8 results on '"Kollmeyer T.M."'
Search Results
2. Polymorphous Low-Grade Neuroepithelial Tumor of the Young (PLNTY): Molecular Profiling Confirms Frequent MAPK Pathway Activation
- Author
-
Kay Minn, Jaime I. Davila, Cristiane M. Ida, Caterina Giannini, Robert B. Jenkins, Kevin C. Halling, Kar-Ming A Fung, Timothy J. Kaufmann, Jessica R. Balcom, Dong Kun Kim, Numrah Fadra, Asha Nair, Derek R. Johnson, Benjamin R. Kipp, Thomas M. Kollmeyer, Ida C.M., Johnson D.R., Nair A.A., Davila J., Kollmeyer T.M., Minn K., Fadra N.M., Balcom J.R., Fung K.-M.A., Kim D.K., Kaufmann T.J., Kipp B.R., Halling K.C., Jenkins R.B., and Giannini C.
- Subjects
Adult ,MAPK/ERK pathway ,NTRK2 ,Mitogen-Activated Protein Kinase Kinase ,Biology ,medicine.disease_cause ,BRAF ,Pathology and Forensic Medicine ,Polymorphous low-grade neuroepithelial tumor ,Cellular and Molecular Neuroscience ,Recurrence ,Seizures ,Chromosome instability ,KIAA1549 ,Gene duplication ,medicine ,Humans ,Receptor, trkB ,Mitogen-Activated Protein Kinase Kinases ,Mutation ,Membrane Glycoproteins ,Chromosome ,Original Articles ,General Medicine ,Aneuploidy ,medicine.disease ,Seizure ,Neoplasms, Neuroepithelial ,Neuroepithelial cell ,Neurology ,FGFR2 ,Cancer research ,Female ,CD34 ,Membrane Glycoprotein ,Neurology (clinical) ,Oligodendroglioma ,Gene Fusion ,Chromosomes, Human, Pair 9 ,V600E ,Human ,Transcription Factors - Abstract
Polymorphous low-grade neuroepithelial tumor of the young (PLNTY) is a recently described epileptogenic tumor characterized by oligodendroglioma-like components, aberrant CD34 expression, and frequent mitogen-activated protein kinase (MAPK) pathway activation. We molecularly profiled 13 cases with diagnostic histopathological features of PLNTY (10 female; median age, 16 years; range, 5–52). Patients frequently presented with seizures (9 of 12 with available history) and temporal lobe tumors (9 of 13). MAPK pathway activating alterations were identified in all 13 cases. Fusions were present in the 7 youngest patients: FGFR2-CTNNA3 (n = 2), FGFR2-KIAA1598 (FGFR2-SHTN1) (n = 1), FGFR2-INA (n = 1), FGFR2-MPRIP (n = 1), QKI-NTRK2 (n = 1), and KIAA1549-BRAF (n = 1). BRAF V600E mutation was present in 6 patients (17 years or older). Two fusion-positive cases additionally harbored TP53/RB1 abnormalities suggesting biallelic inactivation. Copy number changes predominantly involving whole chromosomes were observed in all 10 evaluated cases, with losses of chromosome 10q occurring with FGFR2-KIAA1598 (SHTN1)/CTNNA3 fusions. The KIAA1549-BRAF and QKI-NTRK2 fusions were associated respectively with a 7q34 deletion and 9q21 duplication. This study shows that despite its name, PLNTY also occurs in older adults, who frequently show BRAF V600E mutation. It also expands the spectrum of the MAPK pathway activating alterations associated with PLNTY and demonstrates recurrent chromosomal copy number changes consistent with chromosomal instability.
- Published
- 2021
3. Using germline variants to estimate glioma and subtype risks
- Author
-
Beatrice Melin, Lucie McCoy, Margaret Wrensch, Bradley J. Erickson, Claudia F. Lucchinetti, Kristen L. Drucker, Matt L. Kosel, Terri Rice, Joseph L. Wiemels, Annette M. Molinaro, Caterina Giannini, Paige M. Bracci, Corinne Praska, Robert B. Jenkins, Terry C. Burns, Paul A. Decker, Jeanette E. Eckel-Passow, Helen M. Hansen, Thomas M. Kollmeyer, Melissa L. Bondy, Melike Pekmezci, John K. Wiencke, Daniel H. Lachance, Alissa Caron, Eckel-Passow J.E., Decker P.A., Kosel M.L., Kollmeyer T.M., Molinaro A.M., Rice T., Caron A.A., Drucker K.L., Praska C.E., Pekmezci M., Hansen H.M., Mccoy L.S., Bracci P.M., Erickson B.J., Lucchinetti C.F., Wiemels J.L., Wiencke J.K., Bondy M.L., Melin B., Burns T.C., Giannini C., Lachance D.H., Wrensch M.R., and Jenkins R.B.
