15 results on '"Valerie Larouche"'
Search Results
2. 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
3. CTNI-68. FIREFLY-1 (PNOC026): PHASE 2 STUDY OF PAN-RAF INHIBITOR TOVORAFENIB IN PEDIATRIC AND YOUNG ADULT PATIENTS WITH RAF-ALTERED RECURRENT OR PROGRESSIVE LOW-GRADE GLIOMA OR ADVANCED SOLID TUMORS
- Author
-
Lindsay Kilburn, Daniel Landi, Sarah Leary, David Ziegler, Patricia Baxter, Andrea Franson, Geoffrey McCowage, Angela Waanders, Jasper Van der Lugt, Michal Yalon Oren, Nicolas Gerber, Nicholas Gottardo, Dong-Anh Khuong-Quang, Karsten Nysom, Simon Bailey, Pablo Hernáiz Driever, Sebastien Perreault, Olaf Witt, Seungmin Hahn, Darren Hargrave, Timothy Hassall, Nada Jabado, Hyoung Jin Kang, Valerie Larouche, Helen Toledano, Cassie Kline, Mohamed Abdelbaki, Susan Chi, Sharon Gardner, Nicholas Whipple, Sabine Mueller, Samuel Blackman, Xin Zhao, Daniel Da Costa, Michael Cox, Roger Packer, and Jordan Hansford
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
BACKGROUND RAF alterations are oncogenic drivers found in most pediatric low-grade gliomas (LGGs). Tovorafenib is an investigational, oral, selective, CNS-penetrant, small molecule, type II pan‑RAF inhibitor. METHODS FIREFLY-1 (NCT04775485) is a multicenter phase 2 study evaluating the safety and efficacy of tovorafenib monotherapy. Registrational arm 1 enrolled patients with recurrent/progressive LGG harboring an activating BRAF alteration. Patients aged 6 months–25 years who progressed following ≥ 1 prior line of systemic therapy were eligible. Tovorafenib 420 mg/m2 (≤ 600 mg) was administered weekly (tablet or liquid suspension formulation) until progression or for ≥ 26, 28-day cycles. The primary endpoint (arm 1) was overall response rate, as defined by RANO criteria, per independent review. RESULTS As of April 14, 2022, 25 patients were enrolled to arm 1 and had ≥ 6 months of follow-up. Median age at enrollment was 8 years (range 3–18). Most patients had astrocytomas (92%), 48% with optic pathway involvement. Patients were heavily pretreated (56% with ≥ 3 prior lines of therapy), and 72% previously received MAPK pathway-targeted agents. Tumors harbored BRAF fusions (84%) or BRAF V600E mutations (16%). Per independent assessment, partial responses (1 unconfirmed) were seen in 14 (64%) of 22 evaluable patients, with 6 additional patients having stable disease, and a clinical benefit rate of 91%. Responses were achieved in tumors with BRAF fusions and V600E mutations. Most treatment-emergent adverse events (AEs) were grade 1 or 2 (96%). The most common grade ≥ 3 AEs were anemia (12%), vomiting, increased blood creatinine phosphokinase and maculopapular rash (8% each). Seven patients (28%) required dose modification for treatment-related AEs; no patients discontinued tovorafenib due to AEs. Updated results, including efficacy per RAPNO assessments will be presented. CONCLUSIONS Tovorafenib was generally well tolerated and showed encouraging evidence of antitumor activity in children with pretreated BRAF-altered LGG.
