62 results on '"Trent R. Hummel"'
Search Results
2. Data from Gene Expression Analysis Identifies Potential Biomarkers of Neurofibromatosis Type 1 Including Adrenomedullin
- Author
-
Nancy Ratner, Elizabeth K. Schorry, Bruce J. Aronow, Paul F. Worley, Grier P. Page, Conxi Lázaro, Margaret R. Wallace, Victor F. Mautner, Lan Kluwe, Shyra J. Miller, Walter J. Jessen, and Trent R. Hummel
- Abstract
Purpose: Plexiform neurofibromas (pNF) are Schwann cell tumors found in a third of individuals with neurofibromatosis type 1 (NF1). pNF can undergo transformation to malignant peripheral nerve sheath tumors (MPNST). There are no identified serum biomarkers of pNF tumor burden or transformation to MPNST. Serum biomarkers would be useful to verify NF1 diagnosis, monitor tumor burden, and/or detect transformation.Experimental Design: We used microarray gene expression analysis to define 92 genes that encode putative secreted proteins in neurofibroma Schwann cells, neurofibromas, and MPNST. We validated differential expression by quantitative reverse transcription-PCR, Western blotting, and ELISA assays in cell conditioned medium and control and NF1 patient sera.Results: Of 13 candidate genes evaluated, only adrenomedullin (ADM) was confirmed as differentially expressed and elevated in serum of NF1 patients. ADM protein concentrati on was further elevated in serum of a small sampling of NF1 patients with MPNST. MPNST cell conditioned medium, containing ADM and hepatocyte growth factor, stimulated MPNST migration and endothelial cell proliferation.Conclusions: Thus, microarray analysis identifies potential serum biomarkers for disease, and ADM is a serum biomarker of NF1. ADM serum levels do not seem to correlate with the presence of pNFs but may be a biomarker of transformation to MPNST. Clin Cancer Res; 16(20); 5048–57. ©2010 AACR.
- Published
- 2023
3. Data from A Phase I and Pharmacokinetic Study of Oral Dabrafenib in Children and Adolescent Patients with Recurrent or Refractory BRAF V600 Mutation–Positive Solid Tumors
- Author
-
James A. Whitlock, Mark W. Russo, Steven Green, Noelia Nebot, Lillian Tseng, Andrew D.J. Pearson, Christine A. Pratilas, Eric Bouffet, Violet Shen, Trent R. Hummel, Kenneth J. Cohen, Anne-Isabelle Bertozzi, Isabelle Aerts, Pooja Hingorani, Darren Hargrave, Alberto Broniscer, Ira J. Dunkel, Birgit Geoerger, and Mark W. Kieran
- Abstract
Purpose:The 2-part, phase I/IIa, open-label study (NCT01677741) sought to determine the safety, tolerability, pharmacokinetics, and preliminary activity of dabrafenib in pediatric patients with advanced BRAF V600–mutated cancers.Patients and Methods:This phase I dose-finding part treated patients ages 1 to BRAF V600 mutation–positive tumors with oral dabrafenib 3 to 5.25 mg/kg/day to determine the RP2D based on safety and drug exposure target.Results:Between May 2013 and November 2014, 27 patients [12 male; median age, 9 years (range, 1–17 years)] with BRAF V600–mutant solid tumors recurrent/refractory to treatment (low- or high-grade glioma, Langerhans cell histiocytosis, neuroblastoma, or thyroid cancer) were enrolled. The median treatment duration was 75.6 weeks (range, 5.6–148.7 weeks), with 63% treated for >52 weeks and 52% undergoing treatment at data cutoff date. The most common grade 3/4 adverse events suspected to be related to study drug were maculopapular rash and arthralgia (2 patients each). No dose-limiting toxicities were observed. Pharmacokinetic analyses showed a dose-dependent increase in AUC0–12 and achievement of adult exposure levels at the recommended phase II doses of 5.25 mg/kg/day (age Conclusions:In this first clinical trial in pediatric patients with pretreated BRAF V600–mutant tumors, dabrafenib was well tolerated while achieving target exposure levels; the average treatment duration was >1 year with many patients still on treatment. The phase II component is also closed and will be reported separately.
- Published
- 2023
4. Table S1 from A Phase I and Pharmacokinetic Study of Oral Dabrafenib in Children and Adolescent Patients with Recurrent or Refractory BRAF V600 Mutation–Positive Solid Tumors
- Author
-
James A. Whitlock, Mark W. Russo, Steven Green, Noelia Nebot, Lillian Tseng, Andrew D.J. Pearson, Christine A. Pratilas, Eric Bouffet, Violet Shen, Trent R. Hummel, Kenneth J. Cohen, Anne-Isabelle Bertozzi, Isabelle Aerts, Pooja Hingorani, Darren Hargrave, Alberto Broniscer, Ira J. Dunkel, Birgit Geoerger, and Mark W. Kieran
- Abstract
Global study locations for the phase 1 part of the study
- Published
- 2023
5. Table S4 from A Phase I and Pharmacokinetic Study of Oral Dabrafenib in Children and Adolescent Patients with Recurrent or Refractory BRAF V600 Mutation–Positive Solid Tumors
- Author
-
James A. Whitlock, Mark W. Russo, Steven Green, Noelia Nebot, Lillian Tseng, Andrew D.J. Pearson, Christine A. Pratilas, Eric Bouffet, Violet Shen, Trent R. Hummel, Kenneth J. Cohen, Anne-Isabelle Bertozzi, Isabelle Aerts, Pooja Hingorani, Darren Hargrave, Alberto Broniscer, Ira J. Dunkel, Birgit Geoerger, and Mark W. Kieran
- Abstract
Adverse events, regardless of study drug relationship, by preferred term and maximum grade (>20% overall)
- Published
- 2023
6. Table S3 from A Phase I and Pharmacokinetic Study of Oral Dabrafenib in Children and Adolescent Patients with Recurrent or Refractory BRAF V600 Mutation–Positive Solid Tumors
- Author
-
James A. Whitlock, Mark W. Russo, Steven Green, Noelia Nebot, Lillian Tseng, Andrew D.J. Pearson, Christine A. Pratilas, Eric Bouffet, Violet Shen, Trent R. Hummel, Kenneth J. Cohen, Anne-Isabelle Bertozzi, Isabelle Aerts, Pooja Hingorani, Darren Hargrave, Alberto Broniscer, Ira J. Dunkel, Birgit Geoerger, and Mark W. Kieran
- Abstract
Treatment history
- Published
- 2023
7. Supplement 1 from Gene Expression Analysis Identifies Potential Biomarkers of Neurofibromatosis Type 1 Including Adrenomedullin
- Author
-
Nancy Ratner, Elizabeth K. Schorry, Bruce J. Aronow, Paul F. Worley, Grier P. Page, Conxi Lázaro, Margaret R. Wallace, Victor F. Mautner, Lan Kluwe, Shyra J. Miller, Walter J. Jessen, and Trent R. Hummel
- Abstract
PDF file - 6K, List of the forward and reverse quantitative real-time PCR primers are shown in Supplement 1.
- Published
- 2023
8. Figure S1 from A Phase I and Pharmacokinetic Study of Oral Dabrafenib in Children and Adolescent Patients with Recurrent or Refractory BRAF V600 Mutation–Positive Solid Tumors
- Author
-
James A. Whitlock, Mark W. Russo, Steven Green, Noelia Nebot, Lillian Tseng, Andrew D.J. Pearson, Christine A. Pratilas, Eric Bouffet, Violet Shen, Trent R. Hummel, Kenneth J. Cohen, Anne-Isabelle Bertozzi, Isabelle Aerts, Pooja Hingorani, Darren Hargrave, Alberto Broniscer, Ira J. Dunkel, Birgit Geoerger, and Mark W. Kieran
- Abstract
Study design
- Published
- 2023
9. Table S2 from A Phase I and Pharmacokinetic Study of Oral Dabrafenib in Children and Adolescent Patients with Recurrent or Refractory BRAF V600 Mutation–Positive Solid Tumors
- Author
-
James A. Whitlock, Mark W. Russo, Steven Green, Noelia Nebot, Lillian Tseng, Andrew D.J. Pearson, Christine A. Pratilas, Eric Bouffet, Violet Shen, Trent R. Hummel, Kenneth J. Cohen, Anne-Isabelle Bertozzi, Isabelle Aerts, Pooja Hingorani, Darren Hargrave, Alberto Broniscer, Ira J. Dunkel, Birgit Geoerger, and Mark W. Kieran
- Abstract
Dabrafenib dosing
- Published
- 2023
10. Volumetric endpoints in diffuse intrinsic pontine glioma: comparison to cross-sectional measures and outcome correlations in the International DIPG/DMG Registry
- Author
-
Margot A Lazow, Martijn T Nievelstein, Adam Lane, Pratiti Bandopadhayhay, Mariko DeWire-Schottmiller, Maryam Fouladi, John W Glod, Robert J Greiner, Lindsey M Hoffman, Trent R Hummel, Lindsay Kilburn, Sarah Leary, Jane E Minturn, Roger Packer, David S Ziegler, Brooklyn Chaney, Katie Black, Peter de Blank, and James L Leach
- Subjects
Cancer Research ,Cross-Sectional Studies ,Glutamates ,Oncology ,Diffuse Intrinsic Pontine Glioma ,Brain Stem Neoplasms ,Humans ,Glioma ,Registries ,Neurology (clinical) ,Astrocytoma ,Pediatric Neuro-Oncology - Abstract
Background Cross-sectional tumor measures are traditional clinical trial endpoints; however volumetric measures may better assess tumor growth. We determined the correlation and compared the prognostic impact of cross-sectional and volumetric measures of progressive disease (PD) among patients with DIPG. Methods Imaging and clinical data were abstracted from the International DIPG Registry. Tumor volume and cross-sectional product (CP) were measured with mint Lesion™ software using manual contouring. Correlation between CP and volume (segmented and mathematical [ellipsoid] model) thresholds of PD were assessed by linear regression. Landmark analyses determined differences in survival (via log-rank) between patients classified as PD versus non-PD by CP and volumetric measurements at 1, 3, 5, 7, and 9 months postradiotherapy (RT). Hazard ratios (HR) for survival after these time points were calculated by Cox regression. Results A total of 312 MRIs (46 patients) were analyzed. Comparing change from the previous smallest measure, CP increase of 25% (PD) correlated with a segmented volume increase of 30% (R2 = 0.710), rather than 40% (spherical model extrapolation). CP-determined PD predicted survival at 1 month post-RT (HR = 2.77), but not other time points. Segmented volumetric-determined PD (40% threshold) predicted survival at all imaging timepoints (HRs = 2.57, 2.62, 3.35, 2.71, 16.29), and 30% volumetric PD threshold predicted survival at 1, 3, 5, and 9 month timepoints (HRs = 2.57, 2.62, 4.65, 5.54). Compared to ellipsoid volume, segmented volume demonstrated superior survival associations. Conclusions Segmented volumetric assessments of PD correlated better with survival than CP or ellipsoid volume at most time points. Semiautomated tumor volume likely represents a more accurate, prognostically-relevant measure of disease burden in DIPG.
- Published
- 2022
11. Overcoming barriers to establishing autopsy procurement programs in pediatric patients with central nervous system tumors: a call to develop regional centers
- Author
-
Elizabeth Gilger, Mariko DeWire, Wanda Meriwether, Shiva Senthil Kumar, Dinah Meister, Maureen Gallagher, Trent R. Hummel, Lori Driver, Anthony Asher, Christine Fuller, Natasha Pillay-Smiley, Lindsey Hoffman, Rachid Drissi, Susan Jelinek, Ralph Salloum, Blaise V. Jones, James L. Leach, Lionel M L Chow, Peter de Blank, Satarupa Sengupta, Maryam Fouladi, Heather Ward, Bridget Kikta, Lili Miles, and Craig Erker
- Subjects
Pediatric CNS tumor ,Cancer Research ,medicine.medical_specialty ,Neurology ,business.industry ,Translational research ,Autopsy ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,Procurement ,Oncology ,030220 oncology & carcinogenesis ,Donation ,medicine ,Pediatric Brain Tumor ,Neurology (clinical) ,Medical emergency ,business ,030217 neurology & neurosurgery ,Patient education - Abstract
While autopsy-repository programs with a variety of pediatric central nervous system (CNS) tumor types are a critical resource for preclinical neuro-oncology research, few exist and there is no published guidance on how to develop one. The goal of this prospective Pediatric Brain Tumor Repository (PBTR) study was to develop such a program at Cincinnati Children’s Hospital Medical Center (CCHMC) and then publish the quantitative and experiential data as a guide to support the development of similar programs. Protocols and infrastructure were established—to educate oncologists and families, establish eligibility, obtain consent, address pre- and post-autopsy logistics (e.g., patient and tissue transportation), process and authenticate tissue samples, and collect and analyze data. Of the 129 pediatric CNS tumor patients at CCHMC who died between 2013 and 2018, 109 were eligible for our study. Of these, 74% (81 of 109) were approached for PBTR donation, and 68% (55 of 81) consented. In the final year of the study, approach and consent rates were 93% and 85%, respectively. Median time from death to autopsy (postmortem interval, PMI) was 10 h (range, 1.5–30). In the outpatient setting, PMI increased with distance (from the hospice/home where the patient died to CCHMC). In all patients, PMI appeared to be lower, when consent was obtained more than 24 h before death. Procurement of autopsy specimens need not be a barrier in neuro-oncology research. Regional centers, strict timing-of-consent, patient education, and dedicated staff are all needed to minimize PMI and, thereby, increase the value of the procured tissue for an array of basic and translational research applications.
