: Action items aim to address access and reimbursement barriers, expand patient and provider education, develop core competencies, and support survivorship research through funding and other supports., (Published by Oxford University Press on behalf of the Society for Neuro-Oncology and the European Association of Neuro-Oncology 2022.)
- Published
- 2022
- Full Text
- View/download PDF
29. Disparities and Inequities Among Patients with Central Nervous System Tumor.
- Author
-
Chukwueke UN, Hervey-Jumper S, and Porter A
- Subjects
- Delivery of Health Care, Humans, Socioeconomic Factors, United States, Central Nervous System Neoplasms epidemiology, Central Nervous System Neoplasms therapy, Ethnicity
- Abstract
Since the 2002 Institute of Medicine report, which many cite as a landmark in first defining and calling attention to the concept of health disparities in medicine, much work has been dedicated to characterizing health disparities in medical care with the aim of eliminating them. Importantly, this report, "Unequal Treatment: Confronting Racial and Ethnic Disparities in Health Care," laid bare the differences in quality of health care that are based on race, ethnicity, and socioeconomic status. Here, the authors elaborate on these issues and discuss the role of the neuro-oncologic workforce in potentially mitigating these disparities., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
30. Reply to Stummer, W.; Thomas, C. Comment on "Hosmann et al. 5-ALA Fluorescence Is a Powerful Prognostic Marker during Surgery of Low-Grade Gliomas (WHO Grade II)-Experience at Two Specialized Centers. Cancers 2021, 13 , 2540".
- Author
-
Hosmann A, Millesi M, Wadiura LI, Kiesel B, Mercea PA, Mischkulnig M, Borkovec M, Furtner J, Roetzer T, Wolfsberger S, Phillips JJ, Berghoff AS, Hervey-Jumper S, Berger MS, and Widhalm G
- Abstract
We greatly appreciate Dr. Stummer's and Dr. Thomas's interest in our study and their important comments [...].
- Published
- 2021
- Full Text
- View/download PDF
31. G-CSF secreted by mutant IDH1 glioma stem cells abolishes myeloid cell immunosuppression and enhances the efficacy of immunotherapy.
- Author
-
Alghamri MS, McClellan BL, Avvari RP, Thalla R, Carney S, Hartlage CS, Haase S, Ventosa M, Taher A, Kamran N, Zhang L, Faisal SM, Núñez FJ, Garcia-Fabiani MB, Al-Holou WN, Orringer D, Hervey-Jumper S, Heth J, Patil PG, Eddy K, Merajver SD, Ulintz PJ, Welch J, Gao C, Liu J, Núñez G, Hambardzumyan D, Lowenstein PR, and Castro MG
- Abstract
Mutant isocitrate-dehydrogenase 1 ( mIDH1 ) synthesizes the oncometabolite 2-hydroxyglutarate (2HG), which elicits epigenetic reprogramming of the glioma cells’ transcriptome by inhibiting DNA and histone demethylases. We show that the efficacy of immune-stimulatory gene therapy (TK/Flt3L) is enhanced in mIDH1 gliomas, due to the reprogramming of the myeloid cells’ compartment infiltrating the tumor microenvironment (TME). We uncovered that the immature myeloid cells infiltrating the mIDH1 TME are mainly nonsuppressive neutrophils and preneutrophils. Myeloid cell reprogramming was triggered by granulocyte colony-stimulating factor (G-CSF) secreted by mIDH1 glioma stem/progenitor-like cells. Blocking G-CSF in mIDH1 glioma–bearing mice restores the inhibitory potential of the tumor-infiltrating myeloid cells, accelerating tumor progression. We demonstrate that G-CSF reprograms bone marrow granulopoiesis, resulting in noninhibitory myeloid cells within mIDH1 glioma TME and enhancing the efficacy of immune-stimulatory gene therapy.
- Published
- 2021
- Full Text
- View/download PDF
32. Glioblastoma Surgery Imaging-Reporting and Data System: Validation and Performance of the Automated Segmentation Task.
- Author
-
Bouget D, Eijgelaar RS, Pedersen A, Kommers I, Ardon H, Barkhof F, Bello L, Berger MS, Nibali MC, Furtner J, Fyllingen EH, Hervey-Jumper S, Idema AJS, Kiesel B, Kloet A, Mandonnet E, Müller DMJ, Robe PA, Rossi M, Sagberg LM, Sciortino T, Van den Brink WA, Wagemakers M, Widhalm G, Witte MG, Zwinderman AH, Reinertsen I, De Witt Hamer PC, and Solheim O
- Abstract
For patients with presumed glioblastoma, essential tumor characteristics are determined from preoperative MR images to optimize the treatment strategy. This procedure is time-consuming and subjective, if performed by crude eyeballing or manually. The standardized GSI-RADS aims to provide neurosurgeons with automatic tumor segmentations to extract tumor features rapidly and objectively. In this study, we improved automatic tumor segmentation and compared the agreement with manual raters, describe the technical details of the different components of GSI-RADS, and determined their speed. Two recent neural network architectures were considered for the segmentation task: nnU-Net and AGU-Net. Two preprocessing schemes were introduced to investigate the tradeoff between performance and processing speed. A summarized description of the tumor feature extraction and standardized reporting process is included. The trained architectures for automatic segmentation and the code for computing the standardized report are distributed as open-source and as open-access software. Validation studies were performed on a dataset of 1594 gadolinium-enhanced T1-weighted MRI volumes from 13 hospitals and 293 T1-weighted MRI volumes from the BraTS challenge. The glioblastoma tumor core segmentation reached a Dice score slightly below 90%, a patientwise F1-score close to 99%, and a 95th percentile Hausdorff distance slightly below 4.0 mm on average with either architecture and the heavy preprocessing scheme. A patient MRI volume can be segmented in less than one minute, and a standardized report can be generated in up to five minutes. The proposed GSI-RADS software showed robust performance on a large collection of MRI volumes from various hospitals and generated results within a reasonable runtime.
- Published
- 2021
- Full Text
- View/download PDF
33. Central Nervous System Plasticity Influences Language and Cognitive Recovery in Adult Glioma.
- Author
-
Krishna S, Kakaizada S, Almeida N, Brang D, and Hervey-Jumper S
- Subjects
- Adult, Brain Mapping, Central Nervous System, Cognition, Humans, Language, Quality of Life, Brain Neoplasms complications, Glioma complications
- Abstract
Gliomas exist within the framework of complex neuronal circuitry in which network dynamics influence both tumor biology and cognition. The generalized impairment of cognition or loss of language function is a common occurrence for glioma patients. The interface between intrinsic brain tumors such as gliomas and functional cognitive networks are poorly understood. The ability to communicate effectively is critically important for receiving oncological therapies and maintaining a high quality of life. Although the propensity of gliomas to infiltrate cortical and subcortical structures and disrupt key anatomic language pathways is well documented, there is new evidence offering insight into the network and cellular mechanisms underpinning glioma-related aphasia and aphasia recovery. In this review, we will outline the current understanding of the mechanisms of cognitive dysfunction and recovery, using aphasia as an illustrative model., (© Congress of Neurological Surgeons 2021.)