- Subjects
Male ,Cancer Research ,genotype ,Age at diagnosis ,polygenic ,Germline ,Diffuse Glioma ,0302 clinical medicine ,Risk Factors ,glioma ,Genotype ,80 and over ,Cancer ,Aged, 80 and over ,Brain Neoplasms ,Single Nucleotide ,Middle Aged ,classification ,Oncology ,030220 oncology & carcinogenesis ,Basic and Translational Investigations ,Female ,Adult ,Adolescent ,Oncology and Carcinogenesis ,Single-nucleotide polymorphism ,Biology ,Polymorphism, Single Nucleotide ,Young Adult ,03 medical and health sciences ,Rare Diseases ,Clinical Research ,Glioma ,Genetics ,medicine ,Humans ,Oncology & Carcinogenesis ,Polymorphism ,Aged ,glioblastoma ,Neurosciences ,medicine.disease ,Brain Disorders ,Brain Cancer ,Case-Control Studies ,Cancer research ,Neurology (clinical) ,030217 neurology & neurosurgery ,Glioblastoma - Abstract
BACKGROUND: Twenty-five single nucleotide polymorphisms (SNPs) are associated with adult diffuse glioma risk. We hypothesized that the inclusion of these 25 SNPs with age at diagnosis and sex could estimate risk of glioma as well as identify glioma subtypes. METHODS: Case-control design and multinomial logistic regression were used to develop models to estimate the risk of glioma development while accounting for histologic and molecular subtypes. Case-case design and logistic regression were used to develop models to predict isocitrate dehydrogenase (IDH) mutation status. A total of 1273 glioma cases and 443 controls from Mayo Clinic were used in the discovery set, and 852 glioma cases and 231 controls from UCSF were used in the validation set. All samples were genotyped using a custom Illumina OncoArray. RESULTS: Patients in the highest 5% of the risk score had more than a 14-fold increase in relative risk of developing an IDH mutant glioma. Large differences in lifetime absolute risk were observed at the extremes of the risk score percentile. For both IDH mutant 1p/19q non-codeleted glioma and IDH mutant 1p/19q codeleted glioma, the lifetime risk increased from almost null to 2.3% and almost null to 1.7%, respectively. The SNP-based model that predicted IDH mutation status had a validation concordance index of 0.85. CONCLUSIONS: These results suggest that germline genotyping can provide new tools for the initial management of newly discovered brain lesions. Given the low lifetime risk of glioma, risk scores will not be useful for population screening; however, they may be useful in certain clinically defined high-risk groups.
- Published
- 2019
4. 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
5. 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
6. Adult infiltrating gliomas with WHO 2016 integrated diagnosis: additional prognostic roles of ATRX and TERT
- Author
-
Mitchel S. Berger, Tracy Luks, Robert B. Jenkins, Caterina Giannini, Paige M. Bracci, Margaret Wrensch, Tarik Tihan, Thomas M. Kollmeyer, Hugues Sicotte, Terri Rice, Joseph L. Wiemels, Kyle M. Walsh, Jeanette E. Eckel-Passow, Gobinda Sarkar, Lucie McCoy, Jennie Taylor, Arie Perry, Paul A. Decker, Jennifer Leigh Clarke, Melike Pekmezci, John K. Wiencke, Daniel H. Lachance, Annette M. Molinaro, Helen M. Hansen, Pekmezci M., Rice T., Molinaro A.M., Walsh K.M., Decker P.A., Hansen H., Sicotte H., Kollmeyer T.M., McCoy L.S., Sarkar G., Perry A., Giannini C., Tihan T., Berger M.S., Wiemels J.L., Bracci P.M., Eckel-Passow J.E., Lachance D.H., Clarke J., Taylor J.W., Luks T., Wiencke J.K., Jenkins R.B., and Wrensch M.R.
