25 results on '"Northcott, Paul A"'
Search Results
2. Clinical Outcomes and Patient-Matched Molecular Composition of Relapsed Medulloblastoma.
- Author
-
Kumar, Rahul, Smith, Kyle, Deng, Maximilian, Terhune, Colt, Robinson, Giles, Orr, Brent, Liu, Anthony, Lin, Tong, Billups, Catherine, Chintagumpala, Murali, Bowers, Daniel, Hassall, Timothy, Hansford, Jordan, Khuong-Quang, Dong, Crawford, John, Bendel, Anne, Gururangan, Sridharan, Schroeder, Kristin, Bouffet, Eric, Bartels, Ute, Fisher, Michael, Cohn, Richard, Partap, Sonia, Kellie, Stewart, McCowage, Geoffrey, Paulino, Arnold, Rutkowski, Stefan, Fleischhack, Gudrun, Dhall, Girish, Klesse, Laura, Leary, Sarah, Nazarian, Javad, Kool, Marcel, Wesseling, Pieter, Ryzhova, Marina, Zheludkova, Olga, Golanov, Andrey, McLendon, Roger, Packer, Roger, Dunham, Christopher, Hukin, Juliette, Fouladi, Maryam, Faria, Claudia, Pimentel, Jose, Walter, Andrew, Jabado, Nada, Cho, Yoon-Jae, Perreault, Sebastien, Croul, Sidney, Zapotocky, Michal, Hawkins, Cynthia, Tabori, Uri, Taylor, Michael, Pfister, Stefan, Klimo, Paul, Boop, Frederick, Ellison, David, Merchant, Thomas, Onar-Thomas, Arzu, Korshunov, Andrey, Jones, David, Gajjar, Amar, Ramaswamy, Vijay, and Northcott, Paul
- Subjects
Biomarkers ,Tumor ,Cerebellar Neoplasms ,Child ,Child ,Preschool ,Clinical Trials as Topic ,DNA Methylation ,Disease Progression ,Epigenome ,Epigenomics ,Female ,High-Throughput Nucleotide Sequencing ,Humans ,Infant ,Male ,Medulloblastoma ,Neoplasm Recurrence ,Local ,Retreatment ,Time Factors ,Treatment Outcome - Abstract
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
3. Pattern of Relapse and Treatment Response in WNT-Activated Medulloblastoma
- Author
-
Nobre, Liana, Zapotocky, Michal, Khan, Sara, Fukuoka, Kohei, Fonseca, Adriana, McKeown, Tara, Sumerauer, David, Vicha, Ales, Grajkowska, Wieslawa A, Trubicka, Joanna, Li, Kay Ka Wai, Ng, Ho-Keung, Massimi, Luca, Lee, Ji Yeoun, Kim, Seung-Ki, Zelcer, Shayna, Vasiljevic, Alexandre, Faure-Conter, Cécile, Hauser, Peter, Lach, Boleslaw, van Veelen-Vincent, Marie-Lise, French, Pim J, Van Meir, Erwin G, Weiss, William A, Gupta, Nalin, Pollack, Ian F, Hamilton, Ronald L, Rao, Amulya A Nageswara, Giannini, Caterina, Rubin, Joshua B, Moore, Andrew S, Chambless, Lola B, Vibhakar, Rajeev, Ra, Young Shin, Massimino, Maura, McLendon, Roger E, Wheeler, Helen, Zollo, Massimo, Ferruci, Veronica, Kumabe, Toshihiro, Faria, Claudia C, Sterba, Jaroslav, Jung, Shin, López-Aguilar, Enrique, Mora, Jaume, Carlotti, Carlos G, Olson, James M, Leary, Sarah, Cain, Jason, Krskova, Lenka, Zamecnik, Josef, Hawkins, Cynthia E, Tabori, Uri, Huang, Annie, Bartels, Ute, Northcott, Paul A, Taylor, Michael D, Yip, Stephen, Hansford, Jordan R, Bouffet, Eric, and Ramaswamy, Vijay
- Subjects
Cancer ,Clinical Research ,Evaluation of treatments and therapeutic interventions ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,6.1 Pharmaceuticals ,Adolescent ,Antineoplastic Combined Chemotherapy Protocols ,Biomarkers ,Tumor ,Cerebellar Neoplasms ,Child ,Cyclophosphamide ,Female ,Humans ,Ifosfamide ,Male ,Medulloblastoma ,Middle Aged ,Neoplasm Recurrence ,Local ,Progression-Free Survival ,medulloblastoma ,WNT ,genomics ,cyclophosphamide ,chemotherapy ,prognosis ,survival - Abstract
Over the past decade, wingless-activated (WNT) medulloblastoma has been identified as a candidate for therapy de-escalation based on excellent survival; however, a paucity of relapses has precluded additional analyses of markers of relapse. To address this gap in knowledge, an international cohort of 93 molecularly confirmed WNT MB was assembled, where 5-year progression-free survival is 0.84 (95%, 0.763-0.925) with 15 relapsed individuals identified. Maintenance chemotherapy is identified as a strong predictor of relapse, with individuals receiving high doses of cyclophosphamide or ifosphamide having only one very late molecularly confirmed relapse (p = 0.032). The anatomical location of recurrence is metastatic in 12 of 15 relapses, with 8 of 12 metastatic relapses in the lateral ventricles. Maintenance chemotherapy, specifically cumulative cyclophosphamide doses, is a significant predictor of relapse across WNT MB. Future efforts to de-escalate therapy need to carefully consider not only the radiation dose but also the chemotherapy regimen and the propensity for metastatic relapses.
- Published
- 2020
4. Germline Elongator mutations in Sonic Hedgehog medulloblastoma.
- Author
-
Robinson, Giles, Gudenas, Brian, Smith, Kyle, Forget, Antoine, Kojic, Marija, Garcia-Lopez, Jesus, Hadley, Jennifer, Hamilton, Kayla, Indersie, Emilie, Buchhalter, Ivo, Kerssemakers, Jules, Jäger, Natalie, Sharma, Tanvi, Rausch, Tobias, Kool, Marcel, Sturm, Dominik, Jones, David, Vasilyeva, Aksana, Tatevossian, Ruth, Neale, Geoffrey, Lombard, Bérangère, Loew, Damarys, Nakitandwe, Joy, Rusch, Michael, Bowers, Daniel, Bendel, Anne, Partap, Sonia, Chintagumpala, Murali, Crawford, John, Gottardo, Nicholas, Smith, Amy, Dufour, Christelle, Rutkowski, Stefan, Eggen, Tone, Wesenberg, Finn, Kjaerheim, Kristina, Feychting, Maria, Lannering, Birgitta, Schüz, Joachim, Johansen, Christoffer, Andersen, Tina, Röösli, Martin, Kuehni, Claudia, Grotzer, Michael, Remke, Marc, Puget, Stéphanie, Pajtler, Kristian, Milde, Till, Witt, Olaf, Ryzhova, Marina, Korshunov, Andrey, Orr, Brent, Ellison, David, Brugieres, Laurence, Lichter, Peter, Nichols, Kim, Gajjar, Amar, Wainwright, Brandon, Ayrault, Olivier, Korbel, Jan, Northcott, Paul, Pfister, Stefan, and Waszak, Sebastian
- Subjects
Cerebellar Neoplasms ,Child ,Female ,Germ-Line Mutation ,Humans ,Male ,Medulloblastoma ,Pedigree ,RNA ,Transfer ,Transcriptional Elongation Factors - Abstract
Cancer genomics has revealed many genes and core molecular processes that contribute to human malignancies, but the genetic and molecular bases of many rare cancers remains unclear. Genetic predisposition accounts for 5 to 10% of cancer diagnoses in children1,2, and genetic events that cooperate with known somatic driver events are poorly understood. Pathogenic germline variants in established cancer predisposition genes have been recently identified in 5% of patients with the malignant brain tumour medulloblastoma3. Here, by analysing all protein-coding genes, we identify and replicate rare germline loss-of-function variants across ELP1 in 14% of paediatric patients with the medulloblastoma subgroup Sonic Hedgehog (MBSHH). ELP1 was the most common medulloblastoma predisposition gene and increased the prevalence of genetic predisposition to 40% among paediatric patients with MBSHH. Parent-offspring and pedigree analyses identified two families with a history of paediatric medulloblastoma. ELP1-associated medulloblastomas were restricted to the molecular SHHα subtype4 and characterized by universal biallelic inactivation of ELP1 owing to somatic loss of chromosome arm 9q. Most ELP1-associated medulloblastomas also exhibited somatic alterations in PTCH1, which suggests that germline ELP1 loss-of-function variants predispose individuals to tumour development in combination with constitutive activation of SHH signalling. ELP1 is the largest subunit of the evolutionarily conserved Elongator complex, which catalyses translational elongation through tRNA modifications at the wobble (U34) position5,6. Tumours from patients with ELP1-associated MBSHH were characterized by a destabilized Elongator complex, loss of Elongator-dependent tRNA modifications, codon-dependent translational reprogramming, and induction of the unfolded protein response, consistent with loss of protein homeostasis due to Elongator deficiency in model systems7-9. Thus, genetic predisposition to proteome instability may be a determinant in the pathogenesis of paediatric brain cancers. These results support investigation of the role of protein homeostasis in other cancer types and potential for therapeutic interference.
