15 results on '"Dioval Remonde"'
Search Results
2. Table S6 from Genomic and Phenotypic Characterization of a Broad Panel of Patient-Derived Xenografts Reflects the Diversity of Glioblastoma
- Author
-
Jann N. Sarkaria, Ian F. Parney, Robert B. Jenkins, Caterina Giannini, Nhan L. Tran, Brian P. O'Neill, Fredrick B. Meyer, Terry C. Burns, Erik P. Sulman, Roel G. Verhaak, Jeanette E. Eckel-Passow, Daniel H. LaChance, Andrea Califano, Eric W. Klee, Bianca M. Marin, Qianghu Wang, Michael E. Berens, Harshil D. Dhruv, Huihuang Yan, Paul A. Decker, Lisa Evers, Gobinda Sarkar, Daniel J. Ma, Brett L. Carlson, Sen Peng, Rebecca Grove, Thomas M. Kollmeyer, Alissa Caron, Gaspar J. Kitange, Ann C. Mladek, Mark A. Schroeder, Dioval Remonde, Shulan Tian, and Rachael A. Vaubel
- Abstract
Molecular subtype, MGMT methylation, expressed fusions
- Published
- 2023
3. Table S2 from Genomic and Phenotypic Characterization of a Broad Panel of Patient-Derived Xenografts Reflects the Diversity of Glioblastoma
- Author
-
Jann N. Sarkaria, Ian F. Parney, Robert B. Jenkins, Caterina Giannini, Nhan L. Tran, Brian P. O'Neill, Fredrick B. Meyer, Terry C. Burns, Erik P. Sulman, Roel G. Verhaak, Jeanette E. Eckel-Passow, Daniel H. LaChance, Andrea Califano, Eric W. Klee, Bianca M. Marin, Qianghu Wang, Michael E. Berens, Harshil D. Dhruv, Huihuang Yan, Paul A. Decker, Lisa Evers, Gobinda Sarkar, Daniel J. Ma, Brett L. Carlson, Sen Peng, Rebecca Grove, Thomas M. Kollmeyer, Alissa Caron, Gaspar J. Kitange, Ann C. Mladek, Mark A. Schroeder, Dioval Remonde, Shulan Tian, and Rachael A. Vaubel
- Abstract
Patient characteristics of viable PDX lines
- Published
- 2023
4. Data from Genomic and Phenotypic Characterization of a Broad Panel of Patient-Derived Xenografts Reflects the Diversity of Glioblastoma
- Author
-
Jann N. Sarkaria, Ian F. Parney, Robert B. Jenkins, Caterina Giannini, Nhan L. Tran, Brian P. O'Neill, Fredrick B. Meyer, Terry C. Burns, Erik P. Sulman, Roel G. Verhaak, Jeanette E. Eckel-Passow, Daniel H. LaChance, Andrea Califano, Eric W. Klee, Bianca M. Marin, Qianghu Wang, Michael E. Berens, Harshil D. Dhruv, Huihuang Yan, Paul A. Decker, Lisa Evers, Gobinda Sarkar, Daniel J. Ma, Brett L. Carlson, Sen Peng, Rebecca Grove, Thomas M. Kollmeyer, Alissa Caron, Gaspar J. Kitange, Ann C. Mladek, Mark A. Schroeder, Dioval Remonde, Shulan Tian, and Rachael A. Vaubel
- Abstract
Purpose:Glioblastoma is the most frequent and lethal primary brain tumor. Development of novel therapies relies on the availability of relevant preclinical models. We have established a panel of 96 glioblastoma patient-derived xenografts (PDX) and undertaken its genomic and phenotypic characterization.Experimental Design:PDXs were established from glioblastoma, IDH-wildtype (n = 93), glioblastoma, IDH-mutant (n = 2), diffuse midline glioma, H3 K27M-mutant (n = 1), and both primary (n = 60) and recurrent (n = 34) tumors. Tumor growth rates, histopathology, and treatment response were characterized. Integrated molecular profiling was performed by whole-exome sequencing (WES, n = 83), RNA-sequencing (n = 68), and genome-wide methylation profiling (n = 76). WES data from 24 patient