34 results on '"Stefanie Stallard"'
Search Results
2. Medulloblastoma therapy generates risk of a poorly-prognostic H3 wild-type subgroup of diffuse intrinsic pontine glioma: a report from the International DIPG Registry
- Author
-
Hunter C. Gits, Maia Anderson, Stefanie Stallard, Drew Pratt, Becky Zon, Christopher Howell, Chandan Kumar-Sinha, Pankaj Vats, Katayoon Kasaian, Daniel Polan, Martha Matuszak, Daniel E. Spratt, Marcia Leonard, Tingting Qin, Lili Zhao, James Leach, Brooklyn Chaney, Nancy Yanez Escorza, Jacob Hendershot, Blaise Jones, Christine Fuller, Sarah Leary, Ute Bartels, Eric Bouffet, Torunn I. Yock, Patricia Robertson, Rajen Mody, Sriram Venneti, Arul M. Chinnaiyan, Maryam Fouladi, Nicholas G. Gottardo, and Carl Koschmann
- Subjects
Secondary malignant neoplasm ,Diffuse intrinsic pontine glioma ,Medulloblastoma ,Cranial irradiation ,Brainstem ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract With improved survivorship in medulloblastoma, there has been an increasing incidence of late complications. To date, no studies have specifically addressed the risk of radiation-associated diffuse intrinsic pontine glioma (DIPG) in medulloblastoma survivors. Query of the International DIPG Registry identified six cases of DIPG with a history of medulloblastoma treated with radiotherapy. All patients underwent central radiologic review that confirmed a diagnosis of DIPG. Six additional cases were identified in reports from recent cooperative group medulloblastoma trials (total n = 12; ages 7 to 21 years). From these cases, molecular subgrouping of primary medulloblastomas with available tissue (n = 5) revealed only non-WNT, non-SHH subgroups (group 3 or 4). The estimated cumulative incidence of DIPG after post-treatment medulloblastoma ranged from 0.3–3.9%. Posterior fossa radiation exposure (including brainstem) was greater than 53.0 Gy in all cases with available details. Tumor/germline exome sequencing of three radiation-associated DIPGs revealed an H3 wild-type status and mutational signature distinct from primary DIPG with evidence of radiation-induced DNA damage. Mutations identified in the radiation-associated DIPGs had significant molecular overlap with recurrent drivers of adult glioblastoma (e.g. NRAS, EGFR, and PTEN), as opposed to epigenetic dysregulation in H3-driven primary DIPGs. Patients with radiation-associated DIPG had a significantly worse median overall survival (median 8 months; range 4–17 months) compared to patients with primary DIPG. Here, it is demonstrated that DIPG occurs as a not infrequent complication of radiation therapy in survivors of pediatric medulloblastoma and that radiation-associated DIPGs may present as a poorly-prognostic distinct molecular subgroup of H3 wild-type DIPG. Given the abysmal survival of these cases, these findings provide a compelling argument for efforts to reduce exposure of the brainstem in the treatment of medulloblastoma. Additionally, patients with radiation-associated DIPG may benefit from future therapies targeted to the molecular features of adult glioblastoma rather than primary DIPG.
- Published
- 2018
- Full Text
- View/download PDF
3. Supplementary Data and Methods from Expression of the Androgen Receptor Governs Radiation Resistance in a Subset of Glioblastomas Vulnerable to Antiandrogen Therapy
- Author
-
Daniel R. Wahl, Roy E. Strowd, Theodore S. Lawrence, Corey Speers, Joel R. Eisner, Arul M. Chinnaiyan, Jann N. Sarkaria, Howard Colman, Daniel E. Spratt, Edwina Baskin-Bey, Sriram Venneti, Meredith A. Morgan, Alexander M. Hegedus, Waldemar Debinski, Tarik Bor, Carl Koschmann, Stefanie Stallard, Arvind Rao, Yangyang Yao, Weihua Zhou, Ayesha U. Kothari, Joseph Dresser, Kari Wilder-Romans, Hanshi Sun, Uchechi J. Nna, and Christian K. Werner
- Abstract
Supplementary Figure S1: Additional expression data and comparisons of AR expression by subtype, age, sex, and common molecular alterations. Supplementary Figure S2. Tumor growth plots and mice weights for in vivo studies. Supplementary Figure S3: Heat map of down-regulated genes from the GBM 26 RNA-seq experiment. Supplementary Figure S4: Effects of seviteronel on dsDNA break repair in AR-positive and negative models post-radiation. Supplementary Figure S5: Effects of enzalutamide on dsDNA break repair in AR-positive and negative models post-radiation. Supplementary Figure S6. Effects of enzalutamide and seviteronel on alkaline tail moment in AR-positive models post-radiation. Method: alkaline comet assay.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Table S3A and S3B from Electronic DNA Analysis of CSF Cell-free Tumor DNA to Quantify Multi-gene Molecular Response in Pediatric High-grade Glioma
- Author
-
Carl Koschmann, Tingtin Qin, Hugh J.L. Garton, Cormac O. Maher, Karin M. Muraszko, Patricia L. Robertson, Andrea Franson, Jonathan Schwartz, Rajen Mody, Ian Wolfe, Robert P. Dickson, Stefanie Stallard, Kyle Wierzbicki, Leo Tunkle, Evan Cantor, Clarissa May Babila, Jack Wadden, Ashwath Muruganand, Karthik Ravi, and Amy K. Bruzek
- Abstract
Nanopore Sensitivity/Specificity details
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Data from Electronic DNA Analysis of CSF Cell-free Tumor DNA to Quantify Multi-gene Molecular Response in Pediatric High-grade Glioma
- Author
-
Carl Koschmann, Tingtin Qin, Hugh J.L. Garton, Cormac O. Maher, Karin M. Muraszko, Patricia L. Robertson, Andrea Franson, Jonathan Schwartz, Rajen Mody, Ian Wolfe, Robert P. Dickson, Stefanie Stallard, Kyle Wierzbicki, Leo Tunkle, Evan Cantor, Clarissa May Babila, Jack Wadden, Ashwath Muruganand, Karthik Ravi, and Amy K. Bruzek
- Abstract
All supplementary data except table S3A and S3B
- Published
- 2023
- Full Text
- View/download PDF
6. Data from Electronic DNA Analysis of CSF Cell-free Tumor DNA to Quantify Multi-gene Molecular Response in Pediatric High-grade Glioma
- Author
-
Carl Koschmann, Tingtin Qin, Hugh J.L. Garton, Cormac O. Maher, Karin M. Muraszko, Patricia L. Robertson, Andrea Franson, Jonathan Schwartz, Rajen Mody, Ian Wolfe, Robert P. Dickson, Stefanie Stallard, Kyle Wierzbicki, Leo Tunkle, Evan Cantor, Clarissa May Babila, Jack Wadden, Ashwath Muruganand, Karthik Ravi, and Amy K. Bruzek
- Abstract
Purpose:Pediatric high-grade glioma (pHGG) diagnosis portends poor prognosis and therapeutic monitoring remains difficult. Tumors release cell-free tumor DNA (cf-tDNA) into cerebrospinal fluid (CSF), allowing for potential detection of tumor-associated mutations by CSF sampling. We hypothesized that direct, electronic analysis of cf-tDNA with a handheld platform (Oxford Nanopore MinION) could quantify patient-specific CSF cf-tDNA variant allele fraction (VAF) with improved speed and limit of detection compared with established methods.Experimental Design:We performed ultra-short fragment (100–200 bp) PCR amplification of cf-tDNA for clinically actionable alterations in CSF and tumor samples from patients with pHGG (n = 12) alongside nontumor CSF (n = 6). PCR products underwent rapid amplicon-based sequencing by Oxford Nanopore Technology (Nanopore) with quantification of VAF. Additional comparison to next-generation sequencing (NGS) and droplet digital PCR (ddPCR) was performed.Results:Nanopore demonstrated 85% sensitivity and 100% specificity in CSF samples (n = 127 replicates) with 0.1 femtomole DNA limit of detection and 12-hour results, all of which compared favorably with NGS. Multiplexed analysis provided concurrent analysis of H3.3A (H3F3A) and H3C2 (HIST1H3B) mutations in a nonbiopsied patient and results were confirmed by ddPCR. Serial CSF cf-tDNA sequencing by Nanopore demonstrated correlation of radiological response on a clinical trial, with one patient showing dramatic multi-gene molecular response that predicted long-term clinical response.Conclusions:Nanopore sequencing of ultra-short pHGG CSF cf-tDNA fragments is feasible, efficient, and sensitive with low-input samples thus overcoming many of the barriers restricting wider use of CSF cf-tDNA diagnosis and monitoring in this patient population.
- Published
- 2023
- Full Text
- View/download PDF
7. Electronic DNA Analysis of CSF Cell-free Tumor DNA to Quantify Multi-gene Molecular Response in Pediatric High-grade Glioma
- Author
-
Karthik Ravi, Ian Wolfe, Jonathan Schwartz, Jack Wadden, Leo Tunkle, Ashwath Muruganand, Carl Koschmann, Cormac O. Maher, Rajen Mody, Amy K. Bruzek, Hugh J. L. Garton, Kyle Wierzbicki, Patricia L. Robertson, Clarissa Babila, Andrea Franson, Tingtin Qin, Evan Cantor, Stefanie Stallard, Robert P. Dickson, and Karin M. Muraszko
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Adolescent ,Polymerase Chain Reaction ,Article ,Circulating Tumor DNA ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Cerebrospinal fluid ,law ,Glioma ,Biomarkers, Tumor ,medicine ,Humans ,Digital polymerase chain reaction ,Child ,Polymerase chain reaction ,Brain Neoplasms ,business.industry ,Amplicon ,Prognosis ,medicine.disease ,Molecular biology ,Nanopore ,030104 developmental biology ,Oncology ,Case-Control Studies ,Child, Preschool ,Molecular Response ,Mutation ,Female ,Nanopore sequencing ,Electronics ,business ,030217 neurology & neurosurgery ,Follow-Up Studies - Abstract
Purpose: Pediatric high-grade glioma (pHGG) diagnosis portends poor prognosis and therapeutic monitoring remains difficult. Tumors release cell-free tumor DNA (cf-tDNA) into cerebrospinal fluid (CSF), allowing for potential detection of tumor-associated mutations by CSF sampling. We hypothesized that direct, electronic analysis of cf-tDNA with a handheld platform (Oxford Nanopore MinION) could quantify patient-specific CSF cf-tDNA variant allele fraction (VAF) with improved speed and limit of detection compared with established methods. Experimental Design: We performed ultra-short fragment (100–200 bp) PCR amplification of cf-tDNA for clinically actionable alterations in CSF and tumor samples from patients with pHGG (n = 12) alongside nontumor CSF (n = 6). PCR products underwent rapid amplicon-based sequencing by Oxford Nanopore Technology (Nanopore) with quantification of VAF. Additional comparison to next-generation sequencing (NGS) and droplet digital PCR (ddPCR) was performed. Results: Nanopore demonstrated 85% sensitivity and 100% specificity in CSF samples (n = 127 replicates) with 0.1 femtomole DNA limit of detection and 12-hour results, all of which compared favorably with NGS. Multiplexed analysis provided concurrent analysis of H3.3A (H3F3A) and H3C2 (HIST1H3B) mutations in a nonbiopsied patient and results were confirmed by ddPCR. Serial CSF cf-tDNA sequencing by Nanopore demonstrated correlation of radiological response on a clinical trial, with one patient showing dramatic multi-gene molecular response that predicted long-term clinical response. Conclusions: Nanopore sequencing of ultra-short pHGG CSF cf-tDNA fragments is feasible, efficient, and sensitive with low-input samples thus overcoming many of the barriers restricting wider use of CSF cf-tDNA diagnosis and monitoring in this patient population.