- Published
- 2021
- Full Text
- View/download PDF
34. 5-ALA in Suspected Low-Grade Gliomas: Current Role, Limitations, and New Approaches.
- Author
-
Kiesel B, Freund J, Reichert D, Wadiura L, Erkkilae MT, Woehrer A, Hervey-Jumper S, Berger MS, and Widhalm G
- Abstract
Radiologically suspected low-grade gliomas (LGG) represent a special challenge for the neurosurgeon during surgery due to their histopathological heterogeneity and indefinite tumor margin. Therefore, new techniques are required to overcome these current surgical drawbacks. Intraoperative visualization of brain tumors with assistance of 5-aminolevulinic acid (5-ALA) induced protoporphyrin IX (PpIX) fluorescence is one of the major advancements in the neurosurgical field in the last decades. Initially, this technique was exclusively applied for fluorescence-guided surgery of high-grade glioma (HGG). In the last years, the use of 5-ALA was also extended to other indications such as radiologically suspected LGG. Here, we discuss the current role of 5-ALA for intraoperative visualization of focal malignant transformation within suspected LGG. Furthermore, we discuss the current limitations of the 5-ALA technology in pure LGG which usually cannot be visualized by visible fluorescence. Finally, we introduce new approaches based on fluorescence technology for improved detection of pure LGG tissue such as spectroscopic PpIX quantification fluorescence lifetime imaging of PpIX and confocal microscopy to optimize surgery., Competing Interests: JF receives financial research support by NX Development Corp. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Kiesel, Freund, Reichert, Wadiura, Erkkilae, Woehrer, Hervey-Jumper, Berger and Widhalm.)
- Published
- 2021
- Full Text
- View/download PDF
35. Glioblastoma Surgery Imaging-Reporting and Data System: Standardized Reporting of Tumor Volume, Location, and Resectability Based on Automated Segmentations.
- Author
-
Kommers I, Bouget D, Pedersen A, Eijgelaar RS, Ardon H, Barkhof F, Bello L, Berger MS, Conti Nibali M, Furtner J, Fyllingen EH, Hervey-Jumper S, Idema AJS, Kiesel B, Kloet A, Mandonnet E, Müller DMJ, Robe PA, Rossi M, Sagberg LM, Sciortino T, van den Brink WA, Wagemakers M, Widhalm G, Witte MG, Zwinderman AH, Reinertsen I, Solheim O, and De Witt Hamer PC
- Abstract
Treatment decisions for patients with presumed glioblastoma are based on tumor characteristics available from a preoperative MR scan. Tumor characteristics, including volume, location, and resectability, are often estimated or manually delineated. This process is time consuming and subjective. Hence, comparison across cohorts, trials, or registries are subject to assessment bias. In this study, we propose a standardized Glioblastoma Surgery Imaging Reporting and Data System (GSI-RADS) based on an automated method of tumor segmentation that provides standard reports on tumor features that are potentially relevant for glioblastoma surgery. As clinical validation, we determine the agreement in extracted tumor features between the automated method and the current standard of manual segmentations from routine clinical MR scans before treatment. In an observational consecutive cohort of 1596 adult patients with a first time surgery of a glioblastoma from 13 institutions, we segmented gadolinium-enhanced tumor parts both by a human rater and by an automated algorithm. Tumor features were extracted from segmentations of both methods and compared to assess differences, concordance, and equivalence. The laterality, contralateral infiltration, and the laterality indices were in excellent agreement. The native and normalized tumor volumes had excellent agreement, consistency, and equivalence. Multifocality, but not the number of foci, had good agreement and equivalence. The location profiles of cortical and subcortical structures were in excellent agreement. The expected residual tumor volumes and resectability indices had excellent agreement, consistency, and equivalence. Tumor probability maps were in good agreement. In conclusion, automated segmentations are in excellent agreement with manual segmentations and practically equivalent regarding tumor features that are potentially relevant for neurosurgical purposes. Standard GSI-RADS reports can be generated by open access software.
- Published
- 2021
- Full Text
- View/download PDF
36. 5-ALA Fluorescence Is a Powerful Prognostic Marker during Surgery of Low-Grade Gliomas (WHO Grade II)-Experience at Two Specialized Centers.
- Author
-
Hosmann A, Millesi M, Wadiura LI, Kiesel B, Mercea PA, Mischkulnig M, Borkovec M, Furtner J, Roetzer T, Wolfsberger S, Phillips JJ, Berghoff AS, Hervey-Jumper S, Berger MS, and Widhalm G
- Abstract
The prediction of the individual prognosis of low-grade glioma (LGG) patients is limited in routine clinical practice. Nowadays, 5-aminolevulinic acid (5-ALA) fluorescence is primarily applied for improved intraoperative visualization of high-grade gliomas. However, visible fluorescence is also observed in rare cases despite LGG histopathology and might be an indicator for aggressive tumor behavior. The aim of this study was thus to investigate the value of intraoperative 5-ALA fluorescence for prognosis in LGG patients. We performed a retrospective analysis of patients with newly diagnosed histopathologically confirmed LGG and preoperative 5-ALA administration at two independent specialized centers. In this cohort, we correlated the visible intraoperative fluorescence status with progression-free survival (PFS), malignant transformation-free survival (MTFS) and overall survival (OS). Altogether, visible fluorescence was detected in 7 (12%) of 59 included patients in focal intratumoral areas. At a mean follow-up time of 5.3 ± 2.9 years, patients with fluorescing LGG had significantly shorter PFS (2.3 ± 0.7 vs. 5.0 ± 0.4 years; p = 0.01), MTFS (3.9 ± 0.7 vs. 8.0 ± 0.6 years; p = 0.03), and OS (5.4 ± 1.0 vs. 10.3 ± 0.5 years; p = 0.01) than non-fluorescing tumors. Our data indicate that visible 5-ALA fluorescence during surgery of pure LGG might be an already intraoperatively available marker of unfavorable patient outcome and thus close imaging follow-up might be considered.
