Disease Biology

Literature Summaries

This page contains summaries of research papers reviewed and discussed during the course.

New insights into the genetics of glioblastoma multiforme by familial exome sequencing


Glioblastomas are some of the most difficult tumors to treat and detect, due to both their genetic heterogeneity and the lack of models to determine human specific mutations that lead to the rise of this tumor. In this specific case study of a family, both daughter and son, ages 12 and 10 respectively, developed glioblastomas while the parents were unaffected. Based on the family history, it could be deduced that the parents were carriers of a mutation or certain group of mutations while the children were homozygous as well as heterozygous for these mutations that led to the growth of GBM. Through exome sequencing of leukocyte DNA samples, and Genome Analysis Toolkit and ANNOVAR analyses to ensure the correct nucleotide sequences corresponded to correctly between parents and children, the paper was able to determine 85 single-nucleotide variations that were homozygous in the children and heterozygous in the parents. Determining which genes were the most mutated, the authors were able to narrow down the 73 genes that had SNV mutations to focus on only 10 (MYEOV, AKAP1, F5, OVCH1, CR1, LAMA1, RAI1, PTPRB, CROCC and PSG5). Through KEGG pathway analysis, it was determined that these genes impacted ECM to receptor interaction, focal adhesions, and complement and coagulation cascade sequences. Utilizing STRING, as another level of pathway analysis, the ERG/EGFR signaling pathway appeared to have close proximity to many of the mutated genes in the children. Essentially, by genome sequencing, the authors were better able to understand potential, specific sources of mutations that could lead to the growth of pediatric glioblastomas through this family case study.

Backes, Christina et al. “New insights into the genetics of glioblastoma multiforme by familial exome sequencing.” Oncotarget vol. 6,8 (2015): 5918-31. doi:10.18632/oncotarget.2950


A functional artificial neural network for noninvasive pretreatment evaluation of glioblastoma patients.


Although pretreatment assessments for glioblastomas do exist, they typically involve intracranial biopsies which pose great risk to damaging brain tissue in order to get access to the tumor. However, it is still vital to classify and determine varying aspects of tumors, like genetic sequences, to develop an effective treatment plan. Because of the dangers of intracranial biopsies, especially for the elderly, the development of an Artificial Neural Network system (ANN) becomes a valuable tool that can help with treatment plan development for GBMs, as this paper follows. CRL2 degrades proteins such as HIF1-a, which is important in GBM vascularization, and EGFR, involved in GBM growth and proliferation. The paper suggests that copy number variations of CUL2, which is a gene for a scaffold protein in CRL2 complex, can be used to determine patient survival rates and overall survival rates. Additionally, CUL2 CNVs are associated with non-G-CIMP, IDH1 wildtype GBMs, where IDH mutations is a common biomarker for GBMs development. The developed ANN is capable of processing T1 MRI scans, segmenting and calculating individual tumor SvV, determining CUL2 CNV in extracranial tissue (that is reflective of expression in CNS malignant tissue).

Zander, Eric et al. “A functional artificial neural network for noninvasive pretreatment evaluation of glioblastoma patients.” Neuro-oncology advances vol. 4,1 vdab167. 18 Nov. 2021, doi:10.1093/noajnl/vdab167


Analyzing magnetic resonance imaging data from glioma patients using deep learning


Imaging has become a point of interest in predicting clinical variables, such as tumor characteristics, treatment response, and prognosis. Glioma imaging currently consists of a standardized measure established by the World Health Organization (WHO). However, further imaging techniques are suggested to be used by the authors of this paper, due to the increased knowledge that would be gathered. For example, perfusion weighted imaging, which detects fluid and blood flow, could be used to detect pseudo-progression (through detection of fluid buildup in the tumor and inflammation, due to immune responses, that appears as progression without this imaging technique. Due to the potential of imaging data in predicting important aspects of GBM tumors, the BraTS challenge, which compares different brain tumor segmentation algorithms, allows more development of effective deep-learning imaging algorithms to potentially use for clinical practice in detecting tumor boundaries and characteristics. Essentially, with the advancement of non-invasive methods of GBM analysis and detection, more opportunities become available for glioblastoma treatment and research exploration.