- Published
- 2021
12. Characterizing temporal genomic heterogeneity in pediatric low-grade gliomas
- Author
-
Margot A. Lazow, Lindsey Hoffman, Austin Schafer, Diana S. Osorio, Daniel R. Boué, Sarah Rush, Erin Wright, Adam Lane, Mariko D. DeWire-Schottmiller, Teresa Smolarek, Jared Sipple, Heather Taggert, Jaime Reuss, Ralph Salloum, Trent R. Hummel, Peter de Blank, Natasha Pillay-Smiley, Mary E. Sutton, Anthony Asher, Charles B. Stevenson, Rachid Drissi, Jonathan L. Finlay, Maryam Fouladi, and Christine Fuller
- Subjects
Male ,Proto-Oncogene Proteins B-raf ,0301 basic medicine ,Paired ,Adolescent ,lcsh:RC346-429 ,Pathology and Forensic Medicine ,BRAF ,CDKN2A ,Proto-Oncogene Proteins c-myb ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Recurrence ,Proto-Oncogene Proteins ,Humans ,Receptor, trkB ,Receptor, Fibroblast Growth Factor, Type 1 ,Child ,Pediatric low-grade gliomas ,Cyclin-Dependent Kinase Inhibitor p16 ,lcsh:Neurology. Diseases of the nervous system ,Genome ,Membrane Glycoproteins ,Brain Neoplasms ,Research ,Infant ,Glioma ,Genomics ,Isocitrate Dehydrogenase ,030104 developmental biology ,Tumor evolution ,Child, Preschool ,030220 oncology & carcinogenesis ,Disease Progression ,Trans-Activators ,Female ,Neurology (clinical) ,Neoplasm Grading ,Neoplasm Recurrence, Local ,Gene Deletion - Abstract
Recent discoveries have provided valuable insight into the genomic landscape of pediatric low-grade gliomas (LGGs) at diagnosis, facilitating molecularly targeted treatment. However, little is known about their temporal and therapy-related genomic heterogeneity. An adequate understanding of the evolution of pediatric LGGs’ genomic profiles over time is critically important in guiding decisions about targeted therapeutics and diagnostic biopsy at recurrence. Fluorescence in situ hybridization, mutation-specific immunohistochemistry, and/or targeted sequencing were performed on paired tumor samples from primary diagnostic and subsequent surgeries. Ninety-four tumor samples from 45 patients (41 with two specimens, four with three specimens) from three institutions underwent testing. Conservation of BRAF fusion, BRAFV600E mutation, and FGFR1 rearrangement status was observed in 100%, 98%, and 96% of paired specimens, respectively. No loss or gain of IDH1 mutations or NTRK2, MYB, or MYBL1 rearrangements were detected over time. Histologic diagnosis remained the same in all tumors, with no acquired H3K27M mutations or malignant transformation. Changes in CDKN2A deletion status at recurrence occurred in 11 patients (42%), with acquisition of hemizygous CDKN2A deletion in seven and loss in four. Shorter time to progression and shorter time to subsequent surgery were observed among patients with acquired CDKN2A deletions compared to patients without acquisition of this alteration [median time to progression: 5.5 versus 16.0 months (p = 0.048); median time to next surgery: 17.0 months versus 29.0 months (p = 0.031)]. Most targetable genetic aberrations in pediatric LGGs, including BRAF alterations, are conserved at recurrence and following chemotherapy or irradiation. However, changes in CDKN2A deletion status over time were demonstrated. Acquisition of CDKN2A deletion may define a higher risk subgroup of pediatric LGGs with a poorer prognosis. Given the potential for targeted therapies for tumors harboring CDKN2A deletions, biopsy at recurrence may be indicated in certain patients, especially those with rapid progression.
- Published
- 2020
13. A phase I/II study of ribociclib following radiation therapy in children with newly diagnosed diffuse intrinsic pontine glioma (DIPG)
- Author
-
Blaise V. Jones, Luke Pater, Adam Lane, Trent R. Hummel, Shiva Senthil Kumar, Adam C. Carle, Ralph Salloum, James L. Leach, Phil Dexheimer, Christine Fuller, Charles B. Stevenson, David P. Witte, Maryam Fouladi, Mariko DeWire, Rachid Drissi, Sarah A. Lawson, Lionel M L Chow, Peter de Blank, John C. Breneman, and Xiaoting Zhu
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Neurology ,business.industry ,medicine.medical_treatment ,Ribociclib ,Newly diagnosed ,Neutropenia ,medicine.disease ,Radiation therapy ,03 medical and health sciences ,0302 clinical medicine ,Tolerability ,030220 oncology & carcinogenesis ,Internal medicine ,Glioma ,Toxicity ,medicine ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - Abstract
Cyclin-dependent kinase-retinoblastoma (CDK-RB) pathway is dysregulated in some diffuse intrinsic pontine gliomas (DIPG). We evaluated safety, feasibility, and early efficacy of the CDK4/6-inhibitor ribociclib, administered following radiotherapy in newly-diagnosed DIPG patients. Following radiotherapy, eligible patients received ribociclib in 28-day cycles (350 mg/m2; 21 days on/7 days off). Feasibility endpoints included tolerability for at least 6 courses, and a less than 2-week delay in restarting therapy after 1 dose reduction. Early efficacy was measured by 1-year and median overall survival (OS). Patient/parent-by-proxy reported outcomes measurement information system (PROMIS) assessments were completed prospectively. The study included 10 evaluable patients, 9 DIPG and 1 diffuse midline glioma (DMG)—all 3.7 to 19.8 years of age. The median number of courses was 8 (range 3–14). Three patients required dose reduction for grade-4 neutropenia, and 1 discontinued therapy for hematological toxicity following course 4. The most common grade-3/4 toxicity was myelosuppression. After 2 courses, MRI evaluations in 4 patients revealed increased necrotic volume, associated with new neurological symptoms in 3 patients. The 1-year and median OS for DIPG was 89% and 16.1 months (range 10–30), respectively; the DMG patient died at 6 months post-diagnosis. Five patients donated brain tissue and tumor; 3 were RB+ . Ribociclib administered following radiotherapy is feasible in DIPG and DMG. Increased tumor necrosis may represent a treatment effect. These data warrant further prospective volumetric analyses of tumors with necrosis. Feasibility and stabilization findings support further investigation of ribociclib in combination therapies. NCT02607124.
- Published
- 2020
14. Trametinib-associated Hyponatremia in a Child With Low-grade Glioma is Not Seen Following Treatment With Alternative MEK Inhibitor
- Author
-
Peter de Blank, Maryam Fouladi, Trent R. Hummel, Ralph Salloum, Sarah A. Lawson, Natasha Pillay Smiley, Margot A. Lazow, and Mariko DeWire-Schottmiller
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Pyridones ,medicine.medical_treatment ,Antineoplastic Agents ,Pyrimidinones ,Targeted therapy ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Glioma ,medicine ,Humans ,Endocrine system ,Protein Kinase Inhibitors ,Trametinib ,business.industry ,MEK inhibitor ,Infant ,Binimetinib ,Hematology ,medicine.disease ,chemistry ,030220 oncology & carcinogenesis ,Pediatrics, Perinatology and Child Health ,Benzimidazoles ,Low-Grade Glioma ,business ,Hyponatremia ,030215 immunology - Abstract
Molecularly targeted therapy with MEK inhibitors has been increasingly incorporated into the treatment of pediatric low-grade gliomas, but this promising therapy is associated with distinctive and specific toxicities. Understanding life-threatening MEK inhibitor toxicities and their management is critical to MEK inhibitor safety, especially among young children. This report describes severe hyponatremia associated with trametinib in an infant with progressive low-grade glioma without underlying endocrine dysfunction, which recurred despite significant dose reduction. Therapy with an alternative MEK inhibitor, binimetinib, provided excellent tumor response without hyponatremia, suggesting that some toxicities may be avoided by changing MEK inhibitor agents within the same class.
- Published
- 2020
15. Phase I study of ribociclib and everolimus in children with newly diagnosed DIPG and high-grade glioma: A CONNECT pediatric neuro-oncology consortium report
- Author
-
Mariko DeWire, Margot Lazow, Olivia Campagne, James Leach, Christine Fuller, Shiva Senthil Kumar, Joseph Stanek, Peter de Blank, Trent R Hummel, Natasha Pillay-Smiley, Ralph Salloum, Charles B Stevenson, Patricia Baxter, David Gass, Stewart Goldman, Sarah E S Leary, Adam Carle, Leonie Mikael, Dorothy Crabtree, Brooklyn Chaney, Adam Lane, Rachid Drissi, Clinton F Stewart, and Maryam Fouladi
- Subjects
General Medicine - Abstract
Background Genomic aberrations in the cell cycle and PI3K/Akt/mTOR pathways have been reported in diffuse intrinsic pontine glioma (DIPG) and high-grade glioma (HGG). Dual inhibition of CDK4/6 and mTOR has biologic rationale and minimal overlapping toxicities. This study determined the recommended phase 2 dose (RP2D) of ribociclib and everolimus following radiotherapy in children with DIPG and HGG. Methods Patients were enrolled according to a Rolling-6 design and received ribociclib and everolimus once daily for 21 and 28 days, respectively. All patients with HGG and biopsied DIPG were screened for retinoblastoma protein presence by immunohistochemistry. Pharmacokinetics were analyzed. Results Nineteen patients enrolled (median age: 8 years [range: 2-18]). Three patients enrolled at each dose level 1 and 2 without dose-limiting toxicities (DLT). Thirteen patients were enrolled at dose level 3, with one patient experiencing a DLT (grade 3 infection). One patient came off therapy before cycle 9 due to cardiac toxicity. The most common grade 3/4 toxicities were neutropenia (33%), leucopenia (17%), and lymphopenia (11%). Steady-state everolimus exposures in combination were 1.9 ± 0.9-fold higher than single-agent administration. Median overall survival for 15 patients with DIPG was 13.9 months; median event-free survival for four patients with HGG was 10.5 months. Two longer survivors had tumor molecular profiling identifying CDKN2A/B deletion and CDK4 overexpression. Conclusion The combination of ribociclib and everolimus following radiotherapy in children with newly diagnosed DIPG and HGG was well tolerated, with a RP2D of ribociclib 170 mg/m2 and everolimus 1.5 mg/m2. Results will inform a molecularly guided phase II study underway to evaluate efficacy.
- Published
- 2022
16. A Phase I and Pharmacokinetic Study of Oral Dabrafenib in Children and Adolescent Patients with Recurrent or Refractory BRAF V600 Mutation–Positive Solid Tumors
- Author
-
Eric Bouffet, Trent R. Hummel, Anne Isabelle Bertozzi, Violet Shen, Noelia Nebot, Ira J. Dunkel, Christine A. Pratilas, Andrew D.J. Pearson, James A. Whitlock, Mark W. Kieran, Kenneth J. Cohen, Steven H. Green, Darren Hargrave, Isabelle Aerts, Pooja Hingorani, Alberto Broniscer, Birgit Geoerger, Mark W. Russo, and Lillian Tseng
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,business.industry ,Dabrafenib ,medicine.disease ,Gastroenterology ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Tolerability ,Refractory ,030220 oncology & carcinogenesis ,Glioma ,Internal medicine ,medicine ,Maculopapular rash ,medicine.symptom ,Adverse effect ,business ,Thyroid cancer ,medicine.drug - Abstract
Purpose: The 2-part, phase I/IIa, open-label study (NCT01677741) sought to determine the safety, tolerability, pharmacokinetics, and preliminary activity of dabrafenib in pediatric patients with advanced BRAF V600–mutated cancers. Patients and Methods: This phase I dose-finding part treated patients ages 1 to Results: Between May 2013 and November 2014, 27 patients [12 male; median age, 9 years (range, 1–17 years)] with BRAF V600–mutant solid tumors recurrent/refractory to treatment (low- or high-grade glioma, Langerhans cell histiocytosis, neuroblastoma, or thyroid cancer) were enrolled. The median treatment duration was 75.6 weeks (range, 5.6–148.7 weeks), with 63% treated for >52 weeks and 52% undergoing treatment at data cutoff date. The most common grade 3/4 adverse events suspected to be related to study drug were maculopapular rash and arthralgia (2 patients each). No dose-limiting toxicities were observed. Pharmacokinetic analyses showed a dose-dependent increase in AUC0–12 and achievement of adult exposure levels at the recommended phase II doses of 5.25 mg/kg/day (age Conclusions: In this first clinical trial in pediatric patients with pretreated BRAF V600–mutant tumors, dabrafenib was well tolerated while achieving target exposure levels; the average treatment duration was >1 year with many patients still on treatment. The phase II component is also closed and will be reported separately.