- Published
- 2020
- Full Text
- View/download PDF
8. Everolimus improves the efficacy of dasatinib in PDGFRα-driven glioma
- Author
-
Sabine Mueller, Rodrigo Cartaxo, Viveka Nand Yadav, Rajen Mody, Cassie Kline, Alyssa Paul, Zachary Miklja, Brendan Mullan, Patricia L. Robertson, Ruby Siada, Marcia Leonard, Sriram Venneti, Taylor Garcia, Amy K. Bruzek, Stefanie Stallard, Hugh J. L. Garton, Bernard L. Marini, Carl Koschmann, Chase Thomas, Kyle Wierzbicki, Jann N. Sarkaria, Arul M. Chinnaiyan, Theodore Nicolaides, Daniel R. Wahl, Sarah Leary, Chandan Kumar-Sinha, Chana Glasser, Hemant Parmar, Jessica R. Cummings, Ian Wolfe, Tao Yang, Timothy N. Phoenix, and Manjunath P. Pai
- Subjects
Male ,0301 basic medicine ,Oncology ,medicine.medical_treatment ,Dasatinib ,Gene Expression ,Tyrosine-kinase inhibitor ,Targeted therapy ,Mice ,0302 clinical medicine ,Pregnancy ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Tumor Cells, Cultured ,Medicine ,Molecular Targeted Therapy ,Child ,0303 health sciences ,Brain Neoplasms ,Drug Synergism ,Glioma ,General Medicine ,3. Good health ,Child, Preschool ,030220 oncology & carcinogenesis ,Female ,Research Article ,medicine.drug ,medicine.medical_specialty ,ATP Binding Cassette Transporter, Subfamily B ,Adolescent ,medicine.drug_class ,PDGFRA ,Receptor, Platelet-Derived Growth Factor beta ,03 medical and health sciences ,Pharmacokinetics ,In vivo ,Internal medicine ,Animals ,Humans ,Everolimus ,Adverse effect ,Cell Proliferation ,030304 developmental biology ,business.industry ,medicine.disease ,030104 developmental biology ,Drug Screening Assays, Antitumor ,business - Abstract
BackgroundPediatric and adult high-grade glioma (HGG) frequently harbor PDGFRA alterations. We hypothesized that co-treatment with everolimus may improve the efficacy of dasatinib in PDGFRα-driven glioma through combinatorial synergism and increased tumor accumulation of dasatinib.MethodsDose response, synergism studies, P-gp inhibition and pharmacokinetic studies were performed on in vitro and in vivo human and mouse models of HGG. Six patients with recurrent PDGFRα-driven glioma were treated with dasatinib and everolimus.ResultsDasatinib effectively inhibited the proliferation of mouse and human primary HGG cells with a variety of PDGFRA alterations. Dasatinib exhibited synergy with everolimus in the treatment of HGG cells at low nanomolar concentrations of both agents, with reduction in mTOR signaling that persists after dasatinib treatment alone. Prolonged exposure to everolimus significantly improved the CNS retention of dasatinib and extended survival of PPK tumor bearing mice. Pediatric patients (n=6) with glioma tolerated this combination without significant adverse events. Recurrent patients (n=4) demonstrated median overall survival of 8.5 months.ConclusionEfficacy of dasatinib treatment of PDGFRα-driven HGG is improved with everolimus and suggests a promising route for improving targeted therapy for this patient population.Trial RegistrationClinicalTrials.gov NCT03352427FundingThe authors thank the patients and their families for participation in this study. CK is supported by NIH/NINDS K08-NS099427-01, the University of Michigan Chad Carr Pediatric Brain Tumor Center, the Chad Tough Foundation, Hyundai Hope on Wheels, Catching up With Jack, Prayers from Maria Foundation, U CAN-CER VIVE FOUNDATION, Morgan Behen Golf Classic, and the DIPG Collaborative. The PEDS-MIONCOSEQ study was supported by grant 1UM1HG006508 from the National Institutes of Health Clinical Sequencing Exploratory Research Award (PI: Arul Chinnaiyan).
- Published
- 2020
- Full Text
- View/download PDF
9. CSIG-09. THERAPEUTIC TARGETING OF PRENATAL PONTINE ID1 SIGNALING IN DIFFUSE MIDLINE GLIOMA
- Author
-
Dana Messinger, Micah Harris, Jessica Cummings, Chase Thomas, Tao Yang, Stefan Sweha, Rinette Woo, Robert Siddaway, Martin Burkert, Stefanie Stallard, Tingting Qin, Brendan Mullan, Ruby Siada, Ramya Ravindran, Michael Niculcea, Abigail Dowling, Joshua Bradin, Kevin Ginn, Melissa Gener, Kathleen Dorris, Nicholas Vitanza, Susanne Schmidt, Jasper Spitzer, Jiang Li, Mariella Filbin, Xuhong Cao, Maria Castro, Pedro Lowenstein, Rajen Mody, Arul Chinnaiyan, Pierre-Yves Desprez, Sean McAllister, Matthew Dun, Cynthia Hawkins, Sebastian Waszak, Sriram Venneti, Carl Koschmann, and Viveka Yadav
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
Diffuse midline gliomas (DMG) are highly invasive brain tumors with rare survival beyond two years past diagnosis. The mechanism behind tumor invasion is currently not well understood. Previous reports demonstrate upregulation of the protein ID1 with H3K27M and ACVR1 mutations in DMG, but this has not been confirmed in human tumors or therapeutically targeted. Whole exome, RNA, and ChIP-sequencing were performed on the ID1 locus in DMG tissue. Scratch-assay migration and transwell invasion assays of cultured cells were performed following shRNA-mediated ID1-knockdown. In vitro and in vivo genetic and pharmacologic [cannabidiol (CBD)] inhibition of ID1 on DMG tumor growth was assessed. Additional in vitro experiments were performed to determine a potential mechanism of action for CBD-mediated effects. Self-reported CBD dosing information was collected from DMG patients. We found that increased ID1 expression in human DMG and in utero electroporation (IUE) murine tumors is associated with H3K27M mutation and brainstem location. ChIP-sequencing indicates a similar epigenetically active state at ID1 regulatory regions in human H3K27M-DMG tumors and prenatal pontine cells. Higher ID1-expressing astrocyte-like DMG cells share a transcriptional program with oligo/astrocyte-precursor cells (OAPCs) from the developing human brain and demonstrate upregulation of the migration regulatory protein SPARCL1. Genetic and pharmacologic (CBD) suppression of ID1 decreases tumor cell migration, tumor growth, and to a lesser extent invasion in both murine IUE and multiple patient-derived in vivo DMG models, improving mouse survival. ID1 knockdown significantly decreases the effect of CBD on migration, tumor growth, and invasion. CBD increases reactive oxygen species production, which also affects DMG cell proliferation in a non-ID1 mediated manner. Overall, we find that H3K27M-mediated reactivation of ID1 in DMG results in a SPARCL1+ migratory transcriptional program that is therapeutically targetable with CBD.
- Published
- 2022
- Full Text
- View/download PDF
10. Therapeutic targeting of prenatal pontine ID1 signaling in diffuse midline glioma
- Author
-
Dana Messinger, Micah K Harris, Jessica R Cummings, Chase Thomas, Tao Yang, Stefan R Sweha, Rinette Woo, Robert Siddaway, Martin Burkert, Stefanie Stallard, Tingting Qin, Brendan Mullan, Ruby Siada, Ramya Ravindran, Michael Niculcea, Abigail R Dowling, Joshua Bradin, Kevin F Ginn, Melissa A H Gener, Kathleen Dorris, Nicholas A Vitanza, Susanne V Schmidt, Jasper Spitzer, Jiang Li, Mariella G Filbin, Xuhong Cao, Maria G Castro, Pedro R Lowenstein, Rajen Mody, Arul Chinnaiyan, Pierre-Yves Desprez, Sean McAllister, Matthew D Dun, Cynthia Hawkins, Sebastian M Waszak, Sriram Venneti, Carl Koschmann, and Viveka Nand Yadav
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
Background Diffuse midline gliomas (DMG) are highly invasive brain tumors with rare survival beyond two years past diagnosis and limited understanding of the mechanism behind tumor invasion. Previous reports demonstrate upregulation of the protein ID1 with H3K27M and ACVR1 mutations in DMG, but this has not been confirmed in human tumors or therapeutically targeted. Methods Whole exome, RNA, and ChIP-sequencing was performed on the ID1 locus in DMG tissue. Scratch-assay migration and transwell invasion assays of cultured cells were performed following shRNA-mediated ID1-knockdown. In vitro and in vivo genetic and pharmacologic [cannabidiol (CBD)] inhibition of ID1 on DMG tumor growth was assessed. Patient-reported CBD dosing information was collected. Results Increased ID1 expression in human DMG and in utero electroporation (IUE) murine tumors is associated with H3K27M mutation and brainstem location. ChIP-sequencing indicates ID1 regulatory regions are epigenetically active in human H3K27M-DMG tumors and prenatal pontine cells. Higher ID1-expressing astrocyte-like DMG cells share a transcriptional program with oligo/astrocyte-precursor cells (OAPCs) from the developing human brain and demonstrate upregulation of the migration regulatory protein SPARCL1. Genetic and pharmacologic (CBD) suppression of ID1 decreases tumor cell invasion/migration and tumor growth in H3.3/H3.1K27M PPK-IUE and human DIPGXIIIP* in vivo models of pHGG. The effect of CBD on cell proliferation appears to be non-ID1 mediated. Finally, we collected patient-reported CBD treatment data, finding that a clinical trial to standardize dosing may be beneficial. Conclusions H3K27M-mediated re-activation of ID1 in DMG results in a SPARCL1+ migratory transcriptional program that is therapeutically targetable with CBD.