tumors was compared with derivative models.Results:PDXs recapitulate many key phenotypic and molecular features of patient tumors. Orthotopic PDXs show characteristic tumor morphology and invasion patterns, but largely lack microvascular proliferation and necrosis. PDXs capture common and rare molecular drivers, including alterations of TERT, EGFR, PTEN, TP53, BRAF, and IDH1, most at frequencies comparable with human glioblastoma. However, PDGFRA amplification was absent. RNA-sequencing and genome-wide methylation profiling demonstrated broad representation of glioblastoma molecular subtypes. MGMT promoter methylation correlated with increased survival in response to temozolomide. WES of 24 matched patient tumors showed preservation of most genetic driver alterations, including EGFR amplification. However, in four patient–PDX pairs, driver alterations were gained or lost on engraftment, consistent with clonal selection.Conclusions:Our PDX panel captures the molecular heterogeneity of glioblastoma and recapitulates many salient genetic and phenotypic features. All models and genomic data are openly available to investigators.
- Published
- 2023
5. Table S4 from Genomic and Phenotypic Characterization of a Broad Panel of Patient-Derived Xenografts Reflects the Diversity of Glioblastoma
- Author
-
Jann N. Sarkaria, Ian F. Parney, Robert B. Jenkins, Caterina Giannini, Nhan L. Tran, Brian P. O'Neill, Fredrick B. Meyer, Terry C. Burns, Erik P. Sulman, Roel G. Verhaak, Jeanette E. Eckel-Passow, Daniel H. LaChance, Andrea Califano, Eric W. Klee, Bianca M. Marin, Qianghu Wang, Michael E. Berens, Harshil D. Dhruv, Huihuang Yan, Paul A. Decker, Lisa Evers, Gobinda Sarkar, Daniel J. Ma, Brett L. Carlson, Sen Peng, Rebecca Grove, Thomas M. Kollmeyer, Alissa Caron, Gaspar J. Kitange, Ann C. Mladek, Mark A. Schroeder, Dioval Remonde, Shulan Tian, and Rachael A. Vaubel
- Abstract
Frequency of genomic alterations in PDX compared to TCGA
- Published
- 2023
6. Table S3 from Genomic and Phenotypic Characterization of a Broad Panel of Patient-Derived Xenografts Reflects the Diversity of Glioblastoma
- Author
-
Jann N. Sarkaria, Ian F. Parney, Robert B. Jenkins, Caterina Giannini, Nhan L. Tran, Brian P. O'Neill, Fredrick B. Meyer, Terry C. Burns, Erik P. Sulman, Roel G. Verhaak, Jeanette E. Eckel-Passow, Daniel H. LaChance, Andrea Califano, Eric W. Klee, Bianca M. Marin, Qianghu Wang, Michael E. Berens, Harshil D. Dhruv, Huihuang Yan, Paul A. Decker, Lisa Evers, Gobinda Sarkar, Daniel J. Ma, Brett L. Carlson, Sen Peng, Rebecca Grove, Thomas M. Kollmeyer, Alissa Caron, Gaspar J. Kitange, Ann C. Mladek, Mark A. Schroeder, Dioval Remonde, Shulan Tian, and Rachael A. Vaubel
- Abstract
Short tandem repeat genotype and PDX growth characteristics
- Published
- 2023
7. Table S7 from Genomic and Phenotypic Characterization of a Broad Panel of Patient-Derived Xenografts Reflects the Diversity of Glioblastoma
- Author
-
