A Model System for Proliferation and Characterisation of Stem Cells in Childhood T-ALL.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1370-1370
Author(s):  
Charlotte V. Cox ◽  
Roger S. Evely ◽  
Allison Blair

Abstract T cell acute lymphoblastic leukaemias (T-ALL) are highly aggressive malignancies representing 10–15% of paediatric and 25% adult ALLs. T-ALL was considered to arise as a consequence of clonal expansion of early thymocytes. However, progress towards increasing our understanding of the biology of this disease has been hampered by lack of appropriate culture systems to study primary cells and use of murine model systems that often do not accurately reflect human disease. Traditional xenograft models of leukaemia have involved implantation of malignant cells or immortalised leukaemic cell lines with either intraperitoneal or subcutaneous localisation of leukaemia. These models do not mimic the normal pathophysiology of the disease and may therefore provide misleading data. Since evaluation of new agents in paediatric malignancies is limited by the small number of children eligible for clinical trails, there is a need for a predictive preclinical model of paediatric ALL. We have previously used a long-term suspension culture system to evaluate proliferation of T-ALL cells in vitro and demonstrated these cells had a CD34+/CD4−, CD7− phenotype. T-ALL cells with this phenotype were also capable of engrafting NOD/SCID mice, suggesting the disease may arise in a more primitive cell. In this study we have attempted to further characterise T-ALL cells with long-term proliferative ability in vivo and to investigate the kinetics of engraftment. Unsorted cells and cells sorted for the expression of CD133 and CD7 from 5 T-ALL patients were inoculated into sublethally irradiated NOD/SCID mice. Peripheral blood samples were taken at weekly intervals from the lateral tail vein from two weeks post inoculation onwards. BM samples were analysed from 4 weeks post inoculation and all animals were sacrificed no later than 10 weeks post inoculation. Human CD45+ cells were first detected at day 17-post inoculation (1.54–3.8% CD45+). By week 4, this had increased to 4.4–21% CD45+ in PB samples, while levels in BM aspirates were significantly higher at this stage (24–47% CD45+). This pattern of tissue dissemination closely mimics that observed in the patients. The levels of CD45+ cells continued to rise with time and had exceeded 85% in the BM of animals injected with cells from 3 patients by week 7-post inoculation. FISH and flow cytometric analyses showed the engrafted cells had a similar karyotype and phenotype to the patient at diagnosis and there was no evidence of myeloid cell engraftment. Cells harvested from these animals have been used in secondary, tertiary and quaternary transplants with no loss of NOD/SCID repopulating potential and similar engraftment kinetics. Quinary transplants are currently underway. In the sorted cell populations, only the CD133+/CD7− subfraction was capable of engrafting, 0.5–54% CD45+, with as few as 1.4x103–5x103 cells. There was no engraftment with the other subfractions despite injecting 10 to 1000-fold more cells. These engrafted cells expressed high levels of CD34, CD2, CD4 and CD7 and very low levels of CD133. This phenotype was similar to that of the patients at diagnosis, implying they had differentiated in vivo. These data add to the evidence that T-ALL may arise in a cell with a more primitive phenotype, rather than committed thymocytes. These cells may be the most relevant targets for emerging therapeutic strategies and we describe a robust, reproducible in vivo leukaemia model which could be used to investigate the efficacy of novel agents for the treatment of paediatric T-ALL.

Blood ◽  
1998 ◽  
Vol 92 (11) ◽  
pp. 4325-4335 ◽  
Author(s):  
A. Blair ◽  
D.E. Hogge ◽  
H.J. Sutherland