- Published
- 2022
4. CTNI-04. TRAM-01: A PHASE 2 STUDY OF TRAMETINIB FOR PEDIATRIC PATIENTS WITH NEUROFIBROMATOSIS TYPE 1 AND PLEXIFORM NEUROFIBROMAS
- Author
-
Dorsa Sadat Kiaei, Valerie Larouche, Jean-Claude Décarie, Uri Tabori, Cynthia Hawkins, Sarah Lippe, Benjamin Ellezam, Luis H Ospina, Yves Theoret, Leandra Desjardins, Marie-Elaine Metras, Serge Sultan, Edith Cantin, Marie-Eve Routhier, Chantal Mailloux, Marie-claude Bertrand, Maxime Caru, Tara McKeown, Stephanie Vairy, Geneviève Legault, Eric Bouffet, Vijay Ramaswamy, Hallie Coltin, Lucie Lafay-Cousin, Juliette Hukin, Craig Erker, Nada Jabado, Mathieu Dehaes, and Sébastien Perreault
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
BACKGROUND Plexiform neurofibromas (PN) are observed in up to 50% of patients with neurofibromatosis type 1 (NF1). Trametinib has been used widely to treat PN but limited data has been reported on its efficacy within a clinical trial. METHODS This ongoing multicenter phase II trial includes patients with pediatric low-grade glioma and PN. Patients received daily oral trametinib (MEK inhibitor) for eighteen 28-day cycles. The volumes of PN were centrally quantified using a new semi-automatic 3D segmentation method. RESULTS As of May 15, 2022, 46 patients with PN were enrolled in the study and the recruitment was completed for this study arm. Thirty-four completed treatment and were available for analysis. For these patients, the median age was 10.5 years (range 0.7-19.8). The median volume of PN at baseline was 51cm3 (range 2.6 to 487.6). Among the 34 patients, 28 (82.4%) completed 18 cycles as planned. Two patients discontinued due to adverse reaction, three patients refused to continue treatment and one patient discontinued treatment based on physician decision. Median duration of treatment was 16.8 months (range 2.8 to 16.8). Median duration of follow-up was 30.4 months (range 8.2 to 42). A total of 38 PN were available for volumetric analysis. Using RECIST evaluation, the overall response rate was 13.1%. Volumetric assessment demonstrated an overall response rate of 64.7% (22/34 patients), and 65.8% (25/38 PN) of PN showed a decrease of more than 20% in volume. Median volume change was -30% (range -93.5 to 14.3). Thirty-one patients (91.1%) had durable response without progression (lasting ≥ 1 year). After discontinuation of treatment, one patient underwent surgery and three patients resumed MEK inhibitor. CONCLUSION We report outcome and volumetric quantification of PN treated with trametinib within a large clinical trial. Based on the current results, trametinib is effective and offers durable response.
- Published
- 2022
5. MEDB-49. Relapsed SHH medulloblastomas in young children. Are there alternatives to full-dose craniospinal irradiation?
- Author
-
Craig Erker, Brandon Craig, Simon Bailey, Maura Massimino, Valerie Larouche, Jonathan L Finlay, Cassie Kline, George Michaiel, Ashley Margol, Kenneth Cohen, Chantel Cacciotti, Virginia Harrods, Kathleen Doris, Mohammed AbdelBaki, Nisreen Amayiri, Zhihong Wang, Jordan Hansford, Juliette Hukin, Ralph Salloum, Lindsay Hoffman, Jeffrey Muray, Kevin Ginn, Zapotocky Zapotocky, Lorena Baroni, Vijay Ramaswamy, Stephen Gilheens, Dolli Aguiera, Claire Mazewski, Shafqat Shah, Douglas Strother, Sabine Muller, Amar Gajjar, Paul Northcott, Steve Clifford, Giles Robinson, Eric Bouffet, and Lucie Lafay-Cousin
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
BACKGROUND/RATIONAL: Following initial irradiation sparing therapy, many young children with relapsed medulloblastoma can be salvaged with craniospinal irradiation (CSI). However, the interval to relapse is short and neurocognitive sequelae remain a major concern. The contribution of molecular subgrouping may help refine indications and modalities of salvage strategies in this population. METHOD: From a cohort of 151 young children with molecularly characterized relapsed medulloblastoma, subset analysis of the SHH medulloblastoma was conducted to describe the practice of salvage radiotherapy and associated post-relapse survival (PRS). RESULTS: Sixty-seven SHH medulloblastoma patients (46 M0; 54 GTR; 11 non-ND/MBEN) received salvage therapy with curative intent. Before relapse, 54 (80.6%) received conventional chemotherapy (CC), 13 (19.4%) high-dose chemotherapy (HDC), while seven had additional focal radiotherapy (fRT). Median time to relapse was 11.1 months (range 3.8-41.0) and 43.3% were localized. Thirty patients (16 localized relapse) underwent surgery. Forty-seven (71.2%) received salvage radiotherapy (20 with CC; 10 with HDC; 15 alone, two unknown). CSI and fRT accounted for 82% and 18% respectively. CSI median dose was 36Gy (range 18-39Gy). Ten patients (eight with localized relapse) received CSI doses ≤23.4Gy. Nineteen patients (28.8%) did not receive any radiotherapy (nine HDC; 10 CC only). Radiotherapy was associated with better 3-year PRS (73.0% versus 36.1%; p=0.001). All patients treated with CSI ≤ 23.4Gy were alive at median follow-up of 69 months(24-142). Six of nine patients treated with HDC without irradiation were alive at last follow-up. Sixty-three percent of patients received reduced dose CSI(≤23.4Gy), fRT, or no radiotherapy, and their PRS did not significantly differ from those who received CSI ≥ 30.6Gy (p = 0.54). CONCLUSION: While salvage CSI provided PRS benefit in this SHH medulloblastoma cohort, we report the use of reduced salvage radiotherapy and irradiation avoidance in 63% of the patients, with 60% alive at last follow-up.