Jann N. Sarkaria, Ian F. Parney, Robert B. Jenkins, Caterina Giannini, Nhan L. Tran, Brian P. O'Neill, Fredrick B. Meyer, Terry C. Burns, Erik P. Sulman, Roel G. Verhaak, Jeanette E. Eckel-Passow, Daniel H. LaChance, Andrea Califano, Eric W. Klee, Bianca M. Marin, Qianghu Wang, Michael E. Berens, Harshil D. Dhruv, Huihuang Yan, Paul A. Decker, Lisa Evers, Gobinda Sarkar, Daniel J. Ma, Brett L. Carlson, Sen Peng, Rebecca Grove, Thomas M. Kollmeyer, Alissa Caron, Gaspar J. Kitange, Ann C. Mladek, Mark A. Schroeder, Dioval Remonde, Shulan Tian, and Rachael A. Vaubel
- Abstract
PDX Response to Standard Therapy
- Published
- 2023
8. Table S1 from Genomic and Phenotypic Characterization of a Broad Panel of Patient-Derived Xenografts Reflects the Diversity of Glioblastoma
- Author
-
Jann N. Sarkaria, Ian F. Parney, Robert B. Jenkins, Caterina Giannini, Nhan L. Tran, Brian P. O'Neill, Fredrick B. Meyer, Terry C. Burns, Erik P. Sulman, Roel G. Verhaak, Jeanette E. Eckel-Passow, Daniel H. LaChance, Andrea Califano, Eric W. Klee, Bianca M. Marin, Qianghu Wang, Michael E. Berens, Harshil D. Dhruv, Huihuang Yan, Paul A. Decker, Lisa Evers, Gobinda Sarkar, Daniel J. Ma, Brett L. Carlson, Sen Peng, Rebecca Grove, Thomas M. Kollmeyer, Alissa Caron, Gaspar J. Kitange, Ann C. Mladek, Mark A. Schroeder, Dioval Remonde, Shulan Tian, and Rachael A. Vaubel
- Abstract
Histologic diagnoses for all tumors implanted (WHO grade II-IV)
- Published
- 2023
9. Supplementary Materials from Genomic and Phenotypic Characterization of a Broad Panel of Patient-Derived Xenografts Reflects the Diversity of Glioblastoma
- Author
-
Jann N. Sarkaria, Ian F. Parney, Robert B. Jenkins, Caterina Giannini, Nhan L. Tran, Brian P. O'Neill, Fredrick B. Meyer, Terry C. Burns, Erik P. Sulman, Roel G. Verhaak, Jeanette E. Eckel-Passow, Daniel H. LaChance, Andrea Califano, Eric W. Klee, Bianca M. Marin, Qianghu Wang, Michael E. Berens, Harshil D. Dhruv, Huihuang Yan, Paul A. Decker, Lisa Evers, Gobinda Sarkar, Daniel J. Ma, Brett L. Carlson, Sen Peng, Rebecca Grove, Thomas M. Kollmeyer, Alissa Caron, Gaspar J. Kitange, Ann C. Mladek, Mark A. Schroeder, Dioval Remonde, Shulan Tian, and Rachael A. Vaubel
- Abstract
Figures S1-S8, Supplementary Methods, Supplementary Bibliography
- Published
- 2023
10. Genomic and phenotypic characterization of a broad panel of patient-derived xenografts reflects the diversity of glioblastoma
- Author
-
Shulan Tian, Thomas M. Kollmeyer, Erik P. Sulman, Caterina Giannini, Dioval Remonde, Andrea Califano, Paul A. Decker, Huihuang Yan, Jeanette E. Eckel-Passow, Robert B. Jenkins, Jann N. Sarkaria, Ann C. Mladek, Terry C. Burns, Gaspar J. Kitange, Mark A. Schroeder, Fredric B. Meyer, Brian P. O'Neill, Brett L. Carlson, Rachael A. Vaubel, Alissa Caron, Daniel J. Ma, Lisa Evers, Eric W. Klee, Gobinda Sarkar, Roel G.W. Verhaak, Michael E. Berens, Nhan L. Tran, Harshil Dhruv, Daniel H. Lachance, Qianghu Wang, Rebecca Grove, Sen Peng, Ian F. Parney, Bianca M Marin, Vaubel R.A., Tian S., Remonde D., Schroeder M.A., Mladek A.C., Kitange G.J., Caron A., Kollmeyer T.M., Grove R., Peng S., Carlson B.L., Ma D.J., Sarkar G., Evers L., Decker P.A., Yan H., Dhruv H.D., Berens M.E., Wang Q., Marin B.M., Klee E.W., Califano A., LaChance D.H., Eckel-Passow J.E., Verhaak R.G., Sulman E.P., Burns T.C., Meyer F.B., O'Neill B.P., Tran N.L., Giannini C., Jenkins R.B., Parney I.F., and Sarkaria J.N.