Acute myeloid leukemia (AML) occurs as the result of malignant transformation in a hematopoietic progenitor cell, which proliferates to form an accumulation of AML blasts. Only a minority of these AML cells are capable of proliferation in vitro, suggesting that AML cells may be organized in a hierarchy, with only the most primitive of these cells capable of maintaining the leukemic clone. To further investigate this hypothesis, we have evaluated a strategy for purifying these primitive cells based on surface antigen expression. As an in vitro endpoint, we have determined the phenotype of AML progenitor cells which are capable of producing AML colony-forming cells (CFU) for up to 8 weeks in suspension culture (SC) and compared the phenotype with that of cells which reproduce AML in nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice. AML cells were fluorescence-activated cell sorted (FACS) for coexpression of CD34 and CD71, CD38, and/or HLA-DR and the subfractions were assayed in vitro and in vivo at various cell doses to estimate purification. While the majority of primary AML CFU lacked expression of CD34, most cells capable of producing CFU after 2 to 8 weeks in SC were CD34+/CD71−. HLA-DR expression was heterogeneous on cells producing CFU after 2 to 4 weeks. However, after 6 to 8 weeks in SC, the majority of CFU were derived from CD34+/HLA-DR− cells. Similarly, the majority of cells capable of long-term CFU production from SC were CD34+/CD38−. Most cells that were capable of engrafting NOD/SCID mice were also CD34+/CD71− and CD34+/HLA-DR−. Engraftment was not achieved with CD34+/CD71+ or HLA-DR+subfractions, however, in two patients, both the CD34+and CD34− subfractions were capable of engrafting the NOD/SCID mice. A three-color sorting strategy combining these antigens allowed approximately a 2-log purification of these NOD/SCID leukemia initiating cells, with engraftment achieved using as few as 400 cells in one experiment. Phenotyping studies suggest even higher purification could be achieved by combining lack of CD38 expression with the CD34+/CD71− or CD34+/HLA DR− phenotype. These results suggest that most AML cells capable of long-term proliferation in vitro and in vivo share the CD34+/CD71−/HLA-DR− phenotype with normal stem cells. Our data suggests that in this group of patients the leukemic transformation has occurred in a primitive progenitor, as defined by phenotype, with some degree of subsequent differentiation as defined by functional assays.


2021 ◽  
Vol 13 ◽  
Author(s):  
Shen-Qing Zhang ◽  
Long-Long Cao ◽  
Yun-Yue Liang ◽  
Pu Wang

Clinical studies have found that some Alzheimer’s disease (AD) patients suffer from Cushing’s syndrome (CS). CS is caused by the long-term release of excess glucocorticoids (GCs) from the adrenal gland, which in turn, impair brain function and induce dementia. Thus, we investigated the mechanism of the effect of corticosterone (CORT) on the development and progression of AD in a preclinical model. Specifically, the plasma CORT levels of 9-month-old APP/PS1 Tg mice were abnormally increased, suggesting an association between GCs and AD. Long-term administration of CORT accelerated cognitive dysfunction by increasing the production and deposition of β-amyloid (Aβ). The mechanism of action of CORT treatment involved stimulation of the expression of BACE-1 and presenilin (PS) 1 in in vitro and in vivo. This observation was confirmed in mice with adrenalectomy (ADX), which had lower levels of GCs. Moreover, the glucocorticoid receptor (GR) mediated the effects of CORT on the stimulation of the expression of BACE-1 and PS1 via the PKA and CREB pathways in neuroblastoma N2a cells. In addition to these mechanisms, CORT can induce a cognitive decline in APP/PS1 Tg mice by inducing apoptosis and decreasing the differentiation of neurons.


Blood ◽  
1998 ◽  
Vol 92 (11) ◽  
pp. 4325-4335 ◽  
Author(s):  
A. Blair ◽  
D.E. Hogge ◽  
H.J. Sutherland