- Published
- 2021
- Full Text
- View/download PDF
37. Timing of glioblastoma surgery and patient outcomes: a multicenter cohort study.
- Author
-
Müller DMJ, De Swart ME, Ardon H, Barkhof F, Bello L, Berger MS, Bouwknegt W, Van den Brink WA, Conti Nibali M, Eijgelaar RS, Furtner J, Han SJ, Hervey-Jumper S, Idema AJS, Kiesel B, Kloet A, Mandonnet E, Robe PAJT, Rossi M, Sciortino T, Vandertop WP, Visser M, Wagemakers M, Widhalm G, Witte MG, and De Witt Hamer PC
- Abstract
Background: The impact of time-to-surgery on clinical outcome for patients with glioblastoma has not been determined. Any delay in treatment is perceived as detrimental, but guidelines do not specify acceptable timings. In this study, we relate the time to glioblastoma surgery with the extent of resection and residual tumor volume, performance change, and survival, and we explore the identification of patients for urgent surgery., Methods: Adults with first-time surgery in 2012-2013 treated by 12 neuro-oncological teams were included in this study. We defined time-to-surgery as the number of days between the diagnostic MR scan and surgery. The relation between time-to-surgery and patient and tumor characteristics was explored in time-to-event analysis and proportional hazard models. Outcome according to time-to-surgery was analyzed by volumetric measurements, changes in performance status, and survival analysis with patient and tumor characteristics as modifiers., Results: Included were 1033 patients of whom 729 had a resection and 304 a biopsy. The overall median time-to-surgery was 13 days. Surgery was within 3 days for 235 (23%) patients, and within a month for 889 (86%). The median volumetric doubling time was 22 days. Lower performance status (hazard ratio [HR] 0.942, 95% confidence interval [CI] 0.893-0.994) and larger tumor volume (HR 1.012, 95% CI 1.010-1.014) were independently associated with a shorter time-to-surgery. Extent of resection, residual tumor volume, postoperative performance change, and overall survival were not associated with time-to-surgery., Conclusions: With current decision-making for urgent surgery in selected patients with glioblastoma and surgery typically within 1 month, we found equal extent of resection, residual tumor volume, performance status, and survival after longer times-to-surgery., (© The Author(s) 2021. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.)
- Published
- 2021
- Full Text
- View/download PDF
38. Delivering Equitable Care to Underserved Neuro-oncology Populations.
- Author
-
Porter AB, Chukwueke UN, Mammoser AG, Friday B, and Hervey-Jumper S
- Subjects
- Ethnicity, Humans, Racial Groups, United States epidemiology, Vulnerable Populations, Brain Neoplasms diagnosis, Brain Neoplasms epidemiology, Brain Neoplasms therapy, Medically Underserved Area
- Abstract
It is widely recognized that subspecialized multidisciplinary care improves neuro-oncology outcomes. Optimizing patient outcomes relies on the expertise of the treating physicians, neuroradiology and neuropathology, and supportive services familiar with common neurologic syndromes that occur after brain tumor diagnosis and treatment. Despite an increasing number of providers, patient access to specialized multidisciplinary care and clinical trials remains limited. Barriers to equitable health care exist across the United States, with marginalized communities being impacted disproportionately. Such disparity causes increased morbidity and mortality for patients from backgrounds with various elements of diversity. Limited attention to this inequity has resulted in an incomplete understanding of the spectrum of experiences that patients with neuro-oncologic diseases encounter. Clinical trials represent the highest standard and quality of care in medicine, but inclusion of under-represented and underserved groups consistently lags behind counterpart participants from majority racial and ethnic groups. Through provider education as it pertains to issues from bias and health literacy to increasing clinical trial enrollment and offering opportunities through telemedicine, opportunities for improving access to high-quality neuro-oncologic care are explored.
- Published
- 2021
- Full Text
- View/download PDF
39. The state of neuro-oncology during the COVID-19 pandemic: a worldwide assessment.
- Author
-
Mrugala MM, Ostrom QT, Pressley SM, Taylor JW, Thomas AA, Wefel JS, Coven SL, Acquaye AA, Haynes C, Agnihotri S, Lim M, Peters KB, Sulman EP, Salcido JT, Butowski NA, Hervey-Jumper S, Mansouri A, Oliver KR, Porter AB, Nassiri F, Schiff D, Dunbar EM, Hegi ME, Armstrong TS, van den Bent MJ, Chang SM, Zadeh G, and Chheda MG
- Abstract
Background: It remains unknown how the COVID-19 pandemic has changed neuro-oncology clinical practice, training, and research efforts., Methods: We performed an international survey of practitioners, scientists, and trainees from 21 neuro-oncology organizations across 6 continents, April 24-May 17, 2020. We assessed clinical practice and research environments, institutional preparedness and support, and perceived impact on patients., Results: Of 582 respondents, 258 (45%) were US-based and 314 (55%) international. Ninety-four percent of participants reported changes in their clinical practice. Ninety-five percent of respondents converted at least some practice to telemedicine. Ten percent of practitioners felt the need to see patients in person, specifically because of billing concerns and pressure from their institutions. Sixty-seven percent of practitioners suspended enrollment for at least one clinical trial, including 62% suspending phase III trial enrollments. More than 50% believed neuro-oncology patients were at increased risk for COVID-19. Seventy-one percent of clinicians feared for their own personal safety or that of their families, specifically because of their clinical duties; 20% had inadequate personal protective equipment. While 69% reported increased stress, 44% received no psychosocial support from their institutions. Thirty-seven percent had salary reductions and 63% of researchers temporarily closed their laboratories. However, the pandemic created positive changes in perceived patient satisfaction, communication quality, and technology use to deliver care and mediate interactions with other practitioners., Conclusions: The pandemic has changed treatment schedules and limited investigational treatment options. Institutional lack of support created clinician and researcher anxiety. Communication with patients was satisfactory. We make recommendations to guide clinical and scientific infrastructure moving forward and address the personal challenges of providers and researchers., (© The Author(s) 2021. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.)
- Published
- 2021
- Full Text
- View/download PDF
40. Heme Biosynthesis mRNA Expression Signature: Towards a Novel Prognostic Biomarker in Patients with Diffusely Infiltrating Gliomas.