- Published
- 2022
6. IMMU-13. Dual CTLA4/ PD-1 blockade improves survival for replication-repair deficient high-grade gliomas failing single agent PD-1 inhibition: An IRRDC study
- Author
-
Anirban Das, Daniel Morgenstern, Vanessa Bianchi, Sumedha Sudhaman, Melissa Edwards, Lucie Stengs, Valerie Larouche, David Samuel, An Van Damme, David Gass, David Ziegler, Stefan Bielack, Shayna Zelcer, Michal Yalon, Shlomi Constantini, Tomasz Sarosiek, Witold Libionka, Kim Nichols, Rebecca Loret De Mola, Kevin Bielamowicz, Magnus Sabel, Charlotta Frojd, Matthew D Wood, Joana Cristiano Sous Migueis, Chenue Abongwa, Lee Yi Yen, Duncan Stearns, Enrico Opocher, Kanika Bhatia, Santanu Sen, Eduardo Quiroga Cantero, Palma Solano Paez, Bruce Crooks, Vanan Magimairajan, Alyssa Reddy, Jenny Adamski, Gary Mason, Scott Lindhorst, Melyssa Aronson, Birgit Ertl-Wagner, Cynthia Hawkins, Eric Bouffet, and Uri Tabori
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
BACKGROUND: High-grade gliomas (HGG) with replication-repair deficiency (RRD) harbour high mutation burden (TMB) and are rapidly fatal following chemo-radiation approaches. Although hypermutation results in objective responses and prolonged survival in >30% of patients undergoing PD1-blockade, salvage following failure of PD1-inhibition remains a challenge. METHODS: We performed a real-world study of Ipilimumab (anti-CTLA4) in combination with Nivolumab/Pembrolizumab for patients failing single-agent PD1-inhibition. RESULTS: Among 68 consortium patients with relapsed HGG treated with single-agent PD1-inhibitors, progression was observed in 43 (63%). Ipilimumab was added to 20/43 (46.5%), 14 (32.5%) received best supportive care (BSC), and 9 (21%) received miscellaneous therapies. For patients receiving CTLA4/PD1-inhibition, median age at progression was 12.3-years (IQR: 9; 15.6). Time from anti-PD1 initiation to progression was 8-months (IQR: 3.8; 18.5). Germline predisposition was observed in all patients (CMMRD: 70%, Lynch: 25%, polymerase-proofreading deficiency: 5%). All HGG were hypermutant (median TMB: 182 mutations/Mb; IQR: 15.6; 369.4). Centralized radiology review revealed objective responses in 3/20 (15%, all ultra-hypermutant: 320, 496, 834 mutations/Mb), stable disease in 5 (25%), and 12 (60%) eventually progressed (iRANO). Following failure of PD1-blockade, estimated progression-free and overall survival at 18-months for patients receiving CTLA4/PD1-inhibition were 11% and 25%, respectively. Importantly, survival was superior to patients receiving BSC (median OS 1-year on the anti-CTLA4/PD1combination (range:1-48 months). The combinational immunotherapy resulted in significant autoimmune toxicity in 11/20 (55%), warranting immunosuppressive therapy in all, and treatment abandonment in 6 patients. CONCLUSION: Combined CTLA4/PD1-blockade after failure of single-agent PD1-inhibition revealed objective responses and prolonged survival in an otherwise rapidly-fatal disease. This needs to be assessed in the context of significant autoimmunity, supporting the need for the current prospective trial (NCT04500548), and novel strategies to limit treatment-related toxicity.