- Subjects
Oncology ,Male ,Pathology ,Kaplan-Meier Estimate ,Brain tumor prognosis ,Central Nervous System Neoplasms ,Tumor Status ,0302 clinical medicine ,80 and over ,TERT promoter mutation ,Telomerase ,Cancer ,Aged, 80 and over ,Tumor ,Central Nervous System Neoplasm ,Astrocytoma ,Glioma ,Middle Aged ,Prognosis ,Isocitrate Dehydrogenase ,Isocitrate dehydrogenase ,030220 oncology & carcinogenesis ,Glioma classification ,Female ,Case-Control Studie ,Telomere maintenance ,Human ,Adult ,medicine.medical_specialty ,X-linked Nuclear Protein ,Adolescent ,Prognosi ,Clinical Sciences ,Biology ,ATRX alteration ,World Health Organization ,Article ,Pathology and Forensic Medicine ,Brain tumor prognosi ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Young Adult ,Rare Diseases ,Clinical Research ,Internal medicine ,medicine ,Genetics ,Biomarkers, Tumor ,Humans ,ATRX ,Aged ,Neurology & Neurosurgery ,Proportional hazards model ,Case-control study ,Neurosciences ,medicine.disease ,Brain Disorders ,Brain Cancer ,Orphan Drug ,Case-Control Studies ,Mutation ,Neurology (clinical) ,Oligodendroglioma ,Neoplasm Grading ,030217 neurology & neurosurgery ,Biomarkers - Abstract
© 2017, Springer-Verlag Berlin Heidelberg. The “integrated diagnosis” for infiltrating gliomas in the 2016 revised World Health Organization (WHO) classification of tumors of the central nervous system requires assessment of the tumor for IDH mutations and 1p/19q codeletion. Since TERT promoter mutations and ATRX alterations have been shown to be associated with prognosis, we analyzed whether these tumor markers provide additional prognostic information within each of the five WHO 2016 categories. We used data for 1206 patients from the UCSF Adult Glioma Study, the Mayo Clinic and The Cancer Genome Atlas (TCGA) with infiltrative glioma, grades II-IV for whom tumor status for IDH, 1p/19q codeletion, ATRX, and TERT had been determined. All cases were assigned to one of 5 groups following the WHO 2016 diagnostic criteria based on their morphologic features, and IDH and 1p/19q codeletion status. These groups are: (1) Oligodendroglioma, IDH-mutant and 1p/19q-codeleted; (2) Astrocytoma, IDH-mutant; (3) Glioblastoma, IDH-mutant; (4) Glioblastoma, IDH-wildtype; and (5) Astrocytoma, IDH-wildtype. Within each group, we used univariate and multivariate Cox proportional hazards models to assess associations of overall survival with patient age at diagnosis, grade, and ATRX alteration status and/or TERT promoter mutation status. Among Group 1 IDH-mutant 1p/19q-codeleted oligodendrogliomas, the TERT-WT group had significantly worse overall survival than the TERT-MUT group (HR: 2.72, 95% CI 1.05–7.04, p = 0.04). In both Group 2, IDH-mutant astrocytomas and Group 3, IDH-mutant glioblastomas, neither TERT mutations nor ATRX alterations were significantly associated with survival. Among Group 4, IDH-wildtype glioblastomas, ATRX alterations were associated with favorable outcomes (HR: 0.36, 95% CI 0.17–0.81, p = 0.01). Among Group 5, IDH-wildtype astrocytomas, the TERT-WT group had significantly better overall survival than the TERT-MUT group (HR: 0.48, 95% CI 0.27–0.87), p = 0.02). Thus, we present evidence that in certain WHO 2016 diagnostic groups, testing for TERT promoter mutations or ATRX alterations may provide additional useful prognostic information.
- Published
- 2017
7. Synchronous gemistocytic astrocytoma IDH-mutant and oligodendroglioma IDH-mutant and 1p/19q-codeleted in a patient with CCDC26 polymorphism
- Author
-
Benjamin R. Kipp, Alissa Caron, Rachael A. Vaubel, Haohai Liang, Caterina Giannini, Emily G. Barr Fritcher, Jesse S. Voss, Robert B. Jenkins, Thomas M. Kollmeyer, Vaubel R.A., Kollmeyer T.M., Caron A.A., Barr Fritcher E.G., Voss J.S., Liang H., Jenkins R.B., Giannini C., and Kipp B.R.