- Published
- 2019
17. EPCT-06. Phase I study of ribociclib and everolimus post-radiotherapy in children with newly diagnosed diffuse intrinsic pontine glioma (DIPG) and high-grade glioma (HGG): Updated report from the COllaborative Network for NEuro-Oncology Clinical Trials (CONNECT)
- Author
-
Margot A Lazow, Mariko DeWire, Olivia Campagne, James L Leach, Christine Fuller, Shiva Senthil Kumar, Joseph Stanek, Peter de Blank, Trent R Hummel, Natasha Pillay-Smiley, Ralph Salloum, Charles B Stevenson, Patricia Baxter, David Gass, Stewart Goldman, Sarah E S Leary, Adam Carle, Adam Lane, Rachid Drissi, Clinton Stewart, and Maryam Fouladi
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
BACKGROUND: Dual inhibition of CDK4/6 and mTOR in DIPG and pediatric HGG has strong biologic rationale, given prevalent genetic alterations resulting in upregulated cell cycle and PI3K/mTOR pathways in these diseases, as well as non-overlapping agent toxicities. This study sought to evaluate safety/tolerability and determine the recommended phase 2 dose (RP2D) of ribociclib and everolimus among children with newly diagnosed DIPG and HGG post-radiotherapy. METHODS: Patients were enrolled according to a Rolling-6 design and received oral ribociclib and everolimus once daily for 21 and 28 days, respectively, starting 2-4 weeks post-completion of radiotherapy. All HGG and biopsied DIPG patients were screened for RB protein presence by immunohistochemistry. Pharmacokinetics and survival data were analyzed. RESULTS: Nineteen patients enrolled (median age: 8 years [range: 2-18]). Three patients enrolled at each of dose levels 1 and 2 without dose-limiting toxicities (DLTs). Thirteen patients enrolled at dose level 3, with one patient experiencing a DLT (grade 3 infection). One patient came off therapy prior to cycle 9 due to cardiac toxicity. The most common grade 3/4 toxicities were neutropenia (33%), leucopenia (17%), and lymphopenia (11%). Steady-state everolimus exposures in combination were 1.9±0.9-fold higher than single-agent administration. Median overall survival (OS) for 15 patients with DIPG was 13.9 months, with 12-, 24-, and 36-month OS of 53.3%, 38.9%, and 38.9%. Median event-free survival for four patients with HGG was 10.5 months. Among patients with tumor molecular profiling, two longer survivors (OS: 20, >37 months) had evidence of cell cycle upregulation with CDKN2A/B deletion and CDK4 overexpression identified. CONCLUSIONS: The combination of ribociclib and everolimus was well-tolerated post-radiotherapy in children with newly diagnosed DIPG and HGG, with a RP2D of ribociclib 170 mg/m2 days 1-21 and everolimus 1.5 mg/m2 days 1-28. Results will inform a molecularly-guided phase II study currently underway to evaluate efficacy.
- Published
- 2022
18. NIMG-11. VOLUMETRIC ENDPOINTS IN DIFFUSE INTRINSIC PONTINE GLIOMA (DIPG): COMPARISON TO CROSS-SECTIONAL MEASURES AND CORRELATION WITH OUTCOMES
- Author
-
Trent R. Hummel, Martijn Nievelstein, Lindsey Hoffman, Margot Lazow, Katie Black, Maryam Fouladi, Brooklyn Chaney, Lindsay Kilburn, James L. Leach, Jane E. Minturn, Pratiti Bandopadhayay, Mariko DeWire-Schottmiller, Sarah Leary, David S. Ziegler, John Glod, Adam Lane, Peter de Blank, Roger J. Packer, and Robert J. Greiner
- Subjects
Correlation ,Cancer Research ,Nuclear magnetic resonance ,Oncology ,business.industry ,Medicine ,Neurology (clinical) ,26th Annual Meeting & Education Day of the Society for Neuro-Oncology ,business - Abstract
INTRODUCTION Cross-sectional tumor measures are used as endpoints in clinical trials of DIPG, but may not capture meaningful changes in disease burden. Volumetric measures may provide a more accurate assessment of tumor growth. We measured the correlation between cross-sectional and volumetric measures and compared their prognostic impact to better understand response evaluation in DIPG. METHODS Patients from the International DIPG Registry with diagnostic and post-radiation MRIs were included. Utilizing mint LesionTM software, tumors were manually contoured by an experienced pediatric neuro-radiologist to extrapolate cross-sectional product (CP) and volume measures. Correlation between CP and volume was assessed by linear regression. Landmark analyses were performed to determine differences in overall survival (OS) (via log-rank) between patients classified as progressive disease (PD) versus non-PD according to CP and volumetric measurements at one-, three-, and five-months post-radiation. Imaging consistent with pseudoprogression was designated non-PD. Hazard ratios (HR) for survival after these timepoints were calculated by Cox regression. RESULTS A total of 317 MRIs from 46 patients were analyzed. When comparing change from smallest previous tumor size, CP increase of 25% (PD by RAPNO) correlated with volume increase of 28% (R2=0.685). There was no difference in OS between patients classified as PD versus non-PD by CP at one-month, three-months, or five-months post-radiation (p >0.05). However, significant differences in OS were observed between patients classified as PD versus non-PD by volume (28% increase) at one-month (2.7 vs. 12.8 months, p=0.005), three-months (1.9 vs. 10.7 months, p=0.036), and five-months post-radiation (3.7 vs. 9.1 months, p=0.023). PD by volume, but not by CP, was predictive of survival at all timepoints (HR: 5.0, 2.4, 2.4). CONCLUSIONS Volumetric assessments of PD correlated better with survival than CP at all post-radiation timepoints. Tumor volume likely represents a more accurate, prognostically-relevant measure of disease burden that deserves investigation in future DIPG trials.
- Published
- 2021
19. Overcoming barriers to establishing autopsy procurement programs in pediatric patients with central nervous system tumors: a call to develop regional centers
- Author
-
Mariko, DeWire, Craig, Erker, Trent R, Hummel, Lionel M L, Chow, Peter, de Blank, Ralph, Salloum, Natasha, Pillay-Smiley, Lindsey, Hoffman, Elizabeth, Gilger, Maureen, Gallagher, Lori, Driver, Dinah, Meister, Heather, Ward, Rachid, Drissi, Shiva Senthil, Kumar, Satarupa, Sengupta, Bridget, Kikta, Wanda, Meriwether, Susan, Jelinek, Anthony, Asher, Blaise, Jones, James, Leach, Lili, Miles, Christine, Fuller, and Maryam, Fouladi
- Subjects
Adult ,Central Nervous System Neoplasms ,Male ,Young Adult ,Tissue and Organ Procurement ,Adolescent ,Child, Preschool ,Humans ,Infant ,Female ,Autopsy ,Child - Abstract
While autopsy-repository programs with a variety of pediatric central nervous system (CNS) tumor types are a critical resource for preclinical neuro-oncology research, few exist and there is no published guidance on how to develop one. The goal of this prospective Pediatric Brain Tumor Repository (PBTR) study was to develop such a program at Cincinnati Children's Hospital Medical Center (CCHMC) and then publish the quantitative and experiential data as a guide to support the development of similar programs.Protocols and infrastructure were established-to educate oncologists and families, establish eligibility, obtain consent, address pre- and post-autopsy logistics (e.g., patient and tissue transportation), process and authenticate tissue samples, and collect and analyze data.Of the 129 pediatric CNS tumor patients at CCHMC who died between 2013 and 2018, 109 were eligible for our study. Of these, 74% (81 of 109) were approached for PBTR donation, and 68% (55 of 81) consented. In the final year of the study, approach and consent rates were 93% and 85%, respectively. Median time from death to autopsy (postmortem interval, PMI) was 10 h (range, 1.5-30). In the outpatient setting, PMI increased with distance (from the hospice/home where the patient died to CCHMC). In all patients, PMI appeared to be lower, when consent was obtained more than 24 h before death.Procurement of autopsy specimens need not be a barrier in neuro-oncology research. Regional centers, strict timing-of-consent, patient education, and dedicated staff are all needed to minimize PMI and, thereby, increase the value of the procured tissue for an array of basic and translational research applications.
- Published
- 2020
20. A phase I/II study of ribociclib following radiation therapy in children with newly diagnosed diffuse intrinsic pontine glioma (DIPG)
- Author
-
Mariko, DeWire, Christine, Fuller, Trent R, Hummel, Lionel M L, Chow, Ralph, Salloum, Peter, de Blank, Luke, Pater, Sarah, Lawson, Xiaoting, Zhu, Phil, Dexheimer, Adam C, Carle, Shiva Senthil, Kumar, Rachid, Drissi, Charles B, Stevenson, Adam, Lane, John, Breneman, David, Witte, Blaise V, Jones, James L, Leach, and Maryam, Fouladi
- Subjects
Adult ,Male ,Adolescent ,Maximum Tolerated Dose ,Diffuse Intrinsic Pontine Glioma ,Aminopyridines ,Chemoradiotherapy ,Prognosis ,Young Adult ,Purines ,Child, Preschool ,Brain Stem Neoplasms ,Humans ,Female ,Tissue Distribution ,Child ,Follow-Up Studies - Abstract
Cyclin-dependent kinase-retinoblastoma (CDK-RB) pathway is dysregulated in some diffuse intrinsic pontine gliomas (DIPG). We evaluated safety, feasibility, and early efficacy of the CDK4/6-inhibitor ribociclib, administered following radiotherapy in newly-diagnosed DIPG patients.Following radiotherapy, eligible patients received ribociclib in 28-day cycles (350 mg/mThe study included 10 evaluable patients, 9 DIPG and 1 diffuse midline glioma (DMG)-all 3.7 to 19.8 years of age. The median number of courses was 8 (range 3-14). Three patients required dose reduction for grade-4 neutropenia, and 1 discontinued therapy for hematological toxicity following course 4. The most common grade-3/4 toxicity was myelosuppression. After 2 courses, MRI evaluations in 4 patients revealed increased necrotic volume, associated with new neurological symptoms in 3 patients. The 1-year and median OS for DIPG was 89% and 16.1 months (range 10-30), respectively; the DMG patient died at 6 months post-diagnosis. Five patients donated brain tissue and tumor; 3 were RB+ .Ribociclib administered following radiotherapy is feasible in DIPG and DMG. Increased tumor necrosis may represent a treatment effect. These data warrant further prospective volumetric analyses of tumors with necrosis. Feasibility and stabilization findings support further investigation of ribociclib in combination therapies.NCT02607124.
- Published
- 2020
21. PATH-13. CHARACTERIZING TEMPORAL GENOMIC HETEROGENEITY IN PEDIATRIC LOW-GRADE GLIOMAS
- Author
-
Austin Schafer, Heather Taggert, Trent R. Hummel, Christine Fuller, Peter de Blank, Adam Lane, Jonathan L. Finlay, Daniel R. Boue, Sarah Rush, Ralph Salloum, Jared Sipple, Maryam Fouladi, Mary Sutton, Natasha Pillay-Smiley, Charles B. Stevenson, Diana S Osorio, Lindsey Hoffman, Erin Wright, Anthony Asher, Rachid Drissi, Margot A. Lazow, Jaime Reuss, Teresa A. Smolarek, and Mariko DeWire
- Subjects
Cancer Research ,Oncology ,Time to progression ,Molecular Pathology & Classification ,Low-Grade Glioma ,Neurology (clinical) ,TYROSINE KINASE RECEPTOR B ,Computational biology ,Biology ,Genetic profile - Abstract
BACKGROUND Recent discoveries have provided valuable insight into the genomic landscape of pediatric low-grade gliomas (LGGs) at diagnosis, facilitating molecularly targeted treatment. However, little is known about their temporal and therapy-related genomic heterogeneity. An adequate understanding of the evolution of pediatric LGGs’ genomic profiles over time is critically important in guiding decisions about targeted therapeutics and diagnostic biopsy at recurrence. METHODS Fluorescence in situ hybridization, mutation-specific immunohistochemistry, exome analyses, and/or targeted sequencing were performed on paired tumor samples from diagnostic and subsequent surgeries. RESULTS Ninety-four tumor samples from 45 patients (41 with two specimens, four with three specimens) from three institutions underwent testing. Conservation of BRAF fusion, BRAFV600E mutation, and FGFR1 rearrangement status was observed in 100%, 98%, and 96% of paired specimens, respectively. No loss or gain of IDH1 mutations or NTRK2, MYB, or MYBL1 rearrangements were detected over time. Histologic diagnosis remained the same in all tumors, with no acquired H3K27M mutations or malignant transformation. Changes in CDKN2A deletion status occurred in 10 patients (40%), with acquisition of CDKN2A deletion in seven (including three who received chemotherapy [two with temozolomide] and one who received radiation) and loss in three (including one who received targeted therapy and radiation). A trend toward shorter time to progression was observed among patients with acquired CDKN2A deletions compared to patients without acquisition of this alteration (median: 5.5 versus 14.0 months [p=0.078]). CONCLUSIONS Most targetable genetic aberrations in pediatric LGGs, including BRAF alterations, are conserved at recurrence and following chemotherapy or radiation. However, changes in CDKN2A deletion status over time were demonstrated. Acquisition of CDKN2A deletion may define a higher risk subgroup of pediatric LGGs with a poorer prognosis. Given the potential for targeted therapies for tumors harboring CDKN2A deletions, biopsy at recurrence may be indicated in certain patients, especially those with rapid progression.