Abstract Acute myeloid leukemia (AML) occurs as the result of malignant transformation in a hematopoietic progenitor cell, which proliferates to form an accumulation of AML blasts. Only a minority of these AML cells are capable of proliferation in vitro, suggesting that AML cells may be organized in a hierarchy, with only the most primitive of these cells capable of maintaining the leukemic clone. To further investigate this hypothesis, we have evaluated a strategy for purifying these primitive cells based on surface antigen expression. As an in vitro endpoint, we have determined the phenotype of AML progenitor cells which are capable of producing AML colony-forming cells (CFU) for up to 8 weeks in suspension culture (SC) and compared the phenotype with that of cells which reproduce AML in nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice. AML cells were fluorescence-activated cell sorted (FACS) for coexpression of CD34 and CD71, CD38, and/or HLA-DR and the subfractions were assayed in vitro and in vivo at various cell doses to estimate purification. While the majority of primary AML CFU lacked expression of CD34, most cells capable of producing CFU after 2 to 8 weeks in SC were CD34+/CD71−. HLA-DR expression was heterogeneous on cells producing CFU after 2 to 4 weeks. However, after 6 to 8 weeks in SC, the majority of CFU were derived from CD34+/HLA-DR− cells. Similarly, the majority of cells capable of long-term CFU production from SC were CD34+/CD38−. Most cells that were capable of engrafting NOD/SCID mice were also CD34+/CD71− and CD34+/HLA-DR−. Engraftment was not achieved with CD34+/CD71+ or HLA-DR+subfractions, however, in two patients, both the CD34+and CD34− subfractions were capable of engrafting the NOD/SCID mice. A three-color sorting strategy combining these antigens allowed approximately a 2-log purification of these NOD/SCID leukemia initiating cells, with engraftment achieved using as few as 400 cells in one experiment. Phenotyping studies suggest even higher purification could be achieved by combining lack of CD38 expression with the CD34+/CD71− or CD34+/HLA DR− phenotype. These results suggest that most AML cells capable of long-term proliferation in vitro and in vivo share the CD34+/CD71−/HLA-DR− phenotype with normal stem cells. Our data suggests that in this group of patients the leukemic transformation has occurred in a primitive progenitor, as defined by phenotype, with some degree of subsequent differentiation as defined by functional assays.


2021 ◽  
Vol 12 (3) ◽  
Author(s):  
Haoyu Li ◽  
Qing Liu ◽  
Zihua Chen ◽  
Ming Wu ◽  
Chao Zhang ◽  
...  

AbstractTemozolomide (TMZ) is the internationally recognized and preferred drug for glioma chemotherapy treatment. However, TMZ resistance in glioma appears after long-term use and is an urgent problem that needs to be solved. Circular RNAs (circRNAs) are noncoding RNAs and play an important role in the pathogenesis and progression of tumors. Hsa_circ_0110757 was identified in TMZ-resistant glioma cells by high-throughput sequencing analysis and was derived from reverse splicing of myeloid cell leukemia-1 (Mcl-1) exons. The role of hsa_circ_0110757 in TMZ-resistant glioma was evaluated both in vitro and in vivo. It was found that hsa_circ_0110757 and ITGA1 are more highly expressed in TMZ-resistant glioma than in TMZ-sensitive glioma. The overexpression of hsa_circ_0110757 in glioma patients treated with TMZ was obviously associated with tumor invasion. This study indicates that hsa_circ_0110757 inhibits glioma cell apoptosis by sponging hsa-miR-1298-5p to promote ITGA1 expression. Thus, hsa_circ_0110757/hsa-miR-1298-5p/ITGA could be a potential therapeutic target for reversing the resistance of glioma to TMZ.


Blood ◽  
1997 ◽  
Vol 89 (9) ◽  
pp. 3104-3112 ◽  
Author(s):  
A. Blair ◽  
D.E. Hogge ◽  
L.E. Ailles ◽  
P.M. Lansdorp ◽  
H.J. Sutherland

Abstract Acute myeloid leukaemia (AML) is thought to be maintained by a small population of leukemic progenitor cells. To define the phenotype of such cells with long-term proliferative capacity in vitro and in vivo, we have used the production of leukemic clonogenic cells (CFU) after 2 to 8 weeks in suspension culture as a measure of these cells in vitro and compared their phenotype with that of cells capable of engrafting nonobese diabetic severe combined immune deficient (NOD/SCID) mice. Leukemic blast peripheral blood cells were evaluated for expression of CD34 and Thy-1 (CD90) antigens. The majority of AML blast cells at diagnosis lacked expression of Thy-1. Most primary CFU-blast and the CFU detected at up to 8 weeks from suspension cultures were CD34+/Thy-1−. AML cells that were capable of engrafting NOD/SCID mice were also found to have the CD34+/Thy-1− phenotype. However, significant engraftment was achieved using both CD34+/Thy-1− and CD34− subfractions from one AML M5 patient. These results suggest that while heterogeneity exists between individual patients, the leukemic progenitor cells that are capable of maintaining the disease in vitro and in vivo differ from normal hematopoietic progenitor cells in their lack of expression of Thy-1.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1885-1885
Author(s):  
Charlotte V. Cox ◽  
Roger S. Evely ◽  
Nicholas J. Goulden ◽  
Allison Blair