- Published
- 2020
5. Risk-adapted therapy and biological heterogeneity in pineoblastoma: integrated clinico-pathological analysis from the prospective, multi-center SJMB03 and SJYC07 trials
- Author
-
Liu, Anthony PY, Gudenas, Brian, Lin, Tong, Orr, Brent A, Klimo, Paul, Kumar, Rahul, Bouffet, Eric, Gururangan, Sridharan, Crawford, John R, Kellie, Stewart J, Chintagumpala, Murali, Fisher, Michael J, Bowers, Daniel C, Hassall, Tim, Indelicato, Daniel J, Onar-Thomas, Arzu, Ellison, David W, Boop, Frederick A, Merchant, Thomas E, Robinson, Giles W, Northcott, Paul A, and Gajjar, Amar
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Oncology and Carcinogenesis ,Pediatric Cancer ,Genetics ,Clinical Trials and Supportive Activities ,Biotechnology ,Pediatric ,Clinical Research ,Prevention ,Cancer ,Adolescent ,Age Factors ,Brain Neoplasms ,Child ,Child ,Preschool ,Cohort Studies ,DNA Methylation ,Female ,Humans ,Male ,Pineal Gland ,Pinealoma ,Proto-Oncogene Mas ,Risk Factors ,Survival Rate ,Young Adult ,Pineoblastoma ,Clinical trial ,Molecular subgroups ,DICER1 ,MicroRNA processing ,FOXR2 ,Neurosciences ,Neurology & Neurosurgery - Abstract
Pineoblastoma is a rare embryonal tumor of childhood that is conventionally treated with high-dose craniospinal irradiation (CSI). Multi-dimensional molecular evaluation of pineoblastoma and associated intertumoral heterogeneity is lacking. Herein, we report outcomes and molecular features of children with pineoblastoma from two multi-center, risk-adapted trials (SJMB03 for patients ≥ 3 years; SJYC07 for patients
- Published
- 2020
6. Proteomics, Post-translational Modifications, and Integrative Analyses Reveal Molecular Heterogeneity within Medulloblastoma Subgroups
- Author
-
Archer, Tenley C, Ehrenberger, Tobias, Mundt, Filip, Gold, Maxwell P, Krug, Karsten, Mah, Clarence K, Mahoney, Elizabeth L, Daniel, Colin J, LeNail, Alexander, Ramamoorthy, Divya, Mertins, Philipp, Mani, DR, Zhang, Hailei, Gillette, Michael A, Clauser, Karl, Noble, Michael, Tang, Lauren C, Pierre-François, Jessica, Silterra, Jacob, Jensen, James, Tamayo, Pablo, Korshunov, Andrey, Pfister, Stefan M, Kool, Marcel, Northcott, Paul A, Sears, Rosalie C, Lipton, Jonathan O, Carr, Steven A, Mesirov, Jill P, Pomeroy, Scott L, and Fraenkel, Ernest
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Neurosciences ,Pediatric ,Pediatric Research Initiative ,Cancer ,Pediatric Cancer ,Genetics ,Rare Diseases ,Brain Cancer ,Brain Disorders ,Biotechnology ,Adolescent ,Adult ,Biomarkers ,Tumor ,Brain Neoplasms ,Cell Line ,Tumor ,Child ,Child ,Preschool ,DNA Methylation ,DNA-Activated Protein Kinase ,Female ,Gene Expression Profiling ,Hedgehog Proteins ,Humans ,Infant ,Male ,Medulloblastoma ,Nuclear Proteins ,Protein Processing ,Post-Translational ,Proteome ,Proteomics ,Proto-Oncogene Proteins c-myc ,Sequence Analysis ,RNA ,Young Adult ,MYC ,NU-7441 ,SHH ,mass spectrometry ,medulloblastoma ,multi-omics ,network integration ,phospho-proteomics ,proteo-genomics ,radio sensitization ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
There is a pressing need to identify therapeutic targets in tumors with low mutation rates such as the malignant pediatric brain tumor medulloblastoma. To address this challenge, we quantitatively profiled global proteomes and phospho-proteomes of 45 medulloblastoma samples. Integrated analyses revealed that tumors with similar RNA expression vary extensively at the post-transcriptional and post-translational levels. We identified distinct pathways associated with two subsets of SHH tumors, and found post-translational modifications of MYC that are associated with poor outcomes in group 3 tumors. We found kinases associated with subtypes and showed that inhibiting PRKDC sensitizes MYC-driven cells to radiation. Our study shows that proteomics enables a more comprehensive, functional readout, providing a foundation for future therapeutic strategies.
- Published
- 2018
7. Risk-adapted therapy for young children with medulloblastoma (SJYC07): therapeutic and molecular outcomes from a multicentre, phase 2 trial.
- Author
-
Smith, Kyle, Bowers, Daniel, Bendel, Anne, Fisher, Paul, Partap, Sonia, Crawford, John, Hassall, Tim, Indelicato, Daniel, Boop, Frederick, Klimo, Paul, Sabin, Noah, Patay, Zoltan, Merchant, Thomas, Stewart, Clinton, Orr, Brent, Korbel, Jan, Jones, David, Sharma, Tanvi, Lichter, Peter, Kool, Marcel, Korshunov, Andrey, Pfister, Stefan, Gilbertson, Richard, Sanders, Robert, Onar-Thomas, Arzu, Ellison, David, Gajjar, Amar, Northcott, Paul, Robinson, Giles, Rudneva, Vasilisa, Buchhalter, Ivo, Billups, Catherine, and Waszak, Sebastian
- Subjects
Age Factors ,Antineoplastic Combined Chemotherapy Protocols ,Australia ,Biomarkers ,Tumor ,Cerebellar Neoplasms ,Chemotherapy ,Adjuvant ,Child ,Preschool ,Clinical Decision-Making ,Cranial Irradiation ,DNA Methylation ,Gene Expression Profiling ,Humans ,Infant ,Medulloblastoma ,Neoadjuvant Therapy ,Patient Selection ,Predictive Value of Tests ,Progression-Free Survival ,Radiation Dosage ,Radiotherapy ,Adjuvant ,Risk Assessment ,Risk Factors ,Time Factors ,United States - Abstract
BACKGROUND: Young children with medulloblastoma have a poor overall survival compared with older children, due to use of radiation-sparing therapy in young children. Radiotherapy is omitted or reduced in these young patients to spare them from debilitating long-term side-effects. We aimed to estimate event-free survival and define the molecular characteristics associated with progression-free survival in young patients with medulloblastoma using a risk-stratified treatment strategy designed to defer, reduce, or delay radiation exposure. METHODS: In this multicentre, phase 2 trial, we enrolled children younger than 3 years with newly diagnosed medulloblastoma at six centres in the USA and Australia. Children aged 3-5 years with newly diagnosed, non-metastatic medulloblastoma without any high-risk features were also eligible. Eligible patients were required to start therapy within 31 days from definitive surgery, had a Lansky performance score of at least 30, and did not receive previous radiotherapy or chemotherapy. Patients were stratified postoperatively by clinical and histological criteria into low-risk, intermediate-risk, and high-risk treatment groups. All patients received identical induction chemotherapy (methotrexate, vincristine, cisplatin, and cyclophosphamide), with high-risk patients also receiving an additional five doses of vinblastine. Induction was followed by risk-adapted consolidation therapy: low-risk patients received cyclophosphamide (1500 mg/m2 on day 1), etoposide (100 mg/m2 on days 1 and 2), and carboplatin (area under the curve 5 mg/mL per min on day 2) for two 4-week cycles; intermediate-risk patients received focal radiation therapy (54 Gy with a clinical target volume of 5 mm over 6 weeks) to the tumour bed; and high-risk patients received chemotherapy with targeted intravenous topotecan (area under the curve 120-160 ng-h/mL intravenously on days 1-5) and cyclophosphamide (600 mg/m2 intravenously on days 1-5). After consolidation, all patients received maintenance chemotherapy with cyclophosphamide, topotecan, and erlotinib. The coprimary endpoints were event-free survival and patterns of methylation profiling associated with progression-free survival. Outcome and safety analyses were per protocol (all patients who received at least one dose of induction chemotherapy); biological analyses included all patients with tissue available for methylation profiling. This trial is registered with ClinicalTrials.gov, number NCT00602667, and was closed to accrual on April 19, 2017. FINDINGS: Between Nov 27, 2007, and April 19, 2017, we enrolled 81 patients with histologically confirmed medulloblastoma. Accrual to the low-risk group was suspended after an interim analysis on Dec 2, 2015, when the 1-year event-free survival was estimated to be below the stopping rule boundary. After a median follow-up of 5·5 years (IQR 2·7-7·3), 5-year event-free survival was 31·3% (95% CI 19·3-43·3) for the whole cohort, 55·3% (95% CI 33·3-77·3) in the low-risk cohort (n=23) versus 24·6% (3·6-45·6) in the intermediate-risk cohort (n=32; hazard ratio 2·50, 95% CI 1·19-5·27; p=0·016) and 16·7% (3·4-30·0) in the high-risk cohort (n=26; 3·55, 1·66-7·59; p=0·0011; overall p=0·0021). 5-year progression-free survival by methylation subgroup was 51·1% (95% CI 34·6-67·6) in the sonic hedgehog (SHH) subgroup (n=42), 8·3% (95% CI 0·0-24·0%) in the group 3 subgroup (n=24), and 13·3% (95% CI 0·0-37·6%) in the group 4 subgroup (n=10). Within the SHH subgroup, two distinct methylation subtypes were identified and named iSHH-I and iSHH-II. 5-year progression-free survival was 27·8% (95% CI 9·0-46·6; n=21) for iSHH-I and 75·4% (55·0-95·8; n=21) for iSHH-II. The most common adverse events were grade 3-4 febrile neutropenia (48 patients [59%]), neutropenia (21 [26%]), infection with neutropenia (20 [25%]), leucopenia (15 [19%]), vomiting (15 [19%]), and anorexia (13 [16%]). No treatment-related deaths occurred. INTERPRETATION: The risk-adapted approach did not improve event-free survival in young children with medulloblastoma. However, the methylation subgroup analyses showed that the SHH subgroup had improved progression-free survival compared with the group 3 subgroup. Moreover, within the SHH subgroup, the iSHH-II subtype had improved progression-free survival in the absence of radiation, intraventricular chemotherapy, or high-dose chemotherapy compared with the iSHH-I subtype. These findings support the development of a molecularly driven, risk-adapted, treatment approach in future trials in young children with medulloblastoma. FUNDING: American Lebanese Syrian Associated Charities, St Jude Childrens Research Hospital, NCI Cancer Center, Alexander and Margaret Stewart Trust, Sontag Foundation, and American Association for Cancer Research.