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Mice ,0302 clinical medicine ,Genotype ,Promoter Regions, Genetic ,DNA Modification Methylases ,Aged, 80 and over ,biology ,Brain Neoplasms ,Middle Aged ,Phenotype ,Isocitrate Dehydrogenase ,ErbB Receptors ,Survival Rate ,Oncology ,030220 oncology & carcinogenesis ,DNA methylation ,Female ,medicine.drug ,Adult ,IDH1 ,Brain tumor ,Article ,Young Adult ,03 medical and health sciences ,Glioma ,Exome Sequencing ,Biomarkers, Tumor ,Temozolomide ,medicine ,Animals ,Humans ,PTEN ,Antineoplastic Agents, Alkylating ,Aged ,Neoplasm Staging ,Tumor Suppressor Proteins ,glioblastoma, xenograft ,DNA Methylation ,medicine.disease ,Xenograft Model Antitumor Assays ,DNA Repair Enzymes ,030104 developmental biology ,Mutation ,biology.protein ,Cancer research ,Glioblastoma - Abstract
Purpose: Glioblastoma is the most frequent and lethal primary brain tumor. Development of novel therapies relies on the availability of relevant preclinical models. We have established a panel of 96 glioblastoma patient-derived xenografts (PDX) and undertaken its genomic and phenotypic characterization. Experimental Design: PDXs were established from glioblastoma, IDH-wildtype (n = 93), glioblastoma, IDH-mutant (n = 2), diffuse midline glioma, H3 K27M-mutant (n = 1), and both primary (n = 60) and recurrent (n = 34) tumors. Tumor growth rates, histopathology, and treatment response were characterized. Integrated molecular profiling was performed by whole-exome sequencing (WES, n = 83), RNA-sequencing (n = 68), and genome-wide methylation profiling (n = 76). WES data from 24 patient tumors was compared with derivative models. Results: PDXs recapitulate many key phenotypic and molecular features of patient tumors. Orthotopic PDXs show characteristic tumor morphology and invasion patterns, but largely lack microvascular proliferation and necrosis. PDXs capture common and rare molecular drivers, including alterations of TERT, EGFR, PTEN, TP53, BRAF, and IDH1, most at frequencies comparable with human glioblastoma. However, PDGFRA amplification was absent. RNA-sequencing and genome-wide methylation profiling demonstrated broad representation of glioblastoma molecular subtypes. MGMT promoter methylation correlated with increased survival in response to temozolomide. WES of 24 matched patient tumors showed preservation of most genetic driver alterations, including EGFR amplification. However, in four patient–PDX pairs, driver alterations were gained or lost on engraftment, consistent with clonal selection. Conclusions: Our PDX panel captures the molecular heterogeneity of glioblastoma and recapitulates many salient genetic and phenotypic features. All models and genomic data are openly available to investigators.