- Subjects
0301 basic medicine ,Male ,X-linked Nuclear Protein ,Mutant ,Pathology and Forensic Medicine ,Brain Neoplasm ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Text mining ,medicine ,Gemistocytic Astrocytoma ,Humans ,Brain Neoplasms ,Extramural ,business.industry ,Intracellular Signaling Peptides and Proteins ,Glioma ,Middle Aged ,medicine.disease ,Magnetic Resonance Imaging ,Isocitrate Dehydrogenase ,030104 developmental biology ,Chromosomes, Human, Pair 1 ,Intracellular Signaling Peptides and Protein ,Mutation ,Cancer research ,RNA, Long Noncoding ,Neurology (clinical) ,Oligodendroglioma ,Chromosome Deletion ,business ,Chromosomes, Human, Pair 19 ,030217 neurology & neurosurgery ,Human - Abstract
Lettera - non ha abstract
- Published
- 2017
8. Variants in the CDKN2B and RTEL1 regions are associated with high-grade glioma susceptibility
- Author
-
Margaret Wrensch, Joseph L. Wiemels, Jan C. Buckner, Susan M. Chang, Terri Rice, Amanda L. Rynearson, Tarik Tihan, Michael D. Prados, Paul A. Decker, Charles P. Quesenberry, Mitchel S. Berger, Alexander R. Pico, John K. Wiencke, Joe Patoka, Robert B. Jenkins, Caterina Giannini, Ivan Smirnov, Yuanyuan Xiao, Daniel H. Lachance, Brian P. O'Neill, Ru Fang Yeh, Lucie McCoy, Matthew L. Kosel, Jeffrey S. Chang, Chandralekha Halder, Karla V. Ballman, Thomas M. Kollmeyer, Ping Yang, Wrensch M., Jenkins R.B., Chang J.S., Yeh R.-F., Xiao Y., Decker P.A., Ballman K.V., Berger M., Buckner J.C., Chang S., Giannini C., Halder C., Kollmeyer T.M., Kosel M.L., Lachance D.H., McCoy L., O'Neill B.P., Patoka J., Pico A.R., Prados M., Quesenberry C., Rice T., Rynearson A.L., Smirnov I., Tihan T., Wiemels J., Yang P., and Wiencke J.K.
- Subjects
Adult ,Candidate gene ,Genome-wide association study ,Single-nucleotide polymorphism ,Biology ,DNA Helicase ,Polymorphism, Single Nucleotide ,Regression Analysi ,Article ,03 medical and health sciences ,0302 clinical medicine ,Principal Component Analysi ,Glioma ,CDKN2B ,Haplotype ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Gene ,030304 developmental biology ,Cyclin-Dependent Kinase Inhibitor p15 ,0303 health sciences ,Principal Component Analysis ,Models, Genetic ,Genome, Human ,Case-control study ,DNA Helicases ,Chromosome Mapping ,medicine.disease ,3. Good health ,Haplotypes ,030220 oncology & carcinogenesis ,Case-Control Studies ,Cancer research ,Regression Analysis ,Case-Control Studie ,Chromosomes, Human, Pair 9 ,Human ,Genome-Wide Association Study - Abstract
Margaret Wrensch and colleagues report a genome-wide association and replication study for high-grade glioma. They show that common variants in the CDKN2B and RTEL1 regions are associated with risk of this aggressive brain tumor. The causes of glioblastoma and other gliomas remain obscure1,2. To discover new candidate genes influencing glioma susceptibility, we conducted a principal component–adjusted3 genome-wide association study (GWAS) of 275,895 autosomal variants among 692 adult high-grade glioma cases (622 from the San Francisco Adult Glioma Study (AGS) and 70 from the Cancer Genome Atlas (TCGA))4 and 3,992 controls (602 from AGS and 3,390 from Illumina iControlDB (iControls)). For replication, we analyzed the 13 SNPs with P < 10−6 using independent data from 176 high-grade glioma cases and 174 controls from the Mayo Clinic. On 9p21, rs1412829 near CDKN2B had discovery P = 3.4 × 10−8, replication P = 0.0038 and combined P = 1.85 × 10−10. On 20q13.3, rs6010620 intronic to RTEL1 had discovery P = 1.5 × 10−7, replication P = 0.00035 and combined P = 3.40 × 10−9. For both SNPs, the direction of association was the same in discovery and replication phases.
- Published
- 2009
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.