- Published
- 2018
8. Spectrum and prevalence of genetic predisposition in medulloblastoma: a retrospective genetic study and prospective validation in a clinical trial cohort.
- Author
-
Waszak, Sebastian M, Northcott, Paul A, Buchhalter, Ivo, Robinson, Giles W, Sutter, Christian, Groebner, Susanne, Grund, Kerstin B, Brugières, Laurence, Jones, David TW, Pajtler, Kristian W, Morrissy, A Sorana, Kool, Marcel, Sturm, Dominik, Chavez, Lukas, Ernst, Aurelie, Brabetz, Sebastian, Hain, Michael, Zichner, Thomas, Segura-Wang, Maia, Weischenfeldt, Joachim, Rausch, Tobias, Mardin, Balca R, Zhou, Xin, Baciu, Cristina, Lawerenz, Christian, Chan, Jennifer A, Varlet, Pascale, Guerrini-Rousseau, Lea, Fults, Daniel W, Grajkowska, Wiesława, Hauser, Peter, Jabado, Nada, Ra, Young-Shin, Zitterbart, Karel, Shringarpure, Suyash S, De La Vega, Francisco M, Bustamante, Carlos D, Ng, Ho-Keung, Perry, Arie, MacDonald, Tobey J, Hernáiz Driever, Pablo, Bendel, Anne E, Bowers, Daniel C, McCowage, Geoffrey, Chintagumpala, Murali M, Cohn, Richard, Hassall, Timothy, Fleischhack, Gudrun, Eggen, Tone, Wesenberg, Finn, Feychting, Maria, Lannering, Birgitta, Schüz, Joachim, Johansen, Christoffer, Andersen, Tina V, Röösli, Martin, Kuehni, Claudia E, Grotzer, Michael, Kjaerheim, Kristina, Monoranu, Camelia M, Archer, Tenley C, Duke, Elizabeth, Pomeroy, Scott L, Shelagh, Redmond, Frank, Stephan, Sumerauer, David, Scheurlen, Wolfram, Ryzhova, Marina V, Milde, Till, Kratz, Christian P, Samuel, David, Zhang, Jinghui, Solomon, David A, Marra, Marco, Eils, Roland, Bartram, Claus R, von Hoff, Katja, Rutkowski, Stefan, Ramaswamy, Vijay, Gilbertson, Richard J, Korshunov, Andrey, Taylor, Michael D, Lichter, Peter, Malkin, David, Gajjar, Amar, Korbel, Jan O, and Pfister, Stefan M
- Subjects
Humans ,Medulloblastoma ,Cerebellar Neoplasms ,Genetic Predisposition to Disease ,Risk Factors ,Retrospective Studies ,Prospective Studies ,Reproducibility of Results ,Predictive Value of Tests ,Gene Expression Profiling ,Pedigree ,DNA Mutational Analysis ,DNA Methylation ,Heredity ,Phenotype ,Germ-Line Mutation ,Models ,Genetic ,Adolescent ,Adult ,Child ,Child ,Preschool ,Infant ,Female ,Male ,Young Adult ,Genetic Testing ,Transcriptome ,Biomarkers ,Tumor ,Progression-Free Survival ,Exome Sequencing ,Brain Cancer ,Genetics ,Cancer ,Human Genome ,Pediatric ,Pediatric Cancer ,Rare Diseases ,Brain Disorders ,Aetiology ,2.1 Biological and endogenous factors ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis - Abstract
BackgroundMedulloblastoma is associated with rare hereditary cancer predisposition syndromes; however, consensus medulloblastoma predisposition genes have not been defined and screening guidelines for genetic counselling and testing for paediatric patients are not available. We aimed to assess and define these genes to provide evidence for future screening guidelines.MethodsIn this international, multicentre study, we analysed patients with medulloblastoma from retrospective cohorts (International Cancer Genome Consortium [ICGC] PedBrain, Medulloblastoma Advanced Genomics International Consortium [MAGIC], and the CEFALO series) and from prospective cohorts from four clinical studies (SJMB03, SJMB12, SJYC07, and I-HIT-MED). Whole-genome sequences and exome sequences from blood and tumour samples were analysed for rare damaging germline mutations in cancer predisposition genes. DNA methylation profiling was done to determine consensus molecular subgroups: WNT (MBWNT), SHH (MBSHH), group 3 (MBGroup3), and group 4 (MBGroup4). Medulloblastoma predisposition genes were predicted on the basis of rare variant burden tests against controls without a cancer diagnosis from the Exome Aggregation Consortium (ExAC). Previously defined somatic mutational signatures were used to further classify medulloblastoma genomes into two groups, a clock-like group (signatures 1 and 5) and a homologous recombination repair deficiency-like group (signatures 3 and 8), and chromothripsis was investigated using previously established criteria. Progression-free survival and overall survival were modelled for patients with a genetic predisposition to medulloblastoma.FindingsWe included a total of 1022 patients with medulloblastoma from the retrospective cohorts (n=673) and the four prospective studies (n=349), from whom blood samples (n=1022) and tumour samples (n=800) were analysed for germline mutations in 110 cancer predisposition genes. In our rare variant burden analysis, we compared these against 53 105 sequenced controls from ExAC and identified APC, BRCA2, PALB2, PTCH1, SUFU, and TP53 as consensus medulloblastoma predisposition genes according to our rare variant burden analysis and estimated that germline mutations accounted for 6% of medulloblastoma diagnoses in the retrospective cohort. The prevalence of genetic predispositions differed between molecular subgroups in the retrospective cohort and was highest for patients in the MBSHH subgroup (20% in the retrospective cohort). These estimates were replicated in the prospective clinical cohort (germline mutations accounted for 5% of medulloblastoma diagnoses, with the highest prevalence [14%] in the MBSHH subgroup). Patients with germline APC mutations developed MBWNT and accounted for most (five [71%] of seven) cases of MBWNT that had no somatic CTNNB1 exon 3 mutations. Patients with germline mutations in SUFU and PTCH1 mostly developed infant MBSHH. Germline TP53 mutations presented only in childhood patients in the MBSHH subgroup and explained more than half (eight [57%] of 14) of all chromothripsis events in this subgroup. Germline mutations in PALB2 and BRCA2 were observed across the MBSHH, MBGroup3, and MBGroup4 molecular subgroups and were associated with mutational signatures typical of homologous recombination repair deficiency. In patients with a genetic predisposition to medulloblastoma, 5-year progression-free survival was 52% (95% CI 40-69) and 5-year overall survival was 65% (95% CI 52-81); these survival estimates differed significantly across patients with germline mutations in different medulloblastoma predisposition genes.InterpretationGenetic counselling and testing should be used as a standard-of-care procedure in patients with MBWNT and MBSHH because these patients have the highest prevalence of damaging germline mutations in known cancer predisposition genes. We propose criteria for routine genetic screening for patients with medulloblastoma based on clinical and molecular tumour characteristics.FundingGerman Cancer Aid; German Federal Ministry of Education and Research; German Childhood Cancer Foundation (Deutsche Kinderkrebsstiftung); European Research Council; National Institutes of Health; Canadian Institutes for Health Research; German Cancer Research Center; St Jude Comprehensive Cancer Center; American Lebanese Syrian Associated Charities; Swiss National Science Foundation; European Molecular Biology Organization; Cancer Research UK; Hertie Foundation; Alexander and Margaret Stewart Trust; V Foundation for Cancer Research; Sontag Foundation; Musicians Against Childhood Cancer; BC Cancer Foundation; Swedish Council for Health, Working Life and Welfare; Swedish Research Council; Swedish Cancer Society; the Swedish Radiation Protection Authority; Danish Strategic Research Council; Swiss Federal Office of Public Health; Swiss Research Foundation on Mobile Communication; Masaryk University; Ministry of Health of the Czech Republic; Research Council of Norway; Genome Canada; Genome BC; Terry Fox Research Institute; Ontario Institute for Cancer Research; Pediatric Oncology Group of Ontario; The Family of Kathleen Lorette and the Clark H Smith Brain Tumour Centre; Montreal Children's Hospital Foundation; The Hospital for Sick Children: Sonia and Arthur Labatt Brain Tumour Research Centre, Chief of Research Fund, Cancer Genetics Program, Garron Family Cancer Centre, MDT's Garron Family Endowment; BC Childhood Cancer Parents Association; Cure Search Foundation; Pediatric Brain Tumor Foundation; Brainchild; and the Government of Ontario.