- Published
- 2021
11. Molecular profiling and targeted therapy in pediatric gliomas: review and consensus recommendations
- Author
-
Sarah Leary, Carl Koschmann, Amy K. Bruzek, Rameen Beroukhim, Shakti H. Ramkissoon, Theodore Nicolaides, Susan N. Chi, Bonnie Cole, Zachary Miklja, Brendan Mullan, Cassie Kline-Nunnally, Keith L. Ligon, Maryam Fouladi, Angela C. Gauthier, Sabine Mueller, Bernard L. Marini, D William Parsons, Amy L. Pasternak, Taylor Garcia, Christie Atchison, Pratiti Bandopadhayay, and Stefanie Stallard
- Subjects
Cancer Research ,medicine.medical_specialty ,Consensus ,Standard of care ,medicine.medical_treatment ,Review ,Targeted therapy ,Young Adult ,Glioma ,Pediatric glioma ,Humans ,Medicine ,Profiling (information science) ,Molecular Targeted Therapy ,Child ,Intensive care medicine ,High-Grade Glioma ,Brain Neoplasms ,business.industry ,High-Throughput Nucleotide Sequencing ,Precision medicine ,medicine.disease ,Oncology ,Pediatric brain ,Neurology (clinical) ,business - Abstract
As the field of neuro-oncology makes headway in uncovering the key oncogenic drivers in pediatric glioma, the role of precision diagnostics and therapies continues to rapidly evolve with important implications for the standard of care for clinical management of these patients. Four studies at major academic centers were published in the last year outlining the clinically integrated molecular profiling and targeting of pediatric brain tumors; all 4 demonstrated the feasibility and utility of incorporating sequencing into the care of children with brain tumors, in particular for children and young adults with glioma. Based on synthesis of the data from these studies and others, we provide consensus recommendations for the integration of precision diagnostics and therapeutics into the practice of pediatric neuro-oncology. Our primary consensus recommendation is that next-generation sequencing should be routinely included in the workup of most pediatric gliomas.
- Published
- 2019
- Full Text
- View/download PDF
12. Therapeutic reversal of prenatal pontine ID1 signaling in DIPG
- Author
-
Kevin Ginn, Pierre-Yves Desprez, Micah Harris, Cynthia Hawkins, Michael Niculcea, Chase Thomas, Maria G. Castro, Sriram Venneti, Tao Yang, Melissa Gener, Cummings, Xuhong Cao, Sandra S. McAllister, Stefanie Stallard, Messinger D, Jasper Spitzer, Carl Koschmann, Mariella G. Filbin, Chinnaiyan A, Tingting Qin, Pedro R. Lowenstein, Sebastian M. Waszak, Kathleen Dorris, Jiang Li, Viveka Yadav, Burkert M, Ramya Ravindran, Robert Siddaway, Rinette Woo, Susanne V. Schmidt, Ruby Siada, Rajen Mody, Brendan Mullan, and Nicholas A Vitanza
- Subjects
medicine.anatomical_structure ,Downregulation and upregulation ,Precursor cell ,Glioma ,Cancer research ,Brain tumor ,medicine ,Cell migration ,SPARCL1 ,Biology ,medicine.disease ,Transcription factor ,Astrocyte - Abstract
Diffuse intrinsic pontine glioma (DIPG) is a highly aggressive brain tumor with rare survival beyond two years. This poor prognosis is largely due to the tumor’s highly infiltrative and invasive nature. Previous reports demonstrate upregulation of the transcription factor ID1 with H3K27M and ACVR1 mutations, but this has not been confirmed in human tumors or therapeutically targeted. We developed an in utero electroporation (IUE) murine H3K27M-driven tumor model, which demonstrates increased ID1 expression in H3K27M- and ACVR1-mutated tumor cells. In human tumors, elevated ID1 expression is associated with H3K27M/ACVR1-mutation, brainstem location, and reduced survival. The ID1 promoter demonstrates a similar active epigenetic state in H3K27M tumor cells and murine prenatal hindbrain cells. In the developing human brain, ID1 is expressed highest in oligo/astrocyte-precursor cells (OAPCs). These ID1+/SPARCL1+ cells share a transcriptional program with astrocyte-like (AC-like) DIPG cells, and demonstrate upregulation of gene sets involved with regulation of cell migration. Both genetic and pharmacologic [cannabidiol (CBD)] suppression of ID1 results in decreased DIPG cell invasion/migration in vitro and invasion/tumor growth in multiple in vivo models. CBD reduces proliferation through reactive oxygen species (ROS) production at low micromolar concentrations, which we found to be achievable in the murine brainstem. Further, pediatric high-grade glioma patients treated off-trial with CBD (n=15) demonstrate tumor ID1 reduction and improved overall survival compared to historical controls. Our study identifies that ID1 is upregulated in DIPG through reactivation of a developmental OAPC transcriptional state, and ID1-driven invasiveness of DIPG is therapeutically targetable with CBD.One Sentence SummaryThe transcription factor ID1 is upregulated in a subset of DIPG tumor cells, and ID1-driven invasiveness is therapeutically targetable with CBD.
- Published
- 2021
- Full Text
- View/download PDF
13. Clinical efficacy and predictive biomarkers of ONC201 in H3 K27M-mutant diffuse midline glioma
- Author
-
Carl Koschmann, Abed Rahman Kawakibi, Rohinton Tarapore, Sharon Gardner, Chase Thomas, Rodrigo Cartaxo, Viveka Yadav, Andrew Chi, Sylvia Kurz, Patrick Wen, Isabel Arrillaga, Tracy Batchelor, Nicholas Butowski, Ashley Sumrall, Nicole Shonka, Rebecca Harrison, John De Groot, Minesh Mehta, Yazmin Odia, Matthew Hall, Doured Daghistani, Timothy Cloughesy, Benjamin Ellingson, Michelle Kim, Yoshie Umemura, Hugh Garton, Andrea Franson, Patricia Robertson, Jonathan Schwartz, Bernard Marini, Manjunath Pai, Timothy Phoenix, Sunjong Ji, Evan Cantor, Zachary Miklja, Brendan Mullan, Amy Bruzek, Ruby Siada, Jessica Cummings, Stefanie Stallard, Kyle Wierzbicki, Alyssa Paul, Ian Wolfe, Matthew Dun, Jason Cain, Li Jiang, Mariella Filbin, Pankaj Vats, Chandan Kumar-Sinha, Rajen Mody, Arul Chinnaiyan, Drew Pratt, Sriram Venneti, Guangrong Lu, Sabine Mueller, Adam Resnick, Javad Nazarian, Sebastian Waszak, and Joshua Allen
- Abstract
Patients with diffuse midline glioma (DMG) harboring H3 K27M mutation have no proven therapies beyond radiation. ONC201, a DRD2 antagonist and mitochondrial ClpP agonist, has induced early responses in patients with H3 K27M-mutant DMG. We performed an integrated pre-clinical and clinical assessment of ONC201 treatment, in order to define response rates in H3 K27M-mutant DMG patients and to clarify predictors of response. ONC201 was effective in murine H3 K27M-mutant gliomas with excellent CNS penetration and survival benefit. H3 K27M-mutant DMG patients treated with ONC201 on active clinical trials (n=50) showed significant survival benefit in recurrent and non-recurrent settings, with multiple sustained responses. Tumor sequencing from treated patients demonstrates an EGFR/FOXG1-driven telencephalic gene regulatory network that imparts a critical resistance phenotype to ONC201. Genetic and pharmacologic knockdown of EGFR in H3 K27M-mutant cell cultures results in improved sensitivity to ONC201 and reduced FOXG1 enhancer binding, suggesting possible future combinatorial opportunities.
- Published
- 2020
- Full Text
- View/download PDF
14. Expression of the Androgen Receptor Governs Radiation Resistance in a Subset of Glioblastomas Vulnerable to Antiandrogen Therapy
- Author
-
Joel R. Eisner, Corey Speers, Uchechi J. Nna, Arul M. Chinnaiyan, Joseph Dresser, Sriram Venneti, Ayesha U. Kothari, Jann N. Sarkaria, Waldemar Debinski, Daniel R. Wahl, Alexander M. Hegedus, Kari Wilder-Romans, Hanshi Sun, Theodore S. Lawrence, Daniel E. Spratt, Yangyang Yao, Meredith A. Morgan, Carl Koschmann, Edwina Baskin-Bey, Arvind Rao, Roy E. Strowd, Weihua Zhou, Stefanie Stallard, Tarik Bor, Howard Colman, and Christian K. Werner
- Subjects
0301 basic medicine ,Cancer Research ,Antiandrogens ,medicine.medical_treatment ,Mice, SCID ,Biology ,urologic and male genital diseases ,Article ,03 medical and health sciences ,Prostate cancer ,Mice ,0302 clinical medicine ,Prostate ,medicine ,Animals ,Humans ,Antiandrogen Therapy ,Receptor ,Androgen Antagonists ,medicine.disease ,nervous system diseases ,Androgen receptor ,Steroid hormone ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Cell culture ,Receptors, Androgen ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Glioblastoma - Abstract
New approaches are needed to overcome intrinsic therapy resistance in glioblastoma (GBM). Because GBMs exhibit sexual dimorphism and are reported to express steroid hormone receptors, we reasoned that signaling through the androgen receptor (AR) could mediate therapy resistance in GBM, much as it does in AR-positive prostate and breast cancers. We found that nearly half of GBM cell lines, patient-derived xenografts (PDX), and human tumors expressed AR at the transcript and protein level—with expression levels overlapping those of primary prostate cancer. Analysis of gene expression datasets also revealed that AR expression is higher in GBM patient samples than normal brain tissue. Multiple clinical-grade antiandrogens slowed the growth of and radiosensitized AR-positive GBM cell lines and PDXs in vitro and in vivo. Antiandrogens blocked the ability of AR-positive GBM PDXs to engage adaptive transcriptional programs following radiation and slowed the repair of radiation-induced DNA damage. These results suggest that combining blood–brain barrier permeable antiandrogens with radiation may have promise for patients with AR-positive GBMs.