- Author
-
Mischkulnig M, Kiesel B, Lötsch D, Roetzer T, Borkovec M, Wadiura LI, Roessler K, Hervey-Jumper S, Penninger JM, Berger MS, Widhalm G, and Erhart F
- Abstract
Diffusely infiltrating gliomas are characterized by a variable clinical course, and thus novel prognostic biomarkers are needed. The heme biosynthesis cycle constitutes a fundamental metabolic pathway and might play a crucial role in glioma biology. The aim of this study was thus to investigate the role of the heme biosynthesis mRNA expression signature on prognosis in a large glioma patient cohort. Glioma patients with available sequencing data on heme biosynthesis expression were retrieved from The Cancer Genome Atlas (TCGA). In each patient, the heme biosynthesis mRNA expression signature was calculated and categorized into low, medium, and high expression subgroups. Differences in progression-free and overall survival between these subgroups were investigated including a multivariate analysis correcting for WHO grade, tumor subtype, and patient age and sex. In a total of 693 patients, progression-free and overall survival showed a strictly monotonical decrease with increasing mRNA expression signature subgroups. In detail, median overall survival was 134.2 months in the low, 79.9 months in the intermediate, and 16.5 months in the high mRNA expression signature subgroups, respectively. The impact of mRNA expression signature on progression-free and overall survival was independent of the other analyzed prognostic factors. Our data indicate that the heme biosynthesis mRNA expression signature might serve as an additional novel prognostic marker in patients with diffusely infiltrating gliomas to optimize postoperative management.
- Published
- 2021
- Full Text
- View/download PDF
41. High Interobserver Agreement in the Subjective Classification of 5-Aminolevulinic Acid Fluorescence Levels in Newly Diagnosed Glioblastomas.
- Author
-
Mischkulnig M, Kiesel B, Borkovec M, Wadiura LI, Benner D, Hosmann A, Hervey-Jumper S, Knosp E, Roessler K, Berger MS, and Widhalm G
- Subjects
- Cohort Studies, Humans, Observer Variation, Aminolevulinic Acid, Glioblastoma diagnostic imaging, Glioblastoma surgery
- Abstract
Background and Objectives: Fluorescence-guided resection of glioblastomas (GBM) using 5-aminolevulinic acid (5-ALA) improves intraoperative tumor visualization and is thus widely used nowadays. During resection, different fluorescence levels can usually be distinguished within the same tumor. Recently, we demonstrated that strong, vague, and no fluorescence correspond to distinct histopathological characteristics in newly diagnosed GBM. However, the qualitative fluorescence classification by the neurosurgeon is subjective and currently no comprehensive data on interobserver variability is available. The aim of this study was thus to investigate the interobserver variability in the classification of 5-ALA fluorescence levels in newly diagnosed GBM., Study Design/materials and Methods: A questionnaire investigating the interobserver variability in 5-ALA fluorescence quantification was performed at a nation-wide neurosurgical oncology meeting. The participants involved in the neurosurgical/neurooncological field were asked to categorize 30 cases of 5-ALA fluorescence images derived from GBM resection on a lecture hall screen according to the widely used three-tier fluorescence classification scheme (negative, vague, or strong fluorescence). Additionally, participants were asked for information on their medical background such as specialty, level of training, and experience with 5-ALA fluorescence-guided procedures. Interobserver agreement was defined as the calculated mean κ values for each observer., Results: A total of 36 questionnaires were included in the final analysis. The mean average κ value in fluorescence classification within the entire cohort was 0.71 ± 0.12 and 29 (81%) participants had a substantial or almost perfect interobserver agreement (κ values 0.6-1.0). Interobserver agreement was significantly higher in neurosurgeons (mean κ: 0.83) as compared with non-neurosurgeons involved in the neurooncological field (mean κ: 0.52; P < 0.001). Furthermore, interobserver agreement was significantly higher in participants who had experience with at least 25 5-ALA fluorescence-guided surgeries (mean κ: 0.87) compared with less experienced colleagues (mean κ: 0.82; P = 0.039)., Conclusion: Our study found a high interobserver agreement in the qualitative classification of different 5-ALA fluorescence levels in newly diagnosed GBM. Interobserver agreement increases significantly in more experienced participants and therefore a high level of experience is crucial for reliable intraoperative fluorescence classification. Lasers Surg. Med. © 2020 The Authors. Lasers in Surgery and Medicine published by Wiley Periodicals, Inc., (© 2020 The Authors. Lasers in Surgery and Medicine published by Wiley Periodicals, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
42. Robust Deep Learning-based Segmentation of Glioblastoma on Routine Clinical MRI Scans Using Sparsified Training.
- Author
-
Eijgelaar RS, Visser M, Müller DMJ, Barkhof F, Vrenken H, van Herk M, Bello L, Conti Nibali M, Rossi M, Sciortino T, Berger MS, Hervey-Jumper S, Kiesel B, Widhalm G, Furtner J, Robe PAJT, Mandonnet E, De Witt Hamer PC, de Munck JC, and Witte MG
- Abstract
Purpose: To improve the robustness of deep learning-based glioblastoma segmentation in a clinical setting with sparsified datasets., Materials and Methods: In this retrospective study, preoperative T1-weighted, T2-weighted, T2-weighted fluid-attenuated inversion recovery, and postcontrast T1-weighted MRI from 117 patients (median age, 64 years; interquartile range [IQR], 55-73 years; 76 men) included within the Multimodal Brain Tumor Image Segmentation (BraTS) dataset plus a clinical dataset (2012-2013) with similar imaging modalities of 634 patients (median age, 59 years; IQR, 49-69 years; 382 men) with glioblastoma from six hospitals were used. Expert tumor delineations on the postcontrast images were available, but for various clinical datasets, one or more sequences were missing. The convolutional neural network, DeepMedic, was trained on combinations of complete and incomplete data with and without site-specific data. Sparsified training was introduced, which randomly simulated missing sequences during training. The effects of sparsified training and center-specific training were tested using Wilcoxon signed rank tests for paired measurements., Results: A model trained exclusively on BraTS data reached a median Dice score of 0.81 for segmentation on BraTS test data but only 0.49 on the clinical data. Sparsified training improved performance (adjusted P < .05), even when excluding test data with missing sequences, to median Dice score of 0.67. Inclusion of site-specific data during sparsified training led to higher model performance Dice scores greater than 0.8, on par with a model based on all complete and incomplete data. For the model using BraTS and clinical training data, inclusion of site-specific data or sparsified training was of no consequence., Conclusion: Accurate and automatic segmentation of glioblastoma on clinical scans is feasible using a model