290 results on '"Roger J. Packer"'
Search Results
2. Accuracy of central neuro-imaging review of DIPG compared with histopathology in the International DIPG Registry
- Author
-
Jane E. Minturn, Ayman El-Sheikh, Gustavo Sevlever, Michelle Monje-Deisseroth, Hetal Dholaria, Karen Tsui, Maryam Fouladi, Pratiti Bandopadhayay, Cynthia Hawkins, Scott L Coven, Lindsay Kilburn, Christopher L. Tinkle, David S. Ziegler, Eric Sandler, Yvan Samson, Jordan R. Hansford, Eric Bouffet, Sylvia Cheng, Sridharan Gururangan, Kathleen Dorris, Tim Hassall, Mohamed S. Zaghloul, Carl Koschmann, Sarah Leary, Mercedes Garcia Lombardi, Blaise V. Jones, Paul G. Fisher, Anthony Asher, Rachid Drissi, Blanca Diez, Kenneth J. Cohen, Jie Ma, Adriana Fonseca, Katie Black, Nicholas G. Gottardo, Stewart Goldman, Christine E. Fuller, Tabitha Cooney, Moatasem El-Ayadi, Adam Lane, Brooklyn Chaney, Mariko DeWire, Robert J. Greiner, Ute Bartels, Margot A Lazow, James L. Leach, Lars M. Wagner, and Roger J. Packer
- Subjects
Cancer Research ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Cancer ,Autopsy ,Glioma ,Astrocytoma ,medicine.disease ,Tissue acquisition ,Glutamates ,Oncology ,Neuroimaging ,Biopsy ,medicine ,Medical imaging ,Brain Stem Neoplasms ,Humans ,Histopathology ,Registries ,Neurology (clinical) ,Radiology ,Medical diagnosis ,business ,Pediatric Neuro-Oncology - Abstract
Background Diffuse intrinsic pontine glioma (DIPG) remains a clinico-radiologic diagnosis without routine tissue acquisition. Reliable imaging distinction between DIPG and other pontine tumors with potentially more favorable prognoses and treatment considerations is essential. Methods Cases submitted to the International DIPG registry (IDIPGR) with histopathologic and/or radiologic data were analyzed. Central imaging review was performed on diagnostic brain MRIs (if available) by two neuro-radiologists. Imaging features suggestive of alternative diagnoses included nonpontine origin, Results Among 286 patients with pathology from biopsy and/or autopsy, 23 (8%) had histologic diagnoses inconsistent with DIPG, most commonly nondiffuse low-grade gliomas and embryonal tumors. Among 569 patients with centrally-reviewed diagnostic MRIs, 40 (7%) were classified as non-DIPG, alternative diagnosis suspected. The combined analysis included 151 patients with both histopathology and centrally-reviewed MRI. Of 77 patients with imaging classified as characteristic of DIPG, 76 (99%) had histopathologic diagnoses consistent with DIPG (infiltrating grade II-IV gliomas). Of 57 patients classified as likely DIPG with some unusual imaging features, 55 (96%) had histopathologic diagnoses consistent with DIPG. Of 17 patients with imaging features suggestive of an alternative diagnosis, eight (47%) had histopathologic diagnoses inconsistent with DIPG (remaining patients were excluded due to nonpontine tumor origin). Association between central neuro-imaging review impression and histopathology was significant (p < 0.001), and central neuro-imaging impression was prognostic of overall survival. Conclusions The accuracy and important role of central neuro-imaging review in confirming the diagnosis of DIPG is demonstrated.
- Published
- 2021
- Full Text
- View/download PDF
3. Circulating tumor DNA sequencing provides comprehensive mutation profiling for pediatric central nervous system tumors
- Author
-
Erin R. Bonner, Robin Harrington, Augustine Eze, Miriam Bornhorst, Cassie N. Kline, Heather Gordish-Dressman, Adam Dawood, Biswajit Das, Li Chen, Rini Pauly, P. Mickey Williams, Chris Karlovich, Amanda Peach, D’andra Howell, James Doroshow, Lindsay Kilburn, Roger J. Packer, Sabine Mueller, Javad Nazarian, University of Zurich, and Nazarian, Javad
- Subjects
Cancer Research ,Oncology ,10036 Medical Clinic ,610 Medicine & health ,2730 Oncology ,1306 Cancer Research - Abstract
Molecular profiling of childhood CNS tumors is critical for diagnosis and clinical management, yet tissue access is restricted due to the sensitive tumor location. We developed a targeted deep sequencing platform to detect tumor driver mutations, copy number variations, and heterogeneity in the liquid biome. Here, we present the sensitivity, specificity, and clinical relevance of our minimally invasive platform for tumor mutation profiling in children diagnosed with CNS cancer.
- Published
- 2022
- Full Text
- View/download PDF
4. Characteristics of patients ≥10 years of age with diffuse intrinsic pontine glioma: a report from the International DIPG/DMG Registry
- Author
-
Craig Erker, Adam Lane, Brooklyn Chaney, Sarah Leary, Jane E Minturn, Ute Bartels, Roger J Packer, Kathleen Dorris, Nicholas G Gottardo, Katherine E Warren, Alberto Broniscer, Mark W Kieran, Xiaoting Zhu, Peter White, Phillip J Dexheimer, Katie Black, Anthony Asher, Mariko DeWire, Jordan R Hansford, Sridharan Gururangan, Javad Nazarian, David S Ziegler, Eric Sandler, Allison Bartlett, Stewart Goldman, Chie-Schin Shih, Tim Hassall, Hetal Dholaria, Pratiti Bandopadhayay, Yvan Samson, Michelle Monje, Paul G Fisher, Andrew Dodgshun, Sarah Parkin, Murali Chintagumpala, Karen Tsui, David Gass, Valerie Larouche, Emmett Broxson, Mercedes Garcia Lombardi, Stacie Shiqi Wang, Jie Ma, Cynthia Hawkins, Dima Hamideh, Lars Wagner, Carl Koschmann, Christine Fuller, Rachid Drissi, Blaise V Jones, James Leach, and Maryam Fouladi
- Subjects
Adult ,Cancer Research ,Pediatrics ,medicine.medical_specialty ,IDH1 ,Adolescent ,Population ,Clinical Investigations ,Autopsy ,Astrocytoma ,Young Adult ,Biopsy ,medicine ,Brain Stem Neoplasms ,Humans ,In patient ,Registries ,Young adult ,Child ,education ,ATRX ,education.field_of_study ,medicine.diagnostic_test ,business.industry ,Diffuse Intrinsic Pontine Glioma ,Glioma ,Oncology ,Neurology (clinical) ,business ,Median survival - Abstract
BackgroundDiffuse intrinsic pontine gliomas (DIPG) generally occur in young school-age children, although can occur in adolescents and young adults. The purpose of this study was to describe clinical, radiological, pathologic, and molecular characteristics in patients ≥10 years of age with DIPG enrolled in the International DIPG Registry (IDIPGR).MethodsPatients ≥10 years of age at diagnosis enrolled in the IDIPGR with imaging confirmed DIPG diagnosis were included. The primary outcome was overall survival (OS) categorized as long-term survivors (LTS) (≥24 months) or short-term survivors (STS) (ResultsAmong 1010 patients, 208 (21%) were ≥10 years of age at diagnosis; 152 were eligible with a median age of 12 years (range 10-26.8). Median OS was 13 (2-82) months. The 1-, 3-, and 5-year OS was 59.2%, 5.3%, and 3.3%, respectively. The 18/152 (11.8%) LTS were more likely to be older (P < .01) and present with longer symptom duration (P < .01). Biopsy and/or autopsy were performed in 50 (33%) patients; 77%, 61%, 33%, and 6% of patients tested had H3K27M (H3F3A or HIST1H3B), TP53, ATRX, and ACVR1 mutations/genome alterations, respectively. Two of 18 patients with IDH1 testing were IDH1-mutant and 1 was a LTS. The presence or absence of H3 alterations did not affect survival.ConclusionPatients ≥10 years old with DIPG have a median survival of 13 months. LTS present with longer symptom duration and are likely to be older at presentation compared to STS. ATRX mutation rates were higher in this population than the general DIPG population.
- Published
- 2021
- Full Text
- View/download PDF
5. MR imaging features of diffuse intrinsic pontine glioma and relationship to overall survival: report from the International DIPG Registry
- Author
-
Jane E. Minturn, Ute Bartels, Nicholas G. Gottardo, Jordan R. Hansford, Christine Fuller, Cynthia Hawkins, Renee Doughman, Mariko DeWire-Schottmiller, Sarah Leary, Brooklyn Chaney, Blaise V. Jones, Michelle Monje-Deisseroth, James Roebker, Katherine E. Warren, Hetal Dholaria, Austin Schafer, Rachid Drissi, Adam Lane, Joshua J Baugh, Roger J. Packer, James L. Leach, Paul G. Fisher, Stewart Goldman, Stacie S. Wang, Maryam Fouladi, and David S. Ziegler
- Subjects
Imaging Feature ,Cancer Research ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Diffuse Intrinsic Pontine Glioma ,Magnetic resonance imaging ,Magnetic Resonance Imaging ,Mr imaging ,Text mining ,Oncology ,Basic and Translational Investigations ,medicine ,Overall survival ,Medical imaging ,Brain Stem Neoplasms ,Humans ,Prospective Studies ,Registries ,Neurology (clinical) ,Radiology ,Medical diagnosis ,Prospective cohort study ,business - Abstract
Abtract Background This study describes imaging features of diffuse intrinsic pontine glioma (DIPG) and correlates with overall survival (OS) and histone mutation status in the International DIPG Registry (IDIPGR). Methods Four hundred cases submitted to the IDIPGR with a local diagnosis of DIPG and baseline MRI were evaluated by consensus review of 2 neuroradiologists; 43 cases were excluded (inadequate imaging or alternative diagnoses). Agreement between reviewers, association with histone status, and univariable and multivariable analyses relative to OS were assessed. Results On univariable analysis imaging features significantly associated with worse OS included: extrapontine extension, larger size, enhancement, necrosis, diffusion restriction, and distant disease. On central review, 9.5% of patients were considered not to have DIPG. There was moderate mean agreement of MRI features between reviewers. On multivariable analysis, chemotherapy, age, and distant disease were predictors of OS. There was no difference in OS between wild-type and H3 mutated cases. The only imaging feature associated with histone status was the presence of ill-defined signal infiltrating pontine fibers. Conclusions Baseline imaging features are assessed in the IDIPGR. There was a 9.5% discordance in DIPG diagnosis between local and central review, demonstrating need for central imaging confirmation for prospective trials. Although several imaging features were significantly associated with OS (univariable), only age and distant disease were significant on multivariable analyses. There was limited association of imaging features with histone mutation status, although numbers are small and evaluation exploratory.
- Published
- 2020
- Full Text
- View/download PDF
6. A phase II study of continuous oral mTOR inhibitor everolimus for recurrent, radiographic-progressive neurofibromatosis type 1–associated pediatric low-grade glioma: a Neurofibromatosis Clinical Trials Consortium study
- Author
-
Peter E. Manley, Nathan Robison, Stewart Goldman, Michael Fisher, John P. Perentesis, Alan B. Cantor, Coretta Thomas, Bruce R. Korf, Alyssa Reddy, Mark W. Kieran, Susan N. Chi, Sanjay P. Prabhu, Nicole J. Ullrich, Tomoyuki Mizuno, Jeffrey C. Allen, Alexander A. Vinks, David Viskochil, Gary Cutter, Roger J. Packer, and David H. Gutmann
- Subjects
Oncology ,Cancer Research ,Phases of clinical research ,Clinical endpoint ,Child ,Cancer ,Pediatric ,education.field_of_study ,low-grade glioma ,TOR Serine-Threonine Kinases ,Glioma ,6.1 Pharmaceuticals ,Biotechnology ,medicine.drug ,Pediatric Research Initiative ,medicine.medical_specialty ,Neurofibromatosis 1 ,Neurofibromatoses ,Combination therapy ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Population ,Antineoplastic Agents ,Malignant peripheral nerve sheath tumor ,Neurofibromatosis ,Rare Diseases ,Clinical Research ,Internal medicine ,medicine ,Humans ,Everolimus ,Oncology & Carcinogenesis ,education ,RAD001 ,PI3K/AKT/mTOR pathway ,Sirolimus ,business.industry ,Editorials ,Neurosciences ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,Brain Disorders ,Brain Cancer ,Orphan Drug ,NF1 ,Neurology (clinical) ,PIK3K/mTOR pathway ,business ,Pediatric Neuro-Oncology - Abstract
Background Activation of the mammalian target of rapamycin (mTOR) pathway is observed in neurofibromatosis type 1 (NF1) associated low-grade gliomas (LGGs), but agents that inhibit this pathway, including mTOR inhibitors, have not been studied in this population. We evaluate the efficacy of the orally administered mTOR inhibitor everolimus for radiographically progressive NF1-associated pediatric LGGs. Methods Children with radiologic-progressive, NF1-associated LGG and prior treatment with a carboplatin-containing chemotherapy were prospectively enrolled on this phase II clinical trial to receive daily everolimus. Whole blood was analyzed for everolimus and markers of phosphatidylinositol-3 kinase (PI3K)/mTOR pathway inhibition. Serial MRIs were obtained during treatment. The primary endpoint was progression-free survival at 48 weeks. Results Twenty-three participants (median age, 9.4 y; range, 3.2–21.6 y) were enrolled. All participants were initially evaluable for response; 1 patient was removed from study after development of a malignant peripheral nerve sheath tumor. Fifteen of 22 participants (68%) demonstrated a response, defined as either shrinkage (1 complete response, 2 partial response) or arrest of tumor growth (12 stable disease). Of these, 10/15 remained free of progression (median follow-up, 33 mo). All remaining 22 participants were alive at completion of therapy. Treatment was well tolerated; no patient discontinued therapy due to toxicity. Pharmacokinetic parameters and pre-dose concentrations showed substantial between-subject variability. PI3K/mTOR pathway inhibition markers demonstrating blood mononuclear cell mTOR pathway inactivation was achieved in most participants. Conclusion Individuals with recurrent/progressive NF1-associated LGG demonstrate significant disease stability/shrinkage during treatment with oral everolimus with a well-tolerated toxicity profile. Everolimus is well suited for future consideration as upfront or combination therapy in this patient population.
- Published
- 2020
- Full Text
- View/download PDF
7. The 2021 WHO Classification of Tumors of the Central Nervous System: clinical implications
- Author
-
Roger J. Packer and Patrick Y. Wen
- Subjects
Cancer Research ,medicine.anatomical_structure ,Oncology ,business.industry ,Central nervous system ,Editorials ,medicine ,MEDLINE ,Neurology (clinical) ,Who classification ,Bioinformatics ,business - Published
- 2021
- Full Text
- View/download PDF
8. Two clinically distinct cases of infant hemispheric glioma carrying ZCCHC8:ROS1 fusion and responding to entrectinib
- Author
-
Ludmila Papusha, Margarita Zaytseva, Agnesa Panferova, Alexander Druy, Andge Valiakhmetova, Anton Artemov, Ekaterina Salnikova, Alexey Kislyakov, Evgeny Imyanitov, Alexander Karachunsky, Alexey Maschan, Eugene I Hwang, Galina Novichkova, and Roger J Packer
- Subjects
Cancer Research ,Indazoles ,Lung Neoplasms ,Oncology ,Proto-Oncogene Proteins ,Benzamides ,Humans ,Infant ,Nuclear Proteins ,Neurology (clinical) ,Glioma ,Protein-Tyrosine Kinases ,Carrier Proteins - Published
- 2022
9. Upfront Biology-Guided Therapy in Diffuse Intrinsic Pontine Glioma: Therapeutic, Molecular, and Biomarker Outcomes from PNOC003
- Author
-
Cassie Kline, Payal Jain, Lindsay Kilburn, Erin R. Bonner, Nalin Gupta, John R. Crawford, Anu Banerjee, Roger J. Packer, Javier Villanueva-Meyer, Tracy Luks, Yalan Zhang, Madhuri Kambhampati, Jie Zhang, Sridevi Yadavilli, Bo Zhang, Krutika S. Gaonkar, Jo Lynne Rokita, Adam Kraya, John Kuhn, Winnie Liang, Sara Byron, Michael Berens, Annette Molinaro, Michael Prados, Adam Resnick, Sebastian M. Waszak, Javad Nazarian, Sabine Mueller, University of Zurich, and Mueller, Sabine
- Subjects
Cancer Research ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,610 Medicine & health ,Astrocytoma ,Genomic Instability ,Circulating Tumor DNA ,Young Adult ,Rare Diseases ,Clinical Research ,Genetics ,Brain Stem Neoplasms ,Humans ,1306 Cancer Research ,Oncology & Carcinogenesis ,Child ,Biology ,Cancer ,Pediatric ,screening and diagnosis ,Human Genome ,Diffuse Intrinsic Pontine Glioma ,Neurosciences ,Glioma ,Brain Disorders ,4.1 Discovery and preclinical testing of markers and technologies ,Brain Cancer ,Detection ,Orphan Drug ,Good Health and Well Being ,Oncology ,10036 Medical Clinic ,2730 Oncology ,Female ,Biomarkers - Abstract
Purpose: PNOC003 is a multicenter precision medicine trial for children and young adults with newly diagnosed diffuse intrinsic pontine glioma (DIPG). Patients and Methods: Patients (3–25 years) were enrolled on the basis of imaging consistent with DIPG. Biopsy tissue was collected for whole-exome and mRNA sequencing. After radiotherapy (RT), patients were assigned up to four FDA-approved drugs based on molecular tumor board recommendations. H3K27M-mutant circulating tumor DNA (ctDNA) was longitudinally measured. Tumor tissue and matched primary cell lines were characterized using whole-genome sequencing and DNA methylation profiling. When applicable, results were verified in an independent cohort from the Children's Brain Tumor Network (CBTN). Results: Of 38 patients enrolled, 28 patients (median 6 years, 10 females) were reviewed by the molecular tumor board. Of those, 19 followed treatment recommendations. Median overall survival (OS) was 13.1 months [95% confidence interval (CI), 11.2–18.4] with no difference between patients who followed recommendations and those who did not. H3K27M-mutant ctDNA was detected at baseline in 60% of cases tested and associated with response to RT and survival. Eleven cell lines were established, showing 100% fidelity of key somatic driver gene alterations in the primary tumor. In H3K27-altered DIPGs, TP53 mutations were associated with worse OS (TP53mut 11.1 mo; 95% CI, 8.7–14; TP53wt 13.3 mo; 95% CI, 11.8–NA; P = 3.4e−2), genome instability (P = 3.1e−3), and RT resistance (P = 6.4e−4). The CBTN cohort confirmed an association between TP53 mutation status, genome instability, and clinical outcome. Conclusions: Upfront treatment-naïve biopsy provides insight into clinically relevant molecular alterations and prognostic biomarkers for H3K27-altered DIPGs.
- Published
- 2022
10. 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
11. Implications of new understandings of gliomas in children and adults with NF1: report of a consensus conference
- Author
-
Jaishri O. Blakeley, Cynthia Hawkins, Fausto J. Rodriguez, Stefan M. Pfister, Uri Tabori, Brian R. Rood, Antonio Iavarone, Roger J. Packer, Eric Bouffet, Tobey J. MacDonald, Stephen Albert Johnston, David T.W. Jones, Lindsay Kilburn, David H. Gutmann, Michael Fisher, Eugene Hwang, Yuan Zhu, Vijay Ramaswamy, and Jason Fangusaro
- Subjects
Oncology ,Adult ,Cancer Research ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Neurofibromatosis 1 ,Pilocytic Astrocytomas ,Reviews ,pilocytic astrocytomas ,neurofibromatosis type 1 ,Older patients ,Internal medicine ,Glioma ,medicine ,AcademicSubjects/MED00300 ,Animals ,Humans ,Neurofibromatosis ,Child ,neoplasms ,business.industry ,Brain Neoplasms ,Consensus conference ,molecular-targeted therapy ,medicine.disease ,eye diseases ,nervous system diseases ,gliomas ,Editor's Choice ,Disease Models, Animal ,AcademicSubjects/MED00310 ,Neurology (clinical) ,immunotherapy ,business - Abstract
Gliomas are the most common primary central nervous system tumors occurring in children and adults with neurofibromatosis type 1 (NF1). Over the past decade, discoveries of the molecular basis of low-grade gliomas (LGGs) have led to new approaches for diagnosis and treatments. However, these new understandings have not been fully applied to the management of NF1-associated gliomas. A consensus panel consisting of experts in NF1 and gliomas was convened to review the current molecular knowledge of NF1-associated low-grade “transformed” and high-grade gliomas; insights gained from mouse models of NF1-LGGs; challenges in diagnosing and treating older patients with NF1-associated gliomas; and advances in molecularly targeted treatment and potential immunologic treatment of these tumors. Next steps are recommended to advance the management and outcomes for NF1-associated gliomas.
- Published
- 2020
12. Efficacy of Carboplatin and Isotretinoin in Children With High-risk Medulloblastoma: A Randomized Clinical Trial From the Children’s Oncology Group
- Author
-
Linda Heier, Jeff M. Michalski, Leanne Embry, Yimei Li, Amar Gajjar, Karin S. Walsh, Kyle S. Smith, Alok Jaju, Roger J. Packer, Sarah Leary, James M. Olson, Maryam Fouladi, Jennifer Hadley, Catherine A. Billups, Eugene Hwang, Paul A. Northcott, Peter C. Burger, Ian F. Pollack, Yuanyuan Han, and Rahul Kumar
- Subjects
Oncology ,Cancer Research ,Vincristine ,medicine.medical_specialty ,Randomization ,Subgroup analysis ,law.invention ,chemistry.chemical_compound ,Randomized controlled trial ,law ,Internal medicine ,medicine ,Biomarkers, Tumor ,Humans ,Cerebellar Neoplasms ,Isotretinoin ,Original Investigation ,Medulloblastoma ,business.industry ,Gene Expression Profiling ,medicine.disease ,Carboplatin ,Clinical trial ,chemistry ,business ,medicine.drug - Abstract
Importance Brain tumors are the leading cause of disease-related death in children. Medulloblastoma is the most common malignant embryonal brain tumor, and strategies to increase survival are needed. Objective To evaluate therapy intensification with carboplatin as a radiosensitizer and isotretinoin as a proapoptotic agent in children with high-risk medulloblastoma in a randomized clinical trial and, with a correlative biology study, facilitate planned subgroup analysis according to World Health Organization consensus molecular subgroups of medulloblastoma. Design, Setting, and Participants A randomized clinical phase 3 trial was conducted from March 2007 to September 2018. Analysis was completed in September 2020. Patients aged 3 to 21 years with newly diagnosed high-risk medulloblastoma from Children’s Oncology Group institutions within the US, Canada, Australia, and New Zealand were included. High-risk features included metastasis, residual disease, or diffuse anaplasia. Interventions Patients were randomized to receive 36-Gy craniospinal radiation therapy and weekly vincristine with or without daily carboplatin followed by 6 cycles of maintenance chemotherapy with cisplatin, cyclophosphamide, and vincristine with or without 12 cycles of isotretinoin during and following maintenance. Main Outcomes and Measures The primary clinical trial end point was event-free survival, using the log-rank test to compare arms. The primary biology study end point was molecular subgroup classification by DNA methylation array. Results Of 294 patients with medulloblastoma, 261 were evaluable after central radiologic and pathologic review; median age, 8.6 years (range, 3.3-21.2); 183 (70%) male; 189 (72%) with metastatic disease; 58 (22%) with diffuse anaplasia; and 14 (5%) with greater than 1.5-cm2residual disease. For all participants, the 5-year event-free survival was 62.9% (95% CI, 55.6%-70.2%) and overall survival was 73.4% (95% CI, 66.7%-80.1%). Isotretinoin randomization was closed early owing to futility. Five-year event-free survival was 66.4% (95% CI, 56.4%-76.4%) with carboplatin vs 59.2% (95% CI, 48.8%-69.6%) without carboplatin (P = .11), with the effect exclusively observed in group 3 subgroup patients: 73.2% (95% CI, 56.9%-89.5%) with carboplatin vs 53.7% (95% CI, 35.3%-72.1%) without (P = .047). Five-year overall survival differed by molecular subgroup (P = .006): WNT pathway activated, 100% (95% CI, 100%-100%); SHH pathway activated, 53.6% (95% CI, 33.0%-74.2%); group 3, 73.7% (95% CI, 61.9%-85.5%); and group 4, 76.9% (95% CI, 67.3%-86.5%). Conclusions and Relevance In this randomized clinical trial, therapy intensification with carboplatin improved event-free survival by 19% at 5 years for children with high-risk group 3 medulloblastoma. These findings further support the value of an integrated clinical and molecular risk stratification for medulloblastoma. Trial Registration ClinicalTrials.gov Identifier:NCT00392327
- Published
- 2021
13. Children's Oncology Group Phase III Trial of Reduced-Dose and Reduced-Volume Radiotherapy With Chemotherapy for Newly Diagnosed Average-Risk Medulloblastoma
- Author
-
Catherine A. Billups, Timothy N. Booth, Paul A. Northcott, Stephanie M. Perkins, Jeff M. Michalski, Roger J. Packer, Karin M. Muraszko, Yimei Li, Kristina K. Hardy, Paul Colte, Yuanyuan Han, Rahul Kumar, Kyle S. Smith, Anna J. Janss, L. Gilbert Vezina, Nancy J. Tarbell, Leanne Embry, Maryam Fouladi, Peter C. Burger, Ian F. Pollack, Joel M. Cherlow, Johnnie K. Bass, Amar Gajjar, Patricia Cullen, Thomas J. Fitzgerald, Scott L. Pomeroy, Thomas E. Merchant, and Jennifer Hadley
- Subjects
Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Adolescent ,medicine.medical_treatment ,Newly diagnosed ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Child ,Medulloblastoma ,Average risk ,Chemotherapy ,business.industry ,ORIGINAL REPORTS ,medicine.disease ,Reduced dose ,Radiation therapy ,030220 oncology & carcinogenesis ,Child, Preschool ,Female ,business ,030217 neurology & neurosurgery - Abstract
PURPOSE Children with average-risk medulloblastoma (MB) experience survival rates of ≥ 80% at the expense of adverse consequences of treatment. Efforts to mitigate these effects include deintensification of craniospinal irradiation (CSI) dose and volume. METHODS ACNS0331 (ClinicalTrials.gov identifier: NCT00085735 ) randomly assigned patients age 3-21 years with average-risk MB to receive posterior fossa radiation therapy (PFRT) or involved field radiation therapy (IFRT) following CSI. Young children (3-7 years) were also randomly assigned to receive standard-dose CSI (SDCSI; 23.4 Gy) or low-dose CSI (LDCSI; 18 Gy). Post hoc molecular classification and mutational analysis contextualized outcomes according to known biologic subgroups (Wingless, Sonic Hedgehog, group 3, and group 4) and genetic biomarkers. Neurocognitive changes and ototoxicity were monitored over time. RESULTS Five hundred forty-nine patients were enrolled on study, of which 464 were eligible and evaluable to compare PFRT versus IFRT and 226 for SDCSI versus LDCSI. The five-year event-free survival (EFS) was 82.5% (95% CI, 77.2 to 87.8) and 80.5% (95% CI, 75.2 to 85.8) for the IFRT and PFRT regimens, respectively, and 71.4% (95% CI, 62.8 to 80) and 82.9% (95% CI, 75.6 to 90.2) for the LDCSI and SDCSI regimens, respectively. IFRT was not inferior to PFRT (hazard ratio, 0.97; 94% upper CI, 1.32). LDCSI was inferior to SDCSI (hazard ratio, 1.67%; 80% upper CI, 2.10). Improved EFS was observed in patients with Sonic Hedgehog MB who were randomly assigned to the IFRT arm ( P = .018). Patients with group 4 MB receiving LDCSI exhibited inferior EFS ( P = .047). Children receiving SDCSI exhibited greater late declines in IQ (estimate = 5.87; P = .021). CONCLUSION Reducing the radiation boost volume in average-risk MB is safe and does not compromise survival. Reducing CSI dose in young children with average-risk MB results in inferior outcomes, possibly in a subgroup-dependent manner, but is associated with better neurocognitive outcome. Molecularly informed patient selection warrants further exploration for children with MB to be considered for late-effect sparing approaches.
- Published
- 2021
14. Primary analysis of a phase II trial of dabrafenib plus trametinib (dab + tram) in BRAF V600–mutant pediatric low-grade glioma (pLGG)
- Author
-
Eric Bouffet, Jordan Hansford, Maria Luisa Garré, Junichi Hara, Ashley Plant-Fox, Isabelle Aerts, Franco Locatelli, Jasper Van der Lugt, Ludmila Papusha, Felix Sahm, Uri Tabori, Kenneth J. Cohen, Roger J. Packer, Olaf Witt, Lali Sandalic, Ana Bento Pereira da Silva, Mark W. Russo, and Darren R. Hargrave
- Subjects
Cancer Research ,Oncology - Abstract
LBA2002 Background: LGG is the most common pediatric brain cancer, and BRAF V600 mutation has been detected in ≈17% of cases. Most patients (pts) with pLGG have disease progression and require post-surgical therapy. The standard of care is chemotherapy, such as carboplatin + vincristine (C+V), which may be less effective in BRAF V600–mutant disease; thus, alternative treatment options are needed. Dab + tram showed encouraging efficacy in a Phase I/II study (NCT02124772) in pts with previously treated BRAF V600–mutant pLGG. We describe the results of a randomized Phase II study (NCT02684058) of first-line dab + tram vs C+V in BRAF V600–mutant pLGG. Methods: Pts aged 1 to
- Published
- 2022
- Full Text
- View/download PDF
15. Dabrafenib + trametinib (dab + tram) in relapsed/refractory (r/r) BRAF V600–mutant pediatric high-grade glioma (pHGG): Primary analysis of a phase II trial
- Author
-
Darren R. Hargrave, Keita Terashima, Junichi Hara, Uwe R. Kordes, Santhosh A. Upadhyaya, Felix Sahm, Eric Bouffet, Roger J. Packer, Olaf Witt, Lali Sandalic, Agnieszka Kieloch, Mark W. Russo, and Kenneth J. Cohen
- Subjects
Cancer Research ,Oncology - Abstract
2009 Background: HGGs comprise ≈10% of pediatric central nervous system tumors and are a leading cause of childhood cancer-related death. Overall response rates (ORRs) with current standards of care are low, particularly in the second line, and 2-y overall survival (OS) rates are ≤35%. BRAF V600 mutation is infrequent (≈5% of pHGGs) but associated with improved survival from time of initial diagnosis. In previous trials, dab monotherapy and dab + tram yielded encouraging outcomes in BRAF V600–mutant HGG in pediatric and adult patients (pts), respectively. We describe the results of a Phase II study (NCT02684058) of dab + tram in r/r BRAF V600–mutant pHGG. Methods: Pts aged 1 to
- Published
- 2022
- Full Text
- View/download PDF
16. DIPG-47. TSO500ctDNA sequencing reveals oncogenic mutations and copy number variations in the liquid biome of children with diffuse midline glioma
- Author
-
Erin R Bonner, Robin Harrington, Augustine Eze, Miriam Bornhorst, Cassie N Kline, Adam Dawood, Biswajit Das, Li Chen, Rini Pauly, P Mickey Williams, Chris Karlovich, Amanda Peach, D'Andra Howell, James Doroshow, Lindsay Kilburn, Roger J Packer, Sabine Mueller, and Javad Nazarian
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
BACKGROUND: Molecular profiling of childhood CNS tumors is critical for diagnosis and clinical management, yet tissue access is restricted due to sensitive neuroanatomical locations. Moreover, CNS tumors including diffuse midline glioma (DMG) exhibit mutational heterogeneity and clonal evolution, which cannot be captured by upfront diagnostic biopsy alone. To address the lack of tumor visibility, and tprovide opportunity for longitudinal sampling, we validated and optimized a commercially available deep sequencing platform for analysis of circulating tumor DNA (TSO500ctDNATM). METHODS: In a proof-of-concept study, we defined the sensitivity, specificity, and clinical relevance of our novel ctDNA platform via analysis of paired tissue, CSF, and blood from children with DMG (n=10). Paired samples were assessed for concordance and sequencing results were compared to digital droplet PCR (ddPCR) detection of prognostic H3K27M mutation. RESULTS: DMG associated mutations in genes including H3-3A, H3C2, TP53, and ACVR1 were detected in ctDNA, including in CSF samples with low (
- Published
- 2022
- Full Text
- View/download PDF
17. DIPG-48. MRI volumetric and machine learning based analyses predict survival outcome in pediatric diffuse midline glioma
- Author
-
Erin R Bonner, Xinyang Liu, Carlos Tor-Diez, Madhuri Kambhampati, Augustine Eze, Roger J Packer, Javad Nazarian, Marius George Linguraru, and Miriam Bornhorst
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
INTRODUCTION: Diffuse midline glioma (DMG) is a fatal childhood CNS tumor. Magnetic resonance imaging (MRI) is the gold standard for DMG diagnosis and monitoring of response to therapy. Leveraging novel MRI analytical approaches, including volumetric and machine learning based analyses, may aid in the prediction of patient overall survival (OS) and help to identify high-risk cases. METHODS: T1- and T2-weighted MR images were retrospectively collected from children and young adults diagnosed with DMG (n=43). MRI features, including manually determined 3D tumor volume (T2), T1 contrast-enhancing tumor volume, T1 relative to T2 volume (T1/T2), tumor relative to whole brain volume, tumor average intensity, and tumor heterogeneity (i.e., intensity skewness and kurtosis), were evaluated at upfront diagnosis. MRI features were analyzed to identify significant predictors of OS outcome, which was defined as OS shorter, or longer, than one year from diagnosis. A support vector machine was used to predict OS outcomes using combinations of these features. RESULTS: The presence of T1 contrast-enhancing tumor at diagnosis (p=0.01), and a high T1/T2 ratio (>25%, p=0.009), predicted significantly shorter median OS. Moreover, feature selection identified T2 mean intensity (p
- Published
- 2022
- Full Text
- View/download PDF
18. OTHR-08. Pediatric Neurologic Assessment in Neuro-oncology (pNANO) Scale: A tool to assess neurologic function for Response Assessment in Neuro-oncology (RAPNO)
- Author
-
Fatema Malbari, Craig Erker, Pablo Hernáiz Driever, Natasha Pillay-Smiley, Robert A Avery, Robert Craig Castellino, Antoinette A Y N Schouten-van Meeteren, Alicia C Lenzen, Rick Brandsma, Kim Kramer, Patricia A Baxter, Girish Dhall, Stewart Goldman, Patrick Y Wen, Michael Prados, Roger J Packer, Katherine E Warren, and Lakshmi Nayak
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
Background: The Neurologic Assessment in Neuro-Oncology (NANO) scale, a standardized metric to objectively measure neurologic function in adult brain tumor patients, complements radiographic assessment in evaluating outcomes of neuro-oncology patients in clinical trials and clinical practice. Currently, there is no standardized measure for neurologic function in pediatric neuro-oncology patients despite their distinct clinical presentations and tumor locations. Therefore, we developed a dedicated pediatric NANO (pNANO) scale. Methods: An international group of pediatric neurologists and adult and pediatric neuro-oncologists convened bi-weekly over 5 months to draft the pNANO scale as an objective and quantifiable measure of neurologic function in children that can be administered during routine examination by pediatric-trained providers of any sub-specialty and be utilized together with other indicators to assess response in clinical trials. Results: Ten relevant domains of neurologic function were identified based on common pediatric brain tumor locations: gait, strength, cerebellar function, visual fields, visual acuity, facial strength, level of consciousness, extraocular movements, dysarthria and dysphagia. For each domain, developmental age appropriate levels of function were defined and categorized. Each domain, based on direct observation and testing during any routine neurological exam, can be used for longitudinal monitoring. Conclusions: The pNANO scale has been developed and aims to provide an objective metric of neurologic function for pediatric brain tumor patients. This scale will be tested for reliability, feasibility and inter-observer variability. Consistent evaluation of neurologic function using pNANO along with radiographic assessment will enable more comprehensive and standardized response assessment in pediatric neuro-oncology patients enrolled in clinical trials.
- Published
- 2022
- Full Text
- View/download PDF
19. NF106: A Neurofibromatosis Clinical Trials Consortium Phase II Trial of the MEK Inhibitor Mirdametinib (PD-0325901) in Adolescents and Adults With NF1-Related Plexiform Neurofibromas
- Author
-
Chie Emoto, Nathan Robison, Brigitte C. Widemann, Brian Weiss, Stewart Goldman, Jeffrey C. Allen, James H. Tonsgard, Alexander A. Vinks, Nancy Ratner, Michael Fisher, Jaishri O. Blakeley, Bruce R. Korf, Pamela L. Wolters, Coretta Thomas Robinson, Lloyd J. Edwards, Eva Dombi, Elizabeth K. Schorry, Scott R. Plotkin, Tsuyoshi Fukuda, Gary Cutter, and Roger J. Packer
- Subjects
MAPK/ERK pathway ,Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Neurofibromatosis 1 ,Time Factors ,Adolescent ,Extracellular signal-regulated kinases ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Plexiform neurofibroma ,Internal medicine ,medicine ,Humans ,Neurofibromatosis ,Protein Kinase Inhibitors ,Mitogen-Activated Protein Kinase Kinases ,Neurofibroma, Plexiform ,business.industry ,MEK inhibitor ,Diphenylamine ,medicine.disease ,Magnetic Resonance Imaging ,United States ,Clinical trial ,Treatment Outcome ,030220 oncology & carcinogenesis ,Benzamides ,Female ,business ,030217 neurology & neurosurgery - Abstract
PURPOSE Patients with neurofibromatosis type 1 (NF1) frequently develop plexiform neurofibromas (PNs), which can cause significant morbidity. We performed a phase II trial of the MAPK/ERK kinase inhibitor, mirdametinib (PD-0325901), in patients with NF1 and inoperable PNs. The primary objective was response rate based on volumetric magnetic resonance imaging analysis. METHODS Inclusion criteria included age ≥ 16 years and a PN that was either progressive or causing significant morbidity. First-dose pharmacokinetics were performed. Patients completed patient-reported outcome measures. Patients received mirdametinib by mouth twice a day at 2 mg/m2/dose (maximum dose = 4 mg twice a day) in a 3-week on/1-week off sequence. Each course was 4 weeks in duration. Evaluations were performed after four courses for the first year and then after every six courses. Patients could receive a maximum of 24 total courses. RESULTS Nineteen patients were enrolled, and all 19 received mirdametinib. The median age was 24 years (range, 16-39 years); the median baseline tumor volume was 363.8 mL (range, 3.9-5,161 mL). Eight of the 19 patients (42%) achieved a partial response of the target PN by course 12, and 10 (53%) had stable disease. One patient (5%) developed progressive disease at course 8. Significant and durable decreases were observed in pain ratings. CONCLUSION To our knowledge, this analysis represents the first characterization of the activity and pharmacokinetics of mirdametinib in patients with NF1 and PNs and is the first published response study for MAPK/ERK kinase inhibitors in adults with NF1 and PNs. Mirdametinib given at 2 mg/m2/dose (maximum dose, 4 mg) twice daily in a 3-week on/1-week off sequence resulted in a 42% partial response rate with preliminary evidence of reduction in pain.
- Published
- 2021
20. Clinical outcomes and patient-matched molecular composition of relapsed medulloblastoma
- Author
-
Uri Tabori, Kyle S. Smith, Javad Nazarian, Kristin Schroeder, Cynthia Hawkins, Marcel Kool, Pieter Wesseling, Thomas E. Merchant, Sridharan Gururangan, Sarah Leary, Anthony P. Y. Liu, David W. Ellison, Tong Lin, Dong Anh Khuong-Quang, David T.W. Jones, Michael D. Taylor, José Pimentel, Girish Dhall, Colt Terhune, Sonia Partap, Eric Bouffet, Geoffrey McCowage, Laura J. Klesse, Vijay Ramaswamy, Paul A. Northcott, Tim Hassall, Daniel C. Bowers, Arnold C. Paulino, Gudrun Fleischhack, Jordan R. Hansford, Andrey Korshunov, Claudia C. Faria, Stewart J. Kellie, Sidney E Croul, Stefan Rutkowski, Anne Bendel, Maryam Fouladi, John R. Crawford, Sébastien Perreault, Stefan M. Pfister, Amar Gajjar, Yoon Jae Cho, Roger E. McLendon, Michal Zapotocky, Nada Jabado, Arzu Onar-Thomas, Paul Klimo, Catherine A. Billups, Giles W. Robinson, Juliette Hukin, Maximilian Deng, Andrew W. Walter, Brent A. Orr, Rahul Kumar, Olga Zheludkova, Murali Chintagumpala, Richard J. Cohn, Marina Ryzhova, Ute Bartels, Andrey Golanov, Christopher Dunham, Frederick A. Boop, Roger J. Packer, Michael Fisher, and Repositório da Universidade de Lisboa
- Subjects
Epigenomics ,Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Time Factors ,Molecular composition ,MEDLINE ,Relapsed Medulloblastoma ,Medizin ,Epigenome ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Cerebellar Neoplasms ,Child ,Clinical Trials as Topic ,business.industry ,High-Throughput Nucleotide Sequencing ,Infant ,DNA Methylation ,Treatment Outcome ,030104 developmental biology ,Child, Preschool ,030220 oncology & carcinogenesis ,Retreatment ,Disease Progression ,Female ,Neoplasm Recurrence, Local ,business ,Medulloblastoma - Abstract
© 2021 by American Society of Clinical Oncology. Creative Commons Attribution Non-Commercial No Derivatives 4.0 License: https://creativecommons.org/licenses/by-nc-nd/4.0/, Purpose: We sought to investigate clinical outcomes of relapsed medulloblastoma and to compare molecular features between patient-matched diagnostic and relapsed tumors. Methods: Children and infants enrolled on either SJMB03 (NCT00085202) or SJYC07 (NCT00602667) trials who experienced medulloblastoma relapse were analyzed for clinical outcomes, including anatomic and temporal patterns of relapse and postrelapse survival. A largely independent, paired molecular cohort was analyzed by DNA methylation array and next-generation sequencing. Results: A total of 72 of 329 (22%) SJMB03 and 52 of 79 (66%) SJYC07 patients experienced relapse with significant representation of Group 3 and wingless tumors. Although most patients exhibited some distal disease (79%), 38% of patients with sonic hedgehog tumors experienced isolated local relapse. Time to relapse and postrelapse survival varied by molecular subgroup with longer latencies for patients with Group 4 tumors. Postrelapse radiation therapy among previously nonirradiated SJYC07 patients was associated with long-term survival. Reirradiation was only temporizing for SJMB03 patients. Among 127 patients with patient-matched tumor pairs, 9 (7%) experienced subsequent nonmedulloblastoma CNS malignancies. Subgroup (96%) and subtype (80%) stabilities were largely maintained among the remainder. Rare subgroup divergence was observed from Group 4 to Group 3 tumors, which is coincident with genetic alterations involving MYC, MYCN, and FBXW7. Subgroup-specific patterns of alteration were identified for driver genes and chromosome arms. Conclusion: Clinical behavior of relapsed medulloblastoma must be contextualized in terms of up-front therapies and molecular classifications. Group 4 tumors exhibit slower biological progression. Utility of radiation at relapse is dependent on patient age and prior treatments. Degree and patterns of molecular conservation at relapse vary by subgroup. Relapse tissue enables verification of molecular targets and identification of occult secondary malignancies.
- Published
- 2021
21. CTIM-33. THE REMIND TRIAL: MULTI-ANTIGEN TARGETED T CELLS FOR PEDIATRIC CNS TUMORS
- Author
-
Fahmida Hoq, Christopher A. Lazarski, Anushree Datar, Jennifer Wang, Melanie Grant, Jay Tanna, Nan Zhang, Adriana Pitino, Eugene Hwang, Catherine M. Bollard, Patrick J. Hanley, Brian R. Rood, Maria Fernanda Fortiz, Madeline Terpilowski, Emily K. Reynolds, Roger J. Packer, Lindsay Kilburn, and Haili Lang
- Subjects
Cancer Research ,CD40 ,biology ,business.industry ,T-cell receptor ,Clinical Trials: Immunologic ,Interleukin 10 ,Immune system ,Granulocyte macrophage colony-stimulating factor ,Oncology ,Antigen ,Aldesleukin ,Cancer research ,biology.protein ,Medicine ,Neurology (clinical) ,Interleukin 17 ,business ,medicine.drug - Abstract
BACKGROUND Patients with relapsed CNS malignancies or DIPG face terrible prognoses. We hypothesized that T cells specific for 3 tumor-associated antigens (TAA), WT1, PRAME and survivin, would be safe and elicit anti-tumor immunity. METHODS Patients (n=15) received autologous tumor antigen-associated T cells (TAAT) (up to 4x107/m2) for newly diagnosed DIPG (Group A) or recurrent CNS malignancies (Group B) on a Phase I dose-escalation study (NCT03652545) and were monitored for safety and response. RESULTS/DISCUSSION 15/15 patients who received TAAT completed the 45-day safety monitoring phase with no dose-limiting toxicities. Adverse events were minimal despite multiple pretreatments in Group B. Infused cells were predominantly CD3+ T cells (median 96%; range: 87–99%), with CD4+ and CD8+ comprising 16% (range: 5–87%) and 40% (range: 4–67%) respectively. Specificity for 1–3 TAAs was demonstrated in 13/15 TAAT by a-IFN-γ ELISPOT. Plasma cytokine and proteomic analyses are ongoing but have demonstrated dynamic post-infusion immune cytokine and protein responses. Increases in the inflammatory and immune-stimulatory cytokines IL-1b, IL-6, IL-2 and IL-7 were observed post-infusion in most patients evaluated. Infusion-related increases in regulatory cytokines IL-10 and IL-13 were also observed in 4/7 patients. These results are consistent with an infusion-mediated immune response in vivo. Of 9 patients who have been tested thus far, 29/92 plasma proteins showed significant differences between dose levels 1 and 2, including increased IL-7 (p < 0.0004) and CD40L (p < 0.046) and reduced IL-4 (p < 0.0004). T cell receptor sequencing data on in vivo TAAT persistence is pending. In summary, TAAT have thus far been safe and elicit immune responses in vivo. Clinical and immunologic response assessments are ongoing.
- Published
- 2020
22. JNO special issue: an update on pediatric neuro-oncology
- Author
-
Lindsey Kilburn and Roger J. Packer
- Subjects
Cancer Research ,medicine.medical_specialty ,Oncology ,Neurology ,Pediatric Neuro-Oncology ,business.industry ,Brain Neoplasms ,medicine ,Humans ,Medical physics ,Neurology (clinical) ,business ,Child - Published
- 2020
23. Outcomes of BRAF V600E Pediatric Gliomas Treated With Targeted BRAF Inhibition
- Author
-
Cynthia Hawkins, Josef Zamecnik, David D. Eisenstat, Andres Morales La Madrid, Didier Frappaz, Adela Cañete, Valerie Larouche, Liana Nobre, Sarah Leary, Murali Chintagumpala, Lili-Naz Hazrati, Valentina Iurilli, Normand Laperriere, Peter Hauser, Scott L. Coven, Lenka Krskova, Jonathan L. Finlay, Jordan R. Hansford, Vijay Ramaswamy, Giovanni Morana, Julia Balaguer Guill, Palma Solano, Samantha Mascelli, Gurcharanjeet Kaur, Helen Toledano, Peter B. Dirks, Olaf Witt, Ute Bartels, Bev Wilson, Uri Tabori, Cornelis M. van Tilburg, Sarah Injac, Eduardo Quiroga-Cantero, Cecile Faure Conter, Ira J. Dunkel, Jaroslav Sterba, Duarte Salgado, Magnus Sabel, Diana S Osorio, Till Milde, Matthias A. Karajannis, Inga Harting, Zdenek Pavelka, Tara McKeown, Naureen Mushtaq, Daniel Alderete, Michal Zapotocky, Julie Bennett, Ofelia Cruz, Scott Ryall, Abhishek Bavle, James T. Rutka, Frank Lin, Elizabeth Finch, Frank van Landeghem, Anne Grete Bechensteen, Maria Luisa Garrè, Michael D. Taylor, Alvaro Lassaletta, Derek S. Tsang, Karen Gauvain, Daniel R. Boue, Jack Su, David Sumerauer, Sonika Dahiya, Helena Mörse, Annie Huang, Roger J. Packer, Lorena V Baroni, Martin Kyncl, Miriam Bornhorst, Eric Bouffet, and Mariana Fernandes
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Hematology ,endocrine system diseases ,business.industry ,digestive system diseases ,3. Good health ,BRAF V600E ,03 medical and health sciences ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,Mutation (genetic algorithm) ,Original Reports ,medicine ,business ,skin and connective tissue diseases ,neoplasms ,Chemoradiotherapy - Abstract
PURPOSE Children with pediatric gliomas harboring a BRAF V600E mutation have poor outcomes with current chemoradiotherapy strategies. Our aim was to study the role of targeted BRAF inhibition in these tumors. PATIENTS AND METHODS We collected clinical, imaging, molecular, and outcome information from patients with BRAF V600E–mutated glioma treated with BRAF inhibition across 29 centers from multiple countries. RESULTS Sixty-seven patients were treated with BRAF inhibition (pediatric low-grade gliomas [PLGGs], n = 56; pediatric high-grade gliomas [PHGGs], n = 11) for up to 5.6 years. Objective responses were observed in 80% of PLGGs, compared with 28% observed with conventional chemotherapy ( P < .001). These responses were rapid (median, 4 months) and sustained in 86% of tumors up to 5 years while receiving therapy. After discontinuation of BRAF inhibition, 76.5% (13 of 17) of patients with PLGG experienced rapid progression (median, 2.3 months). However, upon rechallenge with BRAF inhibition, 90% achieved an objective response. Poor prognostic factors in conventional therapies, such as concomitant homozygous deletion of CDKN2A, were not associated with lack of response to BRAF inhibition. In contrast, only 36% of those with PHGG responded to BRAF inhibition, with all but one tumor progressing within 18 months. In PLGG, responses translated to 3-year progression-free survival of 49.6% (95% CI, 35.3% to 69.5%) versus 29.8% (95% CI, 20% to 44.4%) for BRAF inhibition versus chemotherapy, respectively ( P = .02). CONCLUSION Use of BRAF inhibition results in robust and durable responses in BRAF V600E–mutated PLGG. Prospective studies are required to determine long-term survival and functional outcomes with BRAF inhibitor therapy in childhood gliomas.
- Published
- 2020
24. Extensive Molecular and Clinical Heterogeneity in Patients With Histologically Diagnosed CNS-PNET Treated as a Single Entity: A Report From the Children’s Oncology Group Randomized ACNS0332 Trial
- Author
-
Marcel Kool, Sebastian Brabetz, Maryam Fouladi, Eugene Hwang, David T.W. Jones, Linda Heier, Jeff M. Michalski, Chris Williams-Hughes, Lukas Chavez, Alok Jaju, David Capper, Peter C. Burger, James M. Olson, Ian F. Pollack, Stefan M. Pfister, Roger J. Packer, Catherine A. Billups, Yimei Li, Andreas von Deimling, Amar Gajjar, Sarah Leary, and Tianni Zhou
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Cns pnet ,Population ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Single entity ,Internal medicine ,medicine ,In patient ,education ,Isotretinoin ,Pineoblastoma ,Medulloblastoma ,education.field_of_study ,business.industry ,ORIGINAL REPORTS ,medicine.disease ,Carboplatin ,chemistry ,030220 oncology & carcinogenesis ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Purpose Children with histologically diagnosed high-risk medulloblastoma, supratentorial primitive neuroectodermal tumor of the CNS (CNS-PNET), and pineoblastoma (PBL) have had poor survival despite intensive treatment. We included these patients in this Children’s Oncology Group trial. Molecular profiling later revealed tumor heterogeneity that was not detectable at protocol inception. Enrollment of patients with CNS-PNET/PBL was subsequently discontinued, and outcomes for this part of the study are reported here. Patients and Methods In this phase III, four-arm prospective trial, consenting children age 3-22 years with newly diagnosed CNS-PNET were randomly assigned (1:1) to receive carboplatin during radiation and/or adjuvant isotretinoin after standard intensive therapy. Primary outcome measure was event-free survival (EFS) in the intent-to-treat population. Molecular tumor classification was retrospectively completed using DNA methylation profiling. Results Eighty-five participants with institutionally diagnosed CNS-PNETs/PBLs were enrolled. Of 60 patients with sufficient tissue, 31 were nonpineal in location, of which 22 (71%) represented tumors that were not intended for trial inclusion, including 18 high-grade gliomas (HGGs), two atypical teratoid rhabdoid tumors, and two ependymomas. Outcomes across tumor types were strikingly different. Patients with supratentorial embryonal tumors/PBLs exhibited 5-year EFS and overall survival of 62.8% (95% CI, 43.4% to 82.2%) and 78.5% (95% CI, 62.2% to 94.8%), respectively, whereas patients with molecularly classified HGG had EFS and overall survival of 5.6% (95% CI, 0% to 13.0%) and 12.0% (95% CI, 0% to 24.7%), respectively. Neither carboplatin, nor isotretinoin significantly altered outcomes for all patients. Survival for patients with HGG was similar to that of historic studies that avoid craniospinal irradiation and intensive chemotherapy. Conclusion For patients with CNS-PNET/PBL, prognosis is considerably better than previously assumed when molecularly confirmed HGGs are removed. Identification of molecular HGGs may spare affected children from unhelpful intensive treatment. This trial highlights the challenges of a histology-based diagnosis for pediatric brain tumors and indicates that molecular profiling should become a standard component of initial diagnosis.
- Published
- 2018
- Full Text
- View/download PDF
25. Longitudinal assessment of late-onset neurologic conditions in survivors of childhood central nervous system tumors: a Childhood Cancer Survivor Study report
- Author
-
Nicole J. Ullrich, Allison A. King, Leslie L. Robison, Kimberly Whelan, Roger J. Packer, Joseph P. Neglia, Kristy Seidel, Kevin R. Krull, Elizabeth Wells, Gregory T. Armstrong, Marilyn Stovall, Wendy M. Leisenring, Kevin C. Oeffinger, Charles A. Sklar, and Lisa Diller
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Pediatrics ,Adolescent ,Hearing loss ,medicine.medical_treatment ,Clinical Investigations ,Late onset ,Childhood Cancer Survivor Study ,Late Onset Disorders ,Central Nervous System Neoplasms ,Meningioma ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Humans ,Medicine ,Cumulative incidence ,Longitudinal Studies ,Survivors ,Child ,Stroke ,Aged ,business.industry ,Hazard ratio ,Middle Aged ,medicine.disease ,Radiation therapy ,Oncology ,030220 oncology & carcinogenesis ,Physical therapy ,Female ,Neurology (clinical) ,Cranial Irradiation ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Background Survivors of childhood central nervous system (CNS) tumors experience high rates of treatment-related neurologic sequelae. Whether survivors continue to be at increased risk for new events as they age is unknown. Methods Adverse neurologic health conditions in 5-year survivors of CNS tumors from the Childhood Cancer Survivor Study (n = 1876) were evaluated longitudinally at a median 23.0 years from diagnosis (range, 5.1-38.9), median age at last evaluation 30.3 years (range, 6.1-56.4). Multivariable regression estimated hazard ratios (HRs) and 95% CIs. Results From 5 to 30 years post diagnosis, cumulative incidence increased for seizures from 27% to 41%, motor impairment 21% to 35%, and hearing loss 9% to 23%. Risks were elevated compared with siblings (eg, seizures HR: 12.7; 95% CI: 9.6-16.7; motor impairment HR: 7.6; 95% CI: 5.8-9.9; hearing loss HR: 18.4; 95% CI: 13.1-25.9). Regional brain doses of radiation therapy were associated with development of new deficits (eg, frontal ≥50 Gy and motor impairment HR: 2.0; 95% CI: 1.2-3.4). Increased risk for motor impairment was also associated with tumor recurrence (HR: 2.6; 95% CI: 1.8-3.8), development of a meningioma (HR: 2.3; 95% CI: 0.9-5.4), and stroke (HR: 14.9; 95% CI: 10.4-21.4). Seizure risk was doubled by recurrence (HR: 2.3; 95% CI: 1.6-3.2), meningioma (HR: 2.6; 95% CI: 1.1-6.5), and stroke (HR: 2.0; 95% CI: 1.1-3.4). Conclusions CNS tumor survivors remain at risk for new-onset adverse neurologic events across their lifespans at a rate greater than siblings. Cranial radiation, stroke, tumor recurrence, and development of meningioma were independently associated with late-onset adverse neurologic sequelae.
- Published
- 2017
- Full Text
- View/download PDF
26. Spatial heterogeneity in medulloblastoma
- Author
-
David T.W. Jones, Yusanne Ma, Xin Wang, Betty Luu, Cynthia Hawkins, Adrian Ally, Marc Remke, John S. Myseros, Nada Jabado, Adam M. Fontebasso, Volker Hovestadt, Michael D. Taylor, Craig Daniels, Borja L. Holgado, Steffen Albrecht, Uri Tabori, David Malkin, Andrey Korshunov, Diana M. Merino, Stuart Horswell, Rebecca Carlsen, James T. Rutka, Daniel Picard, Marco A. Marra, David Shih, Angela Tam, Patrick Plettner, Roger J. Packer, Gary D. Bader, Erin N. Kiehna, Hamza Farooq, Eloi Mercier, John Peacock, Stephen C. Mack, A. Sorana Morrissy, Sameer Agnihotri, Jonathon Torchia, Vijay Ramaswamy, Young Cheng, Kane Tse, Steven J.M. Jones, Laura K. Donovan, Richard A. Moore, Sunit Das, Annie Huang, Nina Thiessen, James Loukides, Darlene Lee, Yisu Li, Marcel Kool, Charles Swanton, Eric Chuah, Tenzin Gayden, Xiaochong Wu, Michael Mayo, Eric Bouffet, Chelsea Mayoh, Peter Lichter, Noreen Dhalla, Brian R. Rood, Jacek Majewski, Ulrich Schüller, Haiyan I. Li, Richard Corbett, Tina Wong, Paul A. Northcott, Pawel Buczkowicz, Florence M.G. Cavalli, Yuan Yao Thompson, William Long, Maria C. Vladoiu, Peter B. Dirks, Hamid Nikbakht, Livia Garzia, Stefan M. Pfister, Jacqueline E. Schein, Simon Papillon-Cavanagh, Andrew J. Mungall, and Jüri Reimand
- Subjects
Adult ,Male ,0301 basic medicine ,renal cell carcinoma ,DNA Copy Number Variations ,Genome-wide association study ,Biology ,medulloblastoma ,Bioinformatics ,Polymorphism, Single Nucleotide ,Article ,whole exome sequencing ,Genetic Heterogeneity ,glioblastoma multiforme ,03 medical and health sciences ,INDEL Mutation ,Renal cell carcinoma ,Glioma ,Biopsy ,gene expression profiling ,Genetics ,medicine ,Cluster Analysis ,Humans ,Cerebellar Neoplasms ,Child ,Aged ,Aged, 80 and over ,Medulloblastoma ,Principal Component Analysis ,medicine.diagnostic_test ,Reverse Transcriptase Polymerase Chain Reaction ,business.industry ,Genetic heterogeneity ,Middle Aged ,medicine.disease ,3. Good health ,Gene Expression Regulation, Neoplastic ,Gene expression profiling ,030104 developmental biology ,Child, Preschool ,Mutation ,Cancer research ,Female ,Spatial heterogeneity ,Personalized medicine ,Transcriptome ,business ,Genome-Wide Association Study - Abstract
Spatial heterogeneity of transcriptional and genetic markers between physically isolated biopsies of a single tumor poses major barriers to the identification of biomarkers and the development of targeted therapies that will be effective against the entire tumor. We analyzed the spatial heterogeneity of multiregional biopsies from 35 patients, using a combination of transcriptomic and genomic profiles. Medulloblastomas (MBs), but not high-grade gliomas (HGGs), demonstrated spatially homogeneous transcriptomes, which allowed for accurate subgrouping of tumors from a single biopsy. Conversely, somatic mutations that affect genes suitable for targeted therapeutics demonstrated high levels of spatial heterogeneity in MB, malignant glioma, and renal cell carcinoma (RCC). Actionable targets found in a single MB biopsy were seldom clonal across the entire tumor, which brings the efficacy of monotherapies against a single target into question. Clinical trials of targeted therapies for MB should first ensure the spatially ubiquitous nature of the target mutation.
- Published
- 2017
- Full Text
- View/download PDF
27. DIPG-55. PATTERNS OF CEREBROSPINAL FLUID DIVERSION AND SURVIVAL IN CHILDREN WITH DIFFUSE INTRINSIC PONTINE GLIOMA: A REPORT FROM THE INTERNATIONAL DIPG REGISTRY
- Author
-
Elisa Carrasquedo Benito, Eric Bouffet, Tim Hassall, Murali Chintagumpala, Paul G. Fisher, Eric Sandler, Michelle Monje, Scott L Coven, Stewart Goldman, Lindsey Kilburn, Tabitha Cooney, Christopher L. Tinkle, Maryam Fouladi, Katie Black, Katherine E. Warren, Andrew Dodgshun, Kathleen Dorris, Ute Bartels, Hailey Bond, David S. Ziegler, Pratiti Bandopadhayay, Hetal Dholaria, Mariko DeWire-Schottmiller, Sarah Leary, Adam Lane, Blaise V. Jones, Jane E. Minturn, Yvan Samson, Raya Saab, Robert J. Greiner, Carl Koschmann, Anthony Asher, Nicholas G. Gottardo, Jordan R. Hansford, Sara Parkin, Brooklyn Chaney, James L. Leach, Lars M. Wagner, and Roger J. Packer
- Subjects
Cancer Research ,medicine.medical_specialty ,Standard of care ,business.industry ,Incidence (epidemiology) ,Medical record ,Diffuse Midline Glioma/DIPG ,respiratory system ,medicine.disease ,Hydrocephalus ,Exact test ,Cerebrospinal fluid ,Oncology ,Internal medicine ,Cerebrospinal fluid diversion ,medicine ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,Progression-free survival ,business ,human activities - Abstract
There is no standard of care for cerebrospinal (CSF) diversion in children with diffuse intrinsic pontine glioma (DIPG), nor understanding of survival impact. We evaluated CSF diversion characteristics in children with DIPG to determine incidence, indications and potential impact on survival. Data was extracted from subjects registered in the International DIPG registry (IDIPGR). IDIPGR team personnel obtained clinical and radiographic data from the registry database and when appropriate, abstracted additional data from individual medical records. Univariable analyses were performed using the Fisher’s exact test or Wilcoxon rank sum test. Survival was estimated using the Kaplan-Meier method. Evaluable patients (n=457) met criteria for DIPG diagnosis by central radiology review. Ninety-two patients (20%) had permanent CSF diversion. Indications for permanent diversion were hydrocephalus (41%), hydrocephalus and clinical symptoms (35%), and clinical symptoms alone (3%). Those with permanent diversion were significantly younger at diagnosis than those without diversion (median 5.3 years vs 6.9 years, p=0.0002), otherwise no significant differences in gender, race, or treatment were found. The progression-free and overall survival of those with permanent CSF diversion compared to those without permanent diversion was 4.5 and 10.9 months vs 6.9 and 11.2 months, respectively (p=0.001, p= 0.4). There was no significant difference in overall survival in patients with or without permanent CSF diversion among a large cohort of DIPG patients. Patients without permanent diversion had significantly prolonged progression free survival compared to those with permanent diversion. The qualitative risks and benefits of permanent CSF diversion need to be further evaluated.
- Published
- 2020
28. DIPG-46. NON-DIPG PATIENTS ENROLLED IN THE INTERNATIONAL DIPG REGISTRY: HISTOPATHOLOGIC EVALUATION OF CENTRAL NEURO-IMAGING REVIEW
- Author
-
Tabitha Cooney, Lars M. Wagner, Kathleen Dorris, Carl Koschmann, Maryam Fouladi, Anthony Asher, Ayman El-Sheikh, Michelle Monje-Deisseroth, Pratiti Bandopadhayay, Roger J. Packer, Mariko DeWire-Schottmiller, Christine Fuller, Eric Bouffet, Sarah Leary, Kenneth J. Cohen, David S. Ziegler, Tim Hassall, Lindsay Kilburn, Cynthia Hawkins, James L. Leach, Katie Black, Yvan Samson, Karen Tsui, Hetal Dholaria, Paul G. Fisher, Jordan R. Hansford, Sylvia Cheng, Rachid Drissi, Blanca Diez, Adam Lane, Stewart Goldman, Blaise V. Jones, Sridharan Gururangan, Mercedes Garcia Lombardi, Scott L. Coven, Eric Sandler, Robert J. Greiner, Jane E. Minturn, Gustavo Sevlever, Margot A. Lazow, Brooklyn Chaney, Christopher L. Tinkle, Mohamed S. Zaghloul, Ute Bartels, Motasem El-Ayadi, Jie Ma, and Nicholas G. Gottardo
- Subjects
Cancer Research ,medicine.medical_specialty ,medicine.diagnostic_test ,Pilocytic astrocytoma ,business.industry ,Diffuse Midline Glioma/DIPG ,Magnetic resonance imaging ,medicine.disease ,Oncology ,Diffuse Astrocytoma ,Glioma ,Biopsy ,medicine ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Histopathology ,Neurology (clinical) ,Radiology ,Differential diagnosis ,business ,Anaplastic astrocytoma - Abstract
INTRODUCTION The role of diagnostic biopsy in diffuse intrinsic pontine glioma (DIPG) remains in question. Distinguishing radiographically between DIPG and other pontine tumors with more favorable prognosis and different therapy is critically important. METHODS Cases submitted to the International DIPG registry with histopathologic data were analyzed. Central imaging review was performed by two neuro-radiologists; all cases with imaging features or histopathology suggestive of alternative diagnoses were re-reviewed. Imaging features suggestive of alternative diagnoses included non-pontine origin, RESULTS Among 297 patients with pathology from biopsy and/or autopsy available, 27 (9%) had histologic diagnoses not consistent with DIPG, commonly embryonal tumors (n=9) and pilocytic astrocytomas (n=11). 163 patients had diagnostic MRI available for central neuroimaging review. Among 81 patients classified as characteristic of DIPG, 80 (99%) had histopathology consistent with DIPG (diffuse midline glioma, H3K27M-mutant, glioblastoma, anaplastic astrocytoma, diffuse astrocytoma). Among 63 patients classified as likely DIPG, but with unusual imaging features, 59 (94%) had histopathology consistent with DIPG. 19 patients had imaging features suggestive of another diagnosis, including 13 with non-pontine tumor origin; the remaining 6 all had histopathology not consistent with DIPG. Association between central imaging review and histopathology was significant (p CONCLUSIONS The important role and accuracy of central neuroimaging review in diagnosing or excluding DIPG is demonstrated. In patients with pontine tumors for which DIPG is felt unlikely radiographically, biopsy may be considered to guide diagnosis and treatment.
- Published
- 2020
- Full Text
- View/download PDF
29. NFB-17. MEK INHIBITOR BINIMETINIB SHOWS CLINICAL ACTIVITY IN CHILDREN WITH NEUROFIBROMATOSIS TYPE 1- ASSOCIATED PLEXIFORM NEUROFIBROMAS: A REPORT FROM PNOC AND THE NF CLINICAL TRIALS CONSORTIUM
- Author
-
Nicole J. Ullrich, Elizabeth K. Schorry, Jeffrey C. Allen, Jaishri O. Blakeley, Eva Dombi, James H. Tonsgard, Alyssa Reddy, Coretta Thomas, Sabine Mueller, Stewart Goldman, Karin S. Walsh, Lloyd Edwards, Andrea M. Gross, Bruce R. Korf, Wade Clapp, Roger J. Packer, Michael D. Prados, and Michael Fisher
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,MEK inhibitor ,Peripheral nerve stimulation ,Time to treatment ,Binimetinib ,medicine.disease ,Neurofibromatosis ,Clinical trial ,chemistry.chemical_compound ,chemistry ,Plexiform neurofibroma ,Internal medicine ,Medicine ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,business ,Neurofibromatoses - Abstract
BACKGROUND Plexiform neurofibromas (PNs) can cause significant morbidity. In this phase 2 study, we assessed imaging and functional outcomes to the MEK-inhibitor Binimetinib in pediatric patients with PNs. METHODS Children (age 1–17 years) with PN that were progressive or causing significant morbidity were eligible. Binimetinib is dosed twice-daily (starting dose of 32mg/m2) for maximum of 24 four-week courses. Participants with partial response (PR; >20% decrease in PN volume on central MRI review) at cycle 12 may stay on therapy. Participants undergo MRI and functional assessments at baseline and after courses 4, 8, 12, 18 and 24. Functional assessments are based on PN location. RESULTS Here we present 1-year response data. Twenty participants (55% male) with median age 12 years (range 2–16 years) enrolled; 19 are evaluable for response. Median baseline tumor volume was 326 ml (range, 8-6661 ml). Fourteen participants (74%) met criteria for PR, with 11 achieving PR by course 5. Median maximal PN volume reduction was 25.5% (range, 9–54%). As of August 2020, 14 participants received at least 12 cycles of Binimetinib; 10 remain on therapy. Off study reasons include treatment associated toxicities (n=2), subject withdrawal (n=2), non-compliance (n=2), prolonged treatment delay (n=1), and lack of response (n=3). Thirteen participants underwent dose reduction. Institution-reported related grade 3 toxicities included dry skin, weight gain, muscle weakness, rash, paronychia, cellulitis, diarrhea, gastric hemorrhage and CPK increase. CONCLUSIONS Binimetinib appears reasonably well-tolerated and shows promising activity in children with NF1-associated PNs. Outcomes on functional improvement will be reported at the meeting.
- Published
- 2020
30. DIPG-74. RE-IRRADIATION OF DIPG: DATA FROM THE INTERNATIONAL DIPG REGISTRY
- Author
-
Pratiti Bandopadhayay, Yvan Samson, Maryam Fouladi, Katie Black, David S. Ziegler, Stewart Goldman, Mariko DeWire-Schottmiller, Kathleen Dorris, Lars M. Wagner, Sarah Leary, Adam Lane, Eric Sandler, Eric Bouffet, Murali Chintagumpala, Sylvia Cheng, Lucie Lafay-Cousin, Austin Schafer, Scott L Coven, Tim Hassall, Rachid Drissi, Sara Parkin, Lindsey Kilburn, Christopher L. Tinkle, Michelle Monje-Deisseroth, Jie Ma, Katherine E. Warren, Hetal Dholaria, Roger J. Packer, Paul D. Fisher, Andrew Dodgshun, Christine Fuller, Karen Tsui, Mohamed S. Zaghloul, Jordan R. Hansford, Sridharan Gururangan, Mercedes Garcia Lombardi, Brooklyn Chaney, Douglas Strother, Jane E. Minturn, Blaise V. Jones, Kenneth J. Cohen, James L. Leach, Ute Bartels, Motasem El-Ayadi, Raya Saab, Robert J. Greiner, David Gass, Nicholas G. Gottardo, and Carl Koschmann
- Subjects
Cancer Research ,medicine.medical_specialty ,Oncology ,business.industry ,Diffuse Midline Glioma/DIPG ,Medicine ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,Radiology ,business ,urologic and male genital diseases ,female genital diseases and pregnancy complications - Abstract
PURPOSE To review data from DIPG Registry patients recorded to have received a second course of radiation therapy (rRT). METHODS The International DIPG Registry was searched for patients with DIPG who were treated with a known dose of rRT. Doses of rRT, timing from initial diagnosis and primary radiation therapy (pRT), radiographic response to rRT and survival from diagnosis (OS) were evaluated. RESULTS Sixty (11.2%) of 535 Registry patients underwent rRT; dose was provided for 44 patients. Median (range) data from those 44 revealed that rRT was given at 12 (2–65) months from initial diagnosis of DIPG and at 9.6 (1–61) months from completion of pRT at a dose of 26.7 (1.8–74) Gy. After completion of rRT, MRI showed response, progression, stable disease or was not available in 19, 8, 3 and 14 patients, respectively. Median PFS and OS were 11 and 18.1 months, respectively. 475 Registry patients did not undergo rRT; their ages, duration of symptoms, and primary treatment with or without chemotherapy were not significantly different from the rRT cohort. Median PFS and OS for the non-rRT patients were 6.9 and 10 months, respectively. rRT patients were more likely to have had radiographic evidence of tumor necrosis at diagnosis than non-rRT patients. CONCLUSIONS Administration of rRT to patients with DIPG has been inconsistent with respect to timing and dose. Toxicity, response and quality of life data are incomplete, but survival appears to be lengthened with rRT. Prospective clinical trials will elucidate benefits and risks of rRT.
- Published
- 2020
31. Integrated analysis of pediatric low-grade glioma: clinical implications and the path forward
- Author
-
Tobey J. MacDonald and Roger J. Packer
- Subjects
Epigenomics ,Oncology ,Cancer Research ,medicine.medical_specialty ,Brain Neoplasms ,business.industry ,Glioma ,Epigenesis, Genetic ,Internal medicine ,Basic and Translational Investigations ,Path (graph theory) ,medicine ,Humans ,Low-Grade Glioma ,Neurology (clinical) ,Child ,business - Abstract
BACKGROUND: Both genetic and methylation analysis have been shown to provide insight into the diagnosis and prognosis of many brain tumors. However, the implication of methylation profiling and its interaction with genetic alterations in pediatric low-grade gliomas (PLGGs) are unclear. METHODS: We performed a comprehensive analysis of PLGG with long-term clinical follow-up. In total 152 PLGGs were analyzed from a range of pathological subtypes, including 40 gangliogliomas. Complete molecular analysis was compared with genome-wide methylation data and outcome in all patients. For further analysis of specific PLGG groups, including BRAF p.V600E mutant gliomas, we compiled an additional cohort of clinically and genetically defined tumors from 3 large centers. RESULTS: Unsupervised hierarchical clustering revealed 5 novel subgroups of PLGG. These were dominated by nonneoplastic factors such as tumor location and lymphocytic infiltration. Midline PLGG clustered together while deep hemispheric lesions differed from lesions in the periphery. Mutations were distributed throughout these location-driven clusters of PLGG. A novel methylation cluster suggesting high lymphocyte infiltration was confirmed pathologically and exhibited worse progression-free survival compared with PLGG harboring similar molecular alterations (P = 0.008; multivariate analysis: P = 0.035). Although the current methylation classifier revealed low confidence in 44% of cases and failed to add information in most PLGG, it was helpful in reclassifying rare cases. The addition of histopathological and molecular information to specific methylation subgroups such as pleomorphic xanthoastrocytoma–like tumors could stratify these tumors into low and high risk (P = 0.0014). CONCLUSION: The PLGG methylome is affected by multiple nonneoplastic factors. Combined molecular and pathological analysis is key to provide additional information when methylation classification is used for PLGG in the clinical setting.
- Published
- 2020
- Full Text
- View/download PDF
32. Multicenter, Prospective, Phase II and Biomarker Study of High-Dose Bevacizumab as Induction Therapy in Patients With Neurofibromatosis Type 2 and Progressive Vestibular Schwannoma
- Author
-
Matthias A. Karajannis, Jian Campian, Jaishri O. Blakeley, Michael Fisher, Gary Cutter, Roger J. Packer, Coretta Thomas, Tena Rosser, D. Wade Clapp, Scott R. Plotkin, Bruce R. Korf, Nicole J. Ullrich, Jeffrey C. Allen, Alona Muzikansky, James H. Tonsgard, and Dan G. Duda
- Subjects
Adult ,Male ,Neurofibromatosis 2 ,Cancer Research ,medicine.medical_specialty ,Adolescent ,Bevacizumab ,Urology ,Angiogenesis Inhibitors ,Schwannoma ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Original Reports ,Biomarkers, Tumor ,medicine ,otorhinolaryngologic diseases ,Humans ,Prospective Studies ,Neurofibromatosis type 2 ,Child ,Prospective cohort study ,Survival rate ,Vestibular system ,Dose-Response Relationship, Drug ,Errata ,business.industry ,Induction Chemotherapy ,Neuroma, Acoustic ,Middle Aged ,Prognosis ,medicine.disease ,Neuroma ,Survival Rate ,Oncology ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,Female ,business ,Follow-Up Studies ,030215 immunology ,medicine.drug - Abstract
PURPOSE Bevacizumab treatment at 7.5 mg/kg every 3 weeks results in improved hearing in approximately 35%-40% of patients with neurofibromatosis type 2 (NF2) and progressive vestibular schwannomas (VSs). However, the optimal dose is unknown. In this multicenter phase II and biomarker study, we evaluated the efficacy and safety of high-dose bevacizumab in pediatric and adult patients with NF2 with progressive VS. PATIENTS AND METHODS Bevacizumab was given for 6 months at 10 mg/kg every 2 weeks, followed by 18 months at 5 mg/kg every 3 weeks. The primary end point was hearing response defined by word recognition score (WRS) at 6 months. Secondary end points included toxicity, radiographic response, quality of life (QOL), and plasma biomarkers. RESULTS Twenty-two participants with NF2 (median age, 23 years) with progressive hearing loss in the target ear (median baseline WRS, 53%) were enrolled. Nine (41%) of 22 participants achieved a hearing response at 6 months (1 of 7 children and 8 of 15 adults; P = .08). Radiographic response was seen in 7 (32%) of 22 patients with VS at 6 months (7 of 15 adults and 0 of 7 children; P = .05). Common mild to moderate adverse events included hypertension, fatigue, headache, and irregular menstruation. Improvement in NF2-related QOL and reduction in tinnitus-related distress were reported in 30% and 60% of participants, respectively. Paradoxically, high-dose bevacizumab treatment was not associated with a significant decrease in free vascular endothelial growth factor but was associated with increased carbonic anhydrase IX, hepatocyte growth factor, placental growth factor, stromal cell-derived factor 1α, and basic fibroblast growth factor concentrations in plasma. CONCLUSION High-dose bevacizumab seems to be no more effective than standard-dose bevacizumab for treatment of patients with NF2 with hearing loss. In contrast to adults, pediatric participants did not experience tumor shrinkage. However, adult and pediatric participants reported similar improvement in QOL during induction. Novel approaches using bevacizumab should be considered for children with NF2.
- Published
- 2019
33. A pilot precision medicine trial for children with diffuse intrinsic pontine glioma-PNOC003: A report from the Pacific Pediatric Neuro-Oncology Consortium
- Author
-
David A. Solomon, Lindsay Kilburn, Cassie Kline, Anu Banerjee, Javad Nazarian, Payal Jain, Bo Zhang, Nathalene Truffaux, Joanna J. Phillips, Michael D. Prados, Sara A. Byron, Alison Roos, Kellie J. Nazemi, Sara Nasser, Sabine Mueller, Winnie S. Liang, Yuankun Zhu, Angela J. Waanders, John G. Kuhn, Michael E. Berens, Suresh N. Magge, John R. Crawford, Annette M. Molinaro, Roger J. Packer, Eshini Panditharatna, Adam C. Resnick, Nalin Gupta, and Claudia Petritsch
- Subjects
Oncology ,Male ,Cancer Research ,Pilot Projects ,Whole Exome Sequencing ,Circulating Tumor DNA ,Histones ,0302 clinical medicine ,Pediatric Neuro-Oncology ,Treatment plan ,Antineoplastic Combined Chemotherapy Protocols ,Brain Stem Neoplasms ,Molecular Targeted Therapy ,Precision Medicine ,Child ,Exome sequencing ,Cancer ,next generation sequencing ,Pediatric ,genomics-guided clinical trial ,Circulating tumor DNA ,030220 oncology & carcinogenesis ,Child, Preschool ,Female ,Sequence Analysis ,Biotechnology ,Adult ,medicine.medical_specialty ,Adolescent ,Pediatric Cancer ,precision medicine ,Oncology and Carcinogenesis ,DNA sequencing ,03 medical and health sciences ,Young Adult ,Rare Diseases ,Clinical Research ,Internal medicine ,Exome Sequencing ,medicine ,Genetics ,Humans ,Oncology & Carcinogenesis ,Preschool ,Whole genome sequencing ,Whole Genome Sequencing ,business.industry ,Sequence Analysis, RNA ,Diffuse Intrinsic Pontine Glioma ,Human Genome ,Neurosciences ,Precision medicine ,Brain Disorders ,Clinical trial ,Brain Cancer ,Good Health and Well Being ,Feasibility Studies ,RNA ,business - Abstract
This clinical trial evaluated whether whole exome sequencing (WES) and RNA sequencing (RNAseq) of paired normal and tumor tissues could be incorporated into a personalized treatment plan for newly diagnosed patients (
- Published
- 2019
- Full Text
- View/download PDF
34. Differential Expression of Wilms' Tumor Protein in Diffuse Intrinsic Pontine Glioma
- Author
-
Sulgi Lee, Madhuri Kambhampati, Heather Gordish-Dressman, M Isabel Almira-Suarez, Conrad Russell Y. Cruz, Javad Nazarian, Mariarita Santi, Roger J. Packer, Sridevi Yadavilli, and Eugene Hwang
- Subjects
Male ,Adolescent ,medicine.medical_treatment ,Pathology and Forensic Medicine ,Cohort Studies ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Young Adult ,0302 clinical medicine ,Antigen ,Western blot ,Diffuse intrinsic pontine glioma (DIPG) ,Glioma ,Brainstem glioma ,medicine ,Brain Stem Neoplasms ,Humans ,Child ,WT1 Proteins ,Childhood midline glioma ,biology ,medicine.diagnostic_test ,business.industry ,Diffuse Intrinsic Pontine Glioma ,Cancer ,Infant ,General Medicine ,Immunotherapy ,Original Articles ,medicine.disease ,Wilms Tumor Protein ,Gene Expression Regulation, Neoplastic ,Histone ,Neurology ,Child, Preschool ,Mutation ,Cancer research ,biology.protein ,Female ,Wilms tumor protein (WT1) ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are deadly tumors comprising 10%–15% of all childhood CNS cancers. Standard treatment is considered palliative and prognosis is near universal mortality. DIPGs have been classified into genomic subtypes based on histone variants with the lysine to methionine mutation on position 27 of histone tails (K27M). Given the increasing promise of immunotherapy, there have been ongoing efforts to identify tumor-specific antigens to serve as immunologic targets. We evaluated a large cohort of CNS specimens for Wilms’ tumor protein (WT1) expression. These specimens include primary pediatric CNS tumors (n = 38 midline gliomas and n = 3 non-midline gliomas; n = 23 DIPG, n = 10 low-grade gliomas, n = 8 high-grade gliomas), and DIPG primary cells. Here, we report the validation of WT1 as a tumor-associated antigen in DIPGs. We further report that WT1 expression is significantly correlated with specific oncohistone variants, with the highest expression detected in the H3.3K27M subgroup. WT1 expression was absent in all control specimens (n = 21). Western blot assays using DIPG primary cells (n = 6) showed a trend of higher WT1 expression in H3.3K27M cells when compared with H3.1 K27M cells and H3 wildtype cells. Our data are the first indication of the association between WT1 and DIPG, with specific upregulation in those harboring oncohistone H3.3K27M.
- Published
- 2019
35. DIPG-34. PRECLINICAL PRECISION TESTING OF PNOC003 BIOPSY DERIVED MODELS OF DIPG
- Author
-
Bo Zhang, Adam C. Resnick, Michael D. Prados, Javad Nazarian, Cassie Kline, Sabine Mueller, Sridevi Yadavili, Nathalene Truffaux, Yuankun Zhu, Payal Jain, Roger J. Packer, Madhuri Kambhampati, Xiaodong Yang, Jie Zhang, Yuying Zhai, Nalin Gupta, and Eshini Panditharatna
- Subjects
Cancer Research ,medicine.diagnostic_test ,business.industry ,Biology ,Diffuse Intrinsic Pontine Glioma (DIPG) ,Genome ,Oncology ,Tumor progression ,Mutation (genetic algorithm) ,Biopsy ,DNA methylation ,Cancer research ,medicine ,Neurology (clinical) ,Personalized medicine ,Epigenetics ,business ,Gene - Abstract
RATIONALE: One of the major hurdles in developing effective treatment for children with DIPG includes the lack of extensive combinatorial studies targeting major driver oncogenic pathways. Combination of H3K27M and TP53 mutations are found in over 50% of DIPG tumors, however, there is a lack of effective combinatorial precision therapy. METHODS: Tissue specimens obtained from subjects with DIPG were used to generate preclinical models. Subjects were enrolled in a prospective molecular diagnostic clinical trial and specimens were obtained by surgical biopsy both at initial diagnosis, and at the time of progression. RESULTS: Fourteen primary neurosphere cell lines and nine xenograft mouse models were successfully generated. The success rate of generating primary neurospheres and xenograft models was 45% and 75%, respectively. Among the 14 cell lines, one was derived from a biopsy performed after tumor progression, and one from washing a biopsy needle. Whole genome analysis of three biopsy-derived primary neurospheres revealed that these samples shared genomic alterations when compared to the primary tumors. All cell lines retained major oncogenic driver mutations in genes such as H3F3A, HIST1H3B, TP53, PPM1D, PIK3R1, and ACVR1. The needle-wash derived cell line was immortalized with hTERT, and exhibited the most deviation in mutation profiles and copy number alterations compared to primary tumor. Global DNA methylation profiling of seven primary tumor biopsies and matched primary neurospheres revealed similar epigenetic profiles. Primary neurospheres derived from biopsies were treated with the same specialized panel of agents recommended by a personalized medicine tumor board, which was based on the mutational profiles of the original tumor. SIGNIFICANCE: All cell lines exhibited varying levels of sensitivity to single agent indicating patterns of intertumor heterogeneity. The use of these model systems allows testing of combinatorial precision therapy for patients with DIPG, where there is a need for rapid translation into clinical trials.
- Published
- 2019
36. LGG-02. A PHASE II PROSPECTIVE TRIAL OF SELUMETINIB IN CHILDREN WITH RECURRENT/PROGRESSIVE PEDIATRIC LOW-GRADE GLIOMA (PLGG) WITH A FOCUS UPON OPTIC PATHWAY/HYPOTHALAMIC TUMORS AND VISUAL ACUITY OUTCOMES: A PEDIATRIC BRAIN TUMOR CONSORTIUM (PBTC) STUDY, PBTC-029B
- Author
-
Azra H. Ligon, Lindsay Kilburn, Tina Young Poussaint, Ibrahim Qaddoumi, Maryam Fouladi, Michael Fisher, Regina I. Jakacki, Gilbert Vezina, Malcolm G. Smith, Neal I. Lindeman, Clinton F. Stewart, Zoltan Patay, Ashok Panigrahy, David T.W. Jones, Ira J. Dunkel, Patricia Baxter, Stewart Goldman, Girish Dhall, Susa G Kreissman, Sofia Haque, Austin Doyle, Roger J. Packer, David S. Enterline, Jason Fangusaro, Arzu Onar-Thomas, Paul G. Fisher, Jeremy Jones, Blaise V. Jones, Soonme Cha, Benita Tamrazi, Anu Banerjee, Shengjie Wu, Ian F. Pollack, Stefan M. Pfister, and Jessica S Stern
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Visual acuity ,Pediatric Brain Tumor Consortium ,business.industry ,Phases of clinical research ,Low Grade Glioma ,medicine.disease ,Internal medicine ,Glioma ,medicine ,Selumetinib ,Hypothalamic Neoplasm ,Low-Grade Glioma ,Neurology (clinical) ,Progression-free survival ,medicine.symptom ,business - Abstract
BACKGROUND: Pediatric low-grade glioma (pLGG) is the most common CNS tumor of childhood. Progression-free survival (PFS) is much lower than overall survival emphasizing the need for alternative treatments. In addition, many children suffer functional morbidities such as visual and motor disturbances. Recently, there has been an appropriate prioritization of functional outcomes in children with pLGG. METHODS: We present the results of a PBTC phase II trial evaluating selumetinib, (AZD6244, ARRY-142886) a MEK-1/2 inhibitor, in children with recurrent/progressive pLGG on 2 strata, including visual outcomes. RESULTS: Stratum 3 enrolled Neurofibromatosis type 1 (NF1)-associated pLGG. Ten of 25 (40%) eligible patients had partial response (PR), 14/25 (56%) had stable disease (SD) and 1/25 (4%) had progressive disease (PD); 2-year PFS was 96+4%. Ten patients with optic pathway glioma (OPG) were evaluable for visual acuity (VA) at baseline and 1 year. VA improved in 2/10 patients (20%) and was stable in 8/10 (80%). One patient (10%) had improvement in visual fields (VF) and 9 patients (90%) had stable VF. Stratum 4 included patients with non-NF1-associated recurrent/progressive hypothalamic and OPG. Five of 25 (20%) eligible patients had PR, 16/25 (64%) had SD and 4 (16%) had PD; 2-year PFS was 78+8.5%. Nineteen of 25 patients were evaluable for VA. VA improved in 4/19 patients (21%), was stable in 13/19 (68%) and worsened in 2/19 (11%). Five patients (26%) had improved VF and 14 (74%) had stable VF. The most common toxicities included grade 1/2 CPK elevation, diarrhea, hypoalbuminemia and rash. Rare grade 3 toxicities included elevated CPK, rash and paronychia. CONCLUSIONS: Selumetinib was tolerable and effective in treating children with NF1-associated and sporadic recurrent/progressive hypothalamic and OPG based upon radiographic response and PFS. Twenty-seven of 29 (93%) evaluable patients had stable or improved vision based on VA and VF testing.
- Published
- 2019
37. DIPG-33. HARMONIZATION AND CHARACTERIZATION OF POSTMORTEM DONATIONS FOR PEDIATRIC BRAIN TUMORS
- Author
-
Sabine Mueller, Eugene Hwang, Roger J. Packer, Maria-Magdalena Georgescu, Naomi E. Rance, Madhuri Kambhampati, Cheng-Ying Ho, Javad Nazarian, Miriam Bornhorst, Sridevi Yadavilli, Isabel Almira-Suarez, Lindsay Kilburn, Brian R. Rood, and Eshini Panditharatna
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,business.industry ,Brain tumor childhood ,medicine.disease ,Diffuse Intrinsic Pontine Glioma (DIPG) ,Oncology ,Pediatric brain ,Glioma ,Antimitochondrial antibody ,Medicine ,Neurology (clinical) ,business ,Personal Integrity - Abstract
RATIONALE: A major factor contributing to the expanding knowledge of CNS tumors, specifically DMG biology is the selfless gift from patients and their families of postmortem tumor tissue. To facilitate this, we developed a postmortem donation program for collection of tissue and biospecimens for accelerating biology studies of end stage disease for pediatric CNS tumors. METHODS: Our postmortem program entails coordinating tissue donations, compiling clinical data, processing of whole brain, spinal cord, biofluids, and germline controls. Through our protocol, primary and disseminated tumor specimens are designated for molecular characterization and for generation of a matched preclinical model. RESULTS: During the past eight years, we coordinated and accrued over 50 autopsy cases including diffuse midline gliomas, ATRT, ETANTR, CPC from 20 institutions across North America and the United Kingdom. We report that approaching a family at the time of progression has provided the most successful outcome for consent to our program. Patient age range was from 11months to 20 years old. The median time from postmortem donation to tissue processing range was 13 hours with the earliest time point being 4 hours and the longest being 96 hours. All collected specimens were successfully processed and assessed for DNA/RNA integrity using bioanalyzer, attempted primary neurosphere cultures and cryopreserved for future molecular studies. PDX generation from fresh tissue proved more successful compared to primary neurospheres and post mortem time did not seem to have an effect. We have successfully generated models that represent molecular subgroups of DMGs such as H3.3/ H3.1- K27M and H3 WT. All generated preclinical models were validated by STR analysis and immunohistochemical assays using human mitochondrial antibodies. IMPACT: Postmortem tissue donations serve as an invaluable source for access to end-stage disease biology, generation of PDX models, studying patterns of tumor migration, testing combination therapy and preclinical drug testing.
- Published
- 2019
38. DIPG-30. ISOFORM SPECIFIC OVEREXPRESSION OF WILMS’ TUMOR PROTEIN IN DIFFUSE INTRINSIC PONTINE GLIOMAS
- Author
-
Madhuri Kambhampati, Pichai Raman, Sabine Mueller, Sridevi Yadavilli, Adam C. Resnick, Krutika S. Gaonkar, Heather Gordish-Dressman, Oren J. Becher, Conrad Russell Y. Cruz, Eugene Hwang, Roger J. Packer, M Isabel Almira-Suarez, Javad Nazarian, and Sulgi Lee
- Subjects
Gene isoform ,Cancer Research ,Chemistry ,medicine.disease ,medicine.disease_cause ,Diffuse Intrinsic Pontine Glioma (DIPG) ,Pons ,Wilms Tumor Protein ,Oncology ,Glioma ,medicine ,Cancer research ,Immunohistochemistry ,Neurology (clinical) ,Carcinogenesis ,Protein overexpression ,Glioblastoma - Abstract
Diffuse intrinsic pontine glioma (DIPG) is a deadly pediatric brain cancer constituting 10–15% of all central nervous system (CNS) tumors in children. Its anatomical location and infiltrative nature make it one of the most challenging tumors to treat. As such, targeted therapies are gaining more interest in CNS tumors. To identify a suitable therapeutic target, we evaluated publicly available gene expression data of pediatric glioblastoma specimens (n=34, GSE50161), DIPG specimens (n=35, GSE50021) and gene expression data of DIPG specimens from our previous dataset (n=5). Further analysis using 37 pediatric midline glioma specimens collected at autopsy in accordance with Children’s National Health System Institutional Review Board approval, indicated overexpression of one candidate protein, Wilms’ tumor protein (WT1), by immunohistochemistry. Interestingly, our immunohistochemistry and immunofluorescence analysis showed that WT1 in DIPG specimens is localized in the cytoplasm. Real-Time Quantitative Reverse Transcription PCR analysis using fresh frozen tumor specimens also showed that H3.3K27M-DIPG subtypes (n=3) to express higher levels of WT1 protein compared to those with H3.1K27M (n=2) (5.3 fold). More importantly, RNA-seq analysis of DIPG tumors (n=23) revealed specific transcriptome expression of WT1 transcript variant F. Nuclear export signal prediction analysis of the WT1 protein sequence translated by this transcript showed a higher nuclear export signal prediction score compared to other isoforms of WT1. Our findings suggest that WT1, particularly WT1 variant F, is a robust tumor specific antigen in DIPG, which may reveal its biological role in tumorigenesis and may have implications for clinical translation as an immunotherapeutic target.
- Published
- 2019
39. Treatment of pediatric cerebral radiation necrosis: a systematic review
- Author
-
Eugene Hwang, Roger J. Packer, Nicole L. Drezner, Kristina K. Hardy, Elizabeth Wells, Cheng-Ying Ho, and Gilbert Vezina
- Subjects
Cancer Research ,medicine.medical_specialty ,Neurology ,Bevacizumab ,medicine.medical_treatment ,Pediatrics ,law.invention ,Necrosis ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,law ,Internal medicine ,medicine ,Humans ,Adverse effect ,Stroke ,Cerebral Cortex ,Radiotherapy ,Brain Neoplasms ,business.industry ,medicine.disease ,Databases, Bibliographic ,Surgery ,Radiation therapy ,Systematic review ,Oncology ,030220 oncology & carcinogenesis ,Toxicity ,Steroids ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Cerebral radiation necrosis (CRN) is a toxicity of radiation therapy that can result in significant, potentially life-threatening neurologic deficits. Treatment for CRN has included surgical resection, corticosteroids, hyperbaric oxygen therapy (HBOT), and bevacizumab, but no consensus approach has been identified. We reviewed the available literature to evaluate efficacy of treatment approaches. Using methods specified in the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines when possible, we conducted searches of Ovid MEDLINE, Embase and Pubmed to identify studies reporting on outcomes for children (≤21 years old) with CRN. Eligible studies from 1990 to 2014 describing central nervous system (CNS) radiation necrosis with details of both treatment and outcomes were included. Eleven studies meeting criteria were identified. Of the nine studies with total patient denominators, 37 of 806 patients developed CRN (incidence = 4.6 %). Patients received treatment courses of steroids alone (n = 13), steroids with bevacizumab (n = 11) or HBOT (n = 12). Patients who failed to respond to steroids were more likely to be older than steroid-responsive patients (p = 0.009). With the exception of steroid-related adverse events, there was only one report of an adverse event (brainstem stroke) potentially attributable to intervention (bevacizumab). Those who received proton beam RT were both younger (p = 0.001) and had a shorter time to development of CRN (p = 0.079). The most common treatment following steroid initiation was addition of bevacizumab or HBOT, with good success and minimal toxicity. However, randomized controlled trials are needed to establish a definitive treatment algorithm that can be applied to children affected by CRN.
- Published
- 2016
- Full Text
- View/download PDF
40. 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
- Full Text
- View/download PDF
41. Nonrandomized comparison of neurofibromatosis type 1 and non-neurofibromatosis type 1 children who received carboplatin and vincristine for progressive low-grade glioma: A report from the Children's Oncology Group
- Author
-
Gilbert Vezina, David R. Freyer, Roger J. Packer, Caihong Xia, Ian F. Pollack, Timothy N. Booth, Richard Sposto, Joann L. Ater, and Claire Mazewski
- Subjects
Oncology ,congenital, hereditary, and neonatal diseases and abnormalities ,Cancer Research ,medicine.medical_specialty ,Vincristine ,medicine.medical_treatment ,Procarbazine ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Glioma ,medicine ,Neurofibromatosis ,neoplasms ,Univariate analysis ,Chemotherapy ,Pilocytic astrocytoma ,business.industry ,medicine.disease ,eye diseases ,Carboplatin ,nervous system diseases ,chemistry ,030220 oncology & carcinogenesis ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
BACKGROUND To evaluate tumor responses, event-free survival (EFS), overall survival (OS), and toxicity of chemotherapy, children with neurofibromatosis type 1 (NF1) and progressive low-grade glioma were enrolled into the Children's Oncology Group (COG) A9952 protocol and treated with carboplatin and vincristine (CV). METHODS Non-NF1 patients were randomized to CV or thioguanine, procarbazine, 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea, and vincristine in COG A9952. NF1 patients were assigned to CV only. NF1 patients and non-NF1 patients who were treated with CV were compared with respect to baseline characteristics, toxicity, tumor responses, EFS, and OS. RESULTS A total of 127 eligible patients with NF1 were nonrandomly assigned to CV: 42 NF1 patients (33%) had events, and 6 (4.7%) died. The 5-year EFS rate was 69% ± 4% for the CV-NF1 group and 39% ± 4% for the CV–non-NF1 group (P
- Published
- 2016
- Full Text
- View/download PDF
42. MBCL-16. EFFICACY OF CARBOPLATIN GIVEN CONCOMITANTLY WITH RADIATION AND ISOTRETINOIN AS A PRO-APOPTOTIC AGENT IN MAINTENANCE THERAPY IN HIGH-RISK MEDULLOBLASTOMA: A REPORT FROM THE CHILDREN’S ONCOLOGY GROUP
- Author
-
Linda Heier, Amar Gajjar, Jeff M. Michalski, Peter C. Burger, Eugene Hwang, Ian F. Pollack, Alok Jaju, Yimei Li, Catherine A. Billups, Roger J. Packer, Paul A. Northcott, James M. Olson, Kyle S. Smith, Maryam Fouladi, and Sarah Leary
- Subjects
Medulloblastoma ,Oncology ,Cisplatin ,Cancer Research ,medicine.medical_specialty ,Vincristine ,business.industry ,medicine.disease ,Carboplatin ,Metastasis ,chemistry.chemical_compound ,Maintenance therapy ,chemistry ,Internal medicine ,medicine ,Medulloblastoma (Clinical) ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,medicine.symptom ,business ,Anaplasia ,Isotretinoin ,medicine.drug - Abstract
BACKGROUND Metastasis, residual disease, and diffuse anaplasia are high-risk features in medulloblastoma. METHODS This was a randomized phase 3 study. Patients age 3–21 years with high-risk medulloblastoma received (+/-) daily carboplatin with 36Gy craniospinal radiation and weekly Vincristine followed by six cycles of maintenance chemotherapy with Cisplatin, Cyclophosphamide and Vincristine (+/) 12 cycles of isotretinoin during and following maintenance. The primary endpoint was event-free survival, with exact log-rank test to compare arms. Retrospective molecular analysis included DNA methylation and exome sequencing. RESULTS Of 294 medulloblastoma patients enrolled, 261 were eligible by central review of radiology and pathology, median age 8.6 years (range 3.3–21.2), 70% male, 189 (72%) with metastatic disease, 58 (22%) with diffuse anaplasia, 14 (5%) with >1.5cm2 residual disease. The 5-year EFS and OS for all subjects was 63%+4 and 73%+3, respectively. Isotretinoin randomization was closed due to futility. 5-year EFS was 66 + 5 with carboplatin versus 59 + 5 without (p=0.11), with effect exclusively observed in Group 3 subtype: 73%+8 with carboplatin versus 54%+9 without (p CONCLUSIONS Therapy intensification with carboplatin improved survival for high-risk group 3 medulloblastoma. These findings further support an integrated clinical and molecular risk stratification for medulloblastoma.
- Published
- 2020
- Full Text
- View/download PDF
43. THER-08. SGT53 – A NOVEL p53 NANOMEDICINE INDUCES SIGNIFICANT RESPONSES IN CHILDREN WITH RECURRENT MEDULLOBLASTOMA AND CHOROID PLEXUS CARCINOMA: A REPORT OF TWO CASES
- Author
-
Esther H. Chang, Chris P. Leung, Eugene Hwang, Lauren Hancock, Roger J. Packer, Kathleen F. Pirollo, and Lindsay Kilburn
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,business.industry ,Choroid plexus carcinoma ,Recurrent Medulloblastoma ,medicine.disease ,Viral/Gene Therapy and other Novel Therapies ,Oncology ,medicine ,Nanomedicine ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,business - Abstract
BACKGROUND Abnormal p53 function commonly defines high-risk CNS tumors, but functional restoration has eluded investigators. SGT-53 is a targeted nanomedicine encapsulating a plasmid DNA encoding wild-type human p53 with a transferrin receptor-targeting scFv on the nanocomplex surface resulting in efficient delivery across the BBB and robust tumor binding/uptake. Data generated by collaborators demonstrated synergy with irradiation and chemotherapy. REPORT: Two children with recurrent CNS malignancies have been treated with SGT-53, each receiving greater than 50 infusions in combination with irradiation and chemotherapy with no grade 3/4 AEs positively attributed to SGT-53. The first was an 11yo male with recurrent disseminated p53+ SHH medulloblastoma, with bulky intracranial/thoracolumbar disease. He received irradiation followed by biweekly doses of SGT-53 and temozolomide, bevacizumab and irinotecan given one week out of four. This patient exhibited a complete response of all disease on his first follow-up scan 8 weeks after therapy initiation and remained in remission for 8 months. The second patient was a 3yo male with disseminated recurrent choroid plexus carcinoma. He received CSI with SGT-53, followed by SGT-53, temozolomide and irinotecan as described above, again with no related grade 3/4 AEs. He experienced a partial response to all sites of disease and completed therapy after six months, progressing 7.8 months after initiating treatment. CONCLUSIONS SGT-53 was well tolerated in two heavily-pretreated patients despite aggressive combinatorial strategies. The majority of the AEs experienced were mild and manageable. Each patient had significant responses, suggesting that SGT-53 should be evaluated in a clinical trial for similar patients.
- Published
- 2020
44. LGG-04. A PHASE II RE-TREATMENT STUDY OF SELUMETINIB FOR RECURRENT OR PROGRESSIVE PEDIATRIC LOW-GRADE GLIOMA (pLGG): A PEDIATRIC BRAIN TUMOR CONSORTIUM (PBTC) STUDY
- Author
-
Lindsay Kilburn, Roger J. Packer, Ibrahim Qaddoumi, Jason Fangusaro, Girish Dhall, Alicia Lenzen, Sonia Partap, Shengjie Wu, Ian F. Pollack, Maryam Fouladi, Tina Young Poussaint, Ira J. Dunkel, and Arzu Onar-Thomas
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Pediatric Brain Tumor Consortium ,business.industry ,Low Grade Glioma ,Brain tumor childhood ,Progressive Neoplastic Disease ,Treatment study ,Partial response ,Internal medicine ,Selumetinib ,AcademicSubjects/MED00300 ,Medicine ,AcademicSubjects/MED00310 ,Low-Grade Glioma ,Neurology (clinical) ,Progression-free survival ,business - Abstract
The PBTC conducted a re-treatment study (NCT01089101) evaluating selumetinib (AZD6244, ARRY-142886), a MEK I/II inhibitor, in children with recurrent/progressive pLGG. Eligible patients must have previously enrolled on PBTC-029 or PBTC-029B and progressed after coming off treatment with selumetinib. Patients must have maintained stable disease (SD) for ≥12 courses or had a sustained radiographic response (partial or complete) during their first exposure to selumetinib. Thirty-five eligible patients (median age: 13.11 years [range 7.96–25.33]) were enrolled, 57% of whom had optic pathway or hypothalamic target lesions. At the time of submission, median duration of treatment was 18 courses (range 2–48) and 21 subjects remained on therapy. Best responses reported to date are 6/35 (17%) partial response, 22/35 (63%) SD and 7/35 (20%) progressive disease with a 2-year progression-free survival of 75.7 + 8.3%, which met the design parameters for success. The most common attributable toxicities were grade 1 diarrhea, elevated AST, hypoalbuminemia, elevated CPK, maculo-papular rash, fatigue, paronychia, ALT elevation, acneiform rash and grade 2 CPK elevation. Rare grade 3 toxicities included CPK elevation (3), lymphopenia (2), paronychia (2) and ALT elevation (2). There was only one grade 4 CPK elevation. Five patients (14%) required dose reductions due to toxicity. There does not appear to be a notable difference in toxicities observed during initial selumetinib therapy versus re-treatment. In pLGG that has recurred/progressed following treatment with selumetinib, re-treatment with selumetinib appears to be effective with 80% of patients again achieving response or prolonged stable disease. Long-term follow-up is ongoing.
- Published
- 2020
- Full Text
- View/download PDF
45. LGG-26. DIFFUSE LEPTOMENINGEAL GLIONEURONAL TUMOR (DLGNT) IN CHILDREN: DIFFERENT CLINICAL PRESENTATIONS AND OUTCOMES
- Author
-
Eugene Hwang, Ludmila Yasko, Roger J. Packer, Alexander Druy, Ludmila Papusha, Andge Valiakhmetova, Galina Novichkova, and Alexander Karachunsky
- Subjects
Trametinib ,Cancer Research ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Treatment outcome ,Low Grade Glioma ,Magnetic resonance imaging ,Chemotherapy regimen ,Carboplatin ,chemistry.chemical_compound ,Oncology ,chemistry ,Glioneuronal tumor ,Biopsy ,medicine ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,Radiology ,Vemurafenib ,business ,medicine.drug - Abstract
Diffuse leptomeningeal glioneuronal tumor (DLGNT) is an extremely rare disease, newly recognized in the 2016 WHO classification of tumors of the CNS. Most DLGNTs are low-grade neuroepithelial tumors with variable elements of neuronal/neurocytic and glial differentiation, have diffuse leptomeningeal enhancement on MRI, and typically harbor KIAA1549-BRAF fusions. Other alterations, such as the BRAF V600E substitution, are less common. Here, we present three cases of DLGNT with different presentations and outcomes. The first patient is a 2yr-old male with KIAA1549-BRAF fusion, and was treated with Carbo/VCR chemotherapy after a biopsy, with resultant ongoing stable disease for 3.5 years. The second patient, an 8yr-old male had the BRAF V600E point mutation and was treated with conventional chemotherapy (VCR/carboplatin). On progression, he received the BRAF inhibitor vemurafenib, achieving a complete response which last 14 month. The third patient, a 27 month old male, harbored a KIAA1549-BRAF fusion and was treated at diagnosis with the MEK inhibitor trametinib. The tumor has been radiographically stable in the context of clinical improvement for 21 months since the treatment initiation, ongoing 24 month. In summary, we present further evidence of MAPK pathway alterations in children with DLGNT. We describe a range of molecular presentations and clinical outcomes, including one patient treated with conventional chemotherapy with further stabilization of disease during 3.5 years and two patients who were successfully treated with targeted therapy.
- Published
- 2020
- Full Text
- View/download PDF
46. MBCL-15. IMPACT OF MOLECULAR SUBGROUPS ON OUTCOMES FOLLOWING RADIATION TREATMENT RANDOMIZATIONS FOR AVERAGE RISK MEDULLOBLASTOMA: A PLANNED ANALYSIS OF CHILDREN’S ONCOLOGY GROUP (COG) ACNS0331
- Author
-
Paul A. Northcott, Gilbert Vezina, Peter C. Burger, Jeff M. Michalski, Thomas E. Merchant, Thomas J. Fitzgerald, Roger J. Packer, Anna J. Janss, Catherine A. Billups, Yimei Li, Maryam Fouladi, Amar Gajjar, Kyle S. Smith, and Nancy J. Tarbell
- Subjects
Medulloblastoma ,Oncology ,Cancer Research ,medicine.medical_specialty ,Average risk ,business.industry ,medicine.disease ,Cog ,Internal medicine ,medicine ,Medulloblastoma (Clinical) ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,business - Abstract
The COG conducted a randomized trial for average-risk medulloblastoma (AR-MB). Patients age 3–21 years were randomized to a radiation boost to the whole posterior fossa (PFRT) or an involved field volume (IFRT) after receiving CSI. Patients age 3–7 years were also randomized to standard dose CSI (23.4Gy, SDCSI) or low dose CSI (18Gy, LDCSI). 464 evaluable patients were available to compare PFRT vs. IFRT and 226 for SDCSI vs. LDCSI. 380 cases had sufficient tissue for DNA methylation-based molecular classification: 362 confirmed medulloblastoma; 6 non-medulloblastoma; 12 inconclusive. Molecular subgrouping confirmed the following representation amongst the evaluable cohort: 156 Group 4 (43.1%), 76 Group 3 (21.0%), 66 SHH (18.2%), 64 WNT (17.7%). Five-year event-free survival (EFS) estimates were 82.5±2.7% and 80.5±2.7% for IFRT and PFRT, respectively (p=0.44). Five-year EFS estimates were 71.4±4.4% and 82.9±3.7% for LDCSI and SDCSI, respectively (p=0.028). EFS distributions differed significantly by subgroup (p
- Published
- 2020
- Full Text
- View/download PDF
47. GCT-23. MULTI-INSTITUTIONAL ANALYSIS OF TREATMENT MODALITIES IN BASAL GANGLIA AND THALAMIC GERMINOMA
- Author
-
Joseph Stanek, Gregory K. Friedman, Ashley Margol, Jonathan L. Finlay, Tabitha Cooney, Rebecca Ronsley, Ute Bartels, Sabine Mueller, Susan N. Chi, Karen Gauvain, Stephanie Villeneuve, Kee Kiat Yeo, Richard T Graham, Girish Dhall, Jeffrey C. Allen, Mohammad H Abu-Arja, Roger J. Packer, Pournima Navalkele, Juliette Hukin, Mohamed S. AbdelBaki, Andrea Cappellano, Nicholas G. Gottardo, Michael Fisher, Jack Su, Lindsey M Hoffmann, and Amar Gajjar
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Germinoma ,business.industry ,medicine.disease ,Oncology ,Treatment modality ,Germ Cell Tumors ,Basal ganglia ,medicine ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,business - Abstract
BACKGROUND Central nervous system (CNS) germinomas are radiotherapy (RT)-sensitive tumors with excellent survival. Current treatment strategies combine chemotherapy with RT to reduce the field and dose of RT. There is no standard treatment for germinomas originating in the basal ganglia/thalami (BGTG) given their rarity and poorly-defined imaging characteristics. Craniospinal (CSI), whole brain (WBI), whole ventricle (WVI), and focal RT have been previously utilized; however, the optimal strategy remains unclear. METHODS Retrospective multi-institutional analysis was conducted across 18 institutions in four countries. RESULTS For 46 cases with non-metastatic BGTG, the event-free survival (EFS) was 86.9% at both 5 and 10 years, while overall survival (OS) was 100%, and 95.7% respectively at 5 and 10 years. Median RT dose and range for the various treatment volumes were as follows: CSI (n=10): 2340 cGy (1980–3060 cGy), WBI (n=8): 2340 (1800–3000 cGy), WVI (n=14): 2340 cGy (1800–2550 cGy), focal (n=9): 3600 cGy (3060–5400 cGy). There was no statistically significant difference in the EFS based on RT modality (p=0.57), but EFS for subjects with CSI and WBI were both 100%. The three subjects who received chemotherapy alone had significantly lower EFS than those who received chemotherapy and RT (p=0.001), but were salvageable with RT. CONCLUSION In the largest study to date for BGTG, there were no significant differences in outcomes between patients who received CSI, WBI, WVI or focal RT. This group of patients should be included in future prospective clinical trials, and a more limited RT field may be considered.
- Published
- 2020
- Full Text
- View/download PDF
48. LGG-55. OUTCOME OF BRAF V600E PEDIATRIC GLIOMAS TREATED WITH TARGETED BRAF INHIBITION
- Author
-
Ofelia Cruz, Mariana Fernandes, B. Wilson, Abhishek Bavle, Daniel Alderete, Michael D. Taylor, Olaf Witt, Gurcharanjeet Kaur, Jordan R. Hansford, Scott Ryall, Till Milde, Andres Morales La Madrid, Sarah Leary, Zdenek Pavelka, David Sumerauer, Anne Grete Bechensteen, Inga Harting, Liana Nobre, Michal Zapotocky, Eric Bouffet, Jaroslav Sterba, Daniel R. Boue, Jack Su, Scott L. Coven, Maria Luisa Garrè, Matthias A. Karajannis, Helen Toledano, Naureen Mushtaq, Ute Bartels, Sarah Injac, Cecile Faure Conter, Samantha Mascelli, Frank van Landeghem, Annie Huang, Peter Hauser, Derek S Tsang, Helena Mörse, Martin Kyncl, Normad Laperriere, Elizabeth Finch, James T. Rutka, Cornelis M. van Tilburg, Roger J. Packer, Uri Tabori, Julia Balaguer Guill, Magnus Sabel, Jonathan L. Finlay, Peter B. Dirks, Sonika Dahiya, Cynthia Hawkins, Lorena V Baroni, Lili-Naz Hazrati, Eduardo Quiroga-Cantero, Diana S Osorio, Murali Chintagumpala, Palma Solano, Josef Zamecbik, Tara McKeown, Ira J. Dunkel, Alvaro Lassaletta, Julie Bennet, David D. Eisenstat, Valerie Larouche, Karen Gauvain, Lenka Krskova, Duarte Salgado, Vijay Ramaswamy, Giovanni Morana, Frank Lin, Didier Frappaz, Miriam Bornhorst, Adela Cañete, and Valentina Iurilli
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Low Grade Glioma ,medicine.disease ,Chemotherapy regimen ,Discontinuation ,CDKN2A ,Internal medicine ,Glioma ,Concomitant ,Medicine ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,Progression-free survival ,business ,Prospective cohort study - Abstract
Children with pediatric gliomas harboring BRAF V600E mutation have a poor outcome with current chemoradiation strategies. Our aim was to study the role of targeted BRAF inhibition in these tumors. We collected clinical, imaging, molecular and outcome information from BRAF V600E glioma patients treated with BRAFi across 29 centers from multiple countries. Sixty-seven patients were treated with BRAFi (56 pediatric low grade gliomas, PLGG and 11 pediatric high grade gliomas, PHGG) for up to 5.6 years. Objective responses were observed in 80% of PLGGs compared to 28% with conventional chemotherapy (p
- Published
- 2020
49. PATH-14. GENETIC SUSCEPTIBILITY AND OUTCOMES OF PEDIATRIC, ADOLESCENT AND YOUNG ADULT IDH-MUTANT ASTROCYTOMAS
- Author
-
Sabine Mueller, Alberto Bronischer, Miriam Bornhorst, Lindsay Kilburn, Hayk Barseghyan, Eric Vilain, Tobey J. MacDonald, Joyce Turner, Brian R. Rood, Matthew Schniederjan, Jeremy Goecks, Denise Leung Leung, Enrico Opocher, Javad Nazarian, Cheng-Ying Ho, Cynthia Hawkins, Eugene Hwang, Eric Bouffet, Daniel R. Boue, Carl Koschmann, Brent A. Orr, Uri Tabori, Alexander O. Vortmeyer, Rajen Mody, Michal Zapotocky, Roger J. Packer, Surajit Bhattacharya, Asher Marks, Liana Nobre, and David A. Solomon
- Subjects
Genetics ,Cancer Research ,Oncology ,Mutant ,Path (graph theory) ,Genetic predisposition ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,Biology ,Young adult ,Pathology and Molecular Diagnosis - Abstract
INTRODUCTION Previously thought to be rare, recent case series have shown that IDH mutations in young patients are more common than previously described. In this study, we analyzed IDH-mutant tumors to determine clinical significance of these mutations in children, adolescents and young adults. METHODS Through this multi-institution study (10 institutions), we collected 64 IDH1/2-mutant infiltrating astrocytoma specimens from 58 patients aged 4–26 (M:F, 0.4:0.6). Specimens included 46 low-grade (LGG) and 18 high-grade (HGG) astrocytomas. Tumor sequencing data (n=45), germline sequencing data (n=37) and outcome data (n=40) was analyzed. RESULTS Similar to adults, most sequenced tumors had a co-mutation in the TP53 gene, while ATRX mutations were less common and primarily seen in HGGs. Approximately 60% (n=21) of patients with germline data available had a mutation in a cancer predisposition gene. Mismatch repair (MMR) mutations were most common (n=12; MSH6 n=9), followed by TP53mutations (n=7). All patients with MMR gene mutations had HGGs and poor progression free (PFS=10% at 2 years, mean TTP=9 months) and overall (OS CONCLUSION IDH-mutant tumors in pediatric patients are strongly associated with cancer predisposition and increased risk for progression/recurrence or malignant transformation. Routine screening for IDH1/2 mutations in children with grade 2–4 astrocytomas could greatly impact patient management.
- Published
- 2020
50. Impact of Molecular Subgroups on Outcomes Following Radiation Treatment Randomizations for Average Risk Medulloblastoma: A Planned Analysis of Children’s Oncology Group (COG) ACNS0331
- Author
-
Roger J. Packer, T.J. FitzGerald, K. Smith, Anna J. Janss, Stephanie M. Perkins, Scott L. Pomeroy, J. Hadley, Nancy J. Tarbell, Gilbert Vezina, Yuxin Li, Peter C. Burger, Maryam Fouladi, Patsy McGuire Cullen, J.M. Michalski, Thomas E. Merchant, Rajesh Kumar, P. Northcott, T. Booth, Catherine A. Billups, and Amar J. Gajjar
- Subjects
Oncology ,Medulloblastoma ,Cancer Research ,medicine.medical_specialty ,Average risk ,Radiation ,business.industry ,medicine.disease ,Cog ,Internal medicine ,medicine ,Radiology, Nuclear Medicine and imaging ,business - Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.