Abstract The cell of origin of childhood acute lymphoblastic leukaemia (ALL) has been the subject of conflicting reports in recent years. One model suggests that many haemopoietic cell types are susceptible to transformation and the level of commitment of the target cell influences the characteristics of the resulting blast cell population. A second model suggests that primitive haemopoietic cells are the targets for transformation, with some differentiation occurring subsequent to the transformation event. This model suggests a hierarchy of progenitors may exist in ALL. In support of this latter model, we have demonstrated that leukaemic stem cells in B-ALL have a primitive CD34+/CD10−/CD19− phenotype and T-ALL cells with NOD/SCID engrafting capacity are CD34+/CD4−. In this investigation we have attempted to further purify and characterise leukaemic stem cells from children with T-ALL. Cells from 7 patients were sorted for expression of CD34 and CD7 and the sorted subfractions evaluated for long-term proliferative ability in vitro using a serum free suspension culture assay and in the NOD/SCID mouse model. In this group of patients, the CD34+/CD7+ fraction represented 7±6% of cells at sorting, 6±4% were CD34+/CD7− and the majority were CD34−/CD7+ (60±12%). After 3 weeks in culture, the majority of proliferating cells were derived from the CD34+/CD7− subfraction (53±16%). By week 6, >70% of proliferating cells were derived from the CD34+/CD7− subfraction. Unsorted ALL cells and the sorted subfractions from 4 of these patients, were evaluated for their ability to engraft sublethally irradiated NOD/SCID mice. In each case, engraftment was achieved using 105–106 unsorted cells (25–80% CD45+) and with the CD34+/CD7− subfraction only (4–84% CD45+ with 3x103–8x104 cells). There was no engraftment with the other subfractions despite injecting up to 100 fold more cells. The engrafted cells had the same karyotype as the patient at diagnosis and expressed high levels of CD2, CD4 and CD7 implying they had differentiated in vivo. The self-renewal capacity of the CD34+/CD7− cells was evaluated by secondary transplantation. CD45+ cells from NOD/SCIDs engrafted with CD34+/CD7− cells successfully engrafted secondary recipients with equivalent levels of human cell engraftment, demonstrating these cells were capable of self-renewal. These findings suggest that cells with a more primitive phenotype may be the targets for transformation in T-ALL, rather than committed lymphocytes. To further investigate this hypothesis, we sorted cells from 4 of these patients for expression of CD133 and CD7 and evaluated their proliferative ability as described above. Results to date indicate that the CD133+/CD7− fraction represents only 0.35% of nucleated cells at sorting. However, after 3 weeks in culture, 48±9% of proliferating cells were derived from this subfraction and by week 6, 58±20% of cells were derived from the CD133+/CD7− subfraction. In vivo analyses completed in 2 patients to date have shown that only the CD133+/CD7− subfraction was capable of engrafting NOD/SCID mice (0.5–54% CD45+ using 3x103–105 cells). These results demonstrate that T-ALL cells with long-term proliferative and NOD/SCID repopulating capacity express the primitive haemopoietic cell antigens CD133 and CD34 and lack expression of T-lineage markers. These findings add further support to the concept of a common cell of origin for acute leukaemias.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 316-316
Author(s):  
Shih-Shih Chen ◽  
Piers E.M. Patten ◽  
Rita Simone ◽  
Sonia Marsilio ◽  
Jacqueline C. Barrientos ◽  
...  