- Published
- 2020
15. TAMI-79. THERAPEUTIC REVERSAL OF PRENATAL PONTINE ID1 SIGNALING IN DIPG
- Author
-
Carl Koschmann, Micah Harris, Sriram Venneti, Sean D. McAllister, Kevin Ginn, Kathleen Dorris, Viveka Nand Yadav, Jasper Spitzer, Mariella G Filbin, Rajen Mody, Stefanie Stallard, Brendan Mullan, Dana Messinger, Pedro R. Lowenstein, Cynthia Hawkins, Jessica R. Cummings, Jiang Li, Nicholas A Vitanza, Ruby Siada, Tao Yang, Sebastian M Waszak, Melissa Gener, Tingting Qin, Robert Siddaway, Ramya Ravindran, Chase Thomas, Xuhong Cao, Maria G. Castro, Martin Burkert, Susanne Schmidt, Pierre Yves Desprez, Arul M. Chinnaiyan, Michael Niculcea, and Rinette Woo
- Subjects
Cancer Research ,business.industry ,Treatment outcome ,Tumor cells ,Prenatal care ,Brain tissue ,26th Annual Meeting & Education Day of the Society for Neuro-Oncology ,Brain tumor childhood ,Pons ,Oncology ,Medicine ,Tumor growth ,Neurology (clinical) ,business ,Neuroscience ,Chromatin Immunoprecipitation Sequencing - Abstract
Diffuse intrinsic pontine glioma (DIPG) is a highly aggressive pediatric brain tumor with rare survival beyond two years. This poor prognosis is largely due to the tumor's highly infiltrative and invasive nature. Nearly 80% of DMGs harbor K27M mutation in the genes encoding histone H3.1 (H3F3A) or H3.3 (HISTIH3B), often with concurrent ACVR1 mutation. Inhibitor of DNA-binding (ID) proteins are key transcriptional regulators of genes involved in lineage commitment and are associated with invasiveness and poor clinical outcomes in multiple human cancers. Introduction of H3K27M and ACVR1 mutations increase ID1 expression in cultured astrocytes, but this has not been confirmed in human tumors or targeted therapeutically. We developed an in-utero electroporation (IUE) murine H3K27M-driven tumor model, which demonstrates increased ID1 expression in H3K27M- and ACVR1-mutated tumor cells. Exome and transcriptome sequencing analysis of multi-focal DMG tumors (n=52) and normal brain tissue revealed that increased ID1 expression is associated with H3K27M/ACVR1-mutation and brainstem location, and correlates with poor survival in patients. ChIP-sequencing for H3K27ac and H3K27me3 in multiple DMG tumors (n=5) revealed that the ID1 gene is epigenetically active, which matches the epigenetic state of murine prenatal hindbrain cells. Higher ID1-expressing astrocyte-like DIPG cells share a similar transcriptional program with ID1+/SPARCL1+ positive oligo/astrocyte-precursor (OAPC) cells from the developing human brain and demonstrate upregulation of gene sets involved in regulation of cell migration. Both genetic and pharmacologic [cannabidiol (CBD)] suppression of ID1 result in decreased DIPG cell invasion/migration in vitro and invasion/tumor growth in multiple in vivo models. Mechanistically, CBD reduces proliferation through production of reactive oxygen species. Further, DIPG patients treated off-trial with CBD (n=15) displayed reduced ID1 tumor expression and improved overall survival. In summary, ID1 is upregulated in DIPG through K27M-mediated epigenetic reactivation of a developmental OAPC-like transcriptional state, and ID1-driven invasiveness of DIPG is therapeutically targetable with CBD.
- Published
- 2021
- Full Text
- View/download PDF
16. Multi-focal sequencing of a diffuse intrinsic pontine glioma establishes PTEN loss as an early event
- Author
-
Marcia Leonard, Stefanie Stallard, Pedro R. Lowenstein, Carl Koschmann, Sriram Venneti, Xuhong Cao, Valerie P. Opipari, Katayoon Kasaian, Daniel Zamler, Hugh J. L. Garton, Rajen Mody, Karin M. Muraszko, Luigi Franchi, Arul M. Chinnaiyan, Shawn L. Hervey-Jumper, Patricia L. Robertson, Maria G. Castro, and Zishaan Farooqui
- Subjects
0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,Case Report ,Somatic evolution in cancer ,lcsh:RC254-282 ,Germline ,03 medical and health sciences ,0302 clinical medicine ,Glioma ,medicine ,PTEN ,Tensin ,Exome ,PI3K/AKT/mTOR pathway ,biology ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Brainstem - Abstract
Improved molecular understanding is needed for rational treatment of diffuse intrinsic pontine gliomas (DIPG). Here, using multi-focal paired tumor and germline exome DNA and RNA sequencing, we uncovered phosphatase and tensin homolog (PTEN) loss as a clonal mutation in the case of a 6-year-old boy with a diffuse intrinsic pontine glioma, and incorporated copy number alteration analyses to provide a more detailed understanding of clonal evolution in diffuse intrinsic pontine gliomas. As well, using the PedcBioPortal, we found alterations in PTEN in 16 of 326 (4.9%) cases of pediatric high-grade glioma (3 of 154 (1.9%) brainstem) for which full sequencing data was available. Our data strengthens the association with PTEN loss in diffuse intrinsic pontine gliomas and provides further argument for the inclusion of PTEN in future targeted sequencing panels for pediatric diffuse intrinsic pontine gliomas and for the development and optimization of mTOR/PI3K inhibitors with optimal central nervous system penetration.
- Published
- 2017
- Full Text
- View/download PDF
17. Climbing Bloom's taxonomy pyramid: Lessons from a graduate histology course
- Author
-
Joel Purkiss, Michael Hortsch, Stefanie Stallard, Charles Hwang, Nikki L. Bibler Zaidi, and Sara Scott
- Subjects
0301 basic medicine ,Embryology ,Pathology ,medicine.medical_specialty ,Histology ,020205 medical informatics ,media_common.quotation_subject ,02 engineering and technology ,03 medical and health sciences ,Graduate level ,Taxonomy (general) ,Pyramid ,0202 electrical engineering, electronic engineering, information engineering ,Mathematics education ,Medicine ,Set (psychology) ,Association (psychology) ,Multiple choice ,media_common ,Graduate education ,business.industry ,General Medicine ,Aptitude ,030101 anatomy & morphology ,Anatomy ,business - Abstract
Bloom's taxonomy was adopted to create a subject-specific scoring tool for histology multiple-choice questions (MCQs). This Bloom's Taxonomy Histology Tool (BTHT) was used to analyze teacher- and student-generated quiz and examination questions from a graduate level histology course. Multiple-choice questions using histological images were generally assigned a higher BTHT level than simple text questions. The type of microscopy technique (light or electron microscopy) used for these image-based questions did not result in any significant differences in their Bloom's taxonomy scores. The BTHT levels for teacher-generated MCQs correlated positively with higher discrimination indices and inversely with the percent of students answering these questions correctly (difficulty index), suggesting that higher-level Bloom's taxonomy questions differentiate well between higher- and lower-performing students. When examining BTHT scores for MCQs that were written by students in a Multiple-Choice Item Development Assignment (MCIDA) there was no significant correlation between these scores and the students' ability to answer teacher-generated MCQs. This suggests that the ability to answer histology MCQs relies on a different skill set than the aptitude to construct higher-level Bloom's taxonomy questions. However, students significantly improved their average BTHT scores from the midterm to the final MCIDA task, which indicates that practice, experience and feedback increased their MCQ writing proficiency. Anat Sci Educ 10: 456-464. © 2017 American Association of Anatomists.
- Published
- 2017
- Full Text
- View/download PDF
18. DIPG-59. UPREGULATION OF PRENATAL PONTINE ID1 SIGNALING IN DIPG
- Author
-
Micah Harris, Cynthia Hawkins, Sriram Venneti, Ruby Siada, Tingting Qin, Sean D. McAllister, Arul M. Chinnaiyan, Carl Koschmann, Xuhong Cao, Pierre Yves Desprez, Ramya Ravindran, Maria G. Castro, Rajen Mody, Robert Siddaway, Rinette Woo, Pedro R. Lowenstein, Brendan Mullan, Stefanie Stallard, Viveka Nand Yadav, Zachary Miklja, and Amy L. Pasternak
- Subjects
Cancer Research ,business.industry ,Diffuse Midline Glioma/DIPG ,Tumor Cell Invasion ,Brain tumor childhood ,Oncology ,Downregulation and upregulation ,Cancer research ,AcademicSubjects/MED00300 ,Medicine ,AcademicSubjects/MED00310 ,Neurology (clinical) ,business ,Chromatin Immunoprecipitation Sequencing - Abstract
BACKGROUND Diffuse intrinsic pontine gliomas (DIPGs) are lethal pediatric brain tumors with no curative therapies. Inhibitor of DNA binding (ID) proteins are key regulators of gene differentiation during embryogenesis. Previous work has shown that H3F3A and ACVR1 mutations increase ID1 expression in cultured astrocytes, but this has not been validated in human DIPG, nor has the regulation and targetability of ID1 been explored in DIPG. RESULTS Analysis of post-mortem tissue and multiple human datasets showed ID1 to be elevated in DIPG, and to correlate with reduced survival. In a multi-focal autopsy of a DIPG case, we also found ID1 expression to be heterogeneous and to correlate with tumor invasion. Chromatin immunoprecipitation qPCR (ChIP-qPCR) revealed elevated H3K27ac and low H3K27me3 at ID1 regulatory regions (enhancers/promoters) in DIPG tissue compared to normal brain, regardless of H3 or ACVR1 mutation status. Analysis of publicly-available ISH and ChIP-sequencing data of developing murine brains revealed H3K27ac at ID1 enhancers to be elevated in the prenatal hindbrain compared to prenatal forebrain and midbrain, and all postnatal brain regions. ID1 shRNA-mediated knockdown of primary human H3K27M DIPG cells (DIPG007) significantly reduced invasion and migration. We also treated DIPG007 cells with cannabidiol (CBD) and found reduced viability at clinically relevant dosing (IC50=2.4 uM) with dose-dependent reduction in ID1 protein. CONCLUSIONS These findings indicate that a multifactorial (genetic and regionally-based) epigenetic upregulation of ID1 drives DIPG invasiveness and is targetable with CBD. ID1 knockdown and CBD treatment experiments in murine models of DIPG are ongoing.