based on large, heterogeneous, and partially incomplete datasets. Sparsified training may boost the performance of a smaller model based on public and site-specific data. Supplemental material is available for this article. Published under a CC BY 4.0 license., Competing Interests: Disclosures of Conflicts of Interest: R.S.E. disclosed no relevant relationships. M.V. Activities related to the present article: institution receives grant from Netherlands Organization for Scientific Research (NOW) (project number 10-10400-96-14003); institution receives grant from Dutch Cancer Society (VU2014-7113). Activities not related to the present article: disclosed no relevant relationships. Other relationships: disclosed no relevant relationships. D.M.J.M. disclosed no relevant relationships. F.B. Activities related to the present article: institution receives grant from Netherlands Organization for Scientific Research (NOW) for PICTURE project. Activities not related to the present article: author paid as board member of Roche, Bayer, and Merck (DSMB and Steering Committees); author is consultant for IXICO; institution receives grants from EU-H2020, UKMSS, NWO, MRC, HIHR-BRCUCLH; author receives royalties from Springer books; institution paid for educational presentations by Biogen (PML educational website). Other relationships: disclosed no relevant relationships. H.V. Activities related to the present article: disclosed no relevant relationships. Activities not related to the present article: institution is consultant for Merck (multiple sclerosis brain imaging consulting); institution receives grants from Teva, Novartis, Merck (research grants for multiple sclerosis studies). Other relationships: disclosed no relevant relationships. M.v.H. Activities related to the present article: institution receives grant from Dutch Cancer Society. Activities not related to the present article: institution receives grants from MRC Proximity to Discovery, MRC Studentship (PhD student); author paid for lectures by ESTRO (Honorarium -Sept 2018, ESTRO [ATP Meeting] €500 Honorarium -Feb 2019, ESTRO [IGRT Meeting] €500); author receives travel accommodations from IPEM, UKIO, AMC, AIFM, IGT Network, ICR, ESTRO, Elekta, NKI (conference and meeting expenses such as travel, accommodations, etc. IPEM July 2019, UKIO June 2019, July 2018, AMC May 2019, Jan 2019, BIR April 2019, March 2019, AIFM March 2019, IGT Network March 2019, Nov 2018, ICR March 2019, ESTRO Feb 2019, Sept 2018 Elekta June 2018, NKI July 2018). Other relationships: disclosed no relevant relationships. L.B. disclosed no relevant relationships. M.C.N. disclosed no relevant relationships. M.R. disclosed no relevant relationships. T.S. disclosed no relevant relationships. M.S.B. disclosed no relevant relationships. S.H.J. disclosed no relevant relationships. B.K. disclosed no relevant relationships. G.W. disclosed no relevant relationships. J.F. disclosed no relevant relationships. P.A.J.T.R. disclosed no relevant relationships. E.M. disclosed no relevant relationships. P.C.D.W.H. Activities related to the present article: institution receives grant from Dutch Cancer Society (VU2014-7113); BrainLab provided SmartBrush software; ZonMW (This research is part of the program Innovative Medical Devices Initiative with project number 10-10400-96-14003, which is financed by the Netherlands Organization for Scientific Research). Activities not related to the present article: disclosed no relevant relationships. Other relationships: disclosed no relevant relationships. J.C.d.M. Activities related to the present article: institution received grant from ZonMW (ZonMW paid a research grant to VUmc to cover salaries of PhD students involved. Activities not related to the present article: disclosed no relevant relationships. Other relationships: disclosed no relevant relationships. M.G.W. disclosed no relevant relationships., (2020 by the Radiological Society of North America, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
43. Comprehensive analysis of diverse low-grade neuroepithelial tumors with FGFR1 alterations reveals a distinct molecular signature of rosette-forming glioneuronal tumor.
- Author
-
Lucas CG, Gupta R, Doo P, Lee JC, Cadwell CR, Ramani B, Hofmann JW, Sloan EA, Kleinschmidt-DeMasters BK, Lee HS, Wood MD, Grafe M, Born D, Vogel H, Salamat S, Puccetti D, Scharnhorst D, Samuel D, Cooney T, Cham E, Jin LW, Khatib Z, Maher O, Chamyan G, Brathwaite C, Bannykh S, Mueller S, Kline CN, Banerjee A, Reddy A, Taylor JW, Clarke JL, Oberheim Bush NA, Butowski N, Gupta N, Auguste KI, Sun PP, Roland JL, Raffel C, Aghi MK, Theodosopoulos P, Chang E, Hervey-Jumper S, Phillips JJ, Pekmezci M, Bollen AW, Tihan T, Chang S, Berger MS, Perry A, and Solomon DA
- Subjects
- Adolescent, Adult, Aged, Brain Neoplasms classification, Brain Neoplasms genetics, Brain Neoplasms pathology, Child, Female, Humans, Male, Middle Aged, Mutation, Spinal Cord Neoplasms classification, Spinal Cord Neoplasms genetics, Spinal Cord Neoplasms pathology, Young Adult, Neoplasms, Neuroepithelial classification, Neoplasms, Neuroepithelial genetics, Neoplasms, Neuroepithelial pathology, Receptor, Fibroblast Growth Factor, Type 1 genetics
- Abstract
The FGFR1 gene encoding fibroblast growth factor receptor 1 has emerged as a frequently altered oncogene in the pathogenesis of multiple low-grade neuroepithelial tumor (LGNET) subtypes including pilocytic astrocytoma, dysembryoplastic neuroepithelial tumor (DNT), rosette-forming glioneuronal tumor (RGNT), and extraventricular neurocytoma (EVN). These activating FGFR1 alterations in LGNET can include tandem duplication of the exons encoding the intracellular tyrosine kinase domain, in-frame gene fusions most often with TACC1 as the partner, or hotspot missense mutations within the tyrosine kinase domain (either at p.N546 or p.K656). However, the specificity of these different FGFR1 events for the various LGNET subtypes and accompanying genetic alterations are not well defined. Here we performed comprehensive genomic and epigenomic characterization on a diverse cohort of 30 LGNET with FGFR1 alterations. We identified that RGNT harbors a distinct epigenetic signature compared to other LGNET with FGFR1 alterations, and is uniquely characterized by FGFR1 kinase domain hotspot missense mutations in combination with either PIK3CA or PIK3R1 mutation, often with accompanying NF1 or PTPN11 mutation. In contrast, EVN harbors its own distinct epigenetic signature and is characterized by FGFR1-TACC1 fusion as the solitary pathogenic alteration. Additionally, DNT and pilocytic astrocytoma are characterized by either kinase domain tandem duplication or hotspot missense mutations, occasionally with accompanying NF1 or PTPN11 mutation, but lacking the accompanying PIK3CA or PIK3R1 mutation that characterizes RGNT. The glial component of LGNET with FGFR1 alterations typically has a predominantly oligodendroglial morphology, and many of the pilocytic astrocytomas with FGFR1 alterations lack the biphasic pattern, piloid processes, and Rosenthal fibers that characterize pilocytic astrocytomas with BRAF mutation or fusion. Together, this analysis improves the classification and histopathologic stratification of LGNET with FGFR1 alterations.