- Published
- 2020
22. GENE-22. MOLECULAR ALTERATIONS AND USE OF TARGETED THERAPY IN 160 PEDIATRIC PATIENTS WITH CENTRAL NERVOUS SYSTEM (CNS) TUMORS
- Author
-
Peter de Blank, Trent R. Hummel, Christine Fuller, Mariko DeWire, Maryam Fouladi, Rebecca Sisson, Demetri Boswell, Ralph Salloum, and Erin Wright
- Subjects
Cancer Research ,Phosphoinositide 3-kinase ,Neurologic Oncology ,biology ,business.industry ,medicine.medical_treatment ,Central nervous system ,Genetics/Epigentics ,medicine.disease ,Targeted therapy ,medicine.anatomical_structure ,Oncology ,Glioma ,Carcinoma ,Cancer research ,biology.protein ,Medicine ,Choroid plexus ,Neurology (clinical) ,business ,Gene - Abstract
BACKGROUND: Molecular profiling is commonly performed in pediatric CNS tumors, however the clinical utilization of genomic data in pediatric neuro-oncology is not fully characterized. We describe a single institution experience adopting a personalized therapeutic approach based on tumor genetic alterations. METHODS: We retrospectively analyzed genomic data, generated using fluorescent in situ hybridization, single gene, or panel sequencing, of pediatric CNS tumors in patients diagnosed between 2010 and 2018 at Cincinnati Children’s Hospital Medical Center. We aimed to identify common genetic aberrations, characterize targeted therapies used, and disease response. RESULTS: One hundred and sixty patients (median age: 7 years, range: 0–27 years) had tumor sequencing performed yielding 147 evaluable results. Low-grade lesions (LGL) (N=94, 63.9%), were mostly low-grade gliomas and glioneuronal tumors; high-grade lesions (HGL) (N=53, 36.0%) included high-grade gliomas (N=35), embryonal neoplasms (N=12), and choroid plexus carcinomas (N=3). Specific BRAF aberrations were more interrogated in LGL while larger panel sequencing was performed in HGL. Targetable aberrations were found in 99 patients (67.3%), most commonly BRAF alterations in LGL as expected, and aberrations in the RTK-PI3K pathway in HGL. Among 99 patients, 56.5% received therapy informed by sequencing data. Thirty three patients with LGL (35.1%) received targeted therapy after a median of 3 prior therapies and remained on treatment for a median of 7 months (range: 1–24 months). Twenty three patients with HGL (43.4%) received targeted therapy after a median of 2 therapies and remained on treatment for a median of 3 months (range: 1–23 months). Targeted therapy was most frequently stopped due to disease progression, only 5 patients developed toxicity that led to cessation of targeted agents. CONCLUSION: Targetable alterations are frequent in pediatric CNS tumors; when identified they guided therapy in a majority of patients commonly at progression and were used earlier and more often in HGL. GERM CELL TUMORS
- Published
- 2019
23. LGG-31. CHARACTERIZING TEMPORAL GENOMIC HETEROGENEITY IN PEDIATRIC LOW GRADE GLIOMAS: ANALYSIS OF AN EXPANDED MULTI-INSTITUTIONAL COHORT WITH 101 PAIRED TUMOR SAMPLES
- Author
-
Anthony Asher, Austin Schafer, Daniel R. Boue, Natasha Pillay Smiley, Diana S Osorio, Margot A Lazow, Trent R Hummel, Lindsey Hoffman, Peter de Blank, Maryam Fouladi, Rachid Drissi, Adam Lane, Erin Wright, Ralph Salloum, Mariko D DeWire-Schottmiller, Christine Fuller, Charles B Stevenson, Jamie Reuss, Jonathan L. Finlay, Sarah Rush, and Mary E Sutton
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Internal medicine ,Cohort ,medicine ,Low Grade Glioma ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,business - Abstract
INTRODUCTION Recent discoveries have provided valuable insight into the genomic landscape of pediatric low grade gliomas (LGGs) at diagnosis, facilitating molecularly targeted treatment. However, little is known about their temporal and therapy-related genomic heterogeneity. An adequate understanding of the evolution of pediatric LGGs’ genomic profiles over time is critically important in guiding decisions about targeted therapeutics and diagnostic biopsy at recurrence. METHODS Fluorescence in situ hybridization, mutation-specific immunohistochemistry, and exome analyses were performed on paired tumor samples from primary diagnostic and subsequent surgeries. RESULTS 101 tumor samples from 48 patients (43 with 2 specimens, 5 with 3 specimens) from 3 institutions underwent testing. BRAF fusion and BRAFV600E status were conserved in 100% and 97% of paired specimens, respectively. No loss or gain of IDH1 mutations or FGFR1, NTRK2, MYB, or MYBL1 rearrangements were detected over time. Histologic diagnosis remained the same in all tumors, with no acquired H3K27M mutations or malignant transformation. CDKN2A deletions were acquired in 7 patients (including 3 who received chemotherapy [2 with temozolomide] and 1 who received radiation), and were associated with a trend toward shorter time to progression (median: 5.5 vs. 13.0 months [p=0.08]). CONCLUSIONS Most targetable genetic alterations in pediatric LGGs, including BRAF alterations, are conserved at recurrence and following chemotherapy or radiation. However, CDKN2A deletion acquisition was demonstrated and may define a higher risk group. Given potential for targeted therapies for tumors acquiring CDKN2A deletions, performing a biopsy at recurrence may be indicated in certain patients, especially those with rapid progression.
- Published
- 2020
24. LGG-30. TRAMETINIB-ASSOCIATED HYPONATREMIA IN A CHILD WITH LOW GRADE GLIOMA IS NOT SEEN FOLLOWING TREATMENT WITH ALTERNATIVE MEK INHIBITOR
- Author
-
Peter de Blank, Ralph Salloum, Margot A. Lazow, Maryam Fouladi, Natasha Pillay Smiley, Sarah A. Lawson, Trent R. Hummel, and Mariko DeWire-Schottmiller
- Subjects
Oncology ,Trametinib ,Cancer Research ,medicine.medical_specialty ,business.industry ,MEK inhibitor ,Low Grade Glioma ,medicine.disease ,Internal medicine ,medicine ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Low-Grade Glioma ,Neurology (clinical) ,Hyponatremia ,business - Abstract
Molecularly targeted therapy with MEK inhibitors is increasingly being incorporated into the treatment of pediatric low-grade gliomas (LGGs). Trametinib is an orally available MEK1/2 inhibitor that has demonstrated tumor control in LGGs with BRAF alterations. Safe expansion of MEK inhibitor therapy within the pediatric patient population demands adequate understanding of and surveillance for potential MEK-inhibitor specific toxicities, especially among young children. Hyponatremia has been reported in adult patients receiving BRAF/MEK inhibitor combination treatment as well as in two pediatric patients with known diabetes insipidus treated with trametinib monotherapy. To our knowledge, single-agent trametinib has not previously been reported to be associated with hyponatremia in children in the absence of an underlying endocrinopathy. We present a case of hyponatremia associated with trametinib use in an infant with progressive LGG without known endocrine dysfunction, which recurred after significant dose reduction. Therapy with an alternative MEK1/2 inhibitor, binimetinib, provided excellent tumor response without hyponatremia. Hyponatremia is a rare but serious side effect of trametinib, even without underlying pituitary dysfunction. Infants and patients lacking the ability to quickly regulate fluid intake in response to osmolality changes are at particular risk of suffering severe consequences from hyponatremia and should be monitored closely with initiation of trametinib. Switching to a different drug within the same class may offer an alternative to significant dose reduction or discontinuation due to this toxicity.
- Published
- 2020
25. NFB-09. ENROLLMENT AND CLINICAL CHARACTERISTICS OF NEWLY DIAGNOSED, NEUROFIBROMATOSIS TYPE 1 ASSOCIATED OPTIC PATHWAY GLIOMA (NF1-OPG): PRELIMINARY RESULTS FROM AN INTERNATIONAL MULTI-CENTER NATURAL HISTORY STUDY
- Author
-
Grant T. Liu, Raymond G. Areaux, Trent R. Hummel, Duncan Stearns, Aimee Sato, W. Walker Motley, Laura J. Klesse, Steven F Stasheff, Arun Y. Reginald, Tena Rosser, David Van Mater, Adam J. Esbenshade, Mays A. El-Dairi, Emily McCourt, Robert Listernick, Eric Bouffet, Nicole J. Ullrich, Shannon Beres, Maree Flaherty, Miriam Bornhorst, Gary Cutter, Michael Fisher, Jeffrey C. Allen, Jason H. Peragallo, Christopher L. Moertel, Faruk Orge, Gena Heidary, Mark Borchert, Simone L. Ardern-Holmes, Milan P. Ranka, John R. Crawford, Kevin J. Bielamowicz, Henry S. O'Halloran, Nicholas K. Foreman, Robert A. Avery, Kristina Tarczy-Hornoch, Cynthia J. Campen, Paul H. Phillips, David H. Gutmann, Peter de Blank, Nick Hogan, David S. Wolf, Janice Lasky Zeid, Michael C. Brodsky, Sean P. Donahue, and Rosalie E. Ferner
- Subjects
Oncology ,musculoskeletal diseases ,Cancer Research ,medicine.medical_specialty ,business.industry ,Newly diagnosed ,medicine.disease ,Neurofibromatosis ,Internal medicine ,medicine ,AcademicSubjects/MED00300 ,Center (algebra and category theory) ,AcademicSubjects/MED00310 ,Neurology (clinical) ,business ,Optic pathway glioma ,Natural history study - Abstract
INTRODUCTION Because treatment and clinical management decisions for children with NF1-OPG remain challenging, we sought to establish evidence-based guidelines. We prospectively enrolled children with newly-diagnosed NF1-OPGs, and gathered standardized clinical neuro-oncology and ophthalmology assessments. METHODS Only children with NF1 and newly diagnosed OPGs, confirmed by central review, were eligible. Indications for obtaining the initial MRI, as well as factors associated with the decision to treat with chemotherapy or observe without treatment, were obtained. Quantitative visual acuity (VA), other ophthalmic features, and imaging were captured at standard time points. Goal enrollment is 250 subjects. RESULTS One-hundred thirty-three children (52% female) from 20 institutions met inclusion criteria, and were included in this preliminary analysis. Eighty-six percent of subjects were able to perform quantitative VA testing at enrollment. The most common reasons for the diagnostic MRI included screening related to NF1 diagnosis (36.8%), ophthalmologic concerns (29.3%), and non-ophthalmologic concerns (24.8%), such as headache. To date, twenty subjects have initiated treatment with chemotherapy, twelve (9%) at the time of the initial OPG diagnosis. Median age at OPG diagnosis was 3.1 years. Age and sex distribution were similar in subjects immediately entering the observation and treatment arms (median age 3.0 versus 3.5 years, respectively). CONCLUSION Most children with NF1-OPGs are observed at time of their initial OPG diagnosis, rather than treated. Importantly, a large proportion of children are able to complete quantitative VA testing at enrollment. Once enrollment is complete, these data will help to establish evidence-based guidelines for clinical management of NF1-OPGs.
- Published
- 2020
26. EPCT-19. A PHASE I STUDY OF RIBOCICLIB AND EVEROLIMUS FOLLOWING RADIATION THERAPY IN CHILDREN WITH NEWLY DIAGNOSED NON-BIOPSIED DIFFUSE PONTINE GLIOMAS (DIPG) AND RB+ BIOPSIED DIPG AND HIGH GRADE GLIOMAS (HGG)
- Author
-
Natasha Pillay-Smiley, Trent R. Hummel, Christine Fuller, Shiva Senthil Kumar, Clinton F. Stewart, Peter de Blank, Maryam Fouladi, David Gass, Patricia Baxter, Olivia Campagne, James L. Leach, Rachid Drissi, Stewart Goldman, Mariko DeWire, Charles B. Stevenson, Sarah Leary, Adam Lane, and Ralph Salloum
- Subjects
Cancer Research ,medicine.medical_specialty ,Everolimus ,medicine.diagnostic_test ,business.industry ,medicine.medical_treatment ,Phases of clinical research ,Ribociclib ,Newly diagnosed ,Neutropenia ,medicine.disease ,Early Phase Clinical Trials ,Radiation therapy ,Oncology ,Glioma ,Biopsy ,medicine ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,Radiology ,business ,medicine.drug - Abstract
Genomic aberrations in the cell cycle and mTOR pathways have been reported in diffuse pontine gliomas (DIPG) and high-grade gliomas (HGG). Dual inhibition of CDK4/6 (ribociclib) and mTOR (everolimus) has strong biologic rationale, non-overlapping single-agent toxicities, and adult clinical experience. The maximum tolerated dose (MTD) and/or recommended phase two dose (RP2D) of ribociclib and everolimus administered during maintenance therapy following radiotherapy was determined in the phase I study as a rolling 6 design. Ribociclib and everolimus were administered once daily for 21 days and 28 days, respectively starting two-four weeks post completion of radiotherapy. All HGG patients and any DIPG patient who had undergone biopsy were screened for RB protein by immunohistochemistry. Eighteen eligible patients enrolled (median age 8 years; range: 2–18). Six patients enrolled at dose levels 1,2, and 3 without dose limiting toxicities (DLT). Currently, five patients are enrolled at dose level 3 expansion cohort. The median number of cycles are 4.5 (range: 1–20+). Among the expansion cohort, one dose limiting toxicity included a grade 3 infection and one patient required a dose reduction in course 3 due to grade 3 ALT and grade 4 hypokalemia. The most common grade 3/4 adverse events included neutropenia. Preliminary pharmacokinetic studies on 12 patients suggest an impact of ribociclib on everolimus pharmacokinetics. The MTD/RP2D of ribociclib and everolimus following radiotherapy in newly diagnosed DIPG and HGG is anticipated to be 170 mg/m2/day x 21 days and 1.5 mg/ m2/day every 28 days which is equivalent to the adult RP2D.