- Published
- 2022
7. SYST-04. TRAM-01: A PHASE 2 STUDY OF TRAMETINIB FOR PATIENTS WITH PEDIATRIC GLIOMA WITH ACTIVATION OF THE MAPK/ERK PATHWAY
- Author
-
Serge Sultan, Craig Erker, Lucie Lafay-Cousin, Cynthia Hawkins, Nada Jabado, Uri Tabori, Luis H. Ospina, Juliette Hukin, Geneviève Legault, Marie-Eve Routhier, Sébastien Perreault, Maxime Caru, Benjamin Ellezam, Eric Bouffet, Valerie Larouche, Édith Cantin, Sarah Lippé, Marie-Élaine Métras, Jean-Claude Décarie, Stephanie Vairy, Leandra Desjardins, and Yves Théorêt
- Subjects
Trametinib ,MAPK/ERK pathway ,business.industry ,Pediatric glioma ,Cancer research ,Medicine ,Phases of clinical research ,business - Abstract
BACKGROUND Pediatric low-grade gliomas (PLGG) are the most frequent brain tumors in children. It is known that the majority of PLGG have activation of the MAPK/ERK pathway. METHODS This ongoing multicenter phase II trial includes three progressing/refractory PLGG groups: NF1 patients, KIAA1549-BRAF fusion patients and patients with other activation of the MAPK/ERK pathway (excluding V600E). The primary objective was to evaluate the overall response rate based on RANO criteria after daily oral trametinib administration for 18 cycles, lasting 28 days each. Secondary objectives include the assessment of progression-free survival, tolerability of trametinib, serum levels of trametinib and quality of life evaluation during treatment. RESULTS As of February 12 2021, 50 patients have been enrolled (NF1: n=10; KIAA1549-BRAF fusion: n=31; other: n=9 including 5 patients with FGFR1 alterations). Median age is 8.8 years (range 2.4-25.5). Median follow-up is 17.5 months (range 4.7-28.5). Forty-three patients are evaluable. The overall response includes: 4 partial response (PR) (9%), 18 minor response (MR) (42%), 17 stable disease (40%), 4 progressive disease (9%). Median time to response is 5.5 months (range 2.4-13.8). Median duration of response is 6.1 months (range 0.6-26.5). Progression free survival at 12 months is 79.9% (95% CI 68.5-93.6%) and median progression free survival has not yet been reached. Treatment was discontinued for 30 patients: 16 after completing 18 cycles as planned, 5 for progressive disease, 5 for adverse events, 4 for other reasons. A total of 8 patients progressed after discontinuation of treatment including 6 patients (37.5%) that completed 18 cycles. Five of these patients had achieved minor response prior to discontinuation. CONCLUSION Trametinib is a potentially effective targeted therapy for patients with recurrent/refractory PLGG. Treatment was overall well tolerated. This ongoing trial will continue to gather data on response rate, duration of response and safety of trametinib for PLGG.