- Published
- 2020
- Full Text
- View/download PDF
19. DIPG-06. RAPID, ULTRA-DEEP SEQUENCING OF PEDIATRIC DIPG FROM CEREBROSPINAL FLUID USING A NOVEL HAND-HELD ELECTRONIC DNA ANALYSIS PLATFORM
- Author
-
Karin L Muraszko, Amy K. Bruzek, Leo Tunkle, Cormac O. Maher, Tingting Qin, Veena Thamilselvan, Carl Koschmann, Ian Wolfe, Hugh J. L. Garton, Rajen Mody, Patricia L. Robertson, and Stefanie Stallard
- Subjects
Cancer Research ,Oncology ,business.industry ,Hand held ,Medicine ,Ultra deep sequencing ,Neurology (clinical) ,Computational biology ,business ,Diffuse Intrinsic Pontine Glioma (DIPG) - Abstract
INTRODUCTION: Brain tumors release tumor DNA (tDNA) into cerebrospinal fluid (CSF), allowing for detection and serial monitoring of tumor-associated genetic mutations by CSF sampling. For midline and brainstem tumors such as diffuse intrinsic pontine glioma (DIPG), surgical biopsy poses possible severe neurological deficits. As such, liquid biopsy is needed. Current platforms for CSF tDNA analysis are limited by their requirement for assay development for each mutation (digital droplet PCR), or cost and timeliness (Illumina sequencing). We hypothesized that direct, electronic analysis of DNA with a novel hand-held platform (Oxford Nanopore) could provide real-time, ultra-deep (>1,000x reads) sequencing of DIPG CSF tDNA. METHODS: We performed amplicon-based PCR on DNA from normal brain (n=6), normal CSF (n=13), tumor brain (n=3), and tumor CSF (n=10) to amplify wildtype and mutant H3F3A K27M, HIST1H3B K27M, and ACVR1 G328E genes from normal controls and pediatric patients with DIPG. We performed parallel barcoded sequencing of multiple samples run in duplicate or triplicate using NanoPore MinION technology and determined variant allele fraction (VAF) of each amplicon via an expeditious pipeline using MinKNOW, Albacore, MiniMap2, and Integrated Genome Browser. RESULTS: NanoPore sequenced 30 amplicons with average depth of 17,244 reads per amplicon in under 80 minutes. H3F3A K27M VAF was 49.14% (S.D. 0.49) in tumor tissue and 13.63% (S.D. 0.35) in CSF. VAF was 10.42% (S. D. 1.11) for HIST1H3B K27M and 7% (S.D. 0.46) for ACVR1 G328E in CSF. VAF were comparable to biopsy results by Illumina. Sensitivity and specificity of NanoPore were 100% and 98.8%, respectively, with p-values of < 0.05 for normal versus tumor samples. CONCLUSIONS: NanoPore MinION technology rapidly, reliably, and efficiently sequences tumor mutations in pooled tissue and CSF from patients with DIPG with outstanding sensitivity and specificity. NanoPore is more efficient, and cost and time effective than digital droplet PCR and Illumina.
- Published
- 2019
20. EPCT-07. ID1 IS A KEY TRANSCRIPTIONAL REGULATOR OF DIPG INVASION AND IS TARGETABLE WITH CANNABIDIOL
- Author
-
Brendan Mullan, Jessica R. Cummings, Pierre Yves Desprez, Micah Harris, Viveka Nand Yadav, Sriram Venneti, Sean D. McAllister, Maria G. Castro, Arul M. Chinnaiyan, Ramya Ravindran, Cynthia Hawkins, Michael Niculcea, Chase Thomas, Carl Koschmann, Rajen Mody, Tingting Qin, Robert Siddaway, Pedro R. Lowenstein, Stefanie Stallard, Xuhong Cao, Rinette Woo, and Ruby Siada
- Subjects
Cancer Research ,Brain tumor childhood ,Biology ,medicine.disease_cause ,Translational/Early Phase Clinical Trials ,Oncology ,ACVR1 Gene ,Downregulation and upregulation ,medicine ,Transcriptional regulation ,Cancer research ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,Carcinogenesis ,Cannabidiol ,medicine.drug - Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are lethal pediatric brain tumors with no effective therapies beyond radiation. The highly invasive nature of DIPG is key to its aggressive phenotype, but the factors and mechanisms contributing to this aggressive invasion are unknown. Inhibitor of DNA binding (ID) proteins, key regulators of lineage commitment during embryogenesis, are implicated in tumorigenesis in multiple human solid tumors. Prior work showed that recurrent H3F3A and ACVR1 mutations increase ID1 expression in cultured astrocytes. However, the impact and targetability of ID1 have not been explored in human DIPG. Exome and transcriptome sequencing analyses of multi-focal DIPG tumors and normal brain tissue from autopsy (n=52) revealed that ID1 expression is significantly elevated in DIPG samples. Higher ID1 expression correlates with reduced survival in DIPG patients and increased regional invasion in multi-focal autopsy samples. Analyses of developing mouse brain RNA/ChIP-Seq data revealed high ID1 expression and H3K27ac promoter binding in prenatal hindbrain compared to all other prenatal and postnatal brain regions. ChIP-qPCR for H3K27ac and H3K27me3 revealed that ID1 gene regulatory regions are epigenetically poised for upregulation in DIPG tissues compared to normal brain, regardless of H3/ACVR1 mutational status. These data support that the developing pons is regionally poised for ID1 activation. Genetic (shRNA) ID1 knockdown of primary human H3.3K27M-DIPG cells (DIPG007) resulted in significantly reduced invasion/migration and significantly improved survival of K27M-DIPG mice. Knockdown of ID1 in DIPG cells also resulted in down-regulation of the WNK1-NKCC1 pathway, which regulates tumor cell electrolyte homeostasis and migration. Finally, treatment of DIPG007 cells with cannabidiol (CBD) reduced ID1 levels, viability of DIPG cells and significantly improved survival of K27M-DIPG mice. In summary, our findings indicate that multifactorial (genetic and regional) epigenetic upregulation of ID1 drives DIPG invasiveness; and that targeting ID1 with CBD could potentially be an effective therapy for DIPG.
- Published
- 2021
21. DIPG-08. ELECTRONIC SEQUENCING PROVIDES OPTIMIZED QUANTIFICATION OF SERIAL, MULTI-GENE MOLECULAR RESPONSE IN THE CSF OF CHILDREN WITH HIGH-GRADE GLIOMA
- Author
-
Karin M. Muraszko, Ian Wolfe, Evan Cantor, Hugh J. L. Garton, Karthik Ravi, Ashwath Muruganand, Cormac O. Maher, Patrica Robertson, Leo Tunkle, Carl Koschmann, Tingting Qin, Rajen Mody, Stefanie Stallard, Andrea Franson, Clarissa Babila, Kyle Wierzbicki, Amy K. Bruzek, and Jack Wadden
- Subjects
Cancer Research ,Oncology ,Molecular Response ,Diffuse Midline Glioma/DIPG ,Cancer research ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,Biology ,Multi gene ,High-Grade Glioma - Abstract
BACKGROUND For pediatric high-grade glioma (pHGG), non-invasive methods for diagnosis and surveillance are needed. Tumors release DNA (tDNA) into cerebrospinal fluid (CSF), allowing for detection of tumor-associated mutations by CSF sampling. We hypothesized that direct, electronic analysis of tDNA with a novel, hand-held platform (Oxford Nanopore MinION) could quantify patient-specific CSF tDNA variant allele fraction (VAF) with improved speed and limit of detection compared to established methods. METHODS We integrated required multi-timepoint (0, 2, and 6 months) correlate lumbar punctures (LP) in two ongoing pHGG clinical trials. Using Nanopore technology, we performed amplicon-based PCR on CSF tDNA for recurrent mutations from patient samples (n=19) and normal controls. VAF were determined via MinKNOW, Guppy, MiniMap2, and Integrated Genome Browser. RESULTS Nanopore CSF tDNA demonstrated improved sensitivity (91%) when compare to NGS sequencing (50%). Nanopore analysis of serially diluted CSF sample demonstrated significantly lower limit of detection (attomolar) than typical NGS sample requirement (nanomolar). H3K27M mutation was reliably detected with 1,000x depth sequencing, which was achieved in less than 15 minutes of sequencing after amplification. Multiplexed Nanopore analysis of H3F3A and HIST1H3B was employed when H3 status was unknown. Serial CSF tDNA analysis confirmed multi-gene (H3F3A K27M, PIK3CA, and TP53) molecular remission in a 17-year-old with thalamic diffuse midline glioma that correlated with sustained clinical response to ONC201 (14 months and ongoing). CONCLUSIONS Use of a hand-held, electronic DNA analysis platform allows quantification of multi-gene molecular response with improved speed and limit of detection in the CSF of children with high-grade glioma.
- Published
- 2020
22. TAMI-29. MULTIFACTORIAL UPREGULATION OF ID1 DRIVES DIPG INVASIVENESS AND IS THERAPEUTICALLY TARGETABLE
- Author
-
Arul M. Chinnaiyan, Ramya Ravindran, Micah Harris, Pedro R. Lowenstein, Tingting Qin, Carl Koschmann, Viveka Nand Yadav, Jessica R. Cummings, Zachary Miklja, Rinette Woo, Robert Siddaway, Sriram Venneti, Rajen Mody, Maria G. Castro, Sean D. McAllister, Cynthia Hawkins, Stefanie Stallard, Xuhong Cao, Brendan Mullan, Pierre Yves Desprez, Ruby Siada, and Amy L. Pasternak
- Subjects
Cancer Research ,Oncology ,Downregulation and upregulation ,Cancer research ,Tumor Microenvironment/Angiogenesis/Metabolism/Invasion ,Neurology (clinical) - Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are lethal brain tumors with no effective therapies other than radiation. Inhibitor of DNA binding (ID) proteins, key regulators of lineage commitment during embryogenesis, are implicated in tumorigenesis in multiple human cancers. Prior work showed that recurrent H3F3A and ACVR1 mutations increase ID1 expression in cultured astrocytes. However, this has not been validated in human DIPG. The regulation and targetability of ID1 in DIPG has not been explored either. Exome and transcriptome sequencing analysis of multi-focal DIPG tumors and normal brain tissue from autopsy (n=52) revealed that ID1 expression is significantly elevated in DIPG tissues. Higher ID1 expression correlates with reduced survival in DIPG patients and increased regional invasion in multi-focal autopsy samples. Analyses of developing mouse brain RNA/ChiP-Seq data revealed high ID1 expression and H3K27ac promoter binding in prenatal hind brain compared to all other prenatal and postnatal brain regions. ChIP-qPCR for H3K27ac and H3K27me3 revealed that ID1 gene regulatory regions are epigenetically poised for upregulation in DIPG tissues compared to normal brain, regardless of H3/ACVR1 mutational status. These data support that the developing pons is regionally poised for ID1 activation. Genetic (shRNA) ID1 knockdown in primary human H3.3K27M-DIPG cells (DIPG007) resulted in significantly reduced invasion and migration in vitro. Additionally, DIPG-ID1-KO cells showed improved sensitivity to radiation therapy. Phospho-kinase array analysis of DIPG cells revealed that Akt and WNK1 activity were significantly downregulated upon ID1 knockdown, which was previously shown in lung tumors. Treatment of DIPG007 cells with cannabidiol (CBD) reduced ID1 expression levels and viability/proliferation of DIPG cells in vitro. ID1 knockdown and CBD treatment studies in vivo are ongoing. In summary, our findings indicate that multifactorial (genetic and regional) epigenetic upregulation of ID1 drives DIPG invasiveness and targeting ID1 using CBD may be a potential strategy for the treatment of DIPGs.