- Published
- 2020
27. Visual field outcomes in children treated for neurofibromatosis type 1–associated optic pathway gliomas: a multicenter retrospective study
- Author
-
Michael Fisher, Arun Reginald, Trent R. Hummel, Annie Kuo, Robert O. Hoffman, Robert A. Avery, Michael J. Loguidice, Rosalie E. Ferner, Gena Heidary, Christopher J Hammond, Grant T. Liu, Nicole J. Ullrich, David H. Gutmann, Robert Listernick, Kush Kapur, and Simone L. Ardern-Holmes
- Subjects
Optic Nerve Glioma ,Pediatrics ,medicine.medical_specialty ,Neurofibromatosis 1 ,Visual acuity ,medicine.medical_treatment ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,In patient ,Prospective Studies ,Neurofibromatosis ,Child ,Prospective cohort study ,Retrospective Studies ,Chemotherapy ,business.industry ,Retrospective cohort study ,medicine.disease ,Visual field ,Ophthalmology ,Pediatrics, Perinatology and Child Health ,Cohort ,030221 ophthalmology & optometry ,Visual Fields ,medicine.symptom ,business - Abstract
Background Optic pathway gliomas associated with neurofibromatosis type 1 (NF1-OPGs) may adversely affect visual acuity, but data regarding visual field (VF) outcomes after treatment in children are limited. The purpose of this study was to investigate the effects of NF1-OPGs on VF function in a large cohort of children after treatment with chemotherapy. Methods We performed a retrospective, international, multicenter study of VF outcomes in patients treated with chemotherapy for NF1-OPGs. Results A total of 25 participants underwent VF testing using formal perimetric techniques. At the end of treatment, 19 participants (76%) had persistent VF deficits. Formal VF testing was available for 16 participants (64%) at initiation and completion of treatment. Of the 16 children who underwent VF testing at initiation and completion of treatment, 7 (44%) showed stability of VF changes, 3 (19%) showed improvement of VF function, and 6 (38%) had worsening of VFs. Improvement or worsening of VF outcome did not always correlate with visual acuity outcome. Posterior tumor location involving the optic tracts and radiations was associated with more frequent and more profound VF defects. Conclusions In our study cohort, children undergoing initial chemotherapy for NF1-OPGs had a high prevalence of VF loss, which could be independent of visual acuity loss. A larger, prospective study is necessary to fully determine the prevalence of VF loss and the effects of chemotherapy on VF outcomes in children with NF1-OPGs.
- Published
- 2020
28. A molecular biology and phase II study of imetelstat (GRN163L) in children with recurrent or refractory central nervous system malignancies: a pediatric brain tumor consortium study
- Author
-
Stewart Goldman, Kathleen Dorris, Maryam Fouladi, Roger J. Packer, Paul G. Fisher, Vinay M. Daryani, Shaoyu Li, Shiva Senthil Kumar, Tong Lin, James M. Boyett, Tina Young Poussaint, Clinton F. Stewart, Ralph Salloum, Lili Miles, Ian F. Pollack, Charles B. Stevenson, Rachid Drissi, Girish Dhall, and Trent R. Hummel
- Subjects
Male ,Niacinamide ,0301 basic medicine ,Ependymoma ,Cancer Research ,Telomerase ,Indoles ,Time Factors ,Pediatric Brain Tumor Consortium ,Adolescent ,Neutrophils ,Oligonucleotides ,Phases of clinical research ,Antineoplastic Agents ,Neutropenia ,Article ,Central Nervous System Neoplasms ,Young Adult ,03 medical and health sciences ,Imetelstat ,0302 clinical medicine ,Glioma ,Humans ,Medicine ,Lymphocytes ,Child ,business.industry ,Alanine Transaminase ,Recurrent Medulloblastoma ,medicine.disease ,Molecular biology ,Blood Cell Count ,Treatment Outcome ,030104 developmental biology ,Neurology ,Oncology ,Child, Preschool ,030220 oncology & carcinogenesis ,Female ,Neurology (clinical) ,Neoplasm Recurrence, Local ,business - Abstract
Telomerase activation is critical in many cancers including central nervous system (CNS) tumors. Imetelstat is an oligonucleotide that binds to the template region of the RNA component of telomerase, inhibiting its enzymatic activity. We conducted an investigator-sponsored molecular biology (MB) and phase II study to estimate inhibition of tumor telomerase activity and sustained responses by imetelstat in children with recurrent CNS malignancies. In the MB study, patients with recurrent medulloblastoma, high-grade glioma (HGG) or ependymoma undergoing resection received one dose of imetelstat as a 2-h intravenous infusion at 285 mg/m(2), 12-24 h before surgery. Telomerase activity was evaluated in fresh tumor from surgery. Post-surgery and in the phase II study, patients received imetelstat IV (days 1 and 8 q21-days) at 285 mg/m(2). Imetelstat pharmacokinetic and pharmacodynamic studies were performed. Of two evaluable patients on the MB trial, intratumoral telomerase activity was inhibited by 95 % compared to baseline archival tissue in one patient and was inevaluable in one patient. Forty-two patients (40 evaluable for toxicity) were enrolled: 9 medulloblastomas, 18 HGG, 4 ependymomas, 9 diffuse intrinsic pontine gliomas. Most common grade 3/4 toxicities included thrombocytopenia (32.5 %), lymphopenia (17.5 %), neutropenia (12.5 %), ALT (7.5 %) and AST (5 %) elevation. Two patients died of intratumoral hemorrhage secondary to thrombocytopenia leading to premature study closure. No objective responses were observed. Telomerase inhibition was observed in peripheral blood mononuclear cells (PBMCs) for at least 8 days. Imetelstat demonstrated intratumoral and PBMC target inhibition; the regimen proved too toxic in children with recurrent CNS tumors.
- Published
- 2016
29. A Phase I and Pharmacokinetic Study of Oral Dabrafenib in Children and Adolescent Patients with Recurrent or Refractory
- Author
-
Mark W, Kieran, Birgit, Geoerger, Ira J, Dunkel, Alberto, Broniscer, Darren, Hargrave, Pooja, Hingorani, Isabelle, Aerts, Anne-Isabelle, Bertozzi, Kenneth J, Cohen, Trent R, Hummel, Violet, Shen, Eric, Bouffet, Christine A, Pratilas, Andrew D J, Pearson, Lillian, Tseng, Noelia, Nebot, Steven, Green, Mark W, Russo, and James A, Whitlock
- Subjects
Male ,Proto-Oncogene Proteins B-raf ,Adolescent ,Maximum Tolerated Dose ,Imidazoles ,Infant ,Treatment Outcome ,Drug Resistance, Neoplasm ,Child, Preschool ,Neoplasms ,Mutation ,Oximes ,Humans ,Female ,Tissue Distribution ,Patient Safety ,Neoplasm Recurrence, Local ,Child ,Protein Kinase Inhibitors - Abstract
The 2-part, phase I/IIa, open-label study (NCT01677741) sought to determine the safety, tolerability, pharmacokinetics, and preliminary activity of dabrafenib in pediatric patients with advancedThis phase I dose-finding part treated patients ages 1 to18 years withBetween May 2013 and November 2014, 27 patients [12 male; median age, 9 years (range, 1-17 years)] withIn this first clinical trial in pediatric patients with pretreated
- Published
- 2017
30. Increased Prevalence of Developmental Venous Anomalies in Children with Intracranial Neoplasms
- Author
-
Luke L. Linscott, Trent R. Hummel, Blaise V. Jones, James L. Leach, and G. Koberlein
- Subjects
Male ,Pediatrics ,medicine.medical_specialty ,Brain Neoplasms ,business.industry ,Significant difference ,Infant ,Magnetic Resonance Imaging ,Veins ,Radiology report ,Developmental venous anomaly ,Child, Preschool ,Prevalence ,medicine ,Humans ,Population study ,Female ,Radiology, Nuclear Medicine and imaging ,Neurology (clinical) ,Child ,business ,Pediatric population - Abstract
BACKGROUND AND PURPOSE: Developmental venous anomalies are considered variants of venous development that, in and of themselves, are of little clinical import. A possible association between intrinsic brain tumors and developmental venous anomalies has been suggested, but a rigorous investigation has not been performed. The aim of this study was to assess any association between developmental venous anomalies and intrinsic brain neoplasms. MATERIALS AND METHODS: A radiology report text search of terms used to describe developmental venous anomalies was performed on a study population of 580 patients with primary intracranial neoplasms and on a control population of 580 patients without neoplasms from the same time period. All positive results were reviewed to confirm that the report was describing a developmental venous anomaly, and the imaging examination was reviewed to confirm the diagnosis. RESULTS: Fifty-nine of the 580 subjects with brain tumors (10.17%) had a developmental venous anomaly identified by report and confirmed on review of the imaging. Thirty-one of the 580 controls (5.34%) had a developmental venous anomaly identified by report and confirmed on review of the imaging (P = .003). No statistically significant difference was noted in the prevalence of developmental venous anomalies among tumor types. No developmental venous anomaly drained the vascular territory of the tumor, and there was no correlation between the location of the developmental venous anomaly and the location of the neoplasm. CONCLUSIONS: The prevalence of developmental venous anomalies in this pediatric population with intracranial primary neoplasms is significantly greater than in those without neoplasms, suggesting an association that may be related to shared causative factors or susceptibilities to the development of these 2 separate entities.
- Published
- 2015
31. Characterizing temporal genomic heterogeneity in pediatric high-grade gliomas
- Author
-
Diana S Osorio, Ralph Salloum, Jonathan L. Finlay, Xiaodan Yang, Charles B. Stevenson, Claudia L. Kleinman, Nicolas De Jay, Daniel R. Boue, Tenzin Gayden, Jacek Majewski, Rachid Drissi, Mariko DeWire, Leonie G. Mikael, Melissa K. McConechy, Jason Karamchandani, Randal Olshefski, Trent R. Hummel, Christine Fuller, Lionel M.L. Chow, Hamid Nikbakht, Nada Jabado, and Maryam Fouladi
- Subjects
Adult ,Male ,0301 basic medicine ,Adolescent ,medicine.medical_treatment ,PDGFRA ,Biology ,Pediatric high-grade gliomas ,Bioinformatics ,lcsh:RC346-429 ,Epigenesis, Genetic ,Pathology and Forensic Medicine ,Targeted therapy ,Young Adult ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Germline mutation ,Recurrence ,CDKN2A ,medicine ,Humans ,Child ,EP300 ,Exome ,lcsh:Neurology. Diseases of the nervous system ,ATRX ,Retrospective Studies ,Brain Neoplasms ,Research ,Genomics ,Glioma ,DNA Methylation ,3. Good health ,Gene Expression Regulation, Neoplastic ,Histone 3 ,Tumor evolution ,030104 developmental biology ,NF1 ,Child, Preschool ,Mutation ,DNA methylation ,Cancer research ,IDH1 ,Female ,Neurology (clinical) ,Neoplasm Grading ,Neoplasm Recurrence, Local - Abstract
Pediatric high-grade gliomas (pHGGs) are aggressive neoplasms representing approximately 20% of brain tumors in children. Current therapies offer limited disease control, and patients have a poor prognosis. Empiric use of targeted therapy, especially at progression, is increasingly practiced despite a paucity of data regarding temporal and therapy-driven genomic evolution in pHGGs. To study the genetic landscape of pHGGs at recurrence, we performed whole exome and methylation analyses on matched primary and recurrent pHGGs from 16 patients. Tumor mutational profiles identified three distinct subgroups. Group 1 (n = 7) harbored known hotspot mutations in Histone 3 (H3) (K27M or G34V) or IDH1 (H3/IDH1 mutants) and co-occurring TP53 or ACVR1 mutations in tumor pairs across the disease course. Group 2 (n = 7), H3/IDH1 wildtype tumor pairs, harbored novel mutations in chromatin modifiers (ZMYND11, EP300 n = 2), all associated with TP53 alterations, or had BRAF V600E mutations (n = 2) conserved across tumor pairs. Group 3 included 2 tumors with NF1 germline mutations. Pairs from primary and relapsed pHGG samples clustered within the same DNA methylation subgroup. ATRX mutations were clonal and retained in H3G34V and H3/IDH1 wildtype tumors, while different genetic alterations in this gene were observed at diagnosis and recurrence in IDH1 mutant tumors. Mutations in putative drug targets (EGFR, ERBB2, PDGFRA, PI3K) were not always shared between primary and recurrence samples, indicating evolution during progression. Our findings indicate that specific key driver mutations in pHGGs are conserved at recurrence and are prime targets for therapeutic development and clinical trials (e.g. H3 post-translational modifications, IDH1, BRAF V600E). Other actionable mutations are acquired or lost, indicating that re-biopsy at recurrence will provide better guidance for effective targeted therapy of pHGGs. Electronic supplementary material The online version of this article (10.1186/s40478-017-0479-8) contains supplementary material, which is available to authorized users.