Menze, Bjoern et al. “Analyzing magnetic resonance imaging data from glioma patients using deep learning.” Computerized medical imaging and graphics : the official journal of the Computerized Medical Imaging Society vol. 88 (2021): 101828. doi:10.1016/j.compmedimag.2020.101828


Imaging signatures of glioblastoma molecular characteristics: A radiogenomics review


With the emergence of radiogenomics, the study of genetic factors through imaging techniques, new methods of detecting genetic mutations in glioblastomas became accessible. Previously completed studies determined that neuroimaging techniques could differentiate between tumor-core, tumor-infiltrated tissue, necrotic core, vascularization, and metabolite factors. By correlating certain mutations to percentage of tissue types as well as location, it was then possible to determine driver mutations in certain GBMs, such as IDH which presented in the left frontal lobe with clear tumor margins and cysts with low T1, suppressed T2-FLAIR signal intensity (among other specific genetic mutation characteristics). Being able to detect mutations just from neuroimaging techniques implies a non-invasive method of determining potentially targetable mutations in GBM tumors before excessive growth occurs. As a result, radiogenomics for glioblastoma tumors becomes a new frontier that is valuable to explore.

Fathi Kazerooni, Anahita et al. “Imaging signatures of glioblastoma molecular characteristics: A radiogenomics review.” Journal of magnetic resonance imaging : JMRI vol. 52,1 (2020): 54-69. doi:10.1002/jmri.26907


Advances in the Knowledge of the Molecular Biology of Glioblastoma and Its Impact in Patient Diagnosis, Stratification, and Treatment


Glioblastomas are complex tumors with extreme heterogeneity between tumors and within the tumor itself. As a result, a classification system becomes important to understand general targets for treatment, rather than a focus on individual, extremely specific treatment methods. The overarching subgroups are primary and secondary GBMs, which are based on the populations they occur in (age groups) and rate of origination (“de novo” or derived from previous tumors). One such mutation in IDH, the most common and a biomarker for secondary glioblastomas. IDH is a metabolic gene that catalyzes isocitrate to alpha-ketoglutarate (α-KG) and carbon dioxide. IDH1 mutant type catalyzes D-2HG formation, which increases in concentration and, among other negative effects on transcription factors, inhibits EGLN from hydrolyzing HIF1a for ubiquitination. Treatment methods can also utilize immunotherapies and nanoparticles that can bypass the enhanced permeable vessels around glioblastomas to carry treatment drugs. The vast array of potential routes to target GBMs is derived from the continuous goal of classifying the tumor to increase the number of more specific and general categories for this tumor, and thus increasing the number of areas of interest to target.

Delgado-Martín, Belén, and Miguel Ángel Medina. “Advances in the Knowledge of the Molecular Biology of Glioblastoma and Its Impact in Patient Diagnosis, Stratification, and Treatment.” Advanced science (Weinheim, Baden-Wurttemberg, Germany) vol. 7,9 1902971. 12 Mar. 2020, doi:10.1002/advs.201902971


Experimental models and tools to tackle glioblastoma


In order to research and better understand GBMs, different models need to be considered for the most accurate results. While considering such models, researchers must take into account the complexity of glioblastomas: high genetic heterogeneity, multiple driver mutations affecting multiple signaling pathways, spread through the brain parenchyma, and have diverse microenvironments. The current models that exist are GBM cell lines, in vitro engineering tumor-initiating cells, tumor slices, in vivo mouse transplanted tumor-initiating cells, and genetically engineering mice (GEMM), all of which can be produced through selective breeding (GEMM), gene editing, or viral injections that initiate oncogenic expression such as lentivirus or retrovirus. The in vitro models tend to focus on a reductionist method which focuses on one aspect of GBMs, but this method becomes ineffective since it does not reflect a human model. In vivo orthotopic xenografts and GEMM are one of the more efficient models as they allow for the genetic complexity of tumors to develop and take into account the interactions between tumor, its environment, and organisms’ immune system. However, because they are of murine models, the differences in immune system and brain environment result in different outcomes when applying the same treatment to humans, and thus this difference adds another factor to consider during GBM modeling.

Robertson, Faye L et al. “Experimental models and tools to tackle glioblastoma.” Disease models & mechanisms vol. 12,9 dmm040386. 6 Sep. 2019, doi:10.1242/dmm.040386


Advances in the management of glioblastoma


The standard for glioblastoma therapy involves gross total resection where possible, chemoradiotherapy with temozolomide (TMZ) and lomustine, as well as fractionated radiotherapy. Additionally, pre-treatment analysis utilizes various imaging tools such as DWI and MR spectroscopy to detect various features of glioblastomas that could provide increased evidence to guide treatment. Available surgical tools include using fluorescent markers to differentiate between GBM tissue and brain tissue, awake surgery to ensure resection does not inhibit functions, and Raman spectroscopy which can detect differences between IDH mutant and wildtype tumor tissue. Immunotherapy treatment methods utilize genetically engineered cells, vaccines with tumor specific antigens, that can help the immune system detect tumors through surface-cell markers.