Abstract Abstract 316 Chronic lymphocytic leukemia (CLL) clones contain activated/proliferative leukemic cells in lymphoid tissues and resting cells in the periphery. Different subsets of CLL cells have distinct proliferation rates. Recently divided “proliferative” cells have a surface membrane phenotype of CXCR4DIMCD5BRIGHT (CXCR4DIM) and contain higher numbers of CD38+ and Ki-67+ cells. Circulating “resting” CLL cells express CXCR4BRIGHTCD5DIM (CXCR4BR) and genetic signatures of older, quiescent cells that need to home to lymphoid tissues or die. CXCR4DIM and CXR4BR subsets are relatively minor (1–10% of total) components of CLL clones, with the major fraction (≥90%) of CLL cells having intermediate levels of CXCR4 and CD5 (CXCR4INT). Based on these differences, we proposed a model of transitioning CXCR4DIM → CXCR4INT → CXCR4BR CLL cells in the blood. Because higher birth rates correlate with more aggressive disease, and transiting back to solid tissues permits clonal survival and re-activation, this model suggests CXCR4DIM and CXCR4BR subsets as therapeutic targets. Aiming to further understand functional differences in CLL subsets in vitro and in vivo, we found that CLL subsets differ in cell size (CXCR4DIM>CXCR4INT>CXCR4BR), in vivo apoptosis and transmigration in vitro (both CXCR4DIM< CXCR4INT< CXCR4BR). Thus, while more CXCR4BR cells undergo apoptosis, CXCR4BR cells can migrate better to tissues to receive survival signals. In vivo functional differences were then studied in a NOD/SCID/γcnull (NSG) mouse model using pre-activated CLL-derived autologous T cells. Primary CLL blood cells from 1 M-CLL and 2 U-CLL patients were sorted for CXCR4BR, CXCR4INT or CXCR4DIM fractions. Each fraction (5×106 cells) was injected into NSG mice with 5×105 CD3/28-activated autologous T cells. At weeks 2–6 post transfer, blood analyses showed more extensive expansion of CLL B and T cells in mice received CXCR4DIM than in those injected with CXCR4BR or CXCR4INT. At weeks 9–12, mice were sacrificed. Although T cells dominated in blood, spleen and bone marrow of all recipients, a larger fraction of CLL B cells existed in CXCR4BR injected mice, suggesting better long-term CLL cell engraftment capacity of this fraction. Because regulation of T cells plays key roles in CLL cell survival/growth in patients and in the NSG adoptive transfer model, we next analyzed the same fractions for their abilities to activate T cells and elicit help for engraftment and growth. Unactivated CD5+ T cells (1–1.5×105) and B-CLL fractions (3–5×106 cells) were sorted from 6 patient samples (3 U-CLL and 3 M-CLL), injected into mice and followed bi-weekly until week 6. In 5 cases, except one with few CXCR4BR and CXCR4DIM cells, CXCR4DIM injected mice had more extensive T cell growth starting from week 2. Mice injected with CXCR4BR from 2 U-CLL cases also showed T cell expansions, but at comparatively lesser levels and at later time points (from week 4–5). At week 6, CLL B cells were found in spleen and bone marrow in mice with activated T cells; the numbers of CLL B cells correlated with T cell numbers. Also, identical CXCR4 levels were found in CLL cells regardless of origination from CXCR4BR or CXCR4DIM. Notably, no human B or T cells were detected in CXCR4INT injected mice. In fact, adding CXCR4INT cells to CXCR4DIM mice suppressed CXCR4DIM induced T cell expansion and cytokine production. Specifically, mice receiving both CXCR4DIM and CXCR4INT cells had diminished T cell expansion and at least 3 fold reduced serum levels of IFNγ and IL5. Overall, our data confirm the need for activated T cells for CLL B cell growth in mice; suggest superior long term CLL B cell engraftment by CXCR4BR cells with activated T cell support, and identify a greater ability of CXCR4DIM cells to activate autologous T cells, although some U-CLL CXCR4BR cells could do so to a lesser degree. Superior activation of T cells by CXCR4DIM B cells may be due to higher numbers of CD23+, CD25+, CD27+, CD29+ and CD44+ cells in CXCR4DIM fraction that facilitate cellular interactions. Finally, unlike CXCR4BR and CXCR4DIM cells, the major fraction in patient blood, CXCR4INT, inhibited T cell activation. These results indicate previously unappreciated levels of intraclonal CLL cell heterogeneity that may have important clinical relevance, allow more precise biologic analyses, and provide a rationale for preferential therapeutic targeting of these fractions. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 1 (6) ◽  
Author(s):  
Shanaya Ramchandani ◽  
Irum Naz ◽  
Namrata Dhudha ◽  
Manoj Garg