- Published
- 2020
- Full Text
- View/download PDF
23. Abstract 6267: Repurposing antiandrogens to overcome therapy resistance in androgen receptor-positive glioblastoma
- Author
-
Sriram Venneti, Roy E. Strowd, Corey Speers, Stefanie Stallard, Arul Chinnayain, Hanshi Sun, Jann N. Sarkaria, Carl Koschmann, Arvind Rao, Edwina Baskin-Bey, Joseph Dresser, Christian K. Werner, Daniel E. Spratt, Meredith A. Morgan, Alexander M. Hegedus, Howard Colman, Daniel R. Wahl, Weihua Zhou, Theodore S. Lawrence, Uchechi J. Nna, Ayesha U. Kothari, Kari Wilder-Romans, Tarik Bor, Waldemar Debinski, Joel R. Eisner, and Yangyang Yao
- Subjects
Cancer Research ,Oncology ,business.industry ,Antiandrogens ,Cancer research ,medicine ,Androgen Receptor Positive ,Treatment resistance ,medicine.disease ,business ,Repurposing ,Glioblastoma - Abstract
New approaches are needed to overcome intrinsic therapy resistance in glioblastoma (GBM). Because GBMs exhibit sexual dimorphism and are reported to express steroid hormone receptors, we reasoned that signaling through the androgen receptor (AR) could mediate therapy resistance in GBM, as it does in AR-positive prostate and breast cancers. Using RNAseq, immunoblot and immunohistochemistry, we found that nearly half of GBM cell lines, patient-derived xenografts and human tumors express AR transcript and protein with levels that overlap those of primary prostate cancer. AR expression in GBM did not vary by sex, age or common molecular alterations. We identified two cell line models of GBM that expressed AR protein (LN18 and T98G: termed “AR positive”) and two that did not (8MGBA and AM38: termed “AR negative”). Seviteronel, a blood-brain barrier permeable CYP17 lyase inhibitor and antiandrogen slowed growth in AR positive GBM cell lines (GI50 3-4 µM) but not AR negative lines (GI50 > 500 µM) as measured by the colony formation assay. The antiandrogen enzalutamide, which also penetrates the blood brain barrier, similarly preferentially slowed growth in AR positive GBM cell lines. Seviteronel and enzalutamide sensitized AR positive GBM cell lines to radiation with enhancement ratios of 1.3-1.6 as measured by the clonogenic survival assay. Antiandrogens had no effect on the radiosensitivity of AR negative GBM cell lines. Seviteronel treatment did not affect the growth of AR positive T98G xenografts grown in vivo, but did sensitize these tumors to radiation (median time to tripling: 15 d with radiation alone and not reached with radiation combined with seviteronel). Enzalutamide similarly had modest single agent effects on an AR positive GBM patient-derived xenograft (GBM26 from the Mayo Clinic GBM PDX national resource) grown in vivo but sensitized these tumors to radiation (median time to tripling: 25.5 d with radiation alone and 39 d with radiation combined with enzalutamide). RNAseq performed on GBM26 tumors grown in vivo revealed that enzalutamide treatment caused minimal transcriptional changes when given as monotherapy but, when given in combination with radiation, blocked the ability of AR-positive GBMs to engage adaptive transcriptional programs related to multiple DNA repair pathways. We confirmed these mechanistic findings in vitro, as antiandrogens selectively impaired the repair of radiation-induced double strand DNA breaks in AR positive GBM cell lines. These results suggest that AR signaling may mediate therapy resistance in AR positive GBMs, and patients with these tumors could derive clinical benefit from combination therapies involving radiation and blood-brain-barrier permeable antiandrogens. Citation Format: Christian K. Werner, Uchechi Nna, Hanshi Sun, Kari Wilder-Romans, Joseph Dresser, Ayesha Kothari, Weihua Zhou, Yangyang Yao, Arvind Rao, Stefanie Stallard, Carl Koschmann, Tarik Bor, Waldemar Debinski, Alexander Hegedus, Meredith Morgan, Sriram Venneti, Edwina Baskin-Bey, Daniel Spratt, Howard Colman, Jann Sarkaria, Arul Chinnayain, Joel Eisner, Corey Speers, Theodore S. Lawrence, Roy Strowd, Daniel R. Wahl. Repurposing antiandrogens to overcome therapy resistance in androgen receptor-positive glioblastoma [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 6267.
- Published
- 2020
- Full Text
- View/download PDF
24. CSF H3F3A K27M circulating tumor DNA copy number quantifies tumor growth and in vitro treatment response
- Author
-
Ruby Siada, Rintaro Hashizume, Sriram Venneti, Masha G. Savelieff, Bailey Anderson, Hugh J. L. Garton, Benjamin H. Singer, Stefanie Stallard, Zachary Miklja, Karin M. Muraszko, Carl Koschmann, Angel M. Carcaboso, Kaitlin Q. McMurray, Kyle Wierzbicki, Jason Heth, Brendan Mullan, Patricia L. Robertson, Rajen Mody, Amy K. Bruzek, and Taylor Garcia
- Subjects
Male ,Treatment response ,medicine.medical_specialty ,Neurology ,In Vitro Techniques ,Adolescent ,DNA Copy Number Variations ,lcsh:RC346-429 ,Pathology and Forensic Medicine ,Circulating Tumor DNA ,Histones ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Text mining ,Methionine ,medicine ,Humans ,Tumor growth ,Child ,Letter to the Editor ,lcsh:Neurology. Diseases of the nervous system ,Cell Proliferation ,medicine.diagnostic_test ,business.industry ,Cell growth ,Brain Neoplasms ,Lysine ,Magnetic resonance imaging ,Glioma ,Magnetic Resonance Imaging ,In vitro ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Female ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - Published
- 2018
25. Medulloblastoma therapy generates risk of a poorly-prognostic H3 wild-type subgroup of diffuse intrinsic pontine glioma: a report from the International DIPG Registry
- Author
-
Arul M. Chinnaiyan, Katayoon Kasaian, Becky Zon, James L. Leach, Martha M. Matuszak, Torunn I. Yock, Pankaj Vats, Maryam Fouladi, Carl Koschmann, Sriram Venneti, Lili Zhao, Marcia Leonard, Stefanie Stallard, Nancy Yanez Escorza, Hunter C. Gits, Christine Fuller, Christopher Howell, Patricia L. Robertson, Tingting Qin, Daniel F. Polan, Nicholas G. Gottardo, Blaise V. Jones, Jacob Hendershot, Chandan Kumar-Sinha, Eric Bouffet, Maia Anderson, Rajen Mody, Ute Bartels, Daniel E. Spratt, Brooklyn Chaney, Sarah Leary, and Drew Pratt
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Oncology ,Male ,Neurology ,medicine.medical_treatment ,International Cooperation ,lcsh:RC346-429 ,Cohort Studies ,Histones ,0302 clinical medicine ,Medicine ,Brain Stem Neoplasms ,Cumulative incidence ,Exome ,Registries ,Child ,Exome sequencing ,Incidence (epidemiology) ,Glioma ,3. Good health ,030220 oncology & carcinogenesis ,Cranial irradiation ,Female ,Brainstem ,Signal Transduction ,Diffuse intrinsic pontine glioma ,medicine.medical_specialty ,Adolescent ,Statistics, Nonparametric ,Pathology and Forensic Medicine ,Secondary malignant neoplasm ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Young Adult ,Internal medicine ,Humans ,Hedgehog Proteins ,Cerebellar Neoplasms ,lcsh:Neurology. Diseases of the nervous system ,Medulloblastoma ,Radiotherapy ,business.industry ,Research ,medicine.disease ,Radiation therapy ,Wnt Proteins ,Mutation ,Neurology (clinical) ,business ,Complication ,Transcriptome ,030217 neurology & neurosurgery - Abstract
With improved survivorship in medulloblastoma, there has been an increasing incidence of late complications. To date, no studies have specifically addressed the risk of radiation-associated diffuse intrinsic pontine glioma (DIPG) in medulloblastoma survivors. Query of the International DIPG Registry identified six cases of DIPG with a history of medulloblastoma treated with radiotherapy. All patients underwent central radiologic review that confirmed a diagnosis of DIPG. Six additional cases were identified in reports from recent cooperative group medulloblastoma trials (total n = 12; ages 7 to 21 years). From these cases, molecular subgrouping of primary medulloblastomas with available tissue (n = 5) revealed only non-WNT, non-SHH subgroups (group 3 or 4). The estimated cumulative incidence of DIPG after post-treatment medulloblastoma ranged from 0.3–3.9%. Posterior fossa radiation exposure (including brainstem) was greater than 53.0 Gy in all cases with available details. Tumor/germline exome sequencing of three radiation-associated DIPGs revealed an H3 wild-type status and mutational signature distinct from primary DIPG with evidence of radiation-induced DNA damage. Mutations identified in the radiation-associated DIPGs had significant molecular overlap with recurrent drivers of adult glioblastoma (e.g. NRAS, EGFR, and PTEN), as opposed to epigenetic dysregulation in H3-driven primary DIPGs. Patients with radiation-associated DIPG had a significantly worse median overall survival (median 8 months; range 4–17 months) compared to patients with primary DIPG. Here, it is demonstrated that DIPG occurs as a not infrequent complication of radiation therapy in survivors of pediatric medulloblastoma and that radiation-associated DIPGs may present as a poorly-prognostic distinct molecular subgroup of H3 wild-type DIPG. Given the abysmal survival of these cases, these findings provide a compelling argument for efforts to reduce exposure of the brainstem in the treatment of medulloblastoma. Additionally, patients with radiation-associated DIPG may benefit from future therapies targeted to the molecular features of adult glioblastoma rather than primary DIPG. Electronic supplementary material The online version of this article (10.1186/s40478-018-0570-9) contains supplementary material, which is available to authorized users.
