Imaging of glioblastoma recurrence

Author(s):  
Scott E. Forseen ◽  
Bruce C. Gilbert ◽  
Ramon E. Figueroa
2018 ◽  
Vol 20 (suppl_6) ◽  
pp. vi188-vi188
Author(s):  
Marwa Ismail ◽  
Virginia Hill ◽  
Volodymyr Statsevych ◽  
Raymond Huang ◽  
Ramon Correa ◽  
...  

2020 ◽  
pp. clincanres.3580.2020
Author(s):  
Andreas Stadlbauer ◽  
Thomas M. Kinfe ◽  
Ilker Eyüpoglu ◽  
Max Zimmermann ◽  
Melitta Kitzwoegerer ◽  
...  

2017 ◽  
Vol 19 (suppl_6) ◽  
pp. vi177-vi177
Author(s):  
Guangrong Lu ◽  
Ping Zhu ◽  
Rong Shao ◽  
Jay-Jiguang Zhu

2019 ◽  
Vol 68 (1) ◽  
pp. 33-57 ◽  
Author(s):  
Vashendriya V.V. Hira ◽  
Barbara Breznik ◽  
Miloš Vittori ◽  
Annique Loncq de Jong ◽  
Jernej Mlakar ◽  
...  

Glioblastoma is the most aggressive primary brain tumor. Slowly dividing and therapy-resistant glioblastoma stem cells (GSCs) reside in protective peri-arteriolar niches and are held responsible for glioblastoma recurrence. Recently, we showed similarities between GSC niches and hematopoietic stem cell (HSC) niches in bone marrow. Acute myeloid leukemia (AML) cells hijack HSC niches and are transformed into therapy-resistant leukemic stem cells (LSCs). Current clinical trials are focussed on removal of LSCs out of HSC niches to differentiate and to become sensitized to chemotherapy. In the present study, we elaborated further on these similarities by immunohistochemical analyses of 17 biomarkers in paraffin sections of human glioblastoma and human bone marrow. We found all 17 biomarkers to be expressed both in hypoxic peri-arteriolar HSC niches in bone marrow and hypoxic peri-arteriolar GSC niches in glioblastoma. Our findings implicate that GSC niches are being formed in glioblastoma as a copy of HSC niches in bone marrow. These similarities between HSC niches and GSC niches provide a theoretic basis for the development of novel strategies to force GSCs out of their niches, in a similar manner as in AML, to induce GSC differentiation and proliferation to render them more sensitive to anti-glioblastoma therapies.


2020 ◽  
Vol 146 (11) ◽  
pp. 2885-2896
Author(s):  
Zoltan Kraboth ◽  
Bence Galik ◽  
Marton Tompa ◽  
Bela Kajtar ◽  
Peter Urban ◽  
...  

Abstract Purpose Glioblastoma is the most aggressive form of brain tumors. A better understanding of the molecular mechanisms leading to its evolution is essential for the development of treatments more effective than the available modalities. Here, we aim to identify molecular drivers of glioblastoma development and recurrence by analyzing DNA CpG methylation patterns in sequential samples. Methods DNA was isolated from 22 pairs of primary and recurrent formalin-fixed, paraffin-embedded glioblastoma specimens, and subjected to reduced representation bisulfite sequencing. Bioinformatic analyses were conducted to identify differentially methylated sites and pathways, and biostatistics was used to test correlations among clinical and pathological parameters. Results Differentially methylated pathways likely involved in primary tumor development included those of neuronal differentiation, myelination, metabolic processes, synapse organization and endothelial cell proliferation, while pathways differentially active during glioblastoma recurrence involved those associated with cell processes and differentiation, immune response, Wnt regulation and catecholamine secretion and transport. Conclusion DNA CpG methylation analyses in sequential clinical specimens revealed hypomethylation in certain pathways such as neuronal tissue development and angiogenesis likely involved in early tumor development and growth, while suggested altered regulation in catecholamine secretion and transport, Wnt expression and immune response contributing to glioblastoma recurrence. These pathways merit further investigations and may represent novel therapeutic targets.


Cancers ◽  
2020 ◽  
Vol 12 (2) ◽  
pp. 520 ◽  
Author(s):  
Ella L. Kim ◽  
Maxim Sorokin ◽  
Sven Rainer Kantelhardt ◽  
Darius Kalasauskas ◽  
Bettina Sprang ◽  
...  