- Published
- 2020
- Full Text
- View/download PDF
44. TCGA mRNA Expression Analysis of the Heme Biosynthesis Pathway in Diffusely Infiltrating Gliomas: A Comparison of Typically 5-ALA Fluorescent and Non-Fluorescent Gliomas.
- Author
-
Mischkulnig M, Kiesel B, Lötsch D, Roetzer T, Borkovec M, Wadiura LI, Mercea PA, Jaklin FJ, Hervey-Jumper S, Roessler K, Berger MS, Widhalm G, and Erhart F
- Abstract
5-Aminolevulinic acid (5-ALA) is a fluorescent dye that after metabolization to Protoporphyrin IX (PpIX) by the heme biosynthesis pathway typically leads to visible fluorescence in WHO grade IV but not grade II gliomas. The exact mechanism for high PpIX levels in WHO grade IV gliomas and low PpIX levels in WHO grade II gliomas is not fully clarified. To detect relevant changes in mRNA expression, we performed an in-silico analysis of WHO grade II and IV glioma sequencing datasets provided by The Cancer Genome Atlas (TCGA) to investigate mRNA expression levels of relevant heme biosynthesis genes: Solute Carrier Family 15 Member 1 and 2 (SLC15A1 and SLC15A2), Aminolevulinate-Dehydratase (ALAD), Hydroxymethylbilane-Synthase (HMBS), Uroporphyrinogen-III-Synthase (UROS), Uroporphyrinogen-Decarboxylase (UROD), Coproporphyrinogen-Oxidase (CPOX), Protoporphyrinogen-Oxidase (PPOX), ATP-binding Cassette Subfamily B Member 6 (ABCB6)/G Member 2 (ABCG2) and Ferrochelatase (FECH). Altogether, 258 WHO grade II and 166 WHO grade IV samples were investigated. The mRNA expression levels showed significant differences in 8 of 11 examined genes between WHO grade II and IV gliomas. Significant differences in mRNA expression included increases of HMBS, UROD, FECH and PPOX as well as decreases of SLC15A2, ALAD, UROS and ABCB6 in WHO IV gliomas. Since the majority of changes was found in directions that might actually impair PpIX accumulation in WHO grade IV gliomas, additional studies are needed to analyze the corresponding factors of the heme biosynthesis also on protein level.
- Published
- 2020
- Full Text
- View/download PDF
45. Supracerebellar Approach for Resection of a Falcotentorial Dural Arteriovenous Fistula with Pial Tectal Arteriovenous Malformation Component Associated with a Left Parafalcine Meningioma.
- Author
-
Rubio RR, Li X, Orscelik A, Dubnicoff T, Raper D, Hervey-Jumper S, and Abla AA
- Subjects
- Aged, Central Nervous System Vascular Malformations complications, Central Nervous System Vascular Malformations diagnostic imaging, Cerebral Angiography, Cerebral Veins, Embolization, Therapeutic, Female, Humans, Incidental Findings, Intracranial Aneurysm complications, Intracranial Aneurysm diagnostic imaging, Intracranial Arteriovenous Malformations complications, Intracranial Arteriovenous Malformations diagnostic imaging, Meningeal Arteries, Meningeal Neoplasms complications, Meningioma complications, Tectum Mesencephali, Central Nervous System Vascular Malformations surgery, Intracranial Aneurysm surgery, Intracranial Arteriovenous Malformations surgery, Meningeal Neoplasms surgery, Meningioma surgery, Neurosurgical Procedures methods
- Abstract
A 66-year-old woman presented with a 4.5- × 4-cm left posterior parafalcine meningioma and visual loss in her left eye (Video 1). Prior to meningioma embolization, angiography confirmed an incidental high-risk falcotentorial dural arteriovenous fistula (DAVF) with pial tectal arteriovenous malformation (AVM) and flow-related aneurysms of the superior cerebellar artery (SCA) and posterior cerebral artery (PCA). Arterial supply to the AVM/DAVF consisted of branches of the middle meningeal artery, tentorial branches of the internal carotid arteries, and the PCA and SCA. Drainage into the vein of Galen (VG) and venous reflux into the precentral cerebellar vein (PCCV) were identified. The patient underwent transarterial embolization of the DAVF via the left middle meningeal artery using Onyx with a significant decrease of arterial venous shunting. A semi-sitting supracerebellar approach was performed. The subarachnoid space of the tentorium, cerebellar hemispheres, vermis, quadrigeminal, and ambient cisterns was dissected to reveal the boundaries of the lesion. Indocyanine green video angiography was done before and after in situ occlusion to identify the arterial supply and early venous drainage. The vascular lesion was disconnected circumferentially around the edges of the pial portion of the AVM, and the feeders were carefully cauterized and cut. The vessels on the surface of the brainstem were occluded in situ to prevent any parenchymal transgression. Finally, the drainage into the VG and the venous reflux to the PCCV were ligated. Postoperative angiography showed no residual DAVF or AVM and regression of aneurysms. The patient was discharged with no added deficits, and the meningioma was totally resected several months later., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
46. Influence of Corticosteroids and Antiepileptic Drugs on Visible 5-Aminolevulinic Acid Fluorescence in a Series of Initially Suspected Low-Grade Gliomas Including World Health Organization Grade II, III, and IV Gliomas.