- Published
- 2020
11. Molecular profiling of long-term IDH-wildtype glioblastoma survivors
- Author
-
Dioval Remonde, Cristiane M. Ida, Rachael A. Vaubel, Jann N. Sarkaria, Gaspar J. Kitange, Robert B. Jenkins, Hugues Sicotte, Kathryn L. Kolsky, Danielle M. Burgenske, Ryan S. Youland, Daniel H. Lachance, Matthew L. Kosel, Shiv K. Gupta, Alissa Caron, Paul A. Decker, Caterina Giannini, Ian J Parney, Jeanette E. Eckel-Passow, Emily G. Barr Fritcher, Erik P. Sulman, Jesse S. Voss, Jie Yang, Ann C. Mladek, Benjamin R. Kipp, Fredric B. Meyer, Thomas M. Kollmeyer, Burgenske D.M., Yang J., Decker P.A., Kollmeyer T.M., Kosel M.L., Mladek A.C., Caron A.A., Vaubel R.A., Gupta S.K., Kitange G.J., Sicotte H., Youland R.S., Remonde D., Voss J.S., Fritcher E.G.B., Kolsky K.L., Ida C.M., Meyer F.B., Lachance D.H., Parney I.J., Kipp B.R., Giannini C., Sulman E.P., Jenkins R.B., Eckel-Passow J.E., and Sarkaria J.N.
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,copy number alteration ,mutation analysi ,business.industry ,DNA repair ,Angiogenesis ,Copy number analysis ,glioblastoma ,O-6-methylguanine-DNA methyltransferase ,RNA sequencing ,Methylation ,Isocitrate dehydrogenase ,Internal medicine ,Gene expression ,Medicine ,Neurology (clinical) ,methylation ,business ,Gene - Abstract
BackgroundGlioblastoma (GBM) represents an aggressive cancer type with a median survival of only 14 months. With fewer than 5% of patients surviving 5 years, comprehensive profiling of these rare patients could elucidate prognostic biomarkers that may confer better patient outcomes. We utilized multiple molecular approaches to characterize the largest patient cohort of isocitrate dehydrogenase (IDH)–wildtype GBM long-term survivors (LTS) to date.MethodsRetrospective analysis was performed on 49 archived formalin-fixed paraffin embedded tumor specimens from patients diagnosed with GBM at the Mayo Clinic between December 1995 and September 2013. These patient samples were subdivided into 2 groups based on survival (12 LTS, 37 short-term survivors [STS]) and subsequently examined by mutation sequencing, copy number analysis, methylation profiling, and gene expression.ResultsOf the 49 patients analyzed in this study, LTS were younger at diagnosis (P = 0.016), more likely to be female (P = 0.048), and MGMT promoter methylated (UniD, P = 0.01). IDH-wildtype STS and LTS demonstrated classic GBM mutations and copy number changes. Pathway analysis of differentially expressed genes showed LTS enrichment for sphingomyelin metabolism, which has been linked to decreased GBM growth, invasion, and angiogenesis. STS were enriched for DNA repair and cell cycle control networks.ConclusionsWhile our findings largely report remarkable similarity between these LTS and more typical STS, unique attributes were observed in regard to altered gene expression and pathway enrichment. These attributes may be valuable prognostic markers and are worth further examination. Importantly, this study also underscores the limitations of existing biomarkers and classification methods in predicting patient prognosis.