Background: Inevitable recurrence after radiochemotherapy is the major problem in the treatment of glioblastoma, the most prevalent type of adult brain malignancy. Glioblastomas are notorious for a high degree of intratumor heterogeneity manifest through a diversity of cell types and molecular patterns. The current paradigm of understanding glioblastoma recurrence is that cytotoxic therapy fails to target effectively glioma stem cells. Recent advances indicate that therapy-driven molecular evolution is a fundamental trait associated with glioblastoma recurrence. There is a growing body of evidence indicating that intratumor heterogeneity, longitudinal changes in molecular biomarkers and specific impacts of glioma stem cells need to be taken into consideration in order to increase the accuracy of molecular diagnostics still relying on readouts obtained from a single tumor specimen. Methods: This study integrates a multisampling strategy, longitudinal approach and complementary transcriptomic investigations in order to identify transcriptomic traits of recurrent glioblastoma in whole-tissue specimens of glioblastoma or glioblastoma stem cells. In this study, 128 tissue samples of 44 tumors including 23 first diagnosed, 19 recurrent and 2 secondary recurrent glioblastomas were analyzed along with 27 primary cultures of glioblastoma stem cells by RNA sequencing. A novel algorithm was used to quantify longitudinal changes in pathway activities and model efficacy of anti-cancer drugs based on gene expression data. Results: Our study reveals that intratumor heterogeneity of gene expression patterns is a fundamental characteristic of not only newly diagnosed but also recurrent glioblastomas. Evidence is provided that glioblastoma stem cells recapitulate intratumor heterogeneity, longitudinal transcriptomic changes and drug sensitivity patterns associated with the state of recurrence. Conclusions: Our results provide a transcriptional rationale for the lack of significant therapeutic benefit from temozolomide in patients with recurrent glioblastoma. Our findings imply that the spectrum of potentially effective drugs is likely to differ between newly diagnosed and recurrent glioblastomas and underscore the merits of glioblastoma stem cells as prognostic models for identifying alternative drugs and predicting drug response in recurrent glioblastoma. With the majority of recurrent glioblastomas being inoperable, glioblastoma stem cell models provide the means of compensating for the limited availability of recurrent glioblastoma specimens.


2009 ◽  
Vol 6 (11) ◽  
pp. 648-657 ◽  
Author(s):  
Isaac Yang ◽  
Manish K. Aghi

2016 ◽  
Vol 18 (suppl_6) ◽  
pp. vi137-vi138
Author(s):  
James Battiste ◽  
Suriya Jeyapalan ◽  
Edward Pan ◽  
Josie Sewell ◽  
Denise Damek ◽  
...  

2021 ◽  
Author(s):  
Claudia Garcia-Diaz ◽  
Elisabetta Mereu ◽  
Melanie P Clements ◽  
Anni Pöysti ◽  
Felipe Galvez-Cancino ◽  
...  

Glioblastoma recurrence originates from invasive cells at the tumour margin that escape surgical debulking, but their biology remains poorly understood. Here we generated three somatic mouse models recapitulating the main glioblastoma driver mutations to characterise margin cells. We find that, regardless of genetics, tumours converge on a common set of neural-like cellular states. However, bulk and margin display distinct neurogenic patterns and immune microenvironments. The margin is immune-cold and preferentially follows developmental-like trajectories to produce astrocyte-like cells. In contrast, injury-like programmes dominate in the bulk, are associated with immune infiltration and generate lowly-proliferative injured neural progenitor-like (iNPCs) cells. In vivo label-retention approaches further demonstrate that iNPCs account for a significant proportion of dormant glioblastoma cells and are induced by interferon signalling within T-cell niches. These findings indicate that tumour region is a major determinant of glioblastoma cell fate and therapeutic vulnerabilities identified in bulk may not extend to the margin residuum.


2021 ◽  
Author(s):  
Galina Pavlova ◽  
Varvara Kolesnikova ◽  
Nadezhda Samoylenkova ◽  
Sergey Drozd ◽  
Alexander Revishchin ◽  
...  

Abstract Cancer cell reprogramming based on aptamers with antiproliferative properties in combination with small molecules that are used for conversion iPSCs into neurons represents a new approach to reduce the probability of glioblastoma recurrence and tumor resistance to therapy. In this research we tested several combinations of factors on whole cell cultures, derived from tumor tissue after surgical resection, and on cell cultures divided in CD133 enriched and depleted populations, as CD133 marker is believed to be characteristic for glioblastoma stem cells. We showed that CD133+ and CD133- cells have a different response to tested combinations of factors and CD133-positive cells are more stable and possess stemness properties. Thus, affecting these cells will lead to decrease of therapy resistance. Moreover, we found a combination of factors that is able to inhibit proliferation of both CD133+ and CD133- cells. Our results reveal a promising strategy to improve treatment of patients with glioblastoma.


Sign in / Sign up

Export Citation Format

Share Document