- Published
- 2018
9. Deep sequencing of WNT-activated medulloblastomas reveals secondary SHH pathway activation
- Author
-
Iorgulescu, J Bryan, Van Ziffle, Jessica, Stevers, Meredith, Grenert, James P, Bastian, Boris C, Chavez, Lukas, Stichel, Damian, Buchhalter, Ivo, Samuel, David, Nicolaides, Theodore, Banerjee, Anuradha, Mueller, Sabine, Gupta, Nalin, Tihan, Tarik, Bollen, Andrew W, Northcott, Paul A, Kool, Marcel, Pfister, Stefan, Korshunov, Andrey, Perry, Arie, and Solomon, David A
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Adolescent ,Adult ,Cerebellar Neoplasms ,Child ,Cohort Studies ,Disease Progression ,Female ,Gene Frequency ,Hedgehog Proteins ,High-Throughput Nucleotide Sequencing ,Humans ,Male ,Medulloblastoma ,Mutation ,Signal Transduction ,Wnt Proteins ,Young Adult ,Clinical Sciences ,Neurology & Neurosurgery - Published
- 2018
10. Prognostic value of medulloblastoma extent of resection after accounting for molecular subgroup: a retrospective integrated clinical and molecular analysis
- Author
-
Thompson, Eric M, Hielscher, Thomas, Bouffet, Eric, Remke, Marc, Luu, Betty, Gururangan, Sridharan, McLendon, Roger E, Bigner, Darell D, Lipp, Eric S, Perreault, Sebastien, Cho, Yoon-Jae, Grant, Gerald, Kim, Seung-Ki, Lee, Ji Yeoun, Rao, Amulya A Nageswara, Giannini, Caterina, Li, Kay Ka Wai, Ng, Ho-Keung, Yao, Yu, Kumabe, Toshihiro, Tominaga, Teiji, Grajkowska, Wieslawa A, Perek-Polnik, Marta, Low, David CY, Seow, Wan Tew, Chang, Kenneth TE, Mora, Jaume, Pollack, Ian F, Hamilton, Ronald L, Leary, Sarah, Moore, Andrew S, Ingram, Wendy J, Hallahan, Andrew R, Jouvet, Anne, Fèvre-Montange, Michelle, Vasiljevic, Alexandre, Faure-Conter, Cecile, Shofuda, Tomoko, Kagawa, Naoki, Hashimoto, Naoya, Jabado, Nada, Weil, Alexander G, Gayden, Tenzin, Wataya, Takafumi, Shalaby, Tarek, Grotzer, Michael, Zitterbart, Karel, Sterba, Jaroslav, Kren, Leos, Hortobágyi, Tibor, Klekner, Almos, László, Bognár, Pócza, Tímea, Hauser, Peter, Schüller, Ulrich, Jung, Shin, Jang, Woo-Youl, French, Pim J, Kros, Johan M, van Veelen, Marie-Lise C, Massimi, Luca, Leonard, Jeffrey R, Rubin, Joshua B, Vibhakar, Rajeev, Chambless, Lola B, Cooper, Michael K, Thompson, Reid C, Faria, Claudia C, Carvalho, Alice, Nunes, Sofia, Pimentel, José, Fan, Xing, Muraszko, Karin M, López-Aguilar, Enrique, Lyden, David, Garzia, Livia, Shih, David JH, Kijima, Noriyuki, Schneider, Christian, Adamski, Jennifer, Northcott, Paul A, Kool, Marcel, Jones, David TW, Chan, Jennifer A, Nikolic, Ana, Garre, Maria Luisa, Van Meir, Erwin G, Osuka, Satoru, Olson, Jeffrey J, Jahangiri, Arman, Castro, Brandyn A, Gupta, Nalin, Weiss, William A, Moxon-Emre, Iska, Mabbott, Donald J, Lassaletta, Alvaro, Hawkins, Cynthia E, Tabori, Uri, Drake, James, and Kulkarni, Abhaya
- Subjects
Biomedical and Clinical Sciences ,Cardiovascular Medicine and Haematology ,Clinical Sciences ,Oncology and Carcinogenesis ,Brain Disorders ,Rare Diseases ,Brain Cancer ,Pediatric Research Initiative ,Pediatric ,Cancer ,Adult ,Brain Neoplasms ,Canada ,Child ,Child ,Preschool ,Combined Modality Therapy ,Disease Progression ,Disease-Free Survival ,Female ,Humans ,Infant ,Magnetic Resonance Imaging ,Male ,Medulloblastoma ,Prognosis ,Retrospective Studies ,Oncology & Carcinogenesis ,Oncology and carcinogenesis - Abstract
BackgroundPatients with incomplete surgical resection of medulloblastoma are controversially regarded as having a marker of high-risk disease, which leads to patients undergoing aggressive surgical resections, so-called second-look surgeries, and intensified chemoradiotherapy. All previous studies assessing the clinical importance of extent of resection have not accounted for molecular subgroup. We analysed the prognostic value of extent of resection in a subgroup-specific manner.MethodsWe retrospectively identified patients who had a histological diagnosis of medulloblastoma and complete data about extent of resection and survival from centres participating in the Medulloblastoma Advanced Genomics International Consortium. We collected from resections done between April, 1997, and February, 2013, at 35 international institutions. We established medulloblastoma subgroup affiliation by gene expression profiling on frozen or formalin-fixed paraffin-embedded tissues. We classified extent of resection on the basis of postoperative imaging as gross total resection (no residual tumour), near-total resection (30 Gy vs no craniospinal irradiation). The primary analysis outcome was the effect of extent of resection by molecular subgroup and the effects of other clinical variables on overall and progression-free survival.FindingsWe included 787 patients with medulloblastoma (86 with WNT tumours, 242 with SHH tumours, 163 with group 3 tumours, and 296 with group 4 tumours) in our multivariable Cox models of progression-free and overall survival. We found that the prognostic benefit of increased extent of resection for patients with medulloblastoma is attenuated after molecular subgroup affiliation is taken into account. We identified a progression-free survival benefit for gross total resection over sub-total resection (hazard ratio [HR] 1·45, 95% CI 1·07-1·96, p=0·16) but no overall survival benefit (HR 1·23, 0·87-1·72, p=0·24). We saw no progression-free survival or overall survival benefit for gross total resection compared with near-total resection (HR 1·05, 0·71-1·53, p=0·8158 for progression-free survival and HR 1·14, 0·75-1·72, p=0·55 for overall survival). No significant survival benefit existed for greater extent of resection for patients with WNT, SHH, or group 3 tumours (HR 1·03, 0·67-1·58, p=0·89 for sub-total resection vs gross total resection). For patients with group 4 tumours, gross total resection conferred a benefit to progression-free survival compared with sub-total resection (HR 1·97, 1·22-3·17, p=0·0056), especially for those with metastatic disease (HR 2·22, 1·00-4·93, p=0·050). However, gross total resection had no effect on overall survival compared with sub-total resection in patients with group 4 tumours (HR 1·67, 0·93-2·99, p=0·084).InterpretationThe prognostic benefit of increased extent of resection for patients with medulloblastoma is attenuated after molecular subgroup affiliation is taken into account. Although maximum safe surgical resection should remain the standard of care, surgical removal of small residual portions of medulloblastoma is not recommended when the likelihood of neurological morbidity is high because there is no definitive benefit to gross total resection compared with near-total resection.FundingCanadian Cancer Society Research Institute, Terry Fox Research Institute, Canadian Institutes of Health Research, National Institutes of Health, Pediatric Brain Tumor Foundation, and the Garron Family Chair in Childhood Cancer Research.