Cancer is one of the leading causes of mortality, contributing to 9.6 million deaths globally in 2018 alone. Although several cancer treatments exist, they are often associated with severe side effects and high toxicities, leaving room for significant advancements to be made in the field. In recent years, several phytochemicals from plants and natural bioresources have been extracted and tested against various human malignancies using both in vitro and in vivo preclinical model systems. Cardamonin, a chalcone extracted from the Alpinia species, is an example of a natural therapeutic agent that has anti-cancer and anti-inflammatory effects against human cancer cell lines, including breast, lung, colon, and gastric, in both in vitro culture systems as well as xenograft mouse models. Earlier, cardamonin was used as a natural medicine against stomach related issues, diarrhea, insulin resistance, nephroprotection against cisplatin treatment, vasorelaxant and antinociceptive. The compound is well-known to inhibit proliferation, migration, invasion, and induce apoptosis, through the involvement of Wnt/β-catenin, NF-κB, and PI3K/Akt pathways. The good biosafety and pharmacokinetic profiling of cardamonin satisfy it as an attractive molecule for the development of an anticancer agent. The present review has summarized the chemo-preventive ability of cardamonin as an anticancer agent against numerous human malignancies.


Blood ◽  
1992 ◽  
Vol 80 (4) ◽  
pp. 1026-1032 ◽  
Author(s):  
SJ Pirruccello ◽  
JD Jackson ◽  
MS Lang ◽  
J DeBoer ◽  
S Mann ◽  
...  

Abstract OMA-AML-1 was established from a patient with acute myelomonocytic (M4) leukemia at fifth relapse when blasts were greater than 85% CD34+, CD15- . Leukemic cells were established in suspension culture and independently grown as subcutaneous tumors in SCID mice. Cells growing in suspension culture underwent differentiation by phenotypic and morphologic criteria. In contrast, cells grown as subcutaneous solid tumors in SCID mice maintained progenitor cell characteristics with high-density CD34 expression and lack of morphologic differentiation. A tendency toward differentiation to CD15+, CD34- cells in vitro and self- renewal of CD34+, CD15- cells in vivo was consistently demonstrated regardless of whether cells were initially grown in vitro or in vivo. The cell line maintains both a CD34+, CD15- progentitor cell pool and a non-overlapping, CD15+, CD34- differentiating cell compartment after more than 1 year in continuous culture. Cell cycle analysis and cloning experiments were consistent with terminal differentiation occurring in the CD15+, CD34- population. The cell line shows concentration- dependent proliferative responses to interleukin (IL)-3, granulocyte- macrophage colony-stimulating factor (GM-CSF), and IL-6, but not to granulocyte CSF (G-CSF). OMA-AML-1 appears to mimic several features of normal myeloid hematopoiesis and should prove useful for the study of normal and malignant myeloid differentiation.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii223-ii223
Author(s):  
Nicholas Bayley ◽  
Christopher Tse ◽  
Henan Zhu ◽  
Weihong Yan ◽  
Laura Gosa ◽  
...  

Abstract The derivation of model systems from patient tumors is a requisite for reproducible and high throughput translational cancer research. However, not all tumors can form a model and those that do often fail to capture the molecular diversity specific to their cancer. The potential tumor-intrinsic underpinnings remain largely unknown. In gliomas, the brain tumor microenvironment (TME) is increasingly acknowledged as a regulator of tumor proliferation, invasion, and therapy response. The dissimilar environment of in vitro and heterotopic xenograft models could potentially play a role in the limited fidelity of these model systems. Here we established a culture-free workflow and biobank of 144 glioma direct-from-patient orthotopic xenografts (DPDOX) and 51 parallel gliomasphere cultures (GS). Our direct-from-patient workflow enabled the exclusive in vivo establishment of several gliomas – hereafter termed TME-dependent tumors – including low and high grade mtIDH gliomas and histone H3.3 G34 glioblastomas notoriously difficult to culture in vitro. Through molecular profiling of over 75 patient tumors and their matched derivative models, we find that DPDOX tumors preserve a gene expression signature of neural and glial interactions not found in GS and enriched in brain TME-dependent patient tumors. While these patient tumors span a diversity of clinical diagnoses, network-based inferred transcription factor activity suggests that they converge on shared master regulators of self-renewal driving proneural and OPC/NPC-like cellular state enrichment. Integrating multi-omic profiling from TCGA and other publicly available datasets reveals that this expression signature corresponds to a shared DNA methylation signature across disparate epigenetic subgroups. These findings suggest a brain TME dependence in patient tumors across multiple molecular and clinical classifications of glioma which leads to a lack of representation in model systems failing to recapitulate tumor-promoting components of the TME. Further this work provides a resource to guide translational investigations accounting for influences of the model environment.


Sign in / Sign up

Export Citation Format

Share Document