- Published
- 2021
8. NCOG-47. CAN YOUNG CHILDREN WITH RELAPSED MEDULLOBLASTOMA BE SALVAGED AFTER INITIAL IRRADIATION-SPARING APPROACHES?
- Author
-
Kathleen Dorris, Eric Sandler, Lucie Lafay-Cousin, Shahrad Rod Rassekh, Darren Klawinski, Bruce Crooks, Anna L Hoppman, Ashley Margol, Taryn B. Fay-McClymont, Beverly Wilson, Girish Dhall, Jonathan L. Finlay, Dolly Aguilera, Eric Bouffet, Mohamed S. AbdelBaki, Chantel Cacciotti, Virginia L Harrod, Taylor Holly, Juliette Hukin, Vijay Ramaswamy, David D. Eisenstat, Valerie Larouche, Craig Erker, Kenneth J. Cohen, Sébastien Perreault, Jeffrey Knipstein, and Ralph Salloum
- Subjects
Medulloblastoma ,Oncology ,Cancer Research ,medicine.medical_specialty ,Emotional vulnerability ,business.industry ,medicine.medical_treatment ,Relapsed Medulloblastoma ,Salvage therapy ,medicine.disease ,Chemotherapy regimen ,Craniospinal Irradiation ,Radiation therapy ,Ototoxicity ,Internal medicine ,Medicine ,Neurology (clinical) ,business ,Outcome Measures and Neuro-Cognitive Outcomes - Abstract
INTRODUCTION Irradiation-sparing approaches are used in young children with medulloblastoma (MB) given the vulnerability of the developing brain to neurocognitive impairment. Limited data are available following relapse for these patients. We aimed to describe the management and outcomes of young children with MB who relapsed after initial treatment without craniospinal irradiation (CSI). METHODS International retrospective study including patients with MB diagnosed between 1995-2017, ≤ 72 months old, initially treated without CSI, who subsequently relapsed. RESULTS Data are available for 66 patients. The median age at initial diagnosis was 27 months (range, 6-72). At diagnosis, 27 patients had metastatic disease. Initial therapy included conventional chemotherapy or with high-dose chemotherapy (HDC) in 30 and 36 patients, respectively. Eight (12.1%) received upfront focal irradiation. Molecular subgrouping was available for 27 (41%) patients. Ten were SHH, five group 3, six group 4 and six others were non-WNT/non-SHH. The median time from initial diagnosis to relapse was 13 months (range, 3-63). Relapse was local, disseminated, or combined in 39%, 32%, and 29%, respectively. The median time to death from relapse was 18 months. Curative intent therapy was given in 53 patients with irradiation (81%), conventional chemotherapy without HDC (40%), and HDC (25%). For patients who received irradiation, 85% received CSI (median dose 33 Gy, 18-41.4) and 15% focal irradiation. Ten patients received chemotherapy without salvage irradiation. The median follow-up time was 44 months (range, 4-255), 33 (62%) patients who underwent curative-intent therapy were alive, including 8/10 SHH, 2/3 group 3, 2/6 group 4, and 4/5 non-WNT/non-SHH. Three of four patients with SHH and treated without salvage radiotherapy are survivors. The 5-year OS for curative intent was 70%. CONCLUSION A substantial proportion of young children who relapse following irradiation-sparing strategies can be salvaged. A proportion of children with SHH MB can be salvaged without salvage radiotherapy.
- Published
- 2020