- Published
- 2019
12. TMOD-18. THE PATIENT DERIVED XENOGRAFT NATIONAL RESOURCE: A COMPREHENSIVE COLLECTION OF HIGH-GRADE GLIOMA MODELS FOR PRE-CLINICAL AND TRANSLATIONAL STUDIES
- Author
-
Gaspar J. Kitange, Nhan L. Tran, Brett L. Carlson, Andrea Califano, Shulan Tian, Rachael A. Vaubel, Eric W. Klee, Michael E. Berens, C. David James, Mark A. Schroeder, Erik P. Sulman, Daniel H. Lachance, Paul A. Decker, Ann C. Mladek, Thomas M. Kollmeyer, Matthew L. Kosel, Daniel J. Ma, Lisa Evers, Roel G.W. Verhaak, Jeanette E. Eckel-Passow, Sen Peng, Jann N. Sarkaria, Ian F. Parney, Robert B. Jenkins, Caterina Giannini, and Dioval Remonde
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,endocrine system ,business.industry ,Abstracts ,Resource (project management) ,Internal medicine ,Medicine ,Neurology (clinical) ,business ,neoplasms ,Tumor xenograft ,High-Grade Glioma - Abstract
Patient derived xenograft (PDX) models have shown great utility for pre-clinical and translational studies for a range of malignancies. We have established a comprehensive, publically available collection of 95 high-grade glioma flank PDX models. Viable PDX were derived from Glioblastoma, IDH-wildtype (n=91), Glioblastoma, IDH-mutant (n=2), Diffuse Midline Glioma, H3 K27M-mutant (n=1), and Anaplastic Oligodendroglioma (n=1) and include both primary (n=60) and recurrent (n=35) tumors. Comprehensive molecular characterization of PDX is ongoing and, to date, has included whole exome sequencing (WES, n=82), RNA-sequencing (n=40), and genome-wide methylation profiling (n=78) that included MGMT promoter, with data available in cBioPortal. PDX reflected the genetic characteristics of glioblastoma, with the majority harboring TERT promoter mutations, chromosomal gain +7, loss -10 and homozygous deletion of CKDN2A/B. EGFR alterations were frequent (~40%) and included amplification, point mutation, EGFRvIII, and other splice variants. Other common alterations, including amplifications of MET, CDK4, CDK6, MDM2, MDM4 and mutation of TP53, PTEN, NF1, RB1, PIK3CA, PIK3R1 were present at similar frequencies reported by TCGA, with the exception of PDGFRA alterations that were underrepresented in PDX. RNA-sequencing showed representation of each of the glioblastoma gene expression subtypes. To assess preservation of genetic features during xeongrafting, we performed WES on 20 matched patient tumors. The vast majority of genetic driver alterations were shared between patient and PDX, including EGFR, EGFRvIII, CDK4, MDM2, and MET amplifications. In 3 PDX, subclonal selection events were observed, including amplifications of MYCN, CDK6 and an EGFR splice variant, as well as selection against PDGFRA amplification. Overall, our large panel of PDX models reflects the genetic heterogeneity of glioblastoma and largely preserves the genetic features of the primary patient tumors. The PDX National Resource is a powerful tool for neuro-oncology research, with all PDX models and genomic data openly available (http://www.mayo.edu/research/labs/translational-neuro-oncology/mayo-clinic-brain-tumor-patient-derived-xenograft-national-resource).
- Published
- 2018
13. GENO-37MOLECULAR PROFILING OF LONG-TERM SURVIVORS OF GLIOBLASTOMA
- Author
-
Alissa Caron, Jason T. Huse, Paul A. Decker, Jann N. Sarkaria, Ryan S. Youland, Robert B. Jenkins, Daniel H. Lachance, Ann C. Mladek, Hughes Sicotte, Benjamin R. Kipp, Dioval Remonde, Jeanette E. Eckel-Passow, and Caterina Giannini
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,Temozolomide ,biology ,business.industry ,Histology ,medicine.disease ,Tumor registry ,Text mining ,Internal medicine ,Cohort ,medicine ,biology.protein ,PTEN ,Neurology (clinical) ,business ,Abstracts from the 20th Annual Scientific Meeting of the Society for Neuro-Oncology ,ATRX ,medicine.drug ,Glioblastoma - Abstract