- Author
-
Wadiura LI, Mischkulnig M, Hosmann A, Borkovec M, Kiesel B, Rötzer T, Mercea PA, Furtner J, Hervey-Jumper S, Rössler K, Berger MS, and Widhalm G
- Subjects
- Adrenal Cortex Hormones therapeutic use, Adult, Aged, Aminolevulinic Acid, Anticonvulsants therapeutic use, Brain Neoplasms complications, Brain Neoplasms metabolism, Female, Fluorescence, Glioma complications, Glioma metabolism, Humans, Male, Middle Aged, Neurosurgical Procedures, Retrospective Studies, Seizures metabolism, Young Adult, Adrenal Cortex Hormones pharmacology, Anticonvulsants pharmacology, Brain Neoplasms surgery, Glioma surgery, Seizures drug therapy
- Abstract
Objective: 5-aminolevulinic acid (5-ALA) has been increasingly used in recent years to identify anaplastic foci in primarily suspected low-grade gliomas (LGGs). However, 5-ALA fails to visualize a subgroup of focally anaplastic gliomas. Recently, 2 in vitro studies and 1 in vivo study assumed that antiepileptic drugs (AEDs) and dexamethasone have an influence on the 5-ALA metabolism/visible fluorescence in gliomas. The aim of this study was to analyze for the first time the influence of different AEDs and dexamethasone on visible 5-ALA fluorescence in a large cohort of suspected LGG., Methods: We retrospectively analyzed adult patients with resection of radiologically suspected diffusely infiltrating LGG after 5-ALA administration at 2 specialized centers. Clinical data on the intraoperative 5-ALA fluorescence status, preoperative treatment with AED/dexamethasone, and the total daily dose in cases of levetiracetam and dexamethasone intake were noted., Results: Altogether, 110 patients with suspected LGG were included. A significantly higher percentage of visible fluorescence was present in World Health Organization grade III/IV (73%) compared with World Health Organization grade II gliomas (11%; P < 0.001). In the multivariate analysis, we did not find an independent correlation between the visible fluorescence status and intake of dexamethasone/AED. Furthermore, the median daily dose of dexamethasone and levetiracetam did not differ significantly between fluorescing and nonfluorescing gliomas., Conclusions: In the largest series to date, we did not find a drug-related influence of either dexamethasone or different AED on visible 5-ALA fluorescence in suspected LGG. According to our preliminary data, preoperative treatment with these common drugs in neurosurgery can be performed safely before 5-ALA-assisted surgery of suspected LGG., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
47. Association of Maximal Extent of Resection of Contrast-Enhanced and Non-Contrast-Enhanced Tumor With Survival Within Molecular Subgroups of Patients With Newly Diagnosed Glioblastoma.
- Author
-
Molinaro AM, Hervey-Jumper S, Morshed RA, Young J, Han SJ, Chunduru P, Zhang Y, Phillips JJ, Shai A, Lafontaine M, Crane J, Chandra A, Flanigan P, Jahangiri A, Cioffi G, Ostrom Q, Anderson JE, Badve C, Barnholtz-Sloan J, Sloan AE, Erickson BJ, Decker PA, Kosel ML, LaChance D, Eckel-Passow J, Jenkins R, Villanueva-Meyer J, Rice T, Wrensch M, Wiencke JK, Oberheim Bush NA, Taylor J, Butowski N, Prados M, Clarke J, Chang S, Chang E, Aghi M, Theodosopoulos P, McDermott M, and Berger MS
- Subjects
- Adolescent, Adult, Aged, Antineoplastic Agents, Alkylating administration & dosage, Biomarkers, Tumor genetics, Child, Preschool, Cohort Studies, Contrast Media administration & dosage, DNA Methylation drug effects, Female, Glioblastoma genetics, Glioblastoma pathology, Humans, Isocitrate Dehydrogenase administration & dosage, Male, Middle Aged, Ohio epidemiology, Prognosis, Promoter Regions, Genetic drug effects, Retrospective Studies, Temozolomide administration & dosage, Glioblastoma drug therapy, Glioblastoma surgery, Isocitrate Dehydrogenase genetics
- Abstract
Importance: Per the World Health Organization 2016 integrative classification, newly diagnosed glioblastomas are separated into isocitrate dehydrogenase gene 1 or 2 (IDH)-wild-type and IDH-mutant subtypes, with median patient survival of 1.2 and 3.6 years, respectively. Although maximal resection of contrast-enhanced (CE) tumor is associated with longer survival, the prognostic importance of maximal resection within molecular subgroups and the potential importance of resection of non-contrast-enhanced (NCE) disease is poorly understood., Objective: To assess the association of resection of CE and NCE tumors in conjunction with molecular and clinical information to develop a new road map for cytoreductive surgery., Design, Setting, and Participants: This retrospective, multicenter cohort study included a development cohort from the University of California, San Francisco (761 patients diagnosed from January 1, 1997, through December 31, 2017, with 9.6 years of follow-up) and validation cohorts from the Mayo Clinic (107 patients diagnosed from January 1, 2004, through December 31, 2014, with 5.7 years of follow-up) and the Ohio Brain Tumor Study (99 patients with data collected from January 1, 2008, through December 31, 2011, with a median follow-up of 10.9 months). Image accessors were blinded to patient groupings. Eligible patients underwent surgical resection for newly diagnosed glioblastoma and had available survival, molecular, and clinical data and preoperative and postoperative magnetic resonance images. Data were analyzed from November 15, 2018, to March 15, 2019., Main Outcomes and Measures: Overall survival., Results: Among the 761 patients included in the development cohort (468 [61.5%] men; median age, 60 [interquartile range, 51.6-67.7] years), younger patients with IDH-wild-type tumors and aggressive resection of CE and NCE tumors had survival similar to that of patients with IDH-mutant tumors (median overall survival [OS], 37.3 [95% CI, 31.6-70.7] months). Younger patients with IDH-wild-type tumors and reduction of CE tumor but residual NCE tumors fared worse (median OS, 16.5 [95% CI, 14.7-18.3] months). Older patients with IDH-wild-type tumors benefited from reduction of CE tumor (median OS, 12.4 [95% CI, 11.4-14.0] months). The results were validated in the 2 external cohorts. The association between aggressive CE and NCE in patients with IDH-wild-type tumors was not attenuated by the methylation status of the promoter region of the DNA repair enzyme O6-methylguanine-DNA methyltransferase., Conclusions and Relevance: This study confirms an association between maximal resection of CE tumor and OS in patients with glioblastoma across all subgroups. In addition, maximal resection of NCE tumor was associated with longer OS in younger patients, regardless of IDH status, and among patients with IDH-wild-type glioblastoma regardless of the methylation status of the promoter region of the DNA repair enzyme O6-methylguanine-DNA methyltransferase. These conclusions may help reassess surgical strategies for individual patients with newly diagnosed glioblastoma.