9. PDCT-08. SUPERIOR OUTCOME FOR BRAF V600E PEDIATRIC GLIOMAS TREATED WITH TARGETED BRAF INHIBITION
- Author
-
Diana S Osorio, Scott Ryall, Cynthia Hawkins, Helen Toledano, Ute Bartels, Ira J. Dunkel, Til Milde, Julia Balaguer Guill, Uri Tabori, Tara McKeown, Andres Morales La Madrid, Elizabeth Finch, Palma Solano, Magnus Sabel, Michal Zapotocky, Murali Chintagumpala, Zdenek Pavelka, Sarah Leary, Karen Gauvain, Frank Lin, Alvaro Lassaletta, David Sumerauer, Anne Grete Bechensteen, Jonathan L. Finlay, Matthias A. Karajannis, Gurcharanjeet Kaur, Daniel R. Boue, Cornelis M. van Tilburg, Ofelia Cruz, Jack Su, Eric Bouffet, Peter B. Dirks, Roger J. Packer, Bev Wilson, Miriam Bornhorst, Duarte Salgado, Peter Hauser, Maria Luisa Garrè, Julie Bennett, Naureen Mushtaq, Vijay Ramaswamy, Jordan R. Hansford, Annie Huang, Helena Mörse, Sonika Dahiya, Lorena V Baroni, Liana Nobre, Didier Frappaz, David D. Eisenstat, and Valerie Larouche
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,Pediatric Clinical Trials ,business.industry ,medicine.medical_treatment ,medicine.disease ,Chemotherapy regimen ,Discontinuation ,CDKN2A ,Concomitant ,Internal medicine ,Glioma ,medicine ,Neurology (clinical) ,Progression-free survival ,business ,Prospective cohort study - Abstract
BACKGROUND Children with pediatric low grade glioma’s (PLGG) harboring BRAF V600E mutation have poor outcome due to relative resistance to chemo-radiation and higher risk of malignant transformation. However, the role of targeted BRAF inhibition in these tumors is poorly defined. METHODS We assembled an international cohort of children with BRAF V600E mutant gliomas treated with BRAF inhibition, from 29 centers participating in the PLGG taskforce, and collected response, survival and molecular parameters. RESULTS Sixty-seven patients were treated with BRAFi (56 PLGG and 11 high grade gliomas) for a median time of 17.4 months (6 – 61 months), with 13 PLGG treated upfront. Objective responses was observed in 80% of PLGG patients compared to 28% with conventional chemotherapy (p< 0.001). Rapid responses were observed in most PLGG patients (median of 4 months), sustained in 86% of tumors up to 5 years while on therapy. In contrast, only 36% of PHGG responded to BRAFi with all but one tumor progressing within 18 months. Seventeen patients with PLGG discontinued BRAFi and 76.5% (13/17) progressed rapidly after discontinuation (median time 2.3 months). However, upon re-challenge with BRAFi therapy, 90% achieved an objective response. Poor prognostic factors to conventional therapies such as concomitant homozygous deletion of CDKN2A or H3K27M mutation were not associated with lack of response to BRAFi. Overall these responses translated to 2-year progression-free survival of 0.636 (95%CI 0.505–0.802) and 0.43 (95% CI 0.32–0.57) for BRAFi and chemotherapy treated BRAF V600E PLGG respectively (p=0.003). CONCLUSION The use of BRAFi results in objective, robust and durable responses in BRAF V600E PLGG and is associated with favorable survival. Larger prospective studies will be required to determine appropriate regiments, and long-term functional outcomes with BRAFi therapy in childhood gliomas.