- Published
- 2017
32. DIPG-27. CLEE011XUS17T (NCT 02607124): A PHASE I/II STUDY OF RIBOCICLIB (LEE011) FOLLOWING RADIATION THERAPY IN CHILDREN WITH NEWLY DIAGNOSED NON-BIOPSIED DIFFUSE PONTINE GLIOMAS (DIPG) AND RB+ BIOPSIED DIPG AND HIGH GRADE GLIOMAS (HGG)
- Author
-
Mariko DeWire, Maryam Fouladi, Charles B. Stevenson, Luke Pater, Qing Lu, John C. Breneman, Trent R. Hummel, Christine Fuller, James L. Leach, Lionel M.L. Chow, Rachid Drissi, David P. Witte, Adam Lane, and Ralph Salloum
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Nausea ,medicine.medical_treatment ,Phases of clinical research ,Neutropenia ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,Abstracts ,0302 clinical medicine ,Internal medicine ,Glioma ,medicine ,Adverse effect ,Leukopenia ,business.industry ,medicine.disease ,Discontinuation ,Surgery ,Radiation therapy ,Neurology (clinical) ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Genomic sequencing studies have uncovered alterations of checkpoint cell cycle regulators (CDK4/6, CDK2NA) in diffuse intrinsic pontine gliomas (DIPG) and high-grade gliomas (HGG). Ribociclib (LEE011) is an orally bioavailable, small molecule inhibitor of CDK4/CyclinD1 and CDK6/CyclinD3 complexes resulting in hypophosphorylation of RB and cell cycle arrest. In a feasibility, phase II study, patients with newly-diagnosed DIPG or HGG received radiation therapy prior to enrollment. All HGG patients and any DIPG patient who had undergone biopsy were screened for total RB protein by immunohistochemistry. Ribociclib was administered daily for 21 of 28 days at 350 mg/m/day. Feasibilty endpoints included toxic death, Grade 3/4 toxicities possibly related to ribociclib resulting in drug discontinuation, > 2 weeks delay in the start of any course or discontinuation of the protocol therapy even after appropriate dose modification. Efficacy was measured by the overall survival distribution for DIPG and HGG. Of the 10 patients enrolled (median 6.5 years; range:4–20), 9 received treatment (8 DIPG, 1 HGG) and one developed leptomeningeal disease prior to therapy. To date, the median number of courses are 4.5 (range: 1–10). Ribociclib was well-tolerated; three patients required dose reduction for grade 4 neutropenia; no patients have come off therapy for toxicity. The most common adverse events were leukopenia, neutropenia, lymphopenia, nausea, vomiting, and fatigue. MRI evaluations in 2 patients revealed increased T2 FLAIR signal with new neurological symptoms within the first 2 cycles of therapy. These patients were responsive to steroids and remain on study. Six patients remain on active therapy at a median of 9 months post diagnosis (range: 3–12). The preliminary analysis demonstrates that ribociclib, administered post radiation therapy is well-tolerated in children and young adults with DIPG and RB+ HGG. Analyses for efficacy and correlative studies will be presented at the meeting.
- Published
- 2017
33. HGG-06. CHARACTERIZING TEMPORAL GENOMIC HETEROGENEITY IN PEDIATRIC HIGH-GRADE GLIOMAS
- Author
-
Claudia L. Kleinman, De Jay N, Melissa K. McConechy, Nada Jabado, Tenzin Gayden, Mariko DeWire, Leonie G. Mikael, Lionel M.L. Chow, Jason Karamchandani, Jonathan L. Finlay, Christine E. Fuller, Diana S Osorio, Charles B. Stevenson, Ralph Salloum, Jacek Majewski, Nikhbakht H, Trent R Hummel, Maryam Fouladi, Daniel R. Boue, and Rachid Drissi
- Subjects
Cancer Research ,Abstracts ,Text mining ,Oncology ,business.industry ,Neurology (clinical) ,Computational biology ,Biology ,business - Abstract
Pediatric high-grade gliomas (pHGG) are aggressive malignant neoplasms representing approximately 20% of all pediatric central nervous system tumors. Current therapies offer limited disease control, and patients with recurrent disease have a particularly poor prognosis. While the empiric use of targeted therapy, especially at progression, is being increasingly practiced, there is a paucity of data regarding temporal and therapy-driven genomic evolution in pHGGs. To better understand the genetic landscape of pHGGs at recurrence, we performed whole exome, transcriptome and methylation analyses on pHGG matched primary and recurrent tumors from 16 patients, median age of 15 years (range: 4–29). Matched normal tissue, where available, was evaluated to determine mutation somatic status. Genetic mutational results suggested that our cohort could be segregated into 3 groups. Group 1 tumor pairs (n=6) carry driver mutations in histone 3 (H3F3A, HIST1H3B) or IDH1 that were retained between the primary tumor and recurrence. In tumors with H3F3A and IDH1 mutations, their obligate partner TP53 or ATRX mutations were also conserved. Group 2 tumor pairs (n=8) were histone 3 or IDH1 wild-type, but most harbored mutations in genes that are modifiers of histone or chromatin (KMT2D, ZMYND11, EP300, BCOR). TP53 and ATRX driver mutations were present in 50% (n=4) of tumors, and 25% (n=2) had a BRAF V600E mutation. Mutations in other putative drug targets (EGFR, ERBB2, PI3-kinase mutations) were not always shared between primary and recurrent tumors, indicating evolution during tumor progression. Group 3 samples (n=2) carried germline mutations in NF1. Mutational burden, gene ontology analysis and correlation with treatment exposure are ongoing. The finding that key driver mutations, some of which are targetable (e.g. IDH1, BRAF V600E) are conserved at recurrence, while other targetable mutations are acquired, indicates that re-biopsy at recurrence may provide better guidance for effective treatment of pHGG with targeted therapy.
- Published
- 2017
34. Patterns of progression in pediatric patients with high-grade glioma or diffuse intrinsic pontine glioma treated with Bevacizumab-based therapy at diagnosis
- Author
-
Stewart Goldman, Maryam Fouladi, Adam Lane, James L. Leach, Lili Miles, Trent R. Hummel, Ralph Salloum, Mary Sutton, Lionel M.L. Chow, Mariko DeWire, and Charles B. Stevenson
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Neurology ,Adolescent ,Bevacizumab ,Antineoplastic Agents ,Antibodies, Monoclonal, Humanized ,Irinotecan ,Young Adult ,Glioma ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Child ,Retrospective Studies ,medicine.diagnostic_test ,Brain Neoplasms ,business.industry ,Magnetic resonance imaging ,Retrospective cohort study ,Chemoradiotherapy ,medicine.disease ,Surgery ,Oncology ,Child, Preschool ,Disease Progression ,Camptothecin ,Female ,Neurology (clinical) ,Radiology ,Neoplasm Grading ,business ,Progressive disease ,medicine.drug - Abstract
There is a paucity of data regarding patterns of progression in children with high-grade glioma (HGG) or diffuse intrinsic pontine glioma (DIPG) treated with bevacizumab (BVZ) at diagnosis. We performed a retrospective study of 20 children with HGG or DIPG who received BVZ-based therapy at diagnosis on, or according to, a bi-institutional study. Magnetic resonance imaging (MRI) characteristics of first and most recent progressions were reviewed. Comparison was made to a control group of 19 patients who never received BVZ. Imaging definitions of progressive disease (PD) were local: at primary site or within 2 cm, contiguous; diffuse: >2 cm away but contiguous with primary site, ill-defined and infiltrative; distant: new, non-contiguous disease. In the BVZ-treated group, 14 patients had DIPG, six patients had HGG. Median age was 7 years (range: 3-21). Median time to PD and follow-up were 8.8 months (range 4-21) and 11 months (range: 6-25), respectively. Among 14 patients with PD, 8 (57.1 %) had local PD, 6 (42.9 %) had local and diffuse/distant PD, at initial progression. At most recent progression, a median of 10.8 months (range 6-25) from diagnosis, 10 of 14 (71.4 %) had at least diffuse (n = 8), or distant (n = 6) PD. In the comparable control group, 15 patients had PD: 11(73.3 %) local, 4 (26.7 %) local and diffuse/distant PD at first and most recent progressions. Based on these data, we postulate that BVZ may lead to a higher incidence of distant and diffuse disease in newly-diagnosed children with HGG or DIPG who received BVZ-based therapy.
- Published
- 2014
35. A pediatric phase 1 trial of vorinostat and temozolomide in relapsed or refractory primary brain or spinal cord tumors: A children's oncology group phase 1 consortium study
- Author
-
Lars M. Wagner, Maryam Fouladi, Trent R. Hummel, Joel M. Reid, Susan M. Blaney, Brenda J. Weigel, Richard J. Gilbertson, Terzah M. Horton, Ashish M. Ingle, Renee M. McGovern, Matthew M. Ames, and Charlotte H. Ahern
- Subjects
Oncology ,medicine.medical_specialty ,Temozolomide ,business.industry ,Anemia ,Dacarbazine ,Hematology ,Neutropenia ,medicine.disease ,Pediatric cancer ,Refractory ,Pharmacokinetics ,Internal medicine ,Pediatrics, Perinatology and Child Health ,Medicine ,business ,Vorinostat ,medicine.drug - Abstract
Purpose We conducted a pediatric phase I study to estimate the maximum tolerated dose (MTD), dose-limiting toxicities (DLT), and pharmacokinetic properties of vorinostat, a histone deacetylase (HDAC) inhibitor, when given in combination with temozolomide in children with refractory or recurrent CNS malignancies. Patients and Methods Vorinostat, followed by temozolomide approximately 1 hour later, was orally administered, once daily, for 5 consecutive days every 28 days at three dose levels using the rolling six design. Studies of histone accumulation in peripheral blood mononuclear cells were performed on Day 1 at 0, 6, and 24 hours after vorinostat dosing. Vorinostat pharmacokinetics (PK) and serum MGMT promoter status were also assessed. Results Nineteen eligible patients were enrolled and 18 patients were evaluable for toxicity. There were no DLTs observed at dose level 1 or 2. DLTs occurred in four patients at dose level 3: thrombocytopenia (4), neutropenia (3), and leucopenia (1). Non-dose limiting grade 3 or 4 toxicities related to protocol therapy were also hematologic and included neutropenia, lymphopenia, thrombocytopenia, anemia, and leucopenia. Three patients exhibited stable disease and one patient had a partial response. There was no clear relationship between vorinostat dosage and drug exposure over the dose range studied. Accumulation of acetylated H3 histone in PBMC was observed after administration of vorinostat. Conclusion Five-day cycles of vorinostat in combination with temozolomide are well tolerated in children with recurrent CNS malignancies with myelosuppression as the DLT. The recommended phase II combination doses are vorinostat, 300 mg/m2/day and temozolomide, 150 mg/m2/day. Pediatr Blood Cancer 2013;160:1452–1457. © 2013 Wiley Periodicals, Inc.
- Published
- 2013
36. Pharmacotherapeutic Management of Pediatric Gliomas
- Author
-
Maryam Fouladi, David Neal Franz, Trent R. Hummel, and Lionel M.L. Chow
- Subjects
Oncology ,medicine.medical_specialty ,Telomerase ,medicine.medical_treatment ,Angiogenesis Inhibitors ,Pharmacotherapy ,Growth factor receptor ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Adjuvant therapy ,Animals ,Humans ,Pharmacology (medical) ,Molecular Targeted Therapy ,Epidermal growth factor receptor ,PI3K/AKT/mTOR pathway ,Chemotherapy ,Temozolomide ,biology ,Brain Neoplasms ,business.industry ,Glioma ,Pediatrics, Perinatology and Child Health ,Immunology ,biology.protein ,Neoplasm Recurrence, Local ,business ,medicine.drug - Abstract
Primary glial brain tumors account for the majority of primary brain tumors in children. They are classified as low-grade gliomas (LGG) or high-grade gliomas (HGG), based on specific pathologic characteristics of the tumor, resulting in disparate clinical prognoses. Surgery is a mainstay of treatment for HGG, although it is not curative, and adjuvant therapy is required. Temozolomide, an oral imidazotetrazine prodrug, while considered standard of care for adult HGG, has not shown the same degree of benefit in the treatment of pediatric HGG. There are significant biologic differences that exist between adult and pediatric HGG, and targets specifically aimed at the biology in the pediatric population are required. Novel and specific therapies currently being investigated for pediatric HGG include small molecule inhibitors of epidermal growth factor receptor, platelet-derived growth factor receptor, histone deacetylase, the RAS/AKT pathway, telomerase, integrin, insulin-like growth factor receptor, and γ-secretase. Surgery is also the mainstay for LGG. There are defined front-line, multiagent chemotherapy regimens, but there are few proven second-line chemotherapy options for refractory patients. Approaches such as the inhibition of the mammalian target of rapamycin pathway, inhibition of MEK1 and 2, as well as BRAF, are discussed. Further research is required to understand the biology of pediatric gliomas as well as the use of molecularly targeted agents, especially in patients with surgically unresectable tumors.
- Published
- 2012
37. TB-25LIMITED AUTOPSY IN PEDIATRIC BRAIN TUMOR PATIENTS WITH A COMPREHENSIVE MULTI-DISCIPLINARY APPROACH RESULTS IN PARENTAL SATISFACTION AND SCIENTIFIC INNOVATION
- Author
-
Susan Jelinek, Daniel Heinbaugh, Trent R. Hummel, Mandy Bley, Christine Fuller, Lili Miles, Maryam Fouladi, Qing Lu, Rachid Drissi, Lindsey M. Hoffman, Nancy Yanez-Escorza, Lionel M.L. Chow, Blaise V. Jones, Aimee N. Thompson, Biplab Dasgupta, Bridget Kikta, Mariko DeWire, Ralph Salloum, David P. Witte, and James L. Leach
- Subjects
0301 basic medicine ,Cancer Research ,Pediatrics ,medicine.medical_specialty ,Tuberculosis ,Multi disciplinary ,business.industry ,Autopsy ,Brain tumor childhood ,medicine.disease ,03 medical and health sciences ,Abstracts ,030104 developmental biology ,0302 clinical medicine ,Oncology ,medicine ,Pediatric Brain Tumor ,Neurology (clinical) ,business ,Psychiatry ,030217 neurology & neurosurgery - Published
- 2016
38. Clinical Heterogeneity of Desmoplastic Infantile Ganglioglioma
- Author
-
Blaise V. Jones, Lili Miles, Trent R. Hummel, Francesco T. Mangano, and James I. Geller
- Subjects
Male ,Pediatrics ,medicine.medical_specialty ,genetic structures ,Adverse outcomes ,Review Literature as Topic ,Desmoplastic infantile ganglioglioma ,Disease ,Dig ,Clinical heterogeneity ,medicine ,Humans ,Child ,Ganglioglioma ,Retrospective Studies ,Brain Neoplasms ,business.industry ,Clinical course ,Supratentorial Neoplasms ,Retrospective cohort study ,Hematology ,Prognosis ,Combined Modality Therapy ,Magnetic Resonance Imaging ,eye diseases ,Oncology ,Child, Preschool ,Pediatrics, Perinatology and Child Health ,Female ,sense organs ,business - Abstract
Desmoplastic infantile gangliogliomas (DIG) are intracranial tumors described in 1987 as benign lesions of infancy. A literature review and the clinical course of 3 patients reported herein suggest that the initial description should be amended. Nearly 23% of DIG cases occur in children older than 24 months. Approximately 40% of DIG cases require additional medical, radiation, and/or further surgical intervention, and 15% of infants and children develop leptomeningeal spread or die from DIG. Such adverse outcomes, combined with the recognition that DIG represents a heterogeneous disease, underscore the need for an expanded biological and molecular investigation.
- Published
- 2012
39. Malignant peripheral nerve sheath tumor: Transformation in a patient with neurofibromatosis type 2
- Author
-
Nancy Ratner, Joseph G. Pressey, Blaise V. Jones, Laura Agresta, Francesco T. Mangano, Sara Szabo, Richard McMasters, Ralph Salloum, Luke Pater, Trent R. Hummel, and Christine Fuller
- Subjects
Male ,Neurofibromatosis 2 ,Pathology ,medicine.medical_specialty ,Context (language use) ,Malignant peripheral nerve sheath tumor ,Nerve Sheath Neoplasms ,Malignant transformation ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Neurofibroma ,Neurofibromatosis type 2 ,Neurofibromatosis ,Child ,Peripheral Nerve Sheath ,business.industry ,Sarcoma ,Hematology ,medicine.disease ,Cell Transformation, Neoplastic ,Oncology ,030220 oncology & carcinogenesis ,Pediatrics, Perinatology and Child Health ,business ,030215 immunology - Abstract
Malignant peripheral nerve sheath tumor (MPNST) is a rare soft-tissue sarcoma with an unfavorable prognosis and limited therapeutic options. MPNSTs can be sporadic, but are often associated with neurofibromatosis (NF) 1 and usually arise from preexisting neurofibromas. MPNSTs in patients with NF2 have been reported in only exceedingly rare cases, and the mechanisms underlying transformation into an MPNST have not been fully elucidated. Here, we describe the clinicopathological and genomic features of a peripheral nerve sheath tumor (PNST), with a primary diagnosis of a neurofibroma, as it transforms into a high-grade MPNST in the context of NF2.
- Published
- 2018
40. LGG-24. TEMPORAL GENOMIC HETEROGENEITY IN PEDIATRIC LOW-GRADE GLIOMAS
- Author
-
Austin Schafer, Lindsey Hoffman, Lionel M.L. Chow, Erin Wright, Sarah Rush, Nancy Yanez-Escorza, Rachid Drissi, Daniel R. Boue, Peter de Blank, Jamie Reuss, Jonathan L. Finlay, Trent R. Hummel, Mariko DeWire, Christine Fuller, Charles B. Stevenson, Maryam Fouladi, Diana S Osorio, and Ralph Salloum
- Subjects
Pleomorphic xanthoastrocytoma ,Pilomyxoid astrocytoma ,Cancer Research ,business.industry ,medicine.medical_treatment ,medicine.disease ,Chemotherapy regimen ,Genome ,Radiation therapy ,Abstracts ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Mutation (genetic algorithm) ,Cancer research ,Medicine ,Immunohistochemistry ,Neurology (clinical) ,business ,neoplasms ,Exome ,030217 neurology & neurosurgery - Abstract
INTRODUCTION: Pediatric low-grade gliomas (pLGG) represent the most common brain tumors in children. Though malignant transformation is rare, these tumors pose a formidable therapeutic challenge due to anatomy-limiting surgical options and local recurrence. The temporal and therapy-driven genomic evolution in pLGG is largely unknown. METHODS: To characterize the temporal genomic heterogeneity of pLGG, we performed fluorescence in situ hybridization, mutation-specific immunohistochemistry, and exome analyses on paired tumor samples from primary diagnostic and subsequent surgeries of 27 pLGG patients. RESULTS: 59 tumor samples were available for review and included pilocytic / pilomyxoid astrocytoma (PA) (67%), low grade astrocytoma (19%), ganglioglioma (11%), and pleomorphic xanthoastrocytoma (3%). BRAF duplication/rearrangement was detected in 56% of tested samples, limited to PA in all but one patient (ganglioglioma). 58% of samples harbored CDKN2A deletion, which was demonstrable across various histologies. BRAF duplication/rearrangement was found consistently conserved across primary and subsequent surgical samples, whereas CDKN2A status changed in 60% of tested temporal pairs, with deletion acquisition the most frequent finding (40%) at second surgery. Likewise, BRAF V600E mutation in one PA was lost at second surgery. Although CDKN2A status changes were seen following surgery alone, the addition of chemo-and/or radiation therapy was associated with a higher rate of temporal CDKN2A deletion acquisition (50%, versus 33% with surgery alone). CONCLUSION: Temporal genomic heterogeneity may be encountered in a significant proportion of pLGG, with some alterations potentially facilitated by therapy. Recurrence biopsy may provide improved guidance, particularly if targeted therapeutics are being considered.
- Published
- 2018
41. Efficacy and safety results from a phase I/IIa study of dabrafenib in pediatric patients with BRAF V600–mutant relapsed refractory low-grade glioma
- Author
-
Alberto Broniscer, Birgit Geoerger, Kenneth J. Cohen, Lillian Tseng, Lucas Moreno, Nicolas André, Trent R. Hummel, Ira J. Dunkel, Eric Bouffet, Mark W. Kieran, Noelia Nebot, Jordan R. Hansford, James A. Whitlock, Darren Hargrave, Anne-Isabelle Bertozzi-Salamon, Mark W. Russo, Pooja Hingorani, Sarah Leary, Isabelle Aerts, and Kohinoor Dasgupta
- Subjects
Oncology ,Surgical resection ,Cancer Research ,medicine.medical_specialty ,macromolecular substances ,03 medical and health sciences ,0302 clinical medicine ,Glioma ,Internal medicine ,otorhinolaryngologic diseases ,Medicine ,Curative intent ,business.industry ,Dabrafenib ,medicine.disease ,carbohydrates (lipids) ,stomatognathic diseases ,030220 oncology & carcinogenesis ,Relapsed refractory ,bacteria ,Low-Grade Glioma ,Primary treatment ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
10506Background: The primary treatment for many pediatric patients (pts) with low-grade glioma (LGG) remains surgical resection with curative intent. However, pts whose tumors cannot be completely ...
- Published
- 2018
42. Nocardia farcinica Meningitis Masquerading as Central Nervous System Metastasis in a Child With Cerebellar Pilocytic Astrocytoma
- Author
-
Trent R. Hummel, Maryam Fouladi, Rebecca C. Brady, Blaise V. Jones, Charles B. Stevenson, Jennifer Davis, and Andrew J. Kreppel
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Central nervous system ,Nocardia Infections ,Disease ,Astrocytoma ,Nocardia ,Metastasis ,Diagnosis, Differential ,Meningeal Neoplasms ,Medicine ,Humans ,Meningitis ,Cerebellar Neoplasms ,Nocardia farcinica ,Cerebellar Pilocytic Astrocytoma ,biology ,business.industry ,Brain Neoplasms ,Juvenile Pilocytic Astrocytoma ,Hematology ,medicine.disease ,biology.organism_classification ,Prognosis ,medicine.anatomical_structure ,Oncology ,Infectious disease (medical specialty) ,Child, Preschool ,Pediatrics, Perinatology and Child Health ,business - Abstract
Juvenile pilocytic astrocytoma, the most common pediatric central nervous system (CNS) neoplasm, characteristically displays an indolent growth pattern and rarely demonstrates metastatic dissemination. Reports of infections mimicking CNS metastatic disease are also rare and can impact treatment. We report the youngest known case of a child with a CNS Nocardia farcinica infection who had a known cerebellar pilocytic astrocytoma, review other infections that may masquerade as CNS neoplasms, and discuss N. farcinica CNS infections.
- Published
- 2015
43. A pilot study of bevacizumab-based therapy in patients with newly diagnosed high-grade gliomas and diffuse intrinsic pontine gliomas
- Author
-
James L. Leach, David W. Pruitt, Mary Sutton, Lori Backus, Charles B. Stevenson, Ahna L. H. Pai, Stewart Goldman, Adam Lane, Maryam Fouladi, Trent R. Hummel, Renee Doughman, Rachid Drissi, Ralph Salloum, Mariko DeWire, Matthew Sobo, Shiva Senthil Kumar, Lionel M.L. Chow, Lili Miles, and Laurie Grimme
- Subjects
Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Bevacizumab ,Adolescent ,Pilot Projects ,Neutropenia ,Irinotecan ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Maintenance therapy ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Temozolomide ,Brain Stem Neoplasms ,Humans ,Progression-free survival ,Prospective cohort study ,Child ,business.industry ,Chemoradiotherapy ,Glioma ,medicine.disease ,Prognosis ,Surgery ,Dacarbazine ,Survival Rate ,Regimen ,Neurology ,030220 oncology & carcinogenesis ,Child, Preschool ,Camptothecin ,Female ,Neurology (clinical) ,Neoplasm Grading ,business ,030217 neurology & neurosurgery ,medicine.drug ,Follow-Up Studies - Abstract
Although bevacizumab has not proven effective in adults with newly diagnosed high-grade gliomas (HGG), feasibility in newly diagnosed children with diffuse intrinsic pontine gliomas (DIPG) or HGG has not been reported in a prospective study. In a safety and feasibility study, children and young adults with newly diagnosed HGG received radiotherapy (RT) with bevacizumab (10 mg/kg: days 22, 36) and temozolomide (75-90 mg/m(2)/day for 42 days) followed by bevacizumab (10 mg/kg, days 1, 15), irinotecan (125 mg/m(2), days 1, 15) and temozolomide (150 mg/m(2)/day days 1-5). DIPG patients did not receive temozolomide. Telomerase activity, quality of life (QOL), and functional outcomes were assessed. Among 27 eligible patients (15 DIPG, 12 HGG), median age 10 years (range 3-29 years), 6 discontinued therapy for toxicity: 2 during RT (grade 4 thrombocytopenia, grade 3 hepatotoxicity) and 4 during maintenance therapy (grade 3: thrombosis, hypertension, skin ulceration, and wound dehiscence). Commonest ≥grade 3 toxicities included lymphopenia, neutropenia and leukopenia. Grade 3 hypertension occurred in 2 patients. No intracranial hemorrhages occurred. For DIPG patients, median overall survival (OS) was 10.4 months. For HGG patients, 3-year progression free survival and OS were 33 % (SE ± 14 %) and 50 % (SE ± 14 %), respectively. All 3 tested tumor samples, demonstrated histone H3.3K27M (n = 2 DIPG) or G34R (n = 1 HGG) mutations. QOL scores improved over the course of therapy. A bevacizumab-based regimen is feasible and tolerable in newly diagnosed children and young adults with HGG and DIPG.
- Published
- 2015
44. TR-11A MOLECULAR BIOLOGY AND PHASE II STUDY OF IMETELSTAT (GRN163L) IN CHILDREN WITH RECURRENT OR REFRACTORY CENTRAL NERVOUS SYSTEM (CNS) MALIGNANCIES: A PEDIATRIC BRAIN TUMOR CONSORTIUM STUDY
- Author
-
Shaoyu Li, Roger J. Packer, Kathleen Dorris, Tina Toung-Poussaint, James M. Boyett, Arzu Onar-Thomas, Charles B. Stevenson, Trent R. Hummel, Paul G. Fisher, Stewart Goldman, Shiva-Senthil Kumar, Maryam Fouladi, Lili Miles, Rachid Drissi, Girish Dhall, Ralph Salloum, and Tong Lin
- Subjects
Ependymoma ,Medulloblastoma ,Cancer Research ,Telomerase ,business.industry ,Phases of clinical research ,Neutropenia ,Recurrent Medulloblastoma ,medicine.disease ,Molecular biology ,Imetelstat ,Oncology ,Glioma ,medicine ,Neurology (clinical) ,business ,Abstracts from the 3rd Biennial Conference on Pediatric Neuro-Oncology Basic and Translational Research - Abstract
INTRODUCTION: Telomerase activation is critical in many cancers including CNS tumors making telomerase inhibition a potential therapeutic target. Imetelstat is an oligonucleotide that binds to the template region of the RNA component of human telomerase and inhibits its enzymatic activity. METHODS: We conducted a molecular biology (MB) and phase II study to estimate inhibition of tumor telomerase activity and sustained responses by imetelstat in children with recurrent CNS malignancies. In the MB study, patients with recurrent medulloblastoma, high-grade glioma (HGG) or ependymoma undergoing resection received one dose of imetelstat as a 2-hour intravenous infusion at 285 mg/m2, 12-24 hours before surgery. Telomerase activity was evaluated by TRAP assay in fresh tumor from surgery. Post-surgery and in the phase II study, patients received imetelstat IV (days 1 and 8 q 21-days) at 285mg/m2. Imetelstat pharmacokinetic and pharmacodynamics studies were performed during cycle 1. RESULTS: Of 2 evaluable patients on the MB trial, intratumoral telomerase activity was inhibited by 95% compared to baseline archival tissue in one patient (HGG) and was inevaluable in one patient (medulloblastoma) due to tumor hemorrhage. Forty-two patients (40 evaluable for toxicity) were enrolled: 9 medulloblastomas, 18 HGG, 4 ependymomas, 9 diffuse intrinsic pontine gliomas. The most common grade 3/4 toxicities included thrombocytopenia (32.5%), lymphopenia (17.5%), neutropenia (12.5%), ALT (7.5%) and AST (5%) elevation. Two patients died of intratumoral hemorrhage secondary to thrombocytopenia leading to premature study closure. The median number of courses in the phase II trial was 1 (range 1-3). No objective responses were observed. Telomerase inhibition was observed in peripheral blood mononuclear cells (PBMCs) for at least 8 days. CONCLUSION: Imetelstat demonstrated intratumoral and PBMC target inhibition; however, the regimen proved too toxic in children with recurrent CNS tumors.
- Published
- 2015
45. The Use of Magnetic Resonance Imaging Screening for Optic Pathway Gliomas in Children with Neurofibromatosis Type 1
- Author
-
Anne Lovell, Howard M. Saal, Robert B. Hufnagel, Trent R. Hummel, Robert J. Hopkin, Carlos E. Prada, and Elizabeth K. Schorry
- Subjects
musculoskeletal diseases ,Male ,Optic Nerve Glioma ,medicine.medical_specialty ,Neurofibromatosis 1 ,genetic structures ,Vision Disorders ,Comorbidity ,Single Center ,Imaging data ,medicine ,Humans ,Neurofibromatosis ,Child ,Retrospective Studies ,medicine.diagnostic_test ,business.industry ,Optic Nerve Neoplasms ,Brain ,Infant ,Retrospective cohort study ,Magnetic resonance imaging ,medicine.disease ,Optic Nerve Neoplasm ,Magnetic Resonance Imaging ,eye diseases ,Surgery ,Child, Preschool ,Pediatrics, Perinatology and Child Health ,Disease Progression ,Female ,Radiology ,Optic nerve glioma ,business - Abstract
To evaluate the utility of screening brain/orbital magnetic resonance imaging (MRI) in a large population of children with neurofibromatosis type 1 (NF1) over a 20-year period.A retrospective analysis of clinical and imaging data from children with NF1 seen at a single center between 1990 and 2010 was performed.During the 20-year study period, 826 individuals with NF1 (402 females, 424 males) ages 1-9 years were screened for optic pathway gliomas (OPGs) using brain/orbital MRI; 18% were identified with OPGs with a median age at detection of 3 years. Fifteen percent of patients with OPGs had radiologic or clinical progression requiring therapy. Children with chiasmatic and postchiasmatic tumors were more likely to require therapy compared with patients with prechiasmatic OPGs (P.0001). Patients with visual deficits at the time of diagnosis were more likely to experience visual decline despite therapy when compared with patients treated based on radiologic progression (P.012).Our findings confirm that chiasmatic and postchiasmatic OPG in children with NF1 have the highest risk for progression and vision loss. Early identification of OPG by screening MRI prior to the development of vision loss may lead to improved visual outcomes. Children with negative brain and orbital MRI screening at age 15 months or later did not develop symptomatic OPGs.
- Published
- 2014
46. IMMU-11. AN ANALYSIS OF IMMUNOPHENOTYPE, INCLUDING PD-L1 AND PD1 EXPRESSION, IN PEDIATRIC CNS MALIGNANCIES: A PEDIATRIC BRAIN TUMOR CONSORTIUM STUDY
- Author
-
Duane Mitchell, Trent R. Hummel, Terri McClanahan, Maryam Fouladi, Jennifer H. Yearley, Lakshman Annamalai, Changling Yang, Eugene Hwang, Sonia Partap, Mehmet Kocak, Ira J. Dunkel, Arzu Onar-Thomas, Wendy M. Blumenschein, Sridharan Gururangan, and Jason Fangusaro
- Subjects
Ependymoma ,Medulloblastoma ,Cancer Research ,Pathology ,medicine.medical_specialty ,Pediatric Brain Tumor Consortium ,biology ,business.industry ,medicine.medical_treatment ,Immunotherapy ,medicine.disease ,Abstracts ,Immunophenotyping ,Oncology ,PD-L1 ,Pediatric CNS ,medicine ,biology.protein ,Immunohistochemistry ,Neurology (clinical) ,business - Abstract
BACKGROUND: Primary pediatric central nervous system (CNS) malignancies often have poor outcomes despite intensive treatment. Clinical immunotherapeutic trials targeting the programmed cell death receptor (PD-1) and its ligand (PD-L1) have been successful in some cancers. One of the most promising biomarkers for response has been the expression of PD-L1, although this has been not universally predictive and its presence in pediatric CNS tumors remains unknown. METHODS: One hundred formalin-fixed, paraffin embedded samples from children with various pediatric CNS malignancies were collected, including twenty-five of each of the following: ependymoma, high-grade glioma (HGG), supratentorial primitive neuroectodermal tumor (sPNET), and medulloblastoma. Patient characteristics and disease outcomes were also obtained. Chromogenic immunohistochemical staining of PD-L1 and PD1 was completed with prevalence of stained cells scored on a 0–5 scale. ‘Positive’ expression was assigned to scores of three or greater. Gene expression was analyzed using an nCounter Nanostring immunologic gene expression codeset. RESULTS: IHC was successful except in one patient with sPNET and two with HGG. PDL1 expression was robust in a subset of tumors, with ‘positive’ staining in 8 (33%) ependymomas, 4 (17%) high-grade gliomas, 2 (8%) sPNETs, and 0 medulloblastomas; PD1 staining was similarly heterogeneous. RNA extraction and expression analysis was successful in all but one sPNET and one HGG. Hierarchical clustering with respect to genes involved in inflammatory pathways identified distinct subgroups within each tumor type; in particular, medulloblastoma was noted to demonstrate activation of inflammatory genes. PD1/PD-L1 expression data did not correlate with histologic staining. Further analysis of the relationship between clinical information and gene expression is ongoing. CONCLUSIONS: This study demonstrates the tumor-specific PDL1/PD1 expression across tumor types and also provides immunologic phenotypic information that may be useful for future rational immunotherapy development in children with CNS malignancy.
- Published
- 2017
47. Pediatric tectal plate gliomas: a review of clinical outcomes, endocrinopathies, and neuropsychological sequelae
- Author
-
Trent R. Hummel, Mariko DeWire, David W. Pruitt, Adam Lane, Susan R. Rose, Lionel M.L. Chow, Mary Sutton, Charles B. Stevenson, Ahna L. H. Pai, Maryam Fouladi, Blaise V. Jones, David Gass, and Sarah Lawson
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Pediatrics ,Neurology ,Adolescent ,Population ,Brain tumor ,Hypopituitarism ,Neuropsychological Tests ,Endocrine System Diseases ,Neurosurgical Procedures ,medicine ,Brain Stem Neoplasms ,Humans ,education ,Child ,Neoplasm Staging ,Retrospective Studies ,education.field_of_study ,Tectum Mesencephali ,business.industry ,Endoscopic third ventriculostomy ,Infant ,Retrospective cohort study ,Glioma ,medicine.disease ,Prognosis ,Hydrocephalus ,Surgery ,Oncology ,Child, Preschool ,Disease Progression ,Female ,Neurology (clinical) ,business ,Progressive disease ,Follow-Up Studies - Abstract
Pediatric tectal plate gliomas are indolent slow-growing gliomas that often present with increased intracranial pressure or incidentally on routine brain imaging. We investigated clinical outcomes, endocrinopathies, and neuropsychological sequelae associated with tectal plate gliomas. Twenty-six patients with tectal plate glioma were identified in a 20-year retrospective review. Clinical outcomes, treatments, endocrine function, neuropsychological testing outcomes and radiographic imaging were reviewed for possible signs correlating with tumor progression. Among 26 patients, 19 presented with signs or symptoms of increased intracranial pressure (73 %) versus an incidental finding in 7 (27 %). Median follow-up was 46 months (range 8-143 months). Six of 26 (23 %) experienced progressive disease after diagnosis. Five of 26 (19 %) required more than one surgical procedure due to failure of initial endoscopic third ventriculostomy. Seven of 26 had history of endocrine dysfunction, of which, five presented with endocrine dysfunction (precocious puberty or short stature), 1 developed menstrual irregularities after surgical intervention and 1 had preexisting pan hypopituitarism. Of 12 patients with available neuropsychological testing, eleven had at least one indicator of executive functioning in the low-average to impaired range. While tectal plate gliomas have been considered indolent tumors that are rarely progressive, 23 % of patients in our cohort experienced disease progression and required further therapy. Neurocognitive deficits may occur, while endocrine deficiency is uncommon. Regular multidisciplinary oncology follow-up, routine monitoring with MRI and formal neurocognitive evaluation are imperative to provide early recognition of disease progression or recurrent hydrocephalus and to improve school functioning in this population.
- Published
- 2014
48. BK virus nephropathy in a pediatric autologous stem-cell transplant recipient
- Author
-
Benjamin L. Laskin, Jens Goebel, Sonata Jodele, Hong J. Yin, L. Nelson Sanchez-Pinto, and Trent R. Hummel
- Subjects
Graft dysfunction ,BK virus nephropathy ,business.industry ,viruses ,Transplant recipient ,virus diseases ,Hematology ,medicine.disease_cause ,medicine.disease ,Virology ,BK virus ,Nephropathy ,Blood cancer ,Transplantation ,surgical procedures, operative ,Oncology ,Pediatrics, Perinatology and Child Health ,Immunology ,Medicine ,Stem cell ,business - Abstract
BK virus (BKV) is an increasingly identified cause of pathology in immunocompromised transplant recipients. BKV is a well-known cause of graft dysfunction following renal transplantation and has also been reported in the native kidneys of other solid organ recipients. Less commonly, BKV nephropathy occurs in allogeneic stem-cell transplant (SCT) recipients. We now describe the first reported case of BKV nephropathy after pediatric autologous SCT. Pediatr Blood Cancer 2011;56:495–497. © 2010 Wiley-Liss, Inc.
- Published
- 2010
49. Response of NF1-related plexiform neurofibroma to High-Dose Carboplatin
- Author
-
Brian Weiss, Trent R. Hummel, Jun Mo, Alexander J. Towbin, and Adjoa Anyane-Yeboa
- Subjects
Pathology ,medicine.medical_specialty ,endocrine system diseases ,medicine.diagnostic_test ,business.industry ,Treatment outcome ,Magnetic resonance imaging ,Hematology ,urologic and male genital diseases ,medicine.disease ,Carboplatin ,chemistry.chemical_compound ,Oncology ,chemistry ,Plexiform neurofibroma ,Testicular seminoma ,Pediatrics, Perinatology and Child Health ,medicine ,Neurofibroma ,Neurofibromatosis ,business ,neoplasms ,Standard therapy - Abstract
Plexiform neurofibromas (PN) are a hallmark of neurofibromatosis type 1 (NF1). These large nerve tumors can be disfiguring and surgery is the only known standard therapy. Surgical intervention may be suboptimal due to the diffuse nature of PN. Here, we present a case in which we describe the use of high-dose carboplatin to treat the patient's testicular seminoma which resulted in the decrease in size of a PN in a patient with NF1.
- Published
- 2010
50. Favorable response to soft tissue sarcoma therapy in an adolescent with embryonal renal sarcoma
- Author
-
Jeffrey D. Hord and Trent R Hummel
- Subjects
Pathology ,medicine.medical_specialty ,Adolescent ,Urinary system ,medicine.medical_treatment ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Neoplasm ,Chemotherapy ,Kidney ,business.industry ,Soft tissue sarcoma ,Soft tissue ,Sarcoma ,Wilms' tumor ,Hematology ,Neoplasms, Germ Cell and Embryonal ,medicine.disease ,Combined Modality Therapy ,Kidney Neoplasms ,medicine.anatomical_structure ,Oncology ,Pediatrics, Perinatology and Child Health ,Female ,Embryonal rhabdomyosarcoma ,business - Abstract
Embryonal renal sarcomas were first identified in 1995 among banked tumor samples originally classified as adult Wilms tumor. Few long-term remissions were observed when these rare tumors were treated with chemotherapy usually used for childhood Wilms. Data were collected from the medical record of an adolescent female with embryonal renal sarcoma and treated with sarcoma-directed chemotherapy and radiation. At 66 months following diagnosis, the patient has no evidence of tumor but has experienced severe renal dysfunction and ovarian failure. We believe there is a subset of patients with disseminated embryonal renal sarcoma that respond to intense sarcoma-directed therapy.
- Published
- 2004
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.