- Published
- 2020
- Full Text
- View/download PDF
48. Electrical and synaptic integration of glioma into neural circuits.
- Author
-
Venkatesh HS, Morishita W, Geraghty AC, Silverbush D, Gillespie SM, Arzt M, Tam LT, Espenel C, Ponnuswami A, Ni L, Woo PJ, Taylor KR, Agarwal A, Regev A, Brang D, Vogel H, Hervey-Jumper S, Bergles DE, Suvà ML, Malenka RC, and Monje M
- Subjects
- Animals, Brain cytology, Cell Membrane pathology, Cell Proliferation, Gap Junctions pathology, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Heterografts, Humans, Mice, Mice, Inbred NOD, Neurons pathology, Optogenetics, Potassium metabolism, Synaptic Transmission, Tumor Cells, Cultured, Brain physiopathology, Electrical Synapses pathology, Electrophysiological Phenomena, Glioma physiopathology
- Abstract
High-grade gliomas are lethal brain cancers whose progression is robustly regulated by neuronal activity. Activity-regulated release of growth factors promotes glioma growth, but this alone is insufficient to explain the effect that neuronal activity exerts on glioma progression. Here we show that neuron and glioma interactions include electrochemical communication through bona fide AMPA receptor-dependent neuron-glioma synapses. Neuronal activity also evokes non-synaptic activity-dependent potassium currents that are amplified by gap junction-mediated tumour interconnections, forming an electrically coupled network. Depolarization of glioma membranes assessed by in vivo optogenetics promotes proliferation, whereas pharmacologically or genetically blocking electrochemical signalling inhibits the growth of glioma xenografts and extends mouse survival. Emphasizing the positive feedback mechanisms by which gliomas increase neuronal excitability and thus activity-regulated glioma growth, human intraoperative electrocorticography demonstrates increased cortical excitability in the glioma-infiltrated brain. Together, these findings indicate that synaptic and electrical integration into neural circuits promotes glioma progression.
- Published
- 2019
- Full Text
- View/download PDF
49. IDH1-R132H acts as a tumor suppressor in glioma via epigenetic up-regulation of the DNA damage response.
- Author
-
Núñez FJ, Mendez FM, Kadiyala P, Alghamri MS, Savelieff MG, Garcia-Fabiani MB, Haase S, Koschmann C, Calinescu AA, Kamran N, Saxena M, Patel R, Carney S, Guo MZ, Edwards M, Ljungman M, Qin T, Sartor MA, Tagett R, Venneti S, Brosnan-Cashman J, Meeker A, Gorbunova V, Zhao L, Kremer DM, Zhang L, Lyssiotis CA, Jones L, Herting CJ, Ross JL, Hambardzumyan D, Hervey-Jumper S, Figueroa ME, Lowenstein PR, and Castro MG
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins metabolism, Brain Neoplasms genetics, Brain Neoplasms pathology, Cell Differentiation, DNA Methylation genetics, DNA Repair genetics, Disease Models, Animal, Gene Ontology, Genome, Glioma pathology, Histones metabolism, Humans, Mice, Oligodendroglia pathology, Radiation Tolerance, Signal Transduction, Survival Analysis, DNA Damage genetics, Epigenesis, Genetic, Glioma genetics, Isocitrate Dehydrogenase genetics, Mutation genetics, Tumor Suppressor Proteins genetics, Up-Regulation genetics
- Abstract
Patients with glioma whose tumors carry a mutation in isocitrate dehydrogenase 1 (IDH1
R132H ) are younger at diagnosis and live longer. IDH1 mutations co-occur with other molecular lesions, such as 1p/19q codeletion, inactivating mutations in the tumor suppressor protein 53 (TP53 ) gene, and loss-of-function mutations in alpha thalassemia/mental retardation syndrome X-linked gene ( ATRX ). All adult low-grade gliomas (LGGs) harboring ATRX loss also express the IDH1R132H mutation. The current molecular classification of LGGs is based, partly, on the distribution of these mutations. We developed a genetically engineered mouse model harboring IDH1R132H , TP53 and ATRX inactivating mutations, and activated NRAS G12V. Previously, we established that ATRX deficiency, in the context of wild-type IDH1, induces genomic instability, impairs nonhomologous end-joining DNA repair, and increases sensitivity to DNA-damaging therapies. In this study, using our mouse model and primary patient-derived glioma cultures with IDH1 mutations, we investigated the function of IDH1R132H in the context of TP53 and ATRX loss. We discovered that IDH1R132H expression in the genetic context of ATRX and TP53 gene inactivation (i) increases median survival in the absence of treatment, (ii) enhances DNA damage response (DDR) via epigenetic up-regulation of the ataxia-telangiectasia-mutated (ATM) signaling pathway, and (iii) elicits tumor radioresistance. Accordingly, pharmacological inhibition of ATM or checkpoint kinases 1 and 2, essential kinases in the DDR, restored the tumors' radiosensitivity. Translation of these findings to patients with IDH1132H glioma harboring TP53 and ATRX loss could improve the therapeutic efficacy of radiotherapy and, consequently, patient survival., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2019
- Full Text
- View/download PDF
50. Clinical Factors Associated With ICU-Specific Care Following Supratentoral Brain Tumor Resection and Validation of a Risk Prediction Score.
- Author
-
Franko LR, Hollon T, Linzey J, Roark C, Rajajee V, Sheehan K, Teig M, Hervey-Jumper S, Heth J, Orringer D, and Williamson CA
- Subjects
- Adult, Aged, Female, Glasgow Coma Scale, Humans, Karnofsky Performance Status, Length of Stay, Male, Middle Aged, Patient Acuity, Postoperative Complications epidemiology, Retrospective Studies, Risk Factors, Craniotomy statistics & numerical data, Intensive Care Units statistics & numerical data, Supratentorial Neoplasms surgery
- Abstract
Objectives: The postoperative management of patients who undergo brain tumor resection frequently occurs in an ICU. However, the routine admission of all patients to an ICU following surgery is controversial. This study seeks to identify the frequency with which patients undergoing elective supratentorial tumor resection require care, aside from frequent neurologic checks, that is specific to an ICU and to determine the frequency of new complications during ICU admission. Additionally, clinical predictors of ICU-specific care are identified, and a scoring system to discriminate patients most likely to require ICU-specific treatment is validated., Design: Retrospective observational cohort study., Setting: Academic neurosurgical center., Patients: Two-hundred consecutive adult patients who underwent supratentorial brain tumor surgery. An additional 100 consecutive patients were used to validate the prediction score., Interventions: None., Measurements and Main Results: Univariate statistics and multivariable logistic regression were used to identify clinical characteristics associated with ICU-specific treatment. Eighteen patients (9%) received ICU-specific care, and 19 (9.5%) experienced new complications or underwent emergent imaging while in the ICU. Factors significantly associated with ICU-specific care included nonelective admission, preoperative Glasgow Coma Scale, and volume of IV fluids. A simple clinical scoring system that included Karnofsky Performance Status less than 70 (1 point), general endotracheal anesthesia (1 point), and any early postoperative complications (2 points) demonstrated excellent ability to discriminate patients who required ICU-specific care in both the derivation and validation cohorts., Conclusions: Less than 10% of patients required ICU-specific care following supratentorial tumor resection. A simple clinical scoring system may aid clinicians in stratifying the risk of requiring ICU care and could inform triage decisions when ICU bed availability is limited.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.