- Published
- 2019
10. HGG-20. DNA METHYLATION ANALYSIS OF HIGH-GRADE GLIOMA IN PATIENTS WITH MISMATCH REPAIR DEFICIENCIES
- Author
-
David R. Jones, Shlomi Constantini, Kristina A. Cole, David S. Ziegler, Jagadeesh Ramdas, Roula Farah, Gregory Thomas, Maura Massimino, Uri Tabori, Enrico Opocher, Melissa Edwards, Gary Mason, Warren P. Mason, David Samuel, Michal Oren, Duncan Stearns, Vanan Magimairajan, Brittany Campbell, An Van Damme, Michael J. Sullivan, Patrick Tomboc, Alexander Lossos, Bruce Crooks, Richard Saffery, Stefano Chiaravalli, Andrew Dodgshun, Alexandra Sexton-Oates, Jordan R. Hansford, Kohei Fukuoka, Ashraf Shamvil, Syed Ahmer Hamid, Scott Lindhorst, and Valerie Larouche
- Subjects
Cancer Research ,Mutation ,biology ,Methylation ,medicine.disease_cause ,Abstracts ,Histone ,Mitotic cell cycle ,Oncology ,CpG site ,DNA methylation ,medicine ,biology.protein ,Cancer research ,DNA mismatch repair ,Neurology (clinical) ,Gene - Abstract
Patients with constitutional mismatch repair deficiency (CMMRD) are prone to developing high-grade glioma (HGG). These tumours acquire DNA polymerase mutations and become ultra-hypermutant harbouring hundreds of mutations per megabase. The impact of these mutations on methylation profile and the ability of the tool to differentiate MMRD tumours from others is unknown. In order to answer these questions, we performed either 450k/850K methylation analysis on a cohort of 52 CMMRD-HGG and compared them to 148 non-CMMRD HGG and normal brain controls. CMMRD HGG harbouring classic mutations in histone 3 or IDH genes had a methylation profile which clustered closely with non-MMRD tumours harbouring these mutations. Tumours without these alterations exhibited a tendency to hypomethylation with some tumours being extremely hypomethylated in comparison to other HGG. Hypomethylation was unrelated to mutational burden and type of DNA polymerase mutation present. Gene set analysis of methylation patterns revealed enrichment of hypomethylation for cellular pathways involved in cellular metabolism, organelle maintenance, mitotic cell cycle and gene expression. This pattern persisted in subgroup analysis of IDH mutant tumours in patients with and without MMRD. Importantly, this pattern was present in MMRD HGG with mutational burdens
- Published
- 2018
11. LGG-16. PREDICTORS OF OUTCOME IN BRAF-V600E PEDIATRIC GLIOMAS TREATED WITH BRAF INHIBITORS: A REPORT FROM THE PLGG TASKFORCE
- Author
-
Vijay Ramaswamy, Miriam Bornhorst, Murali Chintagumpala, Andres Morales La Madrid, Frank van Landeghem, Maria Luisa Garrè, Abhi Bavle, Palma Solano, Bev Wilson, Sarah Leary, Olaf Witt, Liana Nobre, Jean M. Mulcahy Levy, Nicholas K. Foreman, Naureen Mushtaq, Jack Su, Jordan R. Hansford, Diana S Osorio, Julia Balaguer Guill, David D. Eisenstat, Mariana Fernandes, Cornelis Vantilburg, Gurcharanjeet Kaur, Julie Bennett, Ana Guerreiro Stucklin, Valerie Larouche, Sabine Mueller, Till Milde, Inga Harting, Zdenek Pavelka, Scott Ryall, Eduardo Quiroga-Cantero, Jaroslav Sterba, Josef Zamecnik, Helena Mörse, Michal Zapotocky, Lenka Krskova, Anne Grete Bechensteen, Valentina Iurilli, Eric Bouffet, Didier Frappaz, Michael D. Taylor, Elisabeth Finch, Adela Misove, Uri Tabori, Sonika Dahiya, Cynthia Hawkins, Alvaro Lassaletta, Nisreen Amayiri, Peter Hauser, Tara McKeown, Ofelia Cruz, Samatha Mascelli, Scott L. Coven, Magnus Sabel, Adela Cañete, Lorena Baroni, Helen Toledano, Roger J. Packer, Sarah Injac, Ute Bartels, Jonathan L. Finlay, David Sumerauer, Cecile Faure Conter, Karen Gauvain, David W. Ellison, Peter B. Dirks, Theodore Nicolaides, Duarte Salgado, Daniel Alderete, and Matthias A. Karajannis
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Low Grade Glioma ,medicine.disease ,Chemotherapy regimen ,BRAF V600E ,Internal medicine ,Glioma ,Mutation (genetic algorithm) ,Medicine ,Low-Grade Glioma ,Neurology (clinical) ,business ,neoplasms - Abstract
The BRAF-V600E mutation is found in 15–20% of pediatric low grade gliomas (PLGG) and result in worse outcome and higher risk of transformation to high grade gliomas (PHGG). Although ongoing trials are assessing the role of BRAF inhibitors (BRAFi) in these children, data are still limited. We aimed to report overall response rates and predictors of outcome in childhood BRAF-V600E gliomas. We collected clinical, imaging and molecular information of patients treated with BRAFi outside trials from centers participating in the PLGG taskforce. Response was calculated by RANO criteria and follow up data were collected for all patients. Sixty-six patients were treated with BRAFi (55 PLGG and 11 PHGG); median follow-up time was 1.5 years (0.1-5y). In PLGG, objective response (tumor reduction of >25%) was observed in 77% compared to 15% in a cohort treated with conventional chemotherapy (pCDKN2A deletion was not associated with lack of response, while specific enhancing patterns correlated strongly with response to BRAFi. Two-year PFS for the BRAF-V600E PLGG was 74% vs 47% for BRAFi vs chemotherapy, respectively (p=0.02). Our data reveal rapid, dramatic and sustained response of BRAF-V600E PLGG to BRAFi. These are in contrast to BRAF-V600E PHGG and non-enhancing PLGG. Additional molecular analyses are being performed to identify poor responders and emerging mechanisms of resistance in these tumors.
