188 results on '"J. Gregory Cairncross"'
Search Results
2. Capicua regulates neural stem cell proliferation and lineage specification through control of Ets factors
- Author
-
Shiekh Tanveer Ahmad, Alexandra D. Rogers, Myra J. Chen, Rajiv Dixit, Lata Adnani, Luke S. Frankiw, Samuel O. Lawn, Michael D. Blough, Mana Alshehri, Wei Wu, Marco A. Marra, Stephen M. Robbins, J. Gregory Cairncross, Carol Schuurmans, and Jennifer A. Chan
- Subjects
Science - Abstract
Although Capicua (Cic), a transcriptional repressor, is linked to cancer, little is known about its function in the non-diseased brain. Here, authors show that Cic is strongly expressed in astrocytic and neuronal lineage cells during development and regulates proliferation and lineage selection via de-repression of Ets transcription factors.
- Published
- 2019
- Full Text
- View/download PDF
3. Mutant IDH1 gliomas downregulate phosphocholine and phosphoethanolamine synthesis in a 2-hydroxyglutarate-dependent manner
- Author
-
Pavithra Viswanath, Marina Radoul, Jose Luis Izquierdo-Garcia, Hema Artee Luchman, J. Gregory Cairncross, Russell O. Pieper, Joanna J. Phillips, and Sabrina M. Ronen
- Subjects
IDH1 mutation ,2-Hydroxyglutarate ,Metabolic reprogramming ,Magnetic resonance spectroscopy ,Phosphocholine ,Phosphoethanolamine ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Magnetic resonance spectroscopy (MRS) studies have identified elevated levels of the phospholipid precursor phosphocholine (PC) and phosphoethanolamine (PE) as metabolic hallmarks of cancer. Unusually, however, PC and PE levels are reduced in mutant isocitrate dehydrogenase 1 (IDHmut) gliomas that produce the oncometabolite 2-hydroxyglutarate (2-HG) relative to wild-type IDH1 (IDHwt) gliomas. The goal of this study was to determine the molecular mechanism underlying this unusual metabolic reprogramming in IDHmut gliomas. Methods Steady-state PC and PE were quantified using 31P-MRS. To quantify de novo PC and PE synthesis, we used 13C-MRS and measured flux to 13C-PC and 13C-PE in cells incubated with [1,2-13C]-choline and [1,2-13C]-ethanolamine. The activities of choline kinase (CK) and ethanolamine kinase (EK), the enzymes responsible for PC and PE synthesis, were quantified using 31P-MR-based assays. To interrogate the role of 2-HG, we examined IDHwt cells incubated with 2-HG and, conversely, IDHmut cells treated with the IDHmut inhibitor AGI-5198. To examine the role of hypoxia-inducible factor 1-α (HIF-1α), we silenced HIF-1α using RNA interference. To confirm our findings in vivo and in the clinic, we studied IDHwt and IDHmut orthotopic tumor xenografts and glioma patient biopsies. Results De novo synthesis of PC and PE was reduced in IDHmut cells relative to IDHwt. Concomitantly, CK activity and EK activity were reduced in IDHmut cells. Pharmacological manipulation of 2-HG levels established that 2-HG was responsible for reduced CK activity, EK activity, PC and PE. 2-HG has previously been reported to stabilize levels of HIF-1α, a known regulator of CK activity. Silencing HIF-1α in IDHmut cells restored CK activity, EK activity, PC and PE to IDHwt levels. Our findings were recapitulated in IDHmut orthotopic tumor xenografts and, most importantly, in IDHmut patient biopsies, validating our findings in vivo and in the clinic. Conclusions This study identifies, to our knowledge for the first time, a direct role for 2-HG in the downregulation of CK and EK activity, and thereby, PC and PE synthesis in IDHmut gliomas. These results highlight the unusual reprogramming of phospholipid metabolism in IDHmut gliomas and have implications for the identification of MRS-detectable metabolic biomarkers associated with 2-HG status.
- Published
- 2018
- Full Text
- View/download PDF
4. Hyperpolarized 13C MR imaging detects no lactate production in mutant IDH1 gliomas: Implications for diagnosis and response monitoring
- Author
-
Myriam M. Chaumeil, Marina Radoul, Chloé Najac, Pia Eriksson, Pavithra Viswanath, Michael D. Blough, Charles Chesnelong, H. Artee Luchman, J. Gregory Cairncross, and Sabrina M. Ronen
- Subjects
Glioma ,Isocitrate dehydrogenase 1 (IDH1) mutation ,Metabolic reprogramming ,Hyperpolarized 13C Magnetic Resonance Spectroscopy (MRS) ,Computer applications to medicine. Medical informatics ,R858-859.7 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Metabolic imaging of brain tumors using 13C Magnetic Resonance Spectroscopy (MRS) of hyperpolarized [1-13C] pyruvate is a promising neuroimaging strategy which, after a decade of preclinical success in glioblastoma (GBM) models, is now entering clinical trials in multiple centers. Typically, the presence of GBM has been associated with elevated hyperpolarized [1-13C] lactate produced from [1-13C] pyruvate, and response to therapy has been associated with a drop in hyperpolarized [1-13C] lactate. However, to date, lower grade gliomas had not been investigated using this approach. The most prevalent mutation in lower grade gliomas is the isocitrate dehydrogenase 1 (IDH1) mutation, which, in addition to initiating tumor development, also induces metabolic reprogramming. In particular, mutant IDH1 gliomas are associated with low levels of lactate dehydrogenase A (LDHA) and monocarboxylate transporters 1 and 4 (MCT1, MCT4), three proteins involved in pyruvate metabolism to lactate. We therefore investigated the potential of 13C MRS of hyperpolarized [1-13C] pyruvate for detection of mutant IDH1 gliomas and for monitoring of their therapeutic response. We studied patient-derived mutant IDH1 glioma cells that underexpress LDHA, MCT1 and MCT4, and wild-type IDH1 GBM cells that express high levels of these proteins. Mutant IDH1 cells and tumors produced significantly less hyperpolarized [1-13C] lactate compared to GBM, consistent with their metabolic reprogramming. Furthermore, hyperpolarized [1-13C] lactate production was not affected by chemotherapeutic treatment with temozolomide (TMZ) in mutant IDH1 tumors, in contrast to previous reports in GBM. Our results demonstrate the unusual metabolic imaging profile of mutant IDH1 gliomas, which, when combined with other clinically available imaging methods, could be used to detect the presence of the IDH1 mutation in vivo.
- Published
- 2016
- Full Text
- View/download PDF
5. Precursor States of Brain Tumor Initiating Cell Lines Are Predictive of Survival in Xenografts and Associated with Glioblastoma Subtypes
- Author
-
Carlo Cusulin, Charles Chesnelong, Pinaki Bose, Misha Bilenky, Karen Kopciuk, Jennifer A. Chan, J. Gregory Cairncross, Steven J. Jones, Marco A. Marra, H. Artee Luchman, and Samuel Weiss
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
In glioblastoma multiforme (GBM), brain-tumor-initiating cells (BTICs) with cancer stem cell characteristics have been identified and proposed as primordial cells responsible for disease initiation, recurrence, and therapeutic resistance. However, the extent to which individual, patient-derived BTIC lines reflect the heterogeneity of GBM remains poorly understood. Here we applied a stem cell biology approach and compared self-renewal, marker expression, label retention, and asymmetric cell division in 20 BTIC lines. Through cluster analysis, we identified two subgroups of BTIC lines with distinct precursor states, stem- or progenitor-like, predictive of survival after xenograft. Moreover, stem and progenitor transcriptomic signatures were identified, which showed a strong association with the proneural and mesenchymal subtypes, respectively, in the TCGA cohort. This study proposes a different framework for the study and use of BTIC lines and provides precursor biology insights into GBM.
- Published
- 2015
- Full Text
- View/download PDF
6. Data from Disulfiram when Combined with Copper Enhances the Therapeutic Effects of Temozolomide for the Treatment of Glioblastoma
- Author
-
Donna L. Senger, Stephen M. Robbins, David R. Kaplan, J. Gregory Cairncross, Samuel Weiss, Artee Luchman, Jacob C. Easaw, Jeffrey L. Wrana, Alessandro Datti, David Uehling, Ahmed Aman, Xiuling Wang, Ngoc-Ha Dang, Xiaoguang Hao, Jennifer C. King, Natalie Grinshtein, J. Connor Wells, and Xueqing Lun
- Abstract
Purpose: Glioblastoma is one of the most lethal cancers in humans, and with existing therapy, survival remains at 14.6 months. Current barriers to successful treatment include their infiltrative behavior, extensive tumor heterogeneity, and the presence of a stem-like population of cells, termed brain tumor–initiating cells (BTIC) that confer resistance to conventional therapies.Experimental Design: To develop therapeutic strategies that target BTICs, we focused on a repurposing approach that explored already-marketed (clinically approved) drugs for therapeutic potential against patient-derived BTICs that encompass the genetic and phenotypic heterogeneity of glioblastoma observed clinically.Results: Using a high-throughput in vitro drug screen, we found that montelukast, clioquinol, and disulfiram (DSF) were cytotoxic against a large panel of patient-derived BTICs. Of these compounds, disulfiram, an off-patent drug previously used to treat alcoholism, in the presence of a copper supplement, showed low nanomolar efficacy in BTICs including those resistant to temozolomide and the highly infiltrative quiescent stem-like population. Low dose DSF-Cu significantly augmented temozolomide activity in vitro, and importantly, prolonged in vivo survival in patient-derived BTIC models established from both newly diagnosed and recurrent tumors. Moreover, we found that in addition to acting as a potent proteasome inhibitor, DSF-Cu functionally impairs DNA repair pathways and enhances the effects of DNA alkylating agents and radiation. These observations suggest that DSF-Cu inhibits proteasome activity and augments the therapeutic effects of DNA-damaging agents (temozolomide and radiation).Conclusions: DSF-Cu should be considered as an adjuvant therapy for the treatment of patients with glioblastoma in both newly diagnosed and recurrent settings. Clin Cancer Res; 22(15); 3860–75. ©2016 AACR.