Few patients with glioblastoma survive beyond 5 years. In this project, the molecular features associated with these exceptional long term survivors (LTS) were evaluated. Patients diagnosed with glioblastoma within the past 30 years surviving >= 5 years were identified from the Mayo tumor registry. Of 1176 patients with glioblastoma in the registry, 40 were LTS (median age 50.4 years; median survival 7.8 years). Pathology was reviewed and diagnosis confirmed by a certified neuropathologist (CG). Histology was fibrillary in 30 (75%) patients. Most (23, 58%) tumors were located in the frontal lobes. Gross total resection was achieved in 26 (65%). Adjuvant radiotherapy was delivered in 35 ( >= 60 Gy in 31). Temozolomide was given in 20 (50%) and BCNU in 12 (30%). In the 22 patients with tissue available for analysis (median age 49.2 years; median survival 7.0 years), TCGA molecular subtype breakdown from RNA extracts analyzed by Nanostring was: proneural 11, neural 0, classical 6, mesenchymal 2, and unknown 3 (insufficient RNA). Next-generation sequencing revealed the following mutations: 12 TERT, 12 ATRX, 12 TP53, 10 IDH, 8 PIK3CA/PIK3R1, 8 PTEN, 4 RB1, and 3 EGFR. 1p19q codeletion (determined via OncoScan assay), TERT, and IDH mutations were used to stratify patients into risk categories: 9 TERT-mutation only, 8 IDH-mutation only, 2 triple positive, and 3 triple negative. The frequency of IDH mutation in LTS (8/22) was significantly higher than observed in a contemporaneous cohort of short-surviving glioblastoma (32/470, p < 0.0001). All IDH mutations were in TCGA proneural patients. Long-term glioblastoma survival is enriched for patients with IDH-mutation only gliomas, although approximately half of tumors have molecular features associated with a more typical aggressive clinical course. Future studies are focused on understanding the mechanisms associated with prolonged survival in the TERT-mutation only tumors.
- Published
- 2015
14. GENE-28. CLINICAL AND MOLECULAR ANALYSES OF LONG-TERM SURVIVORS OF GLIOBLASTOMA
- Author
-
Hugues Sicotte, Daniel H. Lachance, Matthew L. Kosel, Jeanette E. Eckel-Passow, Caterina Giannini, Dioval Remonde, Paul A. Decker, Danielle M. Burgenske, Robert B. Jenkins, Alissa Caron, Ryan S. Youland, Jann N. Sarkaria, and Thomas M. Kollmeyer
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.disease ,Term (time) ,Abstracts ,Text mining ,Internal medicine ,medicine ,Neurology (clinical) ,business ,Gene ,Glioblastoma - Abstract
Glioblastoma (GBM) is a uniformly fatal disease with standard treatments conferring a median overall survival of only 14.6 months. Because of limited local control with these frontline therapies, less than 10% of patients will survive five years. The molecular mechanisms that promote improved therapeutic responses in this long-term survivor group remain unknown. Therefore, we strove to identify features that render these typically refractory tumors highly sensitive to radiation and/or temozolomide (TMZ). We identified GBM patients who survived more than five years (long-term survivors; LTS) and who had tissue available for molecular analysis. These patients were frequency matched to patients who survived less than two years (short-term survivors; STS) by IDH mutation, TERT promoter mutation and 1p/19q codeletion. We focused our analyses on patients who were TERT-mutation only (IDH wild-type and non-codeleted), triple-negative and IDH-mutation only (TERT wild-type and non-codeleted). We molecularly characterized patients using the following technology: Illumina EPIC methylation array, RNA sequencing, Oncoscan copy number array, and DNA mutation sequencing. The Cancer Genome Atlas (TCGA) data were used to classify patients into methylation and RNA expression subtypes. We analyzed 22 LTS (9 TERT-mutation only, 3 triple-negative, 8 IDH-mutation only, and 2 triple positive) and 43 STS (35 TERT-mutation only, 5 triple-negative and 3 IDH-mutation only). Generally, LTS were younger at diagnosis (45 vs. 59 years), experienced longer median survival (6.9 years vs. 1.2 years) and median progression-free survival (5.2 years vs. 0.7 years). LTS were also more likely to have gross total resection (68% vs. 56%; p=0.16). Moreover, STS experienced recurrence at a higher rate than LTS (84% vs 59%; p=0.03). All IDH-mutation only patients were classified into LGm1-LGm2 methylation subtypes, regardless of outcome. Similarly, all TERT-mutation only and triple-negative patients were classified as LGm4-LGm6 methylation subtypes, regardless of outcome. Our results suggest differences exist between LTS and STS.