- Published
- 2019
12. LGG-59. REMARKABLE OBJECTIVE RESPONSE AND FAVORABLE SURVIVAL FOR BRAF-V600E CHILDHOOD LOW-GRADE GLIOMAS TO BRAF INHIBITORS COMPARED CONVENTIONAL CHEMOTHERAPY
- Author
-
Sabine Mueller, Didier Frappaz, Scott Ryall, Plgg taskforce, Sonika Dahiya, Cynthia Hawkins, Adela Cañete, Matthias A. Karajannis, Ana Guerreiro Stucklin, Zdenek Pavelka, Palma Solano, Anne Grete Bechensteen, Vijay Ramaswamy, Jonathan L. Finlay, Peter B. Dirks, Kohei Fukuoka, Michal Zapotocky, David D. Eisenstat, Jack Su, David W. Ellison, Valerie Larouche, Eric Bouffet, Theodore Nicolaides, Maria Luisa Garrè, Sarah Leary, Jean M. Mulcahy Levy, Alvaro Lassaletta, Nisreen Amayiri, Peter Hauser, Julie Bennett, Helena Mörse, Ofelia Cruz, Tara McKeown, Elisabeth Finch, Jordan R. Hansford, Helen Toledano, Ute Bartels, David Sumerauer, Uri Tabori, and Magnus Sabel
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,endocrine system diseases ,business.industry ,digestive system diseases ,BRAF V600E ,Abstracts ,Internal medicine ,medicine ,Conventional chemotherapy ,Neurology (clinical) ,business ,neoplasms ,Objective response - Abstract
Activation of the MAPK pathway represents a hallmark of pediatric low-grade glioma (pLGG) and is frequently caused by BRAF alterations. BRAF-V600E represent an aggressive type of pLGG with less than optimal response to conventional chemo-radiation approaches. While clinical trials using BRAF-V600E inhibitors are ongoing, these data are not yet available. We have assembled an international cohort of BRAF-V600E glioma patients treated off-label with BRAF inhibitors as a monotherapy. Complete molecular, clinical and imaging data is being collected and compared to previous chemo-radiation therapies. Ongoing data form the taskforce on 40 BRAF-V600E gliomas from 25 international institutions is summarized below. The most prevalent histologies were ganglioglioma, pilocytic astrocytoma and pleomorphic xanthoastrocytoma, located mainly in the chiasm, brainstem and temporal lobes. Strikingly, 66% of BRAF V600E pLGG patients achieved partial response (PR) to targeted inhibitors versus only 6.6% response to conventional chemotherapy (p
- Published
- 2018
13. AT-03ATYPICAL TERATOID RHABDOID TUMORS IN THE FIRST YEAR OF LIFE: SHOULD WE TREAT?
- Author
-
Donna L. Johnston, Cynthia Hawkins, Lucie Lafay-Cousin, Haocheng Li, Anne-Sophie Carret, Douglas Strother, Shayna Zelcer, Katrin Scheinemann, Adam Fleming, Tanya Renae Brown, David D. Eisenstat, Mary Fossey, Valerie Larouche, Eric Bouffet, Nada Jabado, Beverly Wilson, Annie Huang, Chris Fryer, and Josee Brossard
- Subjects
Oncology ,Abstracts ,Cancer Research ,medicine.medical_specialty ,Text mining ,business.industry ,Internal medicine ,Rhabdoid tumors ,medicine ,First year of life ,Neurology (clinical) ,business - Published
- 2016
14. HG-53HYPERMUTATION AND NEOANTIGEN FORMATION PREDICT RESPONSE TO IMMUNE CHECKPOINT INHIBITION IN CHILDHOOD BIALLELIC MISMATCH REPAIR DEFICIENT GLIOBLASTOMA
- Author
-
Sunil J. Patel, Roula Farah, Michael Osborn, Alyssa Reddy, Daniele Merico, Melyssa Aronson, Nancy Klauber-DeMore, Rachel Laframboise, David Malkin, Gideon Rechavi, Jordan R. Hansford, Vijay Ramaswamy, Shlomi Constantini, Joerg Kruger, Ronit Elhasid, Richard de Borja, Carol Durno, Jeffrey Atkinson, Uri Tabori, Nataliya Zhukova, Lindsay L. Peterson, Roy W. R. Dudley, Valerie Larouche, Nada Jabado, Gary Mason, Vanan Magimairan, Scott Lindhorst, Michal Yalon, Samina Afzal, Eric Bouffet, Andrew Dodgshun, Melissa Galati, Rina Dvir, Michael Walsh, Brittany Campbell, Brian Hy Chung, Peter B. Dirks, Cynthia Hawkins, Michael D. Taylor, Michael J. Sullivan, Adam Shlien, Zane Cohen, Vanja Cabric, and Steffen Albrecht
- Subjects
Abstracts ,Cancer Research ,Cell cycle checkpoint ,Oncology ,Immunology ,medicine ,Cancer research ,DNA mismatch repair ,Neurology (clinical) ,Biology ,medicine.disease ,Immune checkpoint ,Glioblastoma - Published
- 2016
15. Case 2: An adolescent refugee with lower-extremity pain and weakness
- Author
-
Sébastien Bergeron, Richard E. Bélanger, and Valerie Larouche
- Subjects
medicine.medical_specialty ,Weakness ,Physical medicine and rehabilitation ,business.industry ,Refugee ,Pediatrics, Perinatology and Child Health ,Physical therapy ,Medicine ,medicine.symptom ,business ,Clinician’s Corner ,Lower extremity pain - Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.