- Published
- 2016
11. New Brain Tumor Entities Emerge from Molecular Classification of CNS-PNETs.
- Author
-
Sturm, Dominik, Orr, Brent A, Toprak, Umut H, Hovestadt, Volker, Jones, David TW, Capper, David, Sill, Martin, Buchhalter, Ivo, Northcott, Paul A, Leis, Irina, Ryzhova, Marina, Koelsche, Christian, Pfaff, Elke, Allen, Sariah J, Balasubramanian, Gnanaprakash, Worst, Barbara C, Pajtler, Kristian W, Brabetz, Sebastian, Johann, Pascal D, Sahm, Felix, Reimand, Jüri, Mackay, Alan, Carvalho, Diana M, Remke, Marc, Phillips, Joanna J, Perry, Arie, Cowdrey, Cynthia, Drissi, Rachid, Fouladi, Maryam, Giangaspero, Felice, Łastowska, Maria, Grajkowska, Wiesława, Scheurlen, Wolfram, Pietsch, Torsten, Hagel, Christian, Gojo, Johannes, Lötsch, Daniela, Berger, Walter, Slavc, Irene, Haberler, Christine, Jouvet, Anne, Holm, Stefan, Hofer, Silvia, Prinz, Marco, Keohane, Catherine, Fried, Iris, Mawrin, Christian, Scheie, David, Mobley, Bret C, Schniederjan, Matthew J, Santi, Mariarita, Buccoliero, Anna M, Dahiya, Sonika, Kramm, Christof M, von Bueren, André O, von Hoff, Katja, Rutkowski, Stefan, Herold-Mende, Christel, Frühwald, Michael C, Milde, Till, Hasselblatt, Martin, Wesseling, Pieter, Rößler, Jochen, Schüller, Ulrich, Ebinger, Martin, Schittenhelm, Jens, Frank, Stephan, Grobholz, Rainer, Vajtai, Istvan, Hans, Volkmar, Schneppenheim, Reinhard, Zitterbart, Karel, Collins, V Peter, Aronica, Eleonora, Varlet, Pascale, Puget, Stephanie, Dufour, Christelle, Grill, Jacques, Figarella-Branger, Dominique, Wolter, Marietta, Schuhmann, Martin U, Shalaby, Tarek, Grotzer, Michael, van Meter, Timothy, Monoranu, Camelia-Maria, Felsberg, Jörg, Reifenberger, Guido, Snuderl, Matija, Forrester, Lynn Ann, Koster, Jan, Versteeg, Rogier, Volckmann, Richard, van Sluis, Peter, Wolf, Stephan, Mikkelsen, Tom, Gajjar, Amar, Aldape, Kenneth, Moore, Andrew S, Taylor, Michael D, and Jones, Chris
- Subjects
Humans ,Neuroectodermal Tumors ,Central Nervous System Neoplasms ,Trans-Activators ,Proto-Oncogene Proteins ,Tumor Suppressor Proteins ,Repressor Proteins ,Gene Expression Profiling ,Signal Transduction ,DNA Methylation ,Gene Expression Regulation ,Neoplastic ,Amino Acid Sequence ,Molecular Sequence Data ,Child ,Forkhead Transcription Factors ,Neuroblastoma ,Cancer ,Pediatric ,Neurosciences ,Brain Disorders ,Rare Diseases ,Orphan Drug ,Brain Cancer ,Pediatric Research Initiative ,Pediatric Cancer ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
Primitive neuroectodermal tumors of the central nervous system (CNS-PNETs) are highly aggressive, poorly differentiated embryonal tumors occurring predominantly in young children but also affecting adolescents and adults. Herein, we demonstrate that a significant proportion of institutionally diagnosed CNS-PNETs display molecular profiles indistinguishable from those of various other well-defined CNS tumor entities, facilitating diagnosis and appropriate therapy for patients with these tumors. From the remaining fraction of CNS-PNETs, we identify four new CNS tumor entities, each associated with a recurrent genetic alteration and distinct histopathological and clinical features. These new molecular entities, designated "CNS neuroblastoma with FOXR2 activation (CNS NB-FOXR2)," "CNS Ewing sarcoma family tumor with CIC alteration (CNS EFT-CIC)," "CNS high-grade neuroepithelial tumor with MN1 alteration (CNS HGNET-MN1)," and "CNS high-grade neuroepithelial tumor with BCOR alteration (CNS HGNET-BCOR)," will enable meaningful clinical trials and the development of therapeutic strategies for patients affected by poorly differentiated CNS tumors.
- Published
- 2016
12. Embryonal tumor with abundant neuropil and true rosettes (ETANTR), ependymoblastoma, and medulloepithelioma share molecular similarity and comprise a single clinicopathological entity.
- Author
-
Picard, Daniel, Rosenblum, Marc, Antonelli, Manila, Aronica, Eleonora, Schüller, Ulrich, Hasselblatt, Martin, Woehrer, Adelheid, Zheludkova, Olga, Kumirova, Ella, Puget, Stephanie, Taylor, Michael, Giangaspero, Felice, Peter Collins, V, von Deimling, Andreas, Lichter, Peter, Huang, Annie, Pietsch, Torsten, Pfister, Stefan, Kool, Marcel, Korshunov, Andrey, Sturm, Dominik, Ryzhova, Marina, Hovestadt, Volker, Gessi, Marco, Jones, David, Remke, Marc, Northcott, Paul, and Perry, Arie
- Subjects
Brain Neoplasms ,Child ,Child ,Preschool ,Chromosomes ,Human ,Pair 19 ,DNA Copy Number Variations ,DNA Methylation ,Diagnosis ,Differential ,Female ,Genetic Loci ,Humans ,Immunohistochemistry ,In Situ Hybridization ,Fluorescence ,Infant ,Male ,Neoplasm Recurrence ,Local ,Neoplasms ,Germ Cell and Embryonal ,Neuroectodermal Tumors ,Primitive ,Survival Analysis - Abstract
Three histological variants are known within the family of embryonal rosette-forming neuroepithelial brain tumors. These include embryonal tumor with abundant neuropil and true rosettes (ETANTR), ependymoblastoma (EBL), and medulloepithelioma (MEPL). In this study, we performed a comprehensive clinical, pathological, and molecular analysis of 97 cases of these rare brain neoplasms, including genome-wide DNA methylation and copy number profiling of 41 tumors. We identified uniform molecular signatures in all tumors irrespective of histological patterns, indicating that ETANTR, EBL, and MEPL comprise a single biological entity. As such, future WHO classification schemes should consider lumping these variants into a single diagnostic category, such as embryonal tumor with multilayered rosettes (ETMR). We recommend combined LIN28A immunohistochemistry and FISH analysis of the 19q13.42 locus for molecular diagnosis of this tumor category. Recognition of this distinct pediatric brain tumor entity based on the fact that the three histological variants are molecularly and clinically uniform will help to distinguish ETMR from other embryonal CNS tumors and to better understand the biology of these highly aggressive and therapy-resistant pediatric CNS malignancies, possibly leading to alternate treatment strategies.