- Published
- 2017
15. Abstract 1468: Characterization of patient-derived xenograft (PDX) models to evaluate clinical and therapeutic responses of glioblastoma multiforme
- Author
-
Dioval Remonde, Robert B. Jenkins, Nhan L. Tran, Paul A. Decker, Brock Armstrong, Michael E. Berens, Jann N. Sarkaria, Lisa Evers, Brett L. Carlson, Sen Peng, Mark A. Schroeder, and Jeanette E. Eckel Passow
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,Temozolomide ,Bevacizumab ,business.industry ,medicine.medical_treatment ,medicine.disease ,Animal data ,Internal medicine ,medicine ,Histological grades ,business ,Adjuvant ,Tumor xenograft ,Comparative genomic hybridization ,medicine.drug ,Glioblastoma - Abstract
BACKGROUND/PURPOSE The Mayo Clinic has developed a large panel of patient-derived xenografts (PDX) from patients with glioblastoma multiforme (GBM). Here we report initial molecular profiling of the Mayo PDX panel and a comparison of patient and PDX molecular profile and response to therapy. METHODS & MATERIALS Clinical data was retrieved by retrospective chart review. Animal data from all PDX investigations conducted between 2004-2014 were retrieved from experimental logs and subsequently consolidated into a database for analysis. RESULTS From 1999 to 2014, 182 patient tumor samples of varying histological grades were attempted for xenograft with 73 resulting viable PDX lines. Viable xenografts were only produced from WHO grade IV tumor specimens, yielding an overall success rate of 49% for these tumors. GBM patients that produced viable xenografts compared to those that did not exhibited a trend for decreased overall survival (P = 0.18). There was no significant association between successful xenografting and whether tissue was from newly diagnosed (45/93%) or recurrent (20/40; 50%) tumors. Patient age > 45 at diagnosis was correlated with increased PDX viability in GBMs (p = 0.05). Of the viable PDX models analyzed, EGFR mutation was identified in 17 lines, TERT mutation was found in 13 lines, IDH mutation in 1 line, and MGMT hypermethylation in 25 lines. RNAseq was performed on orthotopic tumor samples from 53 PDX models. After excluding contaminating murine sequence reads, expression analysis demonstrated 32 models with a mesenchymal phenotype. Array comparative genomic hybridization was performed on 9 patient samples and derivative early, mid and late passage PDX tumors. Using unbiased hierarchical clustering, there was a high concordance between patient and xenograft models. Within the PDX panel, fractionated radiation (RT) alone and RT combined with temozolomide (TMZ) was tested in orthotopic tumors in 38 lines. The overall median survival benefit (ratio of median survival for treated vs. placebo) in PDX lines treated with RT only was 1.6 (range: 0.9-2.5) and with RT/TMZ was 2.5 (range: 1.1 - 8.9). There was a positive association between observed patient survival and the corresponding survival benefit in the PDX for subjects treated with RT/TMZ (r = 0.2; n = 17). Response to adjuvant TMZ was evaluated in 42 tumor lines, and response to bevacizumab was tested in 33 tumor lines, and correlations with clinical treatment response are being evaluated. CONCLUSIONS: The Mayo GBM PDX panel is widely used in the neuro-oncology community. The initial molecular analysis suggests a good correlation between patient and PDX at the level of genotypic characteristics and therapeutic sensitivity. Citation Format: Dioval A. Remonde, Brett L. Carlson, Mark A. Schroeder, Brock Armstrong, Sen Peng, Lisa Evers, Paul A. Decker, Jeanette Eckel Passow, Michael E. Berens, Nhan L. Tran, Robert B. Jenkins, Jann N. Sarkaria. Characterization of patient-derived xenograft (PDX) models to evaluate clinical and therapeutic responses of glioblastoma multiforme. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1468. doi:10.1158/1538-7445.AM2015-1468
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.