Ma R, Taphoorn MJB, Plaha P, “Advances in the management of glioblastoma.” Journal of Neurology, Neurosurgery & Psychiatry, 2021; 92:1103-1111.


Immunotherapy For Glioblastoma: A Path Forward


Glioblastomas have low mutagenesis, are immunosuppressive, and have a low neoantigen load. Additionally, there is a low concentration of T-Cells, especially CD8 T-Cells which are most effective against tumors. As a result of these factors, immunotherapy becomes difficult to implement as treatment for GBMs, yet it is still highly researched because overcoming these factors could mean an efficient treatment against glioblastomas. Some immunology treatment methods include immune checkpoint inhibitors and tumor vaccines. A combination of the 2 might help to increase immune cell count and response to GBMs. Another method is novel cancer vaccines in which sample cancer cells are treated with oligodeoxynucleotide and allow different protein markers to be accessible for recognition by T-cells. As a result, the T-Cells can then target the GBM tumor based on the different protein receptors and markers recognized from the cancer vaccine.

Stricklin, Leah. “Immunotherapy For Glioblastoma: A Path Forward.” Cell & Gene, 10 Aug. 2020, https://www.cellandgene.com/doc/immunotherapy-for-glioblastoma-a-path-forward-0001.


Immunotherapy for Glioblastoma: Current Progress and Challenges


The environment in the brain was originally thought to be immune-privileged, however lymphatic vessels (lining the dural sinuses) that drain to cervical lymph nodes suggest access for immune cells to reach the brain. Additionally, due to tumorigenesis and excessive mutations, GBM causes BBB integrity loss surrounding the tumor, which makes it easier to pass drugs through the blood brain barrier to the tumor site. However, this also means access for macrophages, which promote tumor growth, and this loss of selective permeability is variable throughout the GBM. However, this presents opportunities for immunotherapy responses to be applied to glioblastomas. CTL4 and PD-1 are immune system checkpoint molecules expressed by GBMs, thus preventing immune system responses against the tumor. Anti-CTLA-4, anti-PD-1 and an IL-12 expressing oncolytic virus counteract the effects of CTL4 and PD-1, and have a 89% cure rate in mouse models. Because tumor-associated macrophages (TAM) benefit GBMs, research has focused on TAM-targeted therapies as well to inhibit macrophage activity in the tumor environment. CAR-T therapies, which engineer patients’ T-cells to express cancer targeting CARS, are being explored to target EGFRvIII, IL13Ra2, and HER2 mutant glioblastomas. Essentially, current immunotherapy responses work to create an ideal immunogenic environment and produce drugs capable of helping the immune system target GBMs.

Yu, Miranda W, and Daniela F Quail. “Immunotherapy for Glioblastoma: Current Progress and Challenges.” Frontiers in immunology vol. 12 676301. 13 May. 2021, doi:10.3389/fimmu.2021.676301


Functional connectivity within glioblastoma impacts overall survival


Evidence has suggested that glioblastomas, due to their heterogeneity, still contain functioning neural tissue. This study focused on strengthening this claim as well as attempting to correlate overall survival with functional connectivity in glioblastomas. The study used predefined functional connectivity regions of interest in primary GBM patients to analyze the presence of functional connectivity within the tumor. Utilizing resting state fMRI scans, researchers found that functional activity was present in GBM tumors and at varying levels between participants. Furthermore, a correlation was established between high functional connectivity and greater survival. This connection can be attributed to the fact that more within-tumor functional connectivity implies the presence of normal, functional tissue. This suggests a decreased amount of extremely mutated neuronal tissue that would not retain its function (and potentially even be more proliferative). As a result, utilizing fMRI scans to analyze functional connectivity within GBM tumors could be extremely helpful in determining which tumors have a lower proliferation and mutagenesis rate, and thus imply higher overall survival.

Daniel, Andy G S et al. “Functional connectivity within glioblastoma impacts overall survival.” Neuro-oncology vol. 23,3 (2021): 412-421. doi:10.1093/neuonc/noaa189