- Published
- 2014
13. Cytogenetic prognostication within medulloblastoma subgroups.
- Author
-
Shih, David, Northcott, Paul, Remke, Marc, Korshunov, Andrey, Ramaswamy, Vijay, Kool, Marcel, Luu, Betty, Yao, Yuan, Wang, Xin, Dubuc, Adrian, Garzia, Livia, Peacock, John, Mack, Stephen, Wu, Xiaochong, Rolider, Adi, Morrissy, A, Cavalli, Florence, Jones, David, Zitterbart, Karel, Faria, Claudia, Schüller, Ulrich, Kren, Leos, Kumabe, Toshihiro, Tominaga, Teiji, Shin Ra, Young, Garami, Miklós, Hauser, Peter, Chan, Jennifer, Robinson, Shenandoah, Bognár, László, Klekner, Almos, Saad, Ali, Albrecht, Steffen, Fontebasso, Adam, Cinalli, Giuseppe, De Antonellis, Pasqualino, Zollo, Massimo, Cooper, Michael, Thompson, Reid, Bailey, Simon, Lindsey, Janet, Di Rocco, Concezio, Massimi, Luca, Michiels, Erna, Scherer, Stephen, Lee, Ji-Yeoun, Cho, Byung-Kyu, Kim, Seung-Ki, Wang, Kyu-Chang, Leonard, Jeffrey, Rubin, Joshua, de Torres, Carmen, Lavarino, Cinzia, Mora, Jaume, Cho, Yoon-Jae, Tabori, Uri, Olson, James, Gajjar, Amar, Packer, Roger, Fan, Xing, Muraszko, Karin, Vibhakar, Rajeev, Eberhart, Charles, Fouladi, Maryam, Lach, Boleslaw, Jung, Shin, Wechsler-Reya, Robert, Fèvre-Montange, Michelle, Jouvet, Anne, Jabado, Nada, Pollack, Ian, Rutkowski, Stefan, Pomeroy, Scott, French, Pim, Kloosterhof, Nanne, Kros, Johan, Van Meir, Erwin, Clifford, Steven, Bourdeaut, Franck, Delattre, Olivier, Doz, François, Hawkins, Cynthia, Malkin, David, Grajkowska, Wieslawa, Perek-Polnik, Marta, Bouffet, Eric, Rutka, James, Pfister, Stefan, Taylor, Michael, Gupta, Nalin, Phillips, Joanna, Weiss, William, and Liau, Linda
- Subjects
Adolescent ,Biomarkers ,Tumor ,Child ,Child ,Preschool ,Chromosomes ,Human ,Pair 11 ,Chromosomes ,Human ,Pair 14 ,Cytogenetics ,Female ,Gene Expression Profiling ,Gene Expression Regulation ,Neoplastic ,Hedgehog Proteins ,Humans ,In Situ Hybridization ,Fluorescence ,Infant ,Kruppel-Like Transcription Factors ,Male ,Medulloblastoma ,Nuclear Proteins ,Predictive Value of Tests ,Prognosis ,Proportional Hazards Models ,Proto-Oncogene Proteins c-myc ,Reproducibility of Results ,Risk Assessment ,Risk Factors ,Tissue Array Analysis ,Wnt Proteins ,Young Adult ,Zinc Finger Protein Gli2 - Abstract
PURPOSE: Medulloblastoma comprises four distinct molecular subgroups: WNT, SHH, Group 3, and Group 4. Current medulloblastoma protocols stratify patients based on clinical features: patient age, metastatic stage, extent of resection, and histologic variant. Stark prognostic and genetic differences among the four subgroups suggest that subgroup-specific molecular biomarkers could improve patient prognostication. PATIENTS AND METHODS: Molecular biomarkers were identified from a discovery set of 673 medulloblastomas from 43 cities around the world. Combined risk stratification models were designed based on clinical and cytogenetic biomarkers identified by multivariable Cox proportional hazards analyses. Identified biomarkers were tested using fluorescent in situ hybridization (FISH) on a nonoverlapping medulloblastoma tissue microarray (n = 453), with subsequent validation of the risk stratification models. RESULTS: Subgroup information improves the predictive accuracy of a multivariable survival model compared with clinical biomarkers alone. Most previously published cytogenetic biomarkers are only prognostic within a single medulloblastoma subgroup. Profiling six FISH biomarkers (GLI2, MYC, chromosome 11 [chr11], chr14, 17p, and 17q) on formalin-fixed paraffin-embedded tissues, we can reliably and reproducibly identify very low-risk and very high-risk patients within SHH, Group 3, and Group 4 medulloblastomas. CONCLUSION: Combining subgroup and cytogenetic biomarkers with established clinical biomarkers substantially improves patient prognostication, even in the context of heterogeneous clinical therapies. The prognostic significance of most molecular biomarkers is restricted to a specific subgroup. We have identified a small panel of cytogenetic biomarkers that reliably identifies very high-risk and very low-risk groups of patients, making it an excellent tool for selecting patients for therapy intensification and therapy de-escalation in future clinical trials.
- Published
- 2014
14. Genome Sequencing of SHH Medulloblastoma Predicts Genotype-Related Response to Smoothened Inhibition
- Author
-
Kool, Marcel, Jones, David TW, Jäger, Natalie, Northcott, Paul A, Pugh, Trevor J, Hovestadt, Volker, Piro, Rosario M, Esparza, L Adriana, Markant, Shirley L, Remke, Marc, Milde, Till, Bourdeaut, Franck, Ryzhova, Marina, Sturm, Dominik, Pfaff, Elke, Stark, Sebastian, Hutter, Sonja, Şeker-Cin, Huriye, Johann, Pascal, Bender, Sebastian, Schmidt, Christin, Rausch, Tobias, Shih, David, Reimand, Jüri, Sieber, Laura, Wittmann, Andrea, Linke, Linda, Witt, Hendrik, Weber, Ursula D, Zapatka, Marc, König, Rainer, Beroukhim, Rameen, Bergthold, Guillaume, van Sluis, Peter, Volckmann, Richard, Koster, Jan, Versteeg, Rogier, Schmidt, Sabine, Wolf, Stephan, Lawerenz, Chris, Bartholomae, Cynthia C, von Kalle, Christof, Unterberg, Andreas, Herold-Mende, Christel, Hofer, Silvia, Kulozik, Andreas E, von Deimling, Andreas, Scheurlen, Wolfram, Felsberg, Jörg, Reifenberger, Guido, Hasselblatt, Martin, Crawford, John R, Grant, Gerald A, Jabado, Nada, Perry, Arie, Cowdrey, Cynthia, Croul, Sydney, Zadeh, Gelareh, Korbel, Jan O, Doz, Francois, Delattre, Olivier, Bader, Gary D, McCabe, Martin G, Collins, V Peter, Kieran, Mark W, Cho, Yoon-Jae, Pomeroy, Scott L, Witt, Olaf, Brors, Benedikt, Taylor, Michael D, Schüller, Ulrich, Korshunov, Andrey, Eils, Roland, Wechsler-Reya, Robert J, Lichter, Peter, Pfister, Stefan M, and Project, on behalf of the ICGC PedBrain Tumor
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Clinical Sciences ,Neurosciences ,Pediatric Research Initiative ,Genetics ,Rare Diseases ,Cancer ,Pediatric ,Brain Cancer ,Pediatric Cancer ,Brain Disorders ,Adolescent ,Adult ,Animals ,Base Sequence ,Biphenyl Compounds ,Cerebellar Neoplasms ,Child ,Child ,Preschool ,DEAD-box RNA Helicases ,DNA Copy Number Variations ,Drug Resistance ,Neoplasm ,Female ,Gene Expression Profiling ,Hedgehog Proteins ,High-Throughput Nucleotide Sequencing ,Humans ,Infant ,Kruppel-Like Transcription Factors ,Male ,Medulloblastoma ,Mice ,Mice ,Inbred NOD ,Mice ,SCID ,Molecular Sequence Data ,N-Myc Proto-Oncogene Protein ,Neoplasm Transplantation ,Nuclear Proteins ,Oncogene Proteins ,Patched Receptors ,Patched-1 Receptor ,Phosphatidylinositol 3-Kinases ,Promoter Regions ,Genetic ,Proto-Oncogene Proteins c-akt ,Pyridines ,Receptors ,Cell Surface ,Receptors ,G-Protein-Coupled ,Repressor Proteins ,Signal Transduction ,Smoothened Receptor ,Telomerase ,Tumor Suppressor Protein p53 ,Young Adult ,Zinc Finger Protein Gli2 ,ICGC PedBrain Tumor Project ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
Smoothened (SMO) inhibitors recently entered clinical trials for sonic-hedgehog-driven medulloblastoma (SHH-MB). Clinical response is highly variable. To understand the mechanism(s) of primary resistance and identify pathways cooperating with aberrant SHH signaling, we sequenced and profiled a large cohort of SHH-MBs (n = 133). SHH pathway mutations involved PTCH1 (across all age groups), SUFU (infants, including germline), and SMO (adults). Children >3 years old harbored an excess of downstream MYCN and GLI2 amplifications and frequent TP53 mutations, often in the germline, all of which were rare in infants and adults. Functional assays in different SHH-MB xenograft models demonstrated that SHH-MBs harboring a PTCH1 mutation were responsive to SMO inhibition, whereas tumors harboring an SUFU mutation or MYCN amplification were primarily resistant.
- Published
- 2014
15. Medulloblastoma Down Under 2013: a report from the third annual meeting of the International Medulloblastoma Working Group.