- Published
- 2023
7. Supplementary Figure 6 from 2-Hydroxyglutarate-Mediated Autophagy of the Endoplasmic Reticulum Leads to an Unusual Downregulation of Phospholipid Biosynthesis in Mutant IDH1 Gliomas
- Author
-
Sabrina M. Ronen, Joanna J. Phillips, Russell O. Pieper, Bo Huang, J. Gregory Cairncross, Hema Artee Luchman, Wei Qiang Ong, Jose Luis Izquierdo-Garcia, Marina Radoul, and Pavithra Viswanath
- Abstract
CQ and BAF restore phospholipid levels and inhibit clonogenicity of IDHmut glioma cells.
- Published
- 2023
8. Supplementary Figure 7 from 2-Hydroxyglutarate-Mediated Autophagy of the Endoplasmic Reticulum Leads to an Unusual Downregulation of Phospholipid Biosynthesis in Mutant IDH1 Gliomas
- Author
-
Sabrina M. Ronen, Joanna J. Phillips, Russell O. Pieper, Bo Huang, J. Gregory Cairncross, Hema Artee Luchman, Wei Qiang Ong, Jose Luis Izquierdo-Garcia, Marina Radoul, and Pavithra Viswanath
- Abstract
CQ and BAF do not affect ER area or phospholipid levels in IDHwt glioma cells.
- Published
- 2023
9. Supplementary Figure 5 from Dual mTORC1/2 Blockade Inhibits Glioblastoma Brain Tumor Initiating Cells In Vitro and In Vivo and Synergizes with Temozolomide to Increase Orthotopic Xenograft Survival
- Author
-
Samuel Weiss, J. Gregory Cairncross, Xueqing Q. Lun, Stephanie A. Nguyen, Owen D.M. Stechishin, and H. Artee Luchman
- Abstract
Oral administration of AZD is compatible with TMZ chemotherapy in vivo.
- Published
- 2023
10. Supplementary Figure 4 from Dual mTORC1/2 Blockade Inhibits Glioblastoma Brain Tumor Initiating Cells In Vitro and In Vivo and Synergizes with Temozolomide to Increase Orthotopic Xenograft Survival
- Author
-
Samuel Weiss, J. Gregory Cairncross, Xueqing Q. Lun, Stephanie A. Nguyen, Owen D.M. Stechishin, and H. Artee Luchman
- Abstract
AZD and TMZ administration do not significantly influence body weights of treated animals.
- Published
- 2023
11. Supplementary Figure 3 from 2-Hydroxyglutarate-Mediated Autophagy of the Endoplasmic Reticulum Leads to an Unusual Downregulation of Phospholipid Biosynthesis in Mutant IDH1 Gliomas
- Author
-
Sabrina M. Ronen, Joanna J. Phillips, Russell O. Pieper, Bo Huang, J. Gregory Cairncross, Hema Artee Luchman, Wei Qiang Ong, Jose Luis Izquierdo-Garcia, Marina Radoul, and Pavithra Viswanath
- Abstract
2-HG induces a reduction in ER area in IDHmut glioma cells.
- Published
- 2023
12. Data from Novel MSH6 Mutations in Treatment-Naïve Glioblastoma and Anaplastic Oligodendroglioma Contribute to Temozolomide Resistance Independently of MGMT Promoter Methylation
- Author
-
Samuel Weiss, J. Gregory Cairncross, Stephen M. Robbins, Donna L. Senger, Xueqing Q. Lun, H. Artee Luchman, Owen D.M. Stechishin, and Stephanie A. Nguyen
- Abstract
Purpose: The current standard of care for glioblastoma (GBM) involves a combination of surgery, radiotherapy, and temozolomide chemotherapy, but this regimen fails to achieve long-term tumor control. Resistance to temozolomide is largely mediated by expression of the DNA repair enzyme MGMT; however, emerging evidence suggests that inactivation of MSH6 and other mismatch repair proteins plays an important role in temozolomide resistance. Here, we investigate endogenous MSH6 mutations in GBM, anaplastic oligodendroglial tumor tissue, and corresponding brain tumor–initiating cell lines (BTIC).Experimental Design:MSH6 sequence and MGMT promoter methylation were determined in human tumor samples and BTICs. Sensitivity to temozolomide was evaluated in vitro using BTICs in the absence and presence of O6-benzylguanine to deplete MGMT. The influence of MGMT and MSH6 status on in vivo sensitivity to temozolomide was evaluated using intracranial BTIC xenografts.Results: We identified 11 previously unreported mutations in MSH6 in nine different glioma samples and six paired BTIC lines from adult patients. In addition, MSH6 mutations were documented in three oligodendrogliomas and two treatment-naïve gliomas, both previously unreported findings. These mutations were found to influence the sensitivity of BTICs to temozolomide both in vitro and in vivo, independent of MGMT promoter methylation status.Conclusions: These data demonstrate that endogenous MSH6 mutations may be present before alkylator therapy and occur in at least two histologic subtypes of adult glial neoplasms, with this report serving as the first to note these mutations in oligodendroglioma. These findings broaden our understanding of the clinical response to temozolomide in gliomas. Clin Cancer Res; 20(18); 4894–903. ©2014 AACR.
- Published
- 2023
13. Supplementary Figure 1 from Dual mTORC1/2 Blockade Inhibits Glioblastoma Brain Tumor Initiating Cells In Vitro and In Vivo and Synergizes with Temozolomide to Increase Orthotopic Xenograft Survival
- Author
-
Samuel Weiss, J. Gregory Cairncross, Xueqing Q. Lun, Stephanie A. Nguyen, Owen D.M. Stechishin, and H. Artee Luchman
- Abstract
Molecular and genetic analysis of BTICs identifies subgroups with endogenous mutations in EGFR and PTEN.
- Published
- 2023
14. Supplementary Figure 3 from Dual mTORC1/2 Blockade Inhibits Glioblastoma Brain Tumor Initiating Cells In Vitro and In Vivo and Synergizes with Temozolomide to Increase Orthotopic Xenograft Survival
- Author
-
Samuel Weiss, J. Gregory Cairncross, Xueqing Q. Lun, Stephanie A. Nguyen, Owen D.M. Stechishin, and H. Artee Luchman
- Abstract
Rapamycin partially inhibits growth of most BTICs irrespective of EGFR or PTEN mutational status.
- Published
- 2023
15. Data from 2-Hydroxyglutarate-Mediated Autophagy of the Endoplasmic Reticulum Leads to an Unusual Downregulation of Phospholipid Biosynthesis in Mutant IDH1 Gliomas
- Author
-
Sabrina M. Ronen, Joanna J. Phillips, Russell O. Pieper, Bo Huang, J. Gregory Cairncross, Hema Artee Luchman, Wei Qiang Ong, Jose Luis Izquierdo-Garcia, Marina Radoul, and Pavithra Viswanath
- Abstract
Tumor metabolism is reprogrammed to meet the demands of proliferating cancer cells. In particular, cancer cells upregulate synthesis of the membrane phospholipids phosphatidylcholine (PtdCho) and phosphatidylethanolamine (PtdE) in order to allow for rapid membrane turnover. Nonetheless, we show here that, in mutant isocitrate dehydrogenase 1 (IDHmut) gliomas, which produce the oncometabolite 2-hydroxyglutarate (2-HG), PtdCho and PtdE biosynthesis is downregulated and results in lower levels of both phospholipids when compared with wild-type IDH1 cells. 2-HG inhibited collagen-4-prolyl hydroxylase activity, leading to accumulation of misfolded procollagen-IV in the endoplasmic reticulum (ER) of both genetically engineered and patient-derived IDHmut glioma models. The resulting ER stress triggered increased expression of FAM134b, which mediated autophagic degradation of the ER (ER-phagy) and a reduction in the ER area. Because the ER is the site of phospholipid synthesis, ER-phagy led to reduced PtdCho and PtdE biosynthesis. Inhibition of ER-phagy via pharmacological or molecular approaches restored phospholipid biosynthesis in IDHmut glioma cells, triggered apoptotic cell death, inhibited tumor growth, and prolonged the survival of orthotopic IDHmut glioma-bearing mice, pointing to a potential therapeutic opportunity. Glioma patient biopsies also exhibited increased ER-phagy and downregulation of PtdCho and PtdE levels in IDHmut samples compared with wild-type, clinically validating our observations. Collectively, this study provides detailed and clinically relevant insights into the functional link between oncometabolite-driven ER-phagy and phospholipid biosynthesis in IDHmut gliomas.Significance: Downregulation of phospholipid biosynthesis via ER-phagy is essential for proliferation and clonogenicity of mutant IDH1 gliomas, a finding with immediate therapeutic implications. Cancer Res; 78(9); 2290–304. ©2018 AACR.
- Published
- 2023
16. Supplementary Figure 6 from Dual mTORC1/2 Blockade Inhibits Glioblastoma Brain Tumor Initiating Cells In Vitro and In Vivo and Synergizes with Temozolomide to Increase Orthotopic Xenograft Survival
- Author
-
Samuel Weiss, J. Gregory Cairncross, Xueqing Q. Lun, Stephanie A. Nguyen, Owen D.M. Stechishin, and H. Artee Luchman
- Abstract
Pharmacological inhibition of EGFR and PI3K/mTORC1/2 signaling in BTICs.
- Published
- 2023
17. Supplementary Figure 2 from 2-Hydroxyglutarate-Mediated Autophagy of the Endoplasmic Reticulum Leads to an Unusual Downregulation of Phospholipid Biosynthesis in Mutant IDH1 Gliomas
- Author
-
Sabrina M. Ronen, Joanna J. Phillips, Russell O. Pieper, Bo Huang, J. Gregory Cairncross, Hema Artee Luchman, Wei Qiang Ong, Jose Luis Izquierdo-Garcia, Marina Radoul, and Pavithra Viswanath
- Abstract
2-HG inhibits PtdCho and PtdE biosynthesis.
- Published
- 2023
18. Data from Dual mTORC1/2 Blockade Inhibits Glioblastoma Brain Tumor Initiating Cells In Vitro and In Vivo and Synergizes with Temozolomide to Increase Orthotopic Xenograft Survival
- Author
-
Samuel Weiss, J. Gregory Cairncross, Xueqing Q. Lun, Stephanie A. Nguyen, Owen D.M. Stechishin, and H. Artee Luchman
- Abstract
Purpose: The EGFR and PI3K/mTORC1/2 pathways are frequently altered in glioblastoma (GBM), but pharmacologic targeting of EGFR and PI3K signaling has failed to demonstrate efficacy in clinical trials. Lack of relevant models has rendered it difficult to assess whether targeting these pathways might be effective in molecularly defined subgroups of GBMs. Here, human brain tumor–initiating cell (BTIC) lines with different combinations of endogenous EGFR wild-type, EGFRvIII, and PTEN mutations were used to investigate response to the EGFR inhibitor gefitinib, mTORC1 inhibitor rapamycin, and dual mTORC1/2 inhibitor AZD8055 alone and in combination with temozolomide (TMZ)Experimental Design:In vitro growth inhibition and cell death induced by gefitinib, rapamycin, AZD8055, and TMZ or combinations in human BTICs were assessed by alamarBlue, neurosphere, and Western blotting assays. The in vivo efficacy of AZD8055 was assessed in subcutaneous and intracranial BTIC xenografts. Kaplan–Meier survival studies were performed with AZD8055 and in combination with TMZ.Results: We confirm that gefitinib and rapamycin have modest effects in most BTIC lines, but AZD8055 was highly effective at inhibiting Akt/mTORC2 activity and dramatically reduced the viability of BTICs regardless of their EGFR and PTEN mutational status. Systemic administration of AZD8055 effectively inhibited tumor growth in subcutaneous BTIC xenografts and mTORC1/2 signaling in orthotopic BTIC xenografts. AZD8055 was synergistic with the alkylating agent TMZ and significantly prolonged animal survival.Conclusion: These data suggest that dual inhibition of mTORC1/2 may be of benefit in GBM, including the subset of TMZ-resistant GBMs. Clin Cancer Res; 20(22); 5756–67. ©2014 AACR.
- Published
- 2023
19. Supplementary Figures S1-S10 Tables S1-S3 from Disulfiram when Combined with Copper Enhances the Therapeutic Effects of Temozolomide for the Treatment of Glioblastoma
- Author
-
Donna L. Senger, Stephen M. Robbins, David R. Kaplan, J. Gregory Cairncross, Samuel Weiss, Artee Luchman, Jacob C. Easaw, Jeffrey L. Wrana, Alessandro Datti, David Uehling, Ahmed Aman, Xiuling Wang, Ngoc-Ha Dang, Xiaoguang Hao, Jennifer C. King, Natalie Grinshtein, J. Connor Wells, and Xueqing Lun
- Abstract
Figure S1: Preclinical assessment of clioquinol. Figure S2: Preclinical assessment of montelukast. Figure S3: Secondary validation of compounds identified via HTS. Figure S4: Sensitivity of BTICs to TMZ. Figure S5: Copper is required for Disulfiram-mediated cell killing. Figure S6: Cell Viability and Apoptosis in the presence of DSF-Cu. Figure S7: Efficacy to DSF-Cu is independent of MGMT expression. Figure S8: Inhibition of proteasome activity by DSF-Cu. Figure S9: DSF-Cu in combination with TMZ prolongs survival in vivo. Figure S10: DSF-Cu mediated apoptosis in vivo. Table S1: Summary of BTIC Molecular Characterization. Table S2: Summary of Compound Priortization. Table S3: Down regulation of DNA repair genes by DSF-Cu.
- Published
- 2023
20. Data Supplement from Novel MSH6 Mutations in Treatment-Naïve Glioblastoma and Anaplastic Oligodendroglioma Contribute to Temozolomide Resistance Independently of MGMT Promoter Methylation
- Author
-
Samuel Weiss, J. Gregory Cairncross, Stephen M. Robbins, Donna L. Senger, Xueqing Q. Lun, H. Artee Luchman, Owen D.M. Stechishin, and Stephanie A. Nguyen
- Abstract
Supplementary Figure 1. Detection of MSH6 somatic mutations by cDNA and genomic DNA sequencing. A-D, cDNA sequencing (A) revealed a heterozygous T1219I mutation that was also confirmed by sequencing the genomic DNA of cell line 119 (B) and the fresh frozen tumor from patient 119 (C). The T1219I mutation was not present in genomic DNA from a blood sample of patient 119 (D) demonstrating it to be of somatic rather than germline origin.
- Published
- 2023
21. Supplementary Figure 1 from 2-Hydroxyglutarate-Mediated Autophagy of the Endoplasmic Reticulum Leads to an Unusual Downregulation of Phospholipid Biosynthesis in Mutant IDH1 Gliomas
- Author
-
Sabrina M. Ronen, Joanna J. Phillips, Russell O. Pieper, Bo Huang, J. Gregory Cairncross, Hema Artee Luchman, Wei Qiang Ong, Jose Luis Izquierdo-Garcia, Marina Radoul, and Pavithra Viswanath
- Abstract
Phospholipid biosynthesis is down-regulated in IDHmut glioma cells.
- Published
- 2023
22. Supplementary Figure 5 from 2-Hydroxyglutarate-Mediated Autophagy of the Endoplasmic Reticulum Leads to an Unusual Downregulation of Phospholipid Biosynthesis in Mutant IDH1 Gliomas
- Author
-
Sabrina M. Ronen, Joanna J. Phillips, Russell O. Pieper, Bo Huang, J. Gregory Cairncross, Hema Artee Luchman, Wei Qiang Ong, Jose Luis Izquierdo-Garcia, Marina Radoul, and Pavithra Viswanath
- Abstract
Silencing FAM134b, Atg5 or Atg7 restores phospholipid biosynthesis in IDHmut glioma cells.
- Published
- 2023
23. Supplementary Figure 4 from 2-Hydroxyglutarate-Mediated Autophagy of the Endoplasmic Reticulum Leads to an Unusual Downregulation of Phospholipid Biosynthesis in Mutant IDH1 Gliomas
- Author
-
Sabrina M. Ronen, Joanna J. Phillips, Russell O. Pieper, Bo Huang, J. Gregory Cairncross, Hema Artee Luchman, Wei Qiang Ong, Jose Luis Izquierdo-Garcia, Marina Radoul, and Pavithra Viswanath
- Abstract
2-HG induces ER-phagy in IDHmut glioma cells.
- Published
- 2023
24. Supplementary Figure 8 from 2-Hydroxyglutarate-Mediated Autophagy of the Endoplasmic Reticulum Leads to an Unusual Downregulation of Phospholipid Biosynthesis in Mutant IDH1 Gliomas
- Author
-
Sabrina M. Ronen, Joanna J. Phillips, Russell O. Pieper, Bo Huang, J. Gregory Cairncross, Hema Artee Luchman, Wei Qiang Ong, Jose Luis Izquierdo-Garcia, Marina Radoul, and Pavithra Viswanath
- Abstract
ER-phagy is linked to phospholipid biosynthesis in BT54 neurospheres (A-C), orthotopic IDHmut tumor xenografts (D-J) and IDHmut patient biopsies (K-Q).
- Published
- 2023
25. Supplementary Figure 2 from Dual mTORC1/2 Blockade Inhibits Glioblastoma Brain Tumor Initiating Cells In Vitro and In Vivo and Synergizes with Temozolomide to Increase Orthotopic Xenograft Survival
- Author
-
Samuel Weiss, J. Gregory Cairncross, Xueqing Q. Lun, Stephanie A. Nguyen, Owen D.M. Stechishin, and H. Artee Luchman
- Abstract
Gefitinib inhibits growth of EGFRvIII and PTENwt BTICs.
- Published
- 2023
26. Supplementary Figure Legends from Dual mTORC1/2 Blockade Inhibits Glioblastoma Brain Tumor Initiating Cells In Vitro and In Vivo and Synergizes with Temozolomide to Increase Orthotopic Xenograft Survival
- Author
-
Samuel Weiss, J. Gregory Cairncross, Xueqing Q. Lun, Stephanie A. Nguyen, Owen D.M. Stechishin, and H. Artee Luchman
- Abstract
Supplementary Figure Legends from Dual mTORC1/2 Blockade Inhibits Glioblastoma Brain Tumor Initiating Cells In Vitro and In Vivo and Synergizes with Temozolomide to Increase Orthotopic Xenograft Survival
- Published
- 2023
27. Supplementary Figure S1 from IDH1 Mutation Induces Reprogramming of Pyruvate Metabolism
- Author
-
Sabrina M. Ronen, Russell O. Pieper, Joanna J. Phillips, J. Gregory Cairncross, Samuel Weiss, H. Artee Luchman, Michael Blough, Myriam M. Chaumeil, Marina Radoul, Larry Cai, Pia Eriksson, Pavithra Viswanath, and Jose L. Izquierdo-Garcia
- Abstract
Effect of 2-HG on cell proliferation of IDH1 wild-type cells for the U87 (A) and NHA (B) models.
- Published
- 2023
28. Supplementary Table S1 from IDH1 Mutation Induces Reprogramming of Pyruvate Metabolism
- Author
-
Sabrina M. Ronen, Russell O. Pieper, Joanna J. Phillips, J. Gregory Cairncross, Samuel Weiss, H. Artee Luchman, Michael Blough, Myriam M. Chaumeil, Marina Radoul, Larry Cai, Pia Eriksson, Pavithra Viswanath, and Jose L. Izquierdo-Garcia
- Abstract
Comparison of PDH activity in the various models used in this study.
- Published
- 2023
29. ABT-888 restores sensitivity in temozolomide resistant glioma cells and xenografts.
- Author
-
Alice L Yuan, Christian B Ricks, Alexandra K Bohm, Xueqing Lun, Lori Maxwell, Shahana Safdar, Shazreh Bukhari, Amanda Gerber, Wajid Sayeed, Elizabeth A Bering, Haley Pedersen, Jennifer A Chan, Yaoqing Shen, Marco Marra, David R Kaplan, Warren Mason, Lindsey D Goodman, Ravesanker Ezhilarasan, Ascher B Kaufmann, Matthew Cabral, Steve M Robbins, Donna L Senger, Daniel P Cahill, Erik P Sulman, J Gregory Cairncross, and Michael D Blough
- Subjects
Medicine ,Science - Abstract
BACKGROUND:Temozolomide (TMZ) is active against glioblastomas (GBM) in which the O6-methylguanine-DNA methyltransferase (MGMT) gene is silenced. However, even in responsive cases, its beneficial effect is undermined by the emergence of drug resistance. Here, we tested whether inhibition of poly (ADP-ribose) polymerase-1 and -2 (PARP) enhanced the effectiveness of TMZ. METHODS:Using patient derived brain tumor initiating cells (BTICs) and orthotopic xenografts as models of newly diagnosed and recurrent high-grade glioma, we assessed the effects of TMZ, ABT-888, and the combination of TMZ and ABT-888 on the viability of BTICs and survival of tumor-bearing mice. We also studied DNA damage repair, checkpoint protein phosphorylation, and DNA replication in mismatch repair (MMR) deficient cells treated with TMZ and TMZ plus ABT-888. RESULTS:Cells and xenografts derived from newly diagnosed MGMT methylated high-grade gliomas were sensitive to TMZ while those derived from unmethylated and recurrent gliomas were typically resistant. ABT-888 had no effect on the viability of BTICs or tumor bearing mice, but co-treatment with TMZ restored sensitivity in resistant cells and xenografts from newly diagnosed unmethylated gliomas and recurrent gliomas with MSH6 mutations. In contrast, the addition of ABT-888 to TMZ had little sensitizing effect on cells and xenografts derived from newly diagnosed methylated gliomas. In a model of acquired TMZ resistance mediated by loss of MMR gene MSH6, re-sensitization to TMZ by ABT-888 was accompanied by persistent DNA strand breaks, re-engagement of checkpoint kinase signaling, and interruption of DNA synthesis. CONCLUSION:In laboratory models, the addition of ABT-888 to TMZ overcame resistance to TMZ.
- Published
- 2018
- Full Text
- View/download PDF
30. Predicting MGMT Methylation Status of Glioblastomas from MRI Texture.
- Author
-
Ilya Levner, Sylvia Drabycz, Gloria Roldan, Paula De Robles, J. Gregory Cairncross, and Joseph Ross Mitchell
- Published
- 2009
- Full Text
- View/download PDF
31. Joint Final Report of EORTC 26951 and RTOG 9402: phase III trials with procarbazine, lomustine, and vincristine chemotherapy for anaplastic oligodendroglial tumors
- Author
-
Andrew B. Lassman, Khê Hoang-Xuan, Mei-Yin C. Polley, Alba A. Brandes, J. Gregory Cairncross, Johan M. Kros, Lynn S. Ashby, Martin J.B. Taphoorn, Luis Souhami, Winand N.M. Dinjens, Nadia N. Laack, Mathilde C.M. Kouwenhoven, Karen L. Fink, Pim J. French, David R. Macdonald, Denis Lacombe, Minhee Won, Thierry Gorlia, Minesh P. Mehta, Martin J. van den Bent, Pathology, Neurology, and CCA - Cancer Treatment and quality of life
- Subjects
Cancer Research ,Oncology ,SDG 3 - Good Health and Well-being ,Clinical Trials, Phase III as Topic ,Brain Neoplasms ,Lomustine ,Vincristine ,Procarbazine ,Antineoplastic Combined Chemotherapy Protocols ,Oligodendroglioma ,Humans ,Neoplasm Recurrence, Local - Abstract
Clinical trials frequently include multiple end points that mature at different times. The initial report, typically based on the basis of the primary end point, may be published when key planned co-primary or secondary analyses are not yet available. Clinical Trial Updates provide an opportunity to disseminate additional results from studies, published in JCO or elsewhere, for which the primary end point has already been reported. Anaplastic oligodendroglial tumors (AOTs) are chemotherapy-sensitive brain tumors. We report the final very long-term survival results from European Organization for the Research and Treatment of Cancer 26951 and Radiation Therapy Oncology Group 9402 phase III trials initiated in 1990s, which both studied radiotherapy with/without neo/adjuvant procarbazine, lomustine, and vincristine (PCV) for newly diagnosed anaplastic oligodendroglial tumors. The median follow-up duration in both was 18-19 years. For European Organization for the Research and Treatment of Cancer 26951, median, 14-year, and probable 20-year overall survival rates without versus with PCV were 2.6 years, 13.4%, and 10.1% versus 3.5 years, 25.1%, and 16.8% (N = 368 overall; hazard ratio [HR] 0.78; 95% CI, 0.63 to 0.98; P = .033), with 1p19q codeletion 9.3 years, 26.2%, and 13.6% versus 14.2 years, 51.0%, and 37.1% (n = 80; HR 0.60; 95% CI, 0.35 to 1.03; P = .063), respectively. For Radiation Therapy Oncology Group 9402, analogous results were 4.8 years, 16.5%, and 11.2% versus 4.8 years, 29.1%, and 24.6% (N = 289 overall; HR 0.79; 95% CI, 0.61 to 1.03; P = .08), with codeletion 7.3 years, 25.0%, and 14.9% versus 13.2 years, 46.1%, and 37% (n = 125; HR 0.61; 95% CI, 0.40 to 0.94; P = .02), respectively. With that, the studies show similar long-term survival even without tumor recurrence in a significant proportion of patients after first-line treatment with radiotherapy/PCV.
- Published
- 2022
32. Multi-Omic Analysis of CIC’s Functional Networks Reveals Novel Interaction Partners and a Potential Role in Mitotic Fidelity
- Author
-
Yuka Takemon, Véronique G. LeBlanc, Jungeun Song, Susanna Y. Chan, Stephen Dongsoo Lee, Diane L. Trinh, Shiekh Tanveer Ahmad, William R. Brothers, Richard D. Corbett, Alessia Gagliardi, Annie Moradian, J. Gregory Cairncross, Stephen Yip, Samuel A. J. R. Aparicio, Jennifer A. Chan, Christopher S. Hughes, Gregg B. Morin, Sharon M. Gorski, Suganthi Chittaranjan, and Marco A. Marra
- Subjects
Cancer Research ,CIC ,genetic networks ,proteomic interactions ,single-cell sequencing ,cell cycle ,mitosis ,splicing ,Oncology - Abstract
CIC encodes a transcriptional repressor and MAPK signalling effector that is inactivated by loss-of-function mutations in several cancer types, consistent with a role as a tumour suppressor. Here, we used bioinformatic, genomic, and proteomic approaches to investigate CIC’s interaction networks. We observed both previously identified and novel candidate interactions between CIC and SWI/SNF complex members, as well as novel interactions between CIC and cell cycle regulators and RNA processing factors. We found that CIC loss is associated with an increased frequency of mitotic defects in human cell lines and an in vivo mouse model and with dysregulated expression of mitotic regulators. We also observed aberrant splicing in CIC-deficient cell lines, predominantly at 3′ and 5′ untranslated regions of genes, including genes involved in MAPK signalling, DNA repair, and cell cycle regulation. Our study thus characterises the complexity of CIC’s functional network and describes the effect of its loss on cell cycle regulation, mitotic integrity, and transcriptional splicing, thereby expanding our understanding of CIC’s potential roles in cancer. In addition, our work exemplifies how multi-omic, network-based analyses can be used to uncover novel insights into the interconnected functions of pleiotropic genes/proteins across cellular contexts.
- Published
- 2023
33. Temozolomide and seizure outcomes in a randomized clinical trial of elderly glioblastoma patients
- Author
-
Chad Winch, Ryo Nishikawa, Claire Phillips, Normand Laperriere, J. Gregory Cairncross, Wilson Roa, Alba A. Brandes, Christopher J. O'Callaghan, Seth Andrew Climans, Johan Menten, Wolfgang Wick, Michael Fay, James Perry, Warren P. Mason, and Keyue Ding
- Subjects
Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,law.invention ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,Randomized controlled trial ,Quality of life ,law ,Internal medicine ,medicine ,Adverse effect ,Chemotherapy ,Temozolomide ,business.industry ,Hazard ratio ,medicine.disease ,Clinical trial ,Neurology ,Oncology ,030220 oncology & carcinogenesis ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Tumor-related epilepsy may respond to chemotherapy. In a previously-published multi-centre randomized clinical trial of 562 elderly glioblastoma patients, temozolomide plus short-course radiotherapy conferred a survival benefit over radiotherapy alone. Seizure outcomes were not reported. We performed an unplanned secondary analysis of this trial’s data. The trial design has been previously reported. Seizures were recorded by clinicians as adverse events and by patients in quality of life questionnaires. A Chi-square test of seizure rates between the two groups (α = 0.05) and a Kaplan–Meier estimator of time-to-first self-reported seizure were planned. Almost all patients were followed until they died. In the radiotherapy alone group, 68 patients (24%) had a documented or self-reported seizure versus 83 patients (30%) in the temozolomide plus radiotherapy group, Chi-square analysis showed no difference (p = 0.15). Patients receiving radiotherapy alone tended to develop seizures earlier than those receiving temozolomide plus radiotherapy (p = 0.054). Patients with seizures had shorter overall survival than those without seizures (hazard ratio 1.24, p = 0.02). This study was not powered to detect differences in seizure outcomes, but temozolomide seemed to have minimal impact on seizure control in elderly patients with glioblastoma. Clinical Trial Registration: NCT00482677 2007-06-05.
- Published
- 2020
34. PARP inhibition suppresses the emergence of temozolomide resistance in a model system
- Author
-
Jenny Liao, J. Gregory Cairncross, Jacob Willms, Alice L. Yuan, Haley Pedersen, Michael D. Blough, Lori Maxwell, Mathieu Meode, Melanie Tan, Elizabeth A. Bering, and Sophie Black
- Subjects
Cancer Research ,Methyltransferase ,Poly ADP ribose polymerase ,Poly (ADP-Ribose) Polymerase-1 ,Apoptosis ,Model system ,Poly(ADP-ribose) Polymerase Inhibitors ,03 medical and health sciences ,0302 clinical medicine ,Temozolomide ,Tumor Cells, Cultured ,medicine ,Humans ,Clinical efficacy ,Antineoplastic Agents, Alkylating ,Cell Proliferation ,business.industry ,Gene Expression Regulation, Neoplastic ,MSH6 ,Neurology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,PARP inhibitor ,Cancer research ,DNA mismatch repair ,Neurology (clinical) ,Glioblastoma ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Temozolomide (TMZ) is a life prolonging DNA alkylating agent active against glioblastomas (GBM) in which the O6-methylguanine-DNA methyltransferase (MGMT) gene is silenced by promoter methylation. Unfortunately acquired TMZ resistance severely undermines its clinical efficacy. Using an in vitro model, we tested whether poly (ADP-ribose) polymerase-1 and -2 (PARP) inhibition could suppress the emergence of resistance to enhance the effectiveness of TMZ. Using the MGMT-methylated GBM line U251N, in which TMZ resistance can be induced, we developed a method to rapidly recreate mechanisms of TMZ resistance seen in GBMs, including MMR mutations and MGMT re-expression. We then assessed whether TMZ resistant U251N sub-clones could be re-sensitized to TMZ by co-treatment with the PARP inhibitor ABT-888, and also whether the emergence of resistance could be suppressed by PARP inhibition. U251N cultures chronically exposed to TMZ developed discrete colonies that expanded during TMZ treatment. These colonies were isolated, expanded further as sub-clones, and assessed for mechanisms of TMZ resistance. Most resistant sub-clones had detectable mutations in one or more mismatch repair (MMR) genes, frequently MSH6, and displayed infrequent re-expression of MGMT. TMZ resistance was associated with isolated poly(ADP-ribose) (pADPr) up-regulation in one sub-clone and was unexplained in several others. TMZ resistant sub-clones regressed during co-treatment with TMZ and ABT-888, and early co-treatment of U251N parental cultures suppressed the emergence of TMZ resistant colonies. In a model of acquired resistance, co-treatment with TMZ and a PARP inhibitor had two important benefits: re-sensitization of TMZ resistant cells and suppression of TMZ resistance.
- Published
- 2020
35. Corrigendum to: Impact of lymphopenia on survival for elderly patients with glioblastoma: A secondary analysis of the CCTG CE.6 (EORTC 26062-22061, TROG 08.02) randomized clinical trial
- Author
-
Andrew J Song, Keyue Ding, Iyad Alnahhas, Normand J Laperriere, James Perry, Warren P Mason, Chad Winch, Chris J O’Callaghan, Johan J Menten, Alba A Brandes, Claire Phillips, Michael F Fay, Ryo Nishikawa, David Osoba, J Gregory Cairncross, Wilson Roa, Wolfgang Wick, and Wenyin Shi
- Published
- 2022
36. Copy-scAT: Deconvoluting single-cell chromatin accessibility of genetic subclones in cancer
- Author
-
Aaron Gillmor, Marco Gallo, Paola Neri, Nizar J. Bahlis, Steven J.M. Jones, Yaoqing Shen, Katrina Ellestad, Divya Singhal, Mathieu Lupien, Michael J. Johnston, Jennifer A. Chan, J. Gregory Cairncross, Ana Nikolic, and Sorana Morrissy
- Subjects
Multidisciplinary ,Cell ,SciAdv r-articles ,Cancer ,Computational biology ,Biology ,medicine.disease ,Chromatin ,medicine.anatomical_structure ,Research Methods ,Cancer cell ,medicine ,Transcriptional regulation ,Biomedicine and Life Sciences ,human activities ,Research Article ,Epigenomics - Abstract
Description, Inferring structural variants in cancer single-cell chromatin accessibility data links genetics to chromatin organization., Single-cell epigenomic assays have tremendous potential to illuminate mechanisms of transcriptional control in functionally diverse cancer cell populations. However, application of these techniques to clinical tumor specimens has been hampered by the current inability to distinguish malignant from nonmalignant cells, which potently confounds data analysis and interpretation. Here, we describe Copy-scAT, an R package that uses single-cell epigenomic data to infer copy number variants (CNVs) that define cancer cells. Copy-scAT enables studies of subclonal chromatin dynamics in complex tumors like glioblastoma. By deploying Copy-scAT, we uncovered potent influences of genetics on chromatin accessibility profiles in individual subclones. Consequently, some genetic subclones were predisposed to acquire stem-like or more differentiated molecular phenotypes, reminiscent of developmental paradigms. Copy-scAT is ideal for studies of the relationships between genetics and epigenetics in malignancies with high levels of intratumoral heterogeneity and to investigate how cancer cells interface with their microenvironment.
- Published
- 2021
37. Impact of lymphopenia on survival for elderly patients with glioblastoma: A secondary analysis of the CCTG CE.6 (EORTC 26062-22061, TROG03.01) randomized clinical trial
- Author
-
Andrew J, Song, Keyue, Ding, Iyad, Alnahhas, Normand J, Laperriere, James, Perry, Warren P, Mason, Chad, Winch, Chris J, O'Callaghan, Johan J, Menten, Alba A, Brandes, Claire, Phillips, Michael F, Fay, Ryo, Nishikawa, David, Osoba, J Gregory, Cairncross, Wilson, Roa, Wolfgang, Wick, and Wenyin, Shi
- Subjects
short-course radiotherapy ,Clinical Investigations ,glioblastoma ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,temozolomide ,lymphopenia ,Corrigendum ,elderly - Abstract
Background Lymphopenia may lead to worse outcomes for glioblastoma patients. This study is a secondary analysis of the CCTG CE.6 trial evaluating the impact of chemotherapy and radiation on lymphopenia, and effects of lymphopenia on overall survival (OS). Methods CCTG CE.6 randomized elderly glioblastoma patients (≥ 65 years) to short-course radiation alone (RT) or short-course radiation with temozolomide (RT + TMZ). Lymphopenia (mild-moderate: grade 1–2; severe: grade 3–4) was defined per CTCAE v3.0, and measured at baseline, 1 week and 4 weeks post-RT. Preselected key factors for analysis included age, sex, ECOG, resection extent, MGMT methylation, Mini-Mental State Examination, and steroid use. Multinomial logistic regression and multivariable Cox regression models were used to identify lymphopenia-associated factors and association with survival. Results Five hundred and sixty-two patients were analyzed (281 RT vs 281 RT+TMZ). At baseline, both arms had similar rates of mild-moderate (21.4% vs 21.4%) and severe (3.2% vs 2.9%) lymphopenia. However, at 4 weeks post-RT, RT+TMZ was more likely to develop lymphopenia (mild-moderate: 27.9% vs 18.2%; severe: 9.3% vs 1.8%; p
- Published
- 2021
38. Capicua regulates neural stem cell proliferation and lineage specification through control of Ets factors
- Author
-
Marco A. Marra, Rajiv Dixit, Mana Alshehri, Alexandra D. Rogers, Michael D. Blough, Stephen M. Robbins, Samuel Lawn, Lata Adnani, Jennifer A. Chan, Carol Schuurmans, Shiekh Tanveer Ahmad, Luke Frankiw, Wei Wu, Myra J. Chen, and J. Gregory Cairncross
- Subjects
Male ,0301 basic medicine ,Cellular differentiation ,General Physics and Astronomy ,02 engineering and technology ,Mice ,Neural Stem Cells ,Stem Cell Research - Nonembryonic - Human ,lcsh:Science ,Cancer ,Mice, Knockout ,Neurons ,Tumor ,Multidisciplinary ,Brain Neoplasms ,Cell Differentiation ,021001 nanoscience & nanotechnology ,Neural stem cell ,Up-Regulation ,Cell biology ,DNA-Binding Proteins ,Oligodendroglia ,Embryo ,Gene Knockdown Techniques ,Neurological ,Knockout mouse ,Female ,Stem Cell Research - Nonembryonic - Non-Human ,Stem cell ,0210 nano-technology ,Lineage (genetic) ,Knockout ,1.1 Normal biological development and functioning ,Science ,Oligodendroglioma ,Primary Cell Culture ,Cell fate determination ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Prosencephalon ,Underpinning research ,Cell Line, Tumor ,Animals ,Humans ,Transcription factor ,Cell Proliferation ,Proto-Oncogene Proteins c-ets ,Mammalian ,Neurosciences ,General Chemistry ,Embryo, Mammalian ,Stem Cell Research ,Xenograft Model Antitumor Assays ,Repressor Proteins ,030104 developmental biology ,Cell culture ,Astrocytes ,lcsh:Q ,Transcription Factors - Abstract
Capicua (Cic) is a transcriptional repressor mutated in the brain cancer oligodendroglioma. Despite its cancer link, little is known of Cic’s function in the brain. We show that nuclear Cic expression is strongest in astrocytes and neurons but weaker in stem cells and oligodendroglial lineage cells. Using a new conditional Cic knockout mouse, we demonstrate that forebrain-specific Cic deletion increases proliferation and self-renewal of neural stem cells. Furthermore, Cic loss biases neural stem cells toward glial lineage selection, expanding the pool of oligodendrocyte precursor cells (OPCs). These proliferation and lineage effects are dependent on de-repression of Ets transcription factors. In patient-derived oligodendroglioma cells, CIC re-expression or ETV5 blockade decreases lineage bias, proliferation, self-renewal, and tumorigenicity. Our results identify Cic as an important regulator of cell fate in neurodevelopment and oligodendroglioma, and suggest that its loss contributes to oligodendroglioma by promoting proliferation and an OPC-like identity via Ets overactivity.
- Published
- 2019
39. CODEL: phase III study of RT, RT + TMZ, or TMZ for newly diagnosed 1p/19q codeleted oligodendroglioma. Analysis from the initial study design
- Author
-
David Schiff, Kurt A. Jaeckle, Evanthia Galanis, Kenneth Aldape, Donald Nordstrom, Stuart A. Grossman, Jeffrey S. Wefel, J. Gregory Cairncross, Michael A. Vogelbaum, Karla V. Ballman, F. Dhermain, Michael Weller, Martin Klein, Patrick J. Flynn, Wolfgang Wick, Paul D. Brown, S. Keith Anderson, Robert B. Jenkins, Caterina Giannini, Jesse G. Dixon, Jeffrey Raizer, Martin J. van den Bent, Jane H. Cerhan, Medical psychology, CCA - Cancer Treatment and quality of life, Neurology, Jaeckle K.A., Ballman K.V., Van Den Bent M., Giannini C., Galanis E., Brown P.D., Jenkins R.B., Cairncross J.G., Wick W., Weller M., Aldape K.D., Dixon J.G., Anderson S.K., Cerhan J.H., Wefel J.S., Klein M., Grossman S.A., Schiff D., Raizer J.J., Dhermain F., Nordstrom D.G., Flynn P.J., and Vogelbaum M.A.
- Subjects
Oncology ,Adult ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,N0577 ,Oligodendroglioma ,Clinical Investigations ,Brain Neoplasm ,SDG 3 - Good Health and Well-being ,Internal medicine ,CODEL ,Clinical endpoint ,1p/19q ,Temozolomide ,Medicine ,Humans ,Adverse effect ,business.industry ,Proportional hazards model ,Brain Neoplasms ,Hazard ratio ,medicine.disease ,Isocitrate Dehydrogenase ,Progression-Free Survival ,Radiation therapy ,Dacarbazine ,codeleted ,Concomitant ,Neurology (clinical) ,business ,medicine.drug ,Human - Abstract
Background We report the analysis involving patients treated on the initial CODEL design. Methods Adults (>18) with newly diagnosed 1p/19q World Health Organization (WHO) grade III oligodendroglioma were randomized to radiotherapy (RT; 5940 centigray ) alone (arm A); RT with concomitant and adjuvant temozolomide (TMZ) (arm B); or TMZ alone (arm C). Primary endpoint was overall survival (OS), arm A versus B. Secondary comparisons were performed for OS and progression-free survival (PFS), comparing pooled RT arms versus TMZ-alone arm. Results Thirty-six patients were randomized equally. At median follow-up of 7.5 years, 83.3% (10/12) TMZ-alone patients progressed, versus 37.5% (9/24) on the RT arms. PFS was significantly shorter in TMZ-alone patients compared with RT patients (hazard ratio [HR] = 3.12; 95% CI: 1.26, 7.69; P = 0.014). Death from disease progression occurred in 3/12 (25%) of TMZ-alone patients and 4/24 (16.7%) on the RT arms. OS did not statistically differ between arms (comparison underpowered). After adjustment for isocitrate dehydrogenase (IDH) status (mutated/wildtype) in a Cox regression model utilizing IDH and RT treatment status as covariables (arm C vs pooled arms A + B), PFS remained shorter for patients not receiving RT (HR = 3.33; 95% CI: 1.31, 8.45; P = 0.011), but not OS ((HR = 2.78; 95% CI: 0.58, 13.22, P = 0.20). Grade 3+ adverse events occurred in 25%, 42%, and 33% of patients (arms A, B, and C). There were no differences between arms in neurocognitive decline comparing baseline to 3 months. Conclusions TMZ-alone patients experienced significantly shorter PFS than patients treated on the RT arms. The ongoing CODEL trial has been redesigned to compare RT + PCV versus RT + TMZ.
- Published
- 2021
40. In the beginning: PDGFA and the genesis of GBM
- Author
-
Michael D. Blough, Pinaki Bose, Mehul Kumar, and J. Gregory Cairncross
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) ,Computational biology ,Biology ,Letters to the Editor - Published
- 2021
41. Proceedings of the Comprehensive Oncology Network Evaluating Rare CNS Tumors (NCI-CONNECT) Oligodendroglioma Workshop
- Author
-
Marta, Penas-Prado, Jing, Wu, Daniel P, Cahill, Daniel J, Brat, Joseph F, Costello, Paul G, Kluetz, J Gregory, Cairncross, Martin, van den Bent, Roel G W, Verhaak, Orwa, Aboud, Peter, Burger, Susan M, Chang, Christine, Cordova, Raymond Y, Huang, Lindsay S, Rowe, Martin J B, Taphoorn, Mark R, Gilbert, Terri S, Armstrong, Kareem, Zaghloul, and Neurology
- Subjects
0301 basic medicine ,Medical education ,business.industry ,Clinical study design ,Cancer ,Review ,medicine.disease ,Call to action ,Data sharing ,Outreach ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,SDG 3 - Good Health and Well-being ,030220 oncology & carcinogenesis ,General partnership ,Health care ,medicine ,Oligodendroglioma ,business - Abstract
Background Oligodendroglioma is a rare primary central nervous system (CNS) tumor with highly variable outcome and for which therapy is usually not curative. At present, little is known regarding the pathways involved with progression of oligodendrogliomas or optimal biomarkers for stratifying risk. Developing new therapies for this rare cancer is especially challenging. To overcome these challenges, the neuro-oncology community must be particularly innovative, seeking multi-institutional and international collaborations, and establishing partnerships with patients and advocacy groups thereby ensuring that each patient enrolled in a study is as informative as possible. Methods The mission of the National Cancer Institute’s NCI-CONNECT program is to address the challenges and unmet needs in rare CNS cancer research and treatment by connecting patients, health care providers, researchers, and advocacy organizations to work in partnership. On November 19, 2018, the program convened a workshop on oligodendroglioma, one of the 12 rare CNS cancers included in its initial portfolio. The purpose of this workshop was to discuss scientific progress and regulatory challenges in oligodendroglioma research and develop a call to action to advance research and treatment for this cancer. Results The recommendations of the workshop include a multifaceted and interrelated approach covering: biology and preclinical models, data sharing and advanced molecular diagnosis and imaging; clinical trial design; and patient outreach and engagement. Conclusions The NCI-CONNECT program is well positioned to address challenges in oligodendroglioma care and research in collaboration with other stakeholders and is developing a list of action items for future initiatives.
- Published
- 2020
42. Single-cell landscapes of primary glioblastomas and matched explants and cell lines show variable retention of inter- and intratumor heterogeneity
- Author
-
Véronique G. LeBlanc, Diane L. Trinh, Shaghayegh Aslanpour, Martha Hughes, Dorothea Livingstone, Dan Jin, Bo Young Ahn, Michael D. Blough, J. Gregory Cairncross, Jennifer A. Chan, John J.P. Kelly, and Marco A. Marra
- Subjects
Cancer Research ,Oncology ,Brain Neoplasms ,Humans ,Genomics ,Neoplasm Recurrence, Local ,Glioblastoma ,Cell Line - Abstract
Glioblastomas (GBMs) are aggressive brain tumors characterized by extensive inter- and intratumor heterogeneity. Patient-derived models, such as organoids and explants, have recently emerged as useful models to study such heterogeneity, although the extent to which they can recapitulate GBM genomic features remains unclear. Here, we analyze bulk exome and single-cell genome and transcriptome profiles of 12 IDH wild-type GBMs, including two recurrent tumors, and of patient-derived explants (PDEs) and gliomasphere (GS) lines derived from these tumors. We find that PDEs are genetically similar to, and variably retain gene expression characteristics of, their parent tumors. Notably, PDEs appear to exhibit similar levels of transcriptional heterogeneity compared with their parent tumors, whereas GS lines tend to be enriched for cells in a more uniform transcriptional state. The approaches and datasets introduced here will provide a valuable resource to help guide experiments using GBM-derived models, especially in the context of studying cellular heterogeneity.
- Published
- 2022
43. Molecular Determinants of Prognosis and Evolution in Diffuse-Lower Grade Astrocytomas
- Author
-
Pinaki Bose, Mehul Kumar, J. Gregory Cairncross, and Michael D. Blough
- Subjects
Chromosome 7 (human) ,Transcriptome ,Isocitrate dehydrogenase ,Mutant ,Gene expression ,Cancer research ,Gene family ,Methylation ,Biology ,Gene - Abstract
Low grade astrocytomas (LGAs) are classified based on the mutational status of the isocitrate dehydrogenase (IDH) gene. While IDH wild-type (WT) LGAs evolve rapidly to glioblastoma, mutant tumors generally have a more indolent course. To identify potential drivers of the differential progression of LGAs, we analyzed transcriptomes from The Cancer Genome Atlas. Compared to mutant LGAs, platelet-derived growth factor (PDGF) signaling is enriched in WT cases, andPDGFAis the top overexpressed gene in the pathway. Putative mechanisms for differentialPDGFAexpression included copy number gains of chromosome 7 in WT cases and methylation of thePDGFApromoter in mutant LGAs. Additionally, we found that highPDGFAexpression is associated with aneuploidy, immunosuppressive features, and worse prognosis, and that WT LGAs use multiple means to inactivate the p53 pathway to progress to GBM. Our work highlights the contribution of PDGF gene family towards the unique behaviour of LGAs.STATEMENT OF SIGNIFICANCEThis study of gene expression in LGAs suggests that differential regulation of the PDGF pathway may underlie the different natural histories ofIDHWT andIDHmutant LGAs including divergent evolutionary trajectories to GBM. This insight may inspire new therapeutic strategies to suppress the transformation of LGAs to higher-grade cancers.
- Published
- 2020
44. Temozolomide and seizure outcomes in a randomized clinical trial of elderly glioblastoma patients
- Author
-
Seth A, Climans, Alba A, Brandes, J Gregory, Cairncross, Keyue, Ding, Michael, Fay, Normand, Laperriere, Johan, Menten, Ryo, Nishikawa, Christopher J, O'Callaghan, James R, Perry, Claire, Phillips, Wilson, Roa, Wolfgang, Wick, Chad, Winch, and Warren P, Mason
- Subjects
Male ,Radiotherapy ,Brain Neoplasms ,Chemoradiotherapy ,Prognosis ,Survival Rate ,Seizures ,Quality of Life ,Temozolomide ,Humans ,Female ,Glioblastoma ,Antineoplastic Agents, Alkylating ,Aged ,Follow-Up Studies - Abstract
Tumor-related epilepsy may respond to chemotherapy. In a previously-published multi-centre randomized clinical trial of 562 elderly glioblastoma patients, temozolomide plus short-course radiotherapy conferred a survival benefit over radiotherapy alone. Seizure outcomes were not reported.We performed an unplanned secondary analysis of this trial's data. The trial design has been previously reported. Seizures were recorded by clinicians as adverse events and by patients in quality of life questionnaires. A Chi-square test of seizure rates between the two groups (α = 0.05) and a Kaplan-Meier estimator of time-to-first self-reported seizure were planned.Almost all patients were followed until they died. In the radiotherapy alone group, 68 patients (24%) had a documented or self-reported seizure versus 83 patients (30%) in the temozolomide plus radiotherapy group, Chi-square analysis showed no difference (p = 0.15). Patients receiving radiotherapy alone tended to develop seizures earlier than those receiving temozolomide plus radiotherapy (p = 0.054). Patients with seizures had shorter overall survival than those without seizures (hazard ratio 1.24, p = 0.02).This study was not powered to detect differences in seizure outcomes, but temozolomide seemed to have minimal impact on seizure control in elderly patients with glioblastoma.NCT00482677 2007-06-05.
- Published
- 2020
45. 2-Hydroxyglutarate-Mediated Autophagy of the Endoplasmic Reticulum Leads to an Unusual Downregulation of Phospholipid Biosynthesis in Mutant IDH1 Gliomas
- Author
-
Marina Radoul, Jose L. Izquierdo-Garcia, Russell O. Pieper, Joanna J. Phillips, Wei Qiang Ong, Bo Huang, Pavithra Viswanath, J. Gregory Cairncross, Hema Artee Luchman, and Sabrina M. Ronen
- Subjects
0301 basic medicine ,Cancer Research ,Biopsy ,Endoplasmic Reticulum ,Mice ,chemistry.chemical_compound ,Models ,2.1 Biological and endogenous factors ,Aetiology ,Phospholipids ,Cancer ,Tumor ,Glioma ,Magnetic Resonance Imaging ,Isocitrate Dehydrogenase ,Cell biology ,Gene Expression Regulation, Neoplastic ,Oncology ,lipids (amino acids, peptides, and proteins) ,Oncology and Carcinogenesis ,Models, Biological ,Article ,Cell Line ,Glutarates ,03 medical and health sciences ,Rare Diseases ,Downregulation and upregulation ,Cell Line, Tumor ,Autophagy ,medicine ,Animals ,Humans ,Gene Silencing ,Oncology & Carcinogenesis ,Phosphatidylethanolamine ,Neoplastic ,Endoplasmic reticulum ,Biological ,medicine.disease ,Brain Disorders ,Brain Cancer ,030104 developmental biology ,Gene Expression Regulation ,chemistry ,Cell culture ,Mutation ,Proteolysis ,Cancer cell ,Unfolded protein response ,Biomarkers - Abstract
Tumor metabolism is reprogrammed to meet the demands of proliferating cancer cells. In particular, cancer cells upregulate synthesis of the membrane phospholipids phosphatidylcholine (PtdCho) and phosphatidylethanolamine (PtdE) in order to allow for rapid membrane turnover. Nonetheless, we show here that, in mutant isocitrate dehydrogenase 1 (IDHmut) gliomas, which produce the oncometabolite 2-hydroxyglutarate (2-HG), PtdCho and PtdE biosynthesis is downregulated and results in lower levels of both phospholipids when compared with wild-type IDH1 cells. 2-HG inhibited collagen-4-prolyl hydroxylase activity, leading to accumulation of misfolded procollagen-IV in the endoplasmic reticulum (ER) of both genetically engineered and patient-derived IDHmut glioma models. The resulting ER stress triggered increased expression of FAM134b, which mediated autophagic degradation of the ER (ER-phagy) and a reduction in the ER area. Because the ER is the site of phospholipid synthesis, ER-phagy led to reduced PtdCho and PtdE biosynthesis. Inhibition of ER-phagy via pharmacological or molecular approaches restored phospholipid biosynthesis in IDHmut glioma cells, triggered apoptotic cell death, inhibited tumor growth, and prolonged the survival of orthotopic IDHmut glioma-bearing mice, pointing to a potential therapeutic opportunity. Glioma patient biopsies also exhibited increased ER-phagy and downregulation of PtdCho and PtdE levels in IDHmut samples compared with wild-type, clinically validating our observations. Collectively, this study provides detailed and clinically relevant insights into the functional link between oncometabolite-driven ER-phagy and phospholipid biosynthesis in IDHmut gliomas. Significance: Downregulation of phospholipid biosynthesis via ER-phagy is essential for proliferation and clonogenicity of mutant IDH1 gliomas, a finding with immediate therapeutic implications. Cancer Res; 78(9); 2290–304. ©2018 AACR.
- Published
- 2018
46. NCOG-12. COGNITIVE FUNCTION (CF) & QUALITY OF LIFE (QOL) IN PATIENTS TREATED WITH PROCARBAZINE, CCNU, & VINCRISTINE (PCV) + RADIOTHERAPY (RT) VS. RT FOR ANAPLASTIC OLIGODENDROGLIOMA (AO) ON NRG RTOG TRIAL 9402
- Author
-
Minhee Won, Terrence P. Cescon, Wilson Roa, Timothy K. Nguyen, Fabio M. Iwamoto, Igor Barani, J. Gregory Cairncross, Minesh P. Mehta, Mei Polley, Jiayi Huang, Jean-Paul Bahary, Luis Souhami, Alan C. Hartford, Jan C. Buckner, Mark V. Mishra, Karen Fink, Laura Donovan, and Anthony T. Pu
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Vincristine ,business.industry ,medicine.medical_treatment ,Anaplastic oligodendroglioma ,Cognition ,26th Annual Meeting & Education Day of the Society for Neuro-Oncology ,Procarbazine ,humanities ,Radiation therapy ,Quality of life ,Internal medicine ,medicine ,In patient ,Neurology (clinical) ,business ,medicine.drug - Abstract
BACKGROUND PCV+RT substantially prolongs survival in AO patients, but long-term CF and QOL implications are unclear. We compared CF and QOL by treatment arm in RTOG 9402 participants and evaluated the impact that baseline characteristics had on CF, QOL, and survival. METHODS CF and QOL were evaluated using the Mini Mental State Exam (MMSE) and Brain-Quality of Life (B-QOL) scale at baseline and annually. Scores were analyzed between treatment arms at each time point for patients with ≥ 10 years of follow-up data. Shared parameter models evaluated MMSE and B-QOL scores and survival for all patients. RESULTS 42/148 (28.4%) participants in PCV+RT and 20/143 (14%) in RT alone arms survived ≥ 10 years. 35/42 and 39/42 (PCV+RT) and 18/20 and 17/20 (RT) participants completed baseline B-QOL and MMSE assessments, respectively. B-QOL scores did not differ between treatment groups at any time-point. Among 16 patients (10 PCV+RT, 6 RT) who completed year 10 MMSE evaluations, mean MMSE score at 10 years was higher in the RT arm (29.83 [95% CI 22.1, 30.0] vs. 26.50 [95% CI 29.4, 30.0], P= 0.04). Change in MMSE and B-QOL scores from baseline did not differ significantly between treatment groups at any time. In shared parameter models including all patients with baseline assessments, MMSE and B-QOL scores decreased over time (MMSE P= 0.0189, B-QOL P= 0.0005), but this did not differ by treatment group (MMSE P= 0.5727, B-QOL P= 0.3592). Younger age and higher KPS predicted better scores (MMSE P < 0.0001, P = 0.0002; B-QOL P = 0.0043, P = 0.0007). PCV+RT predicted better survival in both models. CONCLUSIONS PCV+RT improves survival in AO. Shared parameter models show decrease in MMSE and B-QOL over time. However, relative to RT alone, the addition of PCV did not impact change in CF and QOL over time.
- Published
- 2021
47. PATH-11. PDGFA INITIATES ABERRANT MITOSIS AND MALIGNANT TRANSFORMATION OF NEURAL PROGENITOR CELLS
- Author
-
Cameron J. Grisdale, Michael D. Blough, Hiba Omairi, and J. Gregory Cairncross
- Subjects
Cancer Research ,Oncology ,Path (graph theory) ,Neurology (clinical) ,Biology ,Mitosis ,Neural stem cell ,Malignant transformation ,Cell biology - Abstract
BACKGROUND Imagining ways to prevent or treat glioblastoma (GBM) have been hindered by a lack of understanding of its pathogenesis. Although platelet derived growth factor-A (PDGFA) overexpression may be an early event, critical details of the biology of GBM, and tools to study its initiation have been lacking. Indeed, many PDGF-driven models replicate its microscopic appearance, but not genomic architecture. Recently, we reported an in vitro model of GBM initiation that overcomes this barrier to authenticity. METHODS We used a method developed to establish neural stem cell cultures to investigate the effects of PDGF-A on cells derived from the subventricular zone (SVZ), a putative region where the cells of origins for GBM are derived. We micro-dissect SVZ tissue from p53-null and wild-type adult mice, culture cells in media supplemented with PDGF-A, and assess cell viability, proliferation, mitotic capacity, and genome stability. RESULTS Paradoxical to its canonical role as a growth factor, we observe abrupt and substantial cell death in PDGF-A. Abnormal mitosis was the first observable alteration and occurred immediately in cells of both p53 wild-type and null genotypes: wild-type cells did not survive in PDGF-A, whereas a fraction of null cells evade apoptosis. Evading cells displayed attenuated proliferation accompanied by early chromosomal gains and losses. After approximately 100 days in PDGF-A, surviving cells suddenly proliferate rapidly, acquire growth factor independence, and become tumorigenic in immune-competent mice. Transformed cells continue to display highly abnormal mitotic phenotypes with complex karyotypes similar to GBM, had a neural progenitor cell (NPC) lineage profile, and were resistant to PDGFR-alpha inhibition. CONCLUSION Abnormal mitosis induced by PDGF-A initiates and perpetuates the genome instability that transforms p53-null neural progenitor cells to yield cancers with the types of recurring chromosomal gains and losses that characterize human GBM.
- Published
- 2021
48. Short-Course Radiation plus Temozolomide in Elderly Patients with Glioblastoma
- Author
-
Enrico Franceschi, J. Gregory Cairncross, Florence Laigle-Donadey, Christopher J. O'Callaghan, Keyue Ding, Wilson Roa, Olivier Chinot, James Perry, Michael Tills, John P. Rossiter, Warren P. Mason, Johan Menten, Michael Fay, Alba A. Brandes, Wolfgang Wick, Vassilis Golfinopoulos, Ryo Nishikawa, H Hirte, Claire Phillips, Normand Laperriere, Antje Wick, David Osoba, Brigitta G. Baumert, Chad Winch, Michael Back, L. Fariselli, Arjun Sahgal, Loïc Feuvret, RS: GROW - R3 - Innovative Cancer Diagnostics & Therapy, and Radiotherapie
- Subjects
Male ,medicine.medical_specialty ,Randomization ,medicine.medical_treatment ,ADJUVANT ,Central Nervous System Neoplasms ,MULTIFORME ,03 medical and health sciences ,0302 clinical medicine ,CONCOMITANT ,Temozolomide ,medicine ,Humans ,PROMOTER METHYLATION ,Survival analysis ,Aged ,Aged, 80 and over ,Chemotherapy ,PHASE-3 TRIAL ,business.industry ,Hazard ratio ,Chemoradiotherapy ,General Medicine ,CHEMOTHERAPY ,Survival Analysis ,TUMORS ,Surgery ,Dacarbazine ,Radiation therapy ,030220 oncology & carcinogenesis ,Concomitant ,MALIGNANT ASTROCYTOMA ,Disease Progression ,Quality of Life ,PATTERNS ,Female ,Glioblastoma ,business ,030217 neurology & neurosurgery ,medicine.drug ,RADIOTHERAPY - Abstract
BACKGROUND Glioblastoma is associated with a poor prognosis in the elderly. Survival has been shown to increase among patients 70 years of age or younger when temozolomide chemotherapy is added to standard radiotherapy (60 Gy over a period of 6 weeks). In elderly patients, more convenient shorter courses of radiotherapy are commonly used, but the benefit of adding temozolomide to a shorter course of radiotherapy is unknown.METHODS We conducted a trial involving patients 65 years of age or older with newly diagnosed glioblastoma. Patients were randomly assigned to receive either radiotherapy alone (40 Gy in 15 fractions) or radiotherapy with concomitant and adjuvant temozolomide.RESULTS A total of 562 patients underwent randomization, 281 to each group. The median age was 73 years (range, 65 to 90). The median overall survival was longer with radiotherapy plus temozolomide than with radiotherapy alone (9.3 months vs. 7.6 months; hazard ratio for death, 0.67; 95% confidence interval [CI], 0.56 to 0.80; PCONCLUSIONS In elderly patients with glioblastoma, the addition of temozolomide to short-course radiotherapy resulted in longer survival than short-course radiotherapy alone. Funded by the Canadian Cancer Society Research Institute and others; ClinicalTrials. gov number, NCT00482677.)
- Published
- 2017
49. TMOD-02. A MODEL OF THE INITIATION AND GENOMIC LANDSCAPE OF GLIOBLASTOMA (GBM)
- Author
-
Carmen Binding, Cameron J. Grisdale, Matthaeus Ware, Nick Chahley, Kaitlin Thomas, Haley Pedersen, Michael D. Blough, Hiba Omairi, Jessica DePetro, J. Gregory Cairncross, Alexandra Bohm, Sam Lawn, Shazreh Bukhari, Amanda Gerber, and Cindy Chen
- Subjects
Cancer Research ,Oncology ,Tumor Models ,Cancer research ,medicine ,Neurology (clinical) ,Biology ,medicine.disease ,Glioblastoma - Abstract
IDH-wildtype GBM is the most common variant of this cancer and occurs in older adults. Unfortunately patients’ tumors are either inherently resistant to standard treatment, which includes radio- and chemo-therapy, or acquire resistance during the therapeutic process. Additionally, although effective in other cancers, targeted therapies have yielded disappointing results in GBM, perhaps because the fully developed disease has significant cellular and molecular heterogeneity, allowing the tumour to adapt to treatments. Better insight into managing GBM might result from a detailed knowledge of its initiating events, which have not yet been elucidated. With this in mind, we developed a mouse model of GBM in which the earliest stages can be studied. This ex vivo model recreates GBM by culturing subventricular zone (SVZ) cells, the putative ‘cell of origin’ of GBM in platelet-derived growth factor A (PDGFA). Under this condition SVZ cells from p53 null mice transform, becoming exogenous growth factor independent and tumorigenic in immune-competent mice. In contrast, wildtype SVZ cells do not proliferate in PDGFA and null cells in EGF/FGF do not transform. To discover why p53 null SVZ cells uniquely transform in PDGFA, we performed array comparative genomic hybridization (aCGH) on cells before and after transformation in PDGFA and whole genome sequencing (WGS) on transformed cells and tumours generated from PDGFA-transformed cells. aCGH and WGS revealed that the genomic landscape of transformed cells displayed a striking similarity to that observed in primary human GBM. Specifically, these studies showed that chromosomal alterations are a hallmark of culturing SVZ cells in PDGFA, an intriguing finding considering GBM is also characterized by a specific landscape of copy number alterations. This model may resemble the pathogenesis of human GBM and be leveraged to investigate the early stages of tumorigenesis, further leading to the development of preventative strategies and novel therapeutics.
- Published
- 2019
50. Comprehensive genomic profiling of glioblastoma tumors, BTICs, and xenografts reveals stability and adaptation to growth environments
- Author
-
Yaoqing Shen, Julia L. MacIsaac, Samuel Weiss, Andrew J. Mungall, Donna L. Senger, Yussanne Ma, Sumaiya A. Islam, J. Gregory Cairncross, Eric Y. Zhao, Steven J.M. Jones, David L. Kaplan, Jake Lever, Xueqing Lun, H. Artee Luchman, Jennifer A. Chan, Alice Yijun Wang, Pinaki Bose, Marco A. Marra, Michael S. Kobor, Stephen M. Robbins, Michael D. Blough, Cameron J. Grisdale, Natalie Grinshtein, and Richard A. Moore
- Subjects
Adult ,Male ,Brain tumor ,Somatic hypermutation ,Apoptosis ,Computational biology ,Mice, SCID ,Biology ,Genome ,Transcriptome ,03 medical and health sciences ,Mice ,0302 clinical medicine ,In vivo ,medicine ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Tumor Microenvironment ,Animals ,Humans ,030304 developmental biology ,Epigenomics ,Aged ,Cell Proliferation ,Aged, 80 and over ,0303 health sciences ,Multidisciplinary ,Massive parallel sequencing ,Whole Genome Sequencing ,Brain Neoplasms ,Gene Expression Profiling ,RNA ,Genomics ,DNA Methylation ,Middle Aged ,medicine.disease ,Xenograft Model Antitumor Assays ,3. Good health ,Gene Expression Regulation, Neoplastic ,PNAS Plus ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Case-Control Studies ,Neoplastic Stem Cells ,Female ,Glioblastoma - Abstract
Glioblastoma multiforme (GBM) is the most deadly brain tumor, and currently lacks effective treatment options. Brain tumor-initiating cells (BTICs) and orthotopic xenografts are widely used in investigating GBM biology and new therapies for this aggressive disease. However, the genomic characteristics and molecular resemblance of these models to GBM tumors remain undetermined. We used massively parallel sequencing technology to decode the genomes and transcriptomes of BTICs and xenografts and their matched tumors in order to delineate the potential impacts of the distinct growth environments. Using data generated from whole-genome sequencing of 201 samples and RNA sequencing of 118 samples, we show that BTICs and xenografts resemble their parental tumor at the genomic level but differ at the mRNA expression and epigenomic levels, likely due to the different growth environment for each sample type. These findings suggest that a comprehensive genomic understanding of in vitro and in vivo GBM model systems is crucial for interpreting data from drug screens, and can help control for biases introduced by cell-culture conditions and the microenvironment in mouse models. We also found that lack of MGMT expression in pretreated GBM is linked to hypermutation, which in turn contributes to increased genomic heterogeneity and requires new strategies for GBM treatment.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.