- Published
- 2018
26. DIPG-38. ID1 EXPRESSION CORRELATES WITH H3F3A K27M MUTATION AND EXTRA-PONTINE INVASION IN DIPG
- Author
-
Xuhong Cao, Cynthia Hawkins, Bailey Anderson, Stefanie Stallard, Brendan Mullan, Katayoon Kasaian, Sriram Venneti, Rajen Mody, Maria G. Castro, Pedro R. Lowenstein, Taylor Garcia, Zachary Miklja, Carl Koschmann, Daniel Zamler, Arul M. Chinnaiyan, Robert Siddaway, and Shawn L. Hervey-Jumper
- Subjects
Solid tumour ,Cancer Research ,K27m mutation ,Tumor size ,business.industry ,medicine.disease ,Abstracts ,Oncology ,Glioma ,medicine ,Cancer research ,Neurology (clinical) ,business ,Basic Helix-Loop-Helix Transcription Factors - Abstract
ID1 regulates transcription by interacting with bHLH transcription factors and previous work has shown that over-expression of the recurrent DIPG H3F3A K27M and ACVR1 mutations in cultured astrocytes lead to an increase in ID1 expression; this has not been validated in human DIPG. DNA (exome)/RNA sequencing of 34 DIPGs and 17 normal samples (SickKids) revealed that ID1 expression was significantly increased in tumor as compared to normal (p=0.001). ID1 expression was significantly higher in H3F3A K27M-mutated tumors as compared to normal (p=0.003), but not in ACVR1-mutated tumors. This was confirmed in an analysis of pediatric high-grade gliomas (PedcBioPortal) where ID1 expression was increased in H3F3A K27M-mutated tumors as compared to H3 wildtype (p=0.0055, n=189), but not in ACVR1-mutated tumors as compared to ACVR1 wildtype (p=0.1178, n=114). In an additional patient with DIPG at autopsy, multi-focal sequencing revealed clonal mutations in HIST1H3B K27M and ACVR1 and ID1 expression correlated with tumor size and cerebellar invasion. We identified several genes whose expression in pediatric HGG correlated with that of ID1 and which have been implicated in invasion and/or metastasis in various solid tumors. ChipSeq revealed reduced K27 me3 and elevated K27 acetylation at the ID1 locus in multiple K27M-mutant DIPG cell lines, pointing to an epigenetic control of this phenotype in H3F3A K27M-mutated DIPGs. Based on these data, we propose that epigenetically controlled upregulation of ID1 promotes DIPG invasion in H3F3A K27M-mutated DIPG and represents an optimal therapeutic target.
- Published
- 2018
27. DIPG-23. BRAINSTEM RADIATION EXPOSURE CONFERS SUBSTANTIAL RISK OF DIFFUSE INTRINSIC PONTINE GLIOMA (DIPG) IN MEDULLOBLASTOMA SURVIVORS: A REPORT FROM THE INTERNATIONAL DIPG REGISTRY
- Author
-
Christine Fuller, Drew Pratt, Maryam Fouladi, Sarah Leary, Ute Bartels, Tingting Qin, Carl Koschmann, Daniel F. Polan, Nicholas G. Gottardo, Maia Anderson, Rajen Mody, Katayoon Kasaian, Blaise V. Jones, Hunter C. Gits, Becky Zon, Eric Bouffet, Stefanie Stallard, Daniel E. Spratt, Siriam Venneti, Torunn I. Yock, Marcia Leonard, Christopher Howell, Martha M. Matuszak, James L. Leach, and Patricia L. Robertson
- Subjects
Oncology ,Medulloblastoma ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.disease ,Radiation exposure ,Abstracts ,Internal medicine ,medicine ,Neurology (clinical) ,Brainstem ,business - Abstract
With improved survivorship in medulloblastoma, there has been increasing recognition of the occurrence of secondary malignant brain tumors. To date, no studies have specifically addressed the risk of diffuse intrinsic pontine glioma (DIPG) in medulloblastoma survivors. We queried the International DIPG Registry and identified six cases of DIPG with prior medulloblastoma. Six additional cases were identified in reports from recent cooperative group medulloblastoma trials. Incidence of DIPG after medulloblastoma ranged from 0.3–3.9%. All 12 cases underwent surgical resection followed by craniospinal photon irradiation (range 18–36 Gy) and posterior fossa boost (range 19.8–36 Gy). Posterior fossa exposure was greater than 53 Gy in all cases. Median time to diagnosis of secondary DIPG was 7 years (range 2–11 years). Patients died of secondary DIPG a median of 8 months after diagnosis (range 4–17 months). Molecular subgroup of primary medulloblastomas with available tissue (n=5) revealed only non-WNT, non-SHH subgroups (group 3 or 4). Tumor/germline exome sequencing of three secondary DIPGs demonstrated tumors to be H3.3 wildtype and harbor higher mutational burden than radiation-naïve DIPGs. Mutational signature analysis of secondary DIPGs showed mutations consistent with radiation-induced DNA damage (e.g., insertional event in TP53), as well as mutations in other oncogenic drivers (e.g., NRAS, PI3KCA), suggestive of distinct mutational processes compared with primary DIPGs. In conclusion, we report for the first time that survivors of pediatric medulloblastoma are at risk for the development of secondary DIPG, likely consequent to radiation exposure. This risk highlights the importance of radiation field, volume, and modality in medulloblastoma treatment.
- Published
- 2018
28. PDTM-10. USE OF A NOVEL, HAND-HELD, ELECTRONIC DNA ANALYSIS PLATFORM TO QUANTIFY MULTI-GENE MOLECULAR RESPONSE IN CSF OF PATIENTS WITH HIGH-GRADE GLIOMA
- Author
-
Leo Tunkle, Carl Koschmann, Ian Wolfe, Cormac O. Maher, Tingting Qin, Clarissa Babila, Rajen Mody, Patricia L. Robertson, Hugh J. L. Garton, Ashwath Muruganand, Veena Thamilselvan, Amy K. Bruzek, Karin M. Muraszko, and Stefanie Stallard
- Subjects
Cancer Research ,Mutation ,Pediatric Tumors ,business.industry ,medicine.disease ,medicine.disease_cause ,Multi gene ,law.invention ,chemistry.chemical_compound ,Oncology ,chemistry ,law ,Molecular Response ,Glioma ,Cancer research ,Medicine ,Neurology (clinical) ,business ,Gene ,Polymerase chain reaction ,DNA ,High-Grade Glioma - Abstract
BACKGROUND For midline tumors, surgical biopsy risks neurological injury. Non-invasive methods for diagnosis and surveillance are greatly needed. Tumors release DNA into cerebrospinal fluid (CSF-ctDNA), allowing for potential detection and serial monitoring of tumor-associated genetic mutations by CSF sampling. Current detection platforms are limited by their requirement for assay development for each mutation (digital droplet PCR), or cost and timeliness (Illumina sequencing). We hypothesized that direct, electronic analysis of CSF-ctDNA with a novel, hand-held platform (Oxford Nanopore MinION) could provide real-time, ultra-deep sequencing of patient-specific alterations in CSF-ctDNA. METHODS We established multiple clinical trials for pediatric high-grade glioma with required multi-time point (0, 2, and 6 month) correlate lumbar puncture (LP) at time of MRI, with accrual ongoing. We performed amplicon-based PCR on CSF-ctDNA for recurrent mutations and sequenced patient samples (tumor tissue n=8, tumor CSF n=60) and normal controls (tissue n=5, CSF n=24) using NanoPore technology. Variant allele fractions (VAF) were determined via MinKNOW, Guppy, MiniMap2, and Integrated Genome Browser. RESULTS Sensitivity was 79% and specificity 100% by NanoPore. Time from LP to results was 12 hours. A 17-year-old female presented with a biopsy-proven grade IV thalamic glioma with clonal mutations in H3F3A K27M, PIK3CA E545G, TP53 R158G, and TP53 R248Q. After failing standard treatment, she was enrolled in the ONC201 clinical trial and underwent serial LPs. MRI showed stable tumor at 2 months and 40% decrease at 6 months of treatment. H3K27M VAF increased from baseline at 2 months, but decreased to 1% at 6 months of treatment, results that were confirmed by ddPCR. PIK3CA E545G, TP53 R158G, and TP53 R248Q demonstrated the same decrease in VAF, with p-value of < 0.0001. CONCLUSIONS We demonstrate a rapid, reliable method to detect tumor mutations in CSF, and further show molecular remission of H3K27M glioma by CSF sampling.
- Published
- 2019
- Full Text
- View/download PDF
29. PDTM-29. CSF H3F3A K27M CIRCULATING TUMOR DNA COPY NUMBER QUANTIFIES TUMOR GROWTH AND TREATMENT RESPONSE
- Author
-
Masha G. Savelieff, Kaitlin Q. McMurray, Kyle Wierzbicki, Jason Heth, Brendan Mullan, Zachary Miklja, Amy K. Bruzek, Rintaro Hashizume, Patricia L. Robertson, Karin M. Muraszko, Hugh J. L. Garton, Benjamin H. Singer, Carl Koschmann, Angel M. Carcaboso, Rajen Mody, Stefanie Stallard, Sriram Venneti, and Taylor Garcia
- Subjects
Abstracts ,Cancer Research ,Treatment response ,Oncology ,Circulating tumor DNA ,business.industry ,Cancer research ,Medicine ,Tumor growth ,Neurology (clinical) ,business - Abstract
Primary brain tumors and CNS metastases shed circulating tumor DNA (ctDNA) into the CSF, which can be assessed for tumor-associated mutations. Thus far, there have been no extensive studies using droplet digital PCR (ddPCR) to detect and quantify ctDNA in the CSF of pediatric high-grade brain tumor patients. There are also gaps in our knowledge, including the potential dependence of ctDNA amount on location of sample collection and whether ctDNA can be used to quantify tumor growth and treatment response. To address these questions, we developed a novel H3F3A K27M ddPCR assay and applied it to four pediatric patients with H3F3A K27M-mutant DIPG and GBM. We found that ddPCR was able to detect the K27M mutation in patient CSF and that the closest relation emerged between mutant K27M copies per ng of total DNA (henceforth K27M copies) and contrast-enhancing tumor area on MRI. Multi-focal CSF sampling at autopsy of a DIPG patient exhibited differences in K27M copies by proximity to the tumor. To better understand changes in K27M copies in response to both growth and treatment of DIPG, we developed an in vitro system comprised of astrocytes (NHAs) co-cultured with luciferase-expressing human DIPG cell line DIPG007 as a means to simulate ctDNA release into the CSF. We found that DIPG007 cells released ctDNA into culture media in proportion to their proliferation, even when the media was changed frequently to approximate the constant production and resorption of CSF. Irradiation with 8 Gy resulted in a spike in mutant ctDNA 72–120 hours post-radiotherapy before decreasing. In summary, our study suggests that H3F3A K27M copies in the CSF of children with high-grade brain tumors have a linear relation with contrast-enhancing tumor area and that ddPCR can be used to follow treatment response including ctDNA release shortly after effective therapies.
- Published
- 2018
- Full Text
- View/download PDF
30. PDTM-42. CROSS-PLATFORM PROFILING OF ctDNA USING ddPCR: STANDARDIZATION OF THE LIQUID BIOPSY FOR PEDIATRIC DIFFUSE MIDLINE GLIOMA
- Author
-
Rishi Lulla, Daphne Li, Stefanie Stallard, Carl Koschmann, Javad Nazarian, Eshini Panditharatna, Tina Huang, Amanda Saratsis, and Erin R. Bonner
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Oncology ,Pediatric Tumors ,business.industry ,Glioma ,medicine ,Profiling (information science) ,Neurology (clinical) ,Liquid biopsy ,medicine.disease ,business - Abstract
INTRODUCTION Pediatric diffuse midline glioma (DMG) with histone H3 mutations (80%), are highly morbid tumors with poor response to therapy. We previously detected H3 mutations in circulating tumor DNA (ctDNA) from CSF derived from children with DMG and high grade glioma. Here, we describe a high-throughput, sensitive and specific approach for H3 mutation detection and quantification in plasma and CSF specimens, validated across multiple centers. METHODS DNA extracted from tissue-validated H3.3K27M specimens (4 tumor, 5 CSF, 4 plasma) and pediatric glioma cell lines (1 wild-type, 1 H3.3K27M) were used to standardize ddPCR workflows. ctDNA extracted from 500uL of CSF or plasma was pre-amplified using sequence-specific primers, then analyzed on RainDance and BioRad ddPCR systems using two sets of custom primers and fluorescent LNA probes (Assays A and B). H3 mutation allelic frequency (MAF) was determined across specimens, and validated through parallel analysis of additional matched H3.3K27M tumor tissue, CSF and plasma specimens (n=3). RESULTS ctDNA processing and detection was standardized across three institutions and two platforms with sensitive, specific and reproducible H3 mutation detection. H3K27M mutations were detected in as little as 0.3-2pg input DNA on both platforms, with comparable MAFs across instruments and assays. RainDance yielded greater overall positive droplet number detection (increased sensitivity). Differences in optimal sample input volume between platforms were circumvented by vacuum concentration or dilution to maintain equal input DNA. Assay A yielded superior specificity so was used for subsequent matched specimen analysis. Mutation detection in plasma, as in previous studies, remained challenging due to low [ctDNA], while CSF analysis yielded reliable results across assays and platforms. CONCLUSIONS We demonstrate the utility of liquid biopsy for identifying H3K27M mutations in plasma and CSF via ddPCR with low starting [DNA], representing a rapid, minimally invasive method for diagnosis and therapeutic monitoring of DMG.
- Published
- 2019
- Full Text
- View/download PDF
31. The effect of everolimus on CNS penetration and efficacy of dasatinib in the treatment of PDGFRA-driven glioma
- Author
-
Zachary Miklja, Alyssa Paul, Marcia Leonard, Ruby Siada, Manjunath P. Pai, Amy K. Bruzek, Brendan Mullan, Patricia L. Robertson, Stefanie Stallard, Carl Koschmann, Bernard L. Marini, Timothy N. Phoenix, Viveka Nand Yadav, and Taylor Garcia
- Subjects
Cancer Research ,Everolimus ,business.industry ,PDGFRA ,medicine.disease ,Cns penetration ,Dasatinib ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Glioma ,medicine ,Cancer research ,business ,030215 immunology ,medicine.drug - Abstract
e13508 Background: Pediatric and adult high-grade glioma (HGG) frequently harbor PDGFRA alterations. The CNS penetration of PDGFRA inhibitors, such as dasatinib, is limited by the tumor-efflux protein P-glycoprotein (P-gp). We hypothesized that co-treatment with everolimus, which has been shown to block P-gp, will increase CNS penetration and efficacy of dasatinib in in vitro and in vivo models as well as in human PDGFRA-driven glioma. Methods: Tumors were generated in mice using an intra-uterine electroporation (IUE) model [introduction of TP53, PDGFRA and H3K27M mutations in pre-natal cortex]. Dose response, synergism studies, P-GP inhibition and pharmacodynamics/pharmacokinetic studies were then performed on in vitro and in vivo models employing this IUE system. A phase 2 trial employing dasatinib and everolimus was established for children with HGG and diffuse intrinsic pontine glioma (DIPG) that contain PDGFRA alterations (NCT03352427). Paired CSF/plasma samples (before and after addition of everolimus) were collected from enrolled patients. Results: Dasatinib effectively treated mouse HGG cells with an IC50 of 100 nM. Dose-dependent reduction in PDGFRA and pPDGFRA was found. P-gp inhibitor assay confirmed that everolimus strongly blocks P-gp activity at 1 uM (p = 0.0028 vs untreated). Mice treated with dasatinib and everolimus had extended survival as compared to control. Two-hour exposure to everolimus resulted in sub-IC50 dasatinib concentration in cortex (23 nM) and tumor (65 nM). 24-hour exposure to everolimus resulted in greater cortex (235 nM) and tumor (509 nM) concentrations. Two trial patients, recurrent HGG ( PDGFRA-amplified) and recurrent DIPG ( PDGFRA D842V) respectively, survived 6 months and 9 months (ongoing) after progression, which compares very favorably to historical controls. A paired CSF sample from the PDGFRA-amplified patient showed a 50% increase in CSF dasatinib level after addition of everolimus. Conclusions: Dasatinib treatment of PDGFRA-driven HGG is improved with everolimus blockade of P-gp and represents a novel route for improving CNS penetration and efficacy of therapies for HGG. Clinical trial information: NCT03352427.
- Published
- 2019
- Full Text
- View/download PDF
32. HGG-03. EVEROLIMUS TREATMENT IMPROVES THE CNS PENETRATION AND EFFICACY OF DASATINIB IN THE TREATMENT OF PDGFRA-DRIVEN PEDIATRIC HIGH-GRADE GLIOMA AND DIFFUSE INTRINSIC PONTINE GLIOMA
- Author
-
Alyssa Paul, Taylor Garcia, Zachary Miklja, Marcia Leonard, Brendan Mullan, Viveka Nand Yadav, Patricia L. Robertson, Carl Koschmann, Bernard L. Marini, Amy K. Bruzek, Manjunath P. Pai, Timothy N. Phoenix, and Stefanie Stallard
- Subjects
Cancer Research ,Everolimus ,business.industry ,PDGFRA ,medicine.disease ,digestive system diseases ,Cns penetration ,Dasatinib ,Oncology ,Glioma ,medicine ,Cancer research ,Neurology (clinical) ,High Grade Glioma ,business ,Platelet-Derived Growth Factor alpha Receptor ,Protein p53 ,High-Grade Glioma ,medicine.drug - Abstract
Pediatric high-grade glioma (HGG) and diffuse intrinsic pontine glioma (DIPG) frequently harbor alterations in PDGFRA. The CNS penetration of PDGFRA inhibitors, such as dasatinib, is limited by the tumor-efflux protein P-glycoprotein (P-gp). We hypothesized that co-treatment with everolimus, which has been shown to block P-gp in non-tumor models, would increase CNS penetration and efficacy of dasatinib in PDGFRA-driven HGG and DIPG. Dasatinib effectively treated mouse DIPG cells generated from an intra-uterine electroporation (IUE) model (TP53, PDGFRA and H3K27M mutations), with an IC(50) of 100 nM and a dose-dependent reduction in PDGFRA and pPDGFRA by western blot. Using an in vitro P-gp inhibitor assay, we confirmed that everolimus strongly blocks P-gp activity at 1 uM (p=0.0028 vs untreated, and NS vs complete P-gp block). Treatment studies using the IUE model are ongoing. Brief treatment with everolimus resulted in sub-IC(50) dasatinib average mouse cortex (23 nM) and tumor (65 nM) concentrations by mass spectroscopy, but prolonged (>24 hours) everolimus exposure resulted in improved average cortex (288 nM) and brainstem tumor (360 nM) concentrations. Based on this promising pre-clinical data, we established a phase 2 trial employing dasatinib and everolimus in children with HGG and DIPG that contain PDGFRA alterations (NCT03352427). The first two patients (a recurrent PDGFRA-amplified HGG and a recurrent PDGFRA D842V-mutated DIPG) treated with dasatinib and everolimus after re-irradiation survived 6 months and 9 months (ongoing), respectively, after progression, which compares very favorably to historical controls. Paired CSF samples (before and after addition of everolimus) from the PDGFRA-amplified patient showed a 50% increase in CSF dasatinib level after addition of everolimus. In summary, we demonstrate that dasatinib treatment of PDGFRA-driven pediatric HGG and DIPG is improved with everolimus blockade of P-gp. This represents a novel route for improving the CNS penetration and efficacy of precision therapies for pediatric HGG.
- Published
- 2019
- Full Text
- View/download PDF
33. Climbing Bloom's taxonomy pyramid: Lessons from a graduate histology course
- Author
-
Nikki B, Zaidi, Charles, Hwang, Sara, Scott, Stefanie, Stallard, Joel, Purkiss, and Michael, Hortsch
- Subjects
Male ,Cognition ,Histology ,Students, Medical ,Academic Performance ,Humans ,Female ,Educational Measurement ,Choice Behavior ,Education, Medical, Undergraduate ,Feedback - Abstract
Bloom's taxonomy was adopted to create a subject-specific scoring tool for histology multiple-choice questions (MCQs). This Bloom's Taxonomy Histology Tool (BTHT) was used to analyze teacher- and student-generated quiz and examination questions from a graduate level histology course. Multiple-choice questions using histological images were generally assigned a higher BTHT level than simple text questions. The type of microscopy technique (light or electron microscopy) used for these image-based questions did not result in any significant differences in their Bloom's taxonomy scores. The BTHT levels for teacher-generated MCQs correlated positively with higher discrimination indices and inversely with the percent of students answering these questions correctly (difficulty index), suggesting that higher-level Bloom's taxonomy questions differentiate well between higher- and lower-performing students. When examining BTHT scores for MCQs that were written by students in a Multiple-Choice Item Development Assignment (MCIDA) there was no significant correlation between these scores and the students' ability to answer teacher-generated MCQs. This suggests that the ability to answer histology MCQs relies on a different skill set than the aptitude to construct higher-level Bloom's taxonomy questions. However, students significantly improved their average BTHT scores from the midterm to the final MCIDA task, which indicates that practice, experience and feedback increased their MCQ writing proficiency. Anat Sci Educ 10: 456-464. © 2017 American Association of Anatomists.
- Published
- 2016
34. TMIC-15MYELOID DERIVED SUPPRESSOR CELLS' TRAFFICKING INTO GBM IS REGULATED BY CXCR2 SIGNALING
- Author
-
Alexandra Calinescue, Maria G. Castro, Mariela-Moreno Ayala, Nicholas Raja, Felipe J Nunez, Stefanie Stallard, Anuska V. Andjelkovic, Neha Kamran, and Pedro R. Lowenstein
- Subjects
Cancer Research ,business.industry ,Biology ,Cell biology ,law.invention ,Text mining ,Oncology ,law ,Suppressor T Lymphocyte ,Suppressor ,Neurology (clinical) ,CXC chemokine receptors ,Signal transduction ,business ,Abstracts from the 20th Annual Scientific Meeting of the Society for Neuro-Oncology - Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.