- Author
-
Williamson, Dan, Gajjar, Amar, Gottardo, Nicholas, Hansford, Jordan, McGlade, Jacqueline, Alvaro, Frank, Ashley, David, Bailey, Simon, Baker, David, Bourdeaut, Franck, Cho, Yoon-Jae, Clay, Moira, Clifford, Steven, Cohn, Richard, Cole, Catherine, Dallas, Peter, Downie, Peter, Doz, François, Ellison, David, Endersby, Raelene, Fisher, Paul, Hassall, Timothy, Heath, John, Hii, Hilary, Jones, David, Junckerstorff, Reimar, Kellie, Stewart, Kool, Marcel, Kotecha, Rishi, Lichter, Peter, Laughton, Stephen, Lee, Sharon, McCowage, Geoff, Northcott, Paul, Olson, James, Packer, Roger, Pfister, Stefan, Pietsch, Torsten, Pizer, Barry, Pomeroy, Scott, Remke, Marc, Robinson, Giles, Rutkowski, Stefan, Schoep, Tobias, Shelat, Anang, Stewart, Clinton, Sullivan, Michael, Taylor, Michael, Wainwright, Brandon, Walwyn, Thomas, and Weiss, William
- Subjects
Adolescent ,Animals ,Antineoplastic Agents ,Australia ,Cerebellar Neoplasms ,Child ,Child ,Preschool ,Disease Models ,Animal ,Genomics ,Humans ,International Agencies ,Medulloblastoma ,Mice - Abstract
Medulloblastoma is curable in approximately 70% of patients. Over the past decade, progress in improving survival using conventional therapies has stalled, resulting in reduced quality of life due to treatment-related side effects, which are a major concern in survivors. The vast amount of genomic and molecular data generated over the last 5-10 years encourages optimism that improved risk stratification and new molecular targets will improve outcomes. It is now clear that medulloblastoma is not a single-disease entity, but instead consists of at least four distinct molecular subgroups: WNT/Wingless, Sonic Hedgehog, Group 3, and Group 4. The Medulloblastoma Down Under 2013 meeting, which convened at Bunker Bay, Australia, brought together 50 leading clinicians and scientists. The 2-day agenda included focused sessions on pathology and molecular stratification, genomics and mouse models, high-throughput drug screening, and clinical trial design. The meeting established a global action plan to translate novel biologic insights and drug targeting into treatment regimens to improve outcomes. A consensus was reached in several key areas, with the most important being that a novel classification scheme for medulloblastoma based on the four molecular subgroups, as well as histopathologic features, should be presented for consideration in the upcoming fifth edition of the World Health Organizations classification of tumours of the central nervous system. Three other notable areas of agreement were as follows: (1) to establish a central repository of annotated mouse models that are readily accessible and freely available to the international research community; (2) to institute common eligibility criteria between the Childrens Oncology Group and the International Society of Paediatric Oncology Europe and initiate joint or parallel clinical trials; (3) to share preliminary high-throughput screening data across discovery labs to hasten the development of novel therapeutics. Medulloblastoma Down Under 2013 was an effective forum for meaningful discussion, which resulted in enhancing international collaborative clinical and translational research of this rare disease. This template could be applied to other fields to devise global action plans addressing all aspects of a disease, from improved disease classification, treatment stratification, and drug targeting to superior treatment regimens to be assessed in cooperative international clinical trials.
- Published
- 2014
16. TERT promoter mutations are highly recurrent in SHH subgroup medulloblastoma.
- Author
-
Remke, Marc, Ramaswamy, Vijay, Peacock, John, Shih, David, Koelsche, Christian, Northcott, Paul, Hill, Nadia, Cavalli, Florence, Kool, Marcel, Wang, Xin, Mack, Stephen, Barszczyk, Mark, Morrissy, A, Wu, Xiaochong, Agnihotri, Sameer, Luu, Betty, Jones, David, Garzia, Livia, Dubuc, Adrian, Zhukova, Nataliya, Vanner, Robert, Kros, Johan, French, Pim, Van Meir, Erwin, Vibhakar, Rajeev, Zitterbart, Karel, Chan, Jennifer, Bognár, László, Klekner, Almos, Lach, Boleslaw, Jung, Shin, Saad, Ali, Albrecht, Steffen, Zollo, Massimo, Cooper, Michael, Thompson, Reid, Delattre, Oliver, Bourdeaut, Franck, Doz, François, Garami, Miklós, Hauser, Peter, Carlotti, Carlos, Van Meter, Timothy, Massimi, Luca, Fults, Daniel, Pomeroy, Scott, Kumabe, Toshiro, Ra, Young, Leonard, Jeffrey, Elbabaa, Samer, Mora, Jaume, Rubin, Joshua, Cho, Yoon-Jae, McLendon, Roger, Bigner, Darell, Eberhart, Charles, Fouladi, Maryam, Wechsler-Reya, Robert, Faria, Claudia, Croul, Sidney, Huang, Annie, Bouffet, Eric, Hawkins, Cynthia, Dirks, Peter, Schüller, Ulrich, Pollack, Ian, Rutkowski, Stefan, Meyronet, David, Jouvet, Anne, Fèvre-Montange, Michelle, Jabado, Nada, Perek-Polnik, Marta, Grajkowska, Wieslawa, Kim, Seung-Ki, Rutka, James, Malkin, David, Tabori, Uri, Pfister, Stefan, Korshunov, Andrey, von Deimling, Andreas, Taylor, Michael, Weiss, William, and Liau, Linda
- Subjects
Adolescent ,Adult ,Brain Neoplasms ,Child ,Child ,Preschool ,DNA Mutational Analysis ,Female ,Gene Expression Profiling ,Genotype ,Humans ,Infant ,Male ,Medulloblastoma ,Middle Aged ,Mutation ,Prognosis ,Promoter Regions ,Genetic ,Telomerase - Abstract
Telomerase reverse transcriptase (TERT) promoter mutations were recently shown to drive telomerase activity in various cancer types, including medulloblastoma. However, the clinical and biological implications of TERT mutations in medulloblastoma have not been described. Hence, we sought to describe these mutations and their impact in a subgroup-specific manner. We analyzed the TERT promoter by direct sequencing and genotyping in 466 medulloblastomas. The mutational distributions were determined according to subgroup affiliation, demographics, and clinical, prognostic, and molecular features. Integrated genomics approaches were used to identify specific somatic copy number alterations in TERT promoter-mutated and wild-type tumors. Overall, TERT promoter mutations were identified in 21 % of medulloblastomas. Strikingly, the highest frequencies of TERT mutations were observed in SHH (83 %; 55/66) and WNT (31 %; 4/13) medulloblastomas derived from adult patients. Group 3 and Group 4 harbored this alteration in
- Published
- 2013
17. Subgroup-specific prognostic implications of TP53 mutation in medulloblastoma.
- Author
-
Zhukova, Nataliya, Ramaswamy, Vijay, Remke, Marc, Pfaff, Elke, Shih, David, Martin, Dianna, Castelo-Branco, Pedro, Baskin, Berivan, Ray, Peter, Bouffet, Eric, von Bueren, André, Jones, David, Northcott, Paul, Kool, Marcel, Sturm, Dominik, Pugh, Trevor, Pomeroy, Scott, Cho, Yoon-Jae, Pietsch, Torsten, Gessi, Marco, Rutkowski, Stefan, Bognar, Laszlo, Klekner, Almos, Cho, Byung-Kyu, Kim, Seung-Ki, Wang, Kyu-Chang, Eberhart, Charles, Fevre-Montange, Michelle, Fouladi, Maryam, French, Pim, Kros, Max, Grajkowska, Wieslawa, Hauser, Peter, Jabado, Nada, Jouvet, Anne, Jung, Shin, Kumabe, Toshihiro, Lach, Boleslaw, Leonard, Jeffrey, Rubin, Joshua, Massimi, Luca, Pollack, Ian, Shin Ra, Young, Van Meir, Erwin, Zitterbart, Karel, Schüller, Ulrich, Hill, Rebecca, Lindsey, Janet, Schwalbe, Ed, Bailey, Simon, Ellison, David, Hawkins, Cynthia, Malkin, David, Clifford, Steven, Korshunov, Andrey, Pfister, Stefan, Taylor, Michael, Tabori, Uri, Gupta, Nalin, Weiss, William, and Liau, Linda
- Subjects
Adolescent ,Adult ,Cerebellar Neoplasms ,Child ,Child ,Preschool ,Female ,Gene Expression Profiling ,Genes ,p53 ,Humans ,Infant ,Male ,Medulloblastoma ,Middle Aged ,Mutation ,Prognosis ,Young Adult - Abstract
PURPOSE: Reports detailing the prognostic impact of TP53 mutations in medulloblastoma offer conflicting conclusions. We resolve this issue through the inclusion of molecular subgroup profiles. PATIENTS AND METHODS: We determined subgroup affiliation, TP53 mutation status, and clinical outcome in a discovery cohort of 397 medulloblastomas. We subsequently validated our results on an independent cohort of 156 medulloblastomas. RESULTS: TP53 mutations are enriched in wingless (WNT; 16%) and sonic hedgehog (SHH; 21%) medulloblastomas and are virtually absent in subgroups 3 and 4 tumors (P < .001). Patients with SHH/TP53 mutant tumors are almost exclusively between ages 5 and 18 years, dramatically different from the general SHH distribution (P < .001). Children with SHH/TP53 mutant tumors harbor 56% germline TP53 mutations, which are not observed in children with WNT/TP53 mutant tumors. Five-year overall survival (OS; ± SE) was 41% ± 9% and 81% ± 5% for patients with SHH medulloblastomas with and without TP53 mutations, respectively (P < .001). Furthermore, TP53 mutations accounted for 72% of deaths in children older than 5 years with SHH medulloblastomas. In contrast, 5-year OS rates were 90% ± 9% and 97% ± 3% for patients with WNT tumors with and without TP53 mutations (P = .21). Multivariate analysis revealed that TP53 status was the most important risk factor for SHH medulloblastoma. Survival rates in the validation cohort mimicked the discovery results, revealing that poor survival of TP53 mutations is restricted to patients with SHH medulloblastomas (P = .012) and not WNT tumors. CONCLUSION: Subgroup-specific analysis reconciles prior conflicting publications and confirms that TP53 mutations are enriched among SHH medulloblastomas, in which they portend poor outcome and account for a large proportion of treatment failures in these patients.
- Published
- 2013
18. LIN28A immunoreactivity is a potent diagnostic marker of embryonal tumor with multilayered rosettes (ETMR).
- Author
-
Picard, Daniel, Rosenblum, Marc, Giangaspero, Felice, Aronica, Eleonora, Schüller, Ulrich, Hasselblatt, Martin, Collins, V, von Deimling, Andreas, Lichter, Peter, Huang, Annie, Pfister, Stefan, Kool, Marcel, Korshunov, Andrey, Ryzhova, Marina, Jones, David, Northcott, Paul, van Sluis, Peter, Volckmann, Richard, Koster, Jan, Versteeg, Rogier, Cowdrey, Cynthia, and Perry, Arie
- Subjects
Adolescent ,Biomarkers ,Tumor ,Brain Neoplasms ,Child ,Child ,Preschool ,DNA-Binding Proteins ,Diagnosis ,Differential ,Gene Expression Profiling ,Humans ,MicroRNAs ,Neoplasms ,Germ Cell and Embryonal ,Neuroectodermal Tumors ,Primitive ,Neuropil ,RNA-Binding Proteins ,Rhabdoid Tumor - Abstract
Embryonal tumor with multilayered rosettes (ETMR, previously known as ETANTR) is a highly aggressive embryonal CNS tumor, which almost exclusively affects infants and is associated with a dismal prognosis. Accurate diagnosis is of critical clinical importance because of its poor response to current treatment protocols and its distinct biology. Amplification of the miRNA cluster at 19q13.42 has been identified previously as a genetic hallmark for ETMR, but an immunohistochemistry-based assay for clinical routine diagnostics [such as INI-1 for atypical teratoid rhabdoid tumor (AT/RT)] is still lacking. In this study, we screened for an ETMR-specific marker using a gene-expression profiling dataset of more than 1,400 brain tumors and identified LIN28A as a highly specific marker for ETMR. The encoded protein binds small RNA and has been implicated in stem cell pluripotency, metabolism and tumorigenesis. Using an LIN28A specific antibody, we carried out immunohistochemical analysis of LIN28A in more than 800 childhood brain-tumor samples and confirmed its high specificity for ETMR. Strong LIN28A immunoexpression was found in all 37 ETMR samples tested, whereas focal reactivity was only present in a small (6/50) proportion of AT/RT samples. All other pediatric brain tumors were completely LIN28A-negative. In summary, we established LIN28A immunohistochemistry as a highly sensitive and specific, rapid, inexpensive diagnostic tool for routine pathological verification of ETMR.
- Published
- 2012
19. Subgroup-specific structural variation across 1,000 medulloblastoma genomes.
- Author
-
Northcott, Paul, Shih, David, Peacock, John, Garzia, Livia, Morrissy, A, Zichner, Thomas, Stütz, Adrian, Korshunov, Andrey, Reimand, Jüri, Schumacher, Steven, Beroukhim, Rameen, Ellison, David, Marshall, Christian, Lionel, Anath, Mack, Stephen, Dubuc, Adrian, Yao, Yuan, Ramaswamy, Vijay, Luu, Betty, Rolider, Adi, Cavalli, Florence, Wang, Xin, Remke, Marc, Wu, Xiaochong, Chiu, Readman, Chu, Andy, Chuah, Eric, Corbett, Richard, Hoad, Gemma, Jackman, Shaun, Li, Yisu, Lo, Allan, Mungall, Karen, Nip, Ka, Qian, Jenny, Raymond, Anthony, Thiessen, Nina, Varhol, Richard, Birol, Inanc, Moore, Richard, Mungall, Andrew, Holt, Robert, Kawauchi, Daisuke, Roussel, Martine, Kool, Marcel, Jones, David, Witt, Hendrick, Fernandez-L, Africa, Kenney, Anna, Wechsler-Reya, Robert, Dirks, Peter, Aviv, Tzvi, Grajkowska, Wieslawa, Perek-Polnik, Marta, Haberler, Christine, Delattre, Olivier, Reynaud, Stéphanie, Doz, François, Pernet-Fattet, Sarah, Cho, Byung-Kyu, Kim, Seung-Ki, Wang, Kyu-Chang, Scheurlen, Wolfram, Eberhart, Charles, Fèvre-Montange, Michelle, Jouvet, Anne, Pollack, Ian, Fan, Xing, Muraszko, Karin, Gillespie, G, Di Rocco, Concezio, Massimi, Luca, Michiels, Erna, Kloosterhof, Nanne, French, Pim, Kros, Johan, Olson, James, Ellenbogen, Richard, Zitterbart, Karel, Kren, Leos, Thompson, Reid, Cooper, Michael, Lach, Boleslaw, McLendon, Roger, Bigner, Darell, Fontebasso, Adam, Albrecht, Steffen, Jabado, Nada, Lindsey, Janet, Bailey, Simon, Van Meir, Erwin, Fouladi, Maryam, Nakamura, Hideo, Cinalli, Giuseppe, Garami, Miklós, Hauser, Peter, Saad, Ali, Iolascon, Achille, Jung, Shin, and Carlotti, Carlos
- Subjects
Carrier Proteins ,Cerebellar Neoplasms ,Child ,DNA Copy Number Variations ,Gene Duplication ,Genes ,myc ,Genome ,Human ,Genomic Structural Variation ,Genomics ,Hedgehog Proteins ,Humans ,Medulloblastoma ,NF-kappa B ,Nerve Tissue Proteins ,Oncogene Proteins ,Fusion ,Proteins ,RNA ,Long Noncoding ,Signal Transduction ,Transforming Growth Factor beta ,Translocation ,Genetic - Abstract
Medulloblastoma, the most common malignant paediatric brain tumour, is currently treated with nonspecific cytotoxic therapies including surgery, whole-brain radiation, and aggressive chemotherapy. As medulloblastoma exhibits marked intertumoural heterogeneity, with at least four distinct molecular variants, previous attempts to identify targets for therapy have been underpowered because of small samples sizes. Here we report somatic copy number aberrations (SCNAs) in 1,087 unique medulloblastomas. SCNAs are common in medulloblastoma, and are predominantly subgroup-enriched. The most common region of focal copy number gain is a tandem duplication of SNCAIP, a gene associated with Parkinsons disease, which is exquisitely restricted to Group 4α. Recurrent translocations of PVT1, including PVT1-MYC and PVT1-NDRG1, that arise through chromothripsis are restricted to Group 3. Numerous targetable SCNAs, including recurrent events targeting TGF-β signalling in Group 3, and NF-κB signalling in Group 4, suggest future avenues for rational, targeted therapy.
- Published
- 2012
20. Medulloblastoma exome sequencing uncovers subtype-specific somatic mutations.
- Author
-
Pugh, Trevor J, Weeraratne, Shyamal Dilhan, Archer, Tenley C, Pomeranz Krummel, Daniel A, Auclair, Daniel, Bochicchio, James, Carneiro, Mauricio O, Carter, Scott L, Cibulskis, Kristian, Erlich, Rachel L, Greulich, Heidi, Lawrence, Michael S, Lennon, Niall J, McKenna, Aaron, Meldrim, James, Ramos, Alex H, Ross, Michael G, Russ, Carsten, Shefler, Erica, Sivachenko, Andrey, Sogoloff, Brian, Stojanov, Petar, Tamayo, Pablo, Mesirov, Jill P, Amani, Vladimir, Teider, Natalia, Sengupta, Soma, Francois, Jessica Pierre, Northcott, Paul A, Taylor, Michael D, Yu, Furong, Crabtree, Gerald R, Kautzman, Amanda G, Gabriel, Stacey B, Getz, Gad, Jäger, Natalie, Jones, David TW, Lichter, Peter, Pfister, Stefan M, Roberts, Thomas M, Meyerson, Matthew, Pomeroy, Scott L, and Cho, Yoon-Jae
- Subjects
Humans ,Medulloblastoma ,Cerebellar Neoplasms ,DNA Helicases ,Histone-Lysine N-Methyltransferase ,Intracellular Signaling Peptides and Proteins ,DNA-Binding Proteins ,Receptors ,Cell Surface ,Neoplasm Proteins ,Proto-Oncogene Proteins ,Nuclear Proteins ,Transcription Factors ,Repressor Proteins ,Signal Transduction ,Protein Structure ,Tertiary ,Mutation ,Genome ,Human ,Models ,Molecular ,Child ,Tumor Suppressor Protein p53 ,Wnt Proteins ,beta Catenin ,TCF Transcription Factors ,Hedgehog Proteins ,DEAD-box RNA Helicases ,Promoter Regions ,Genetic ,LIM Domain Proteins ,Exome ,Patched Receptors ,Patched-1 Receptor ,Histone Methyltransferases ,Brain Disorders ,Pediatric Cancer ,Cancer ,Human Genome ,Biotechnology ,Pediatric ,Brain Cancer ,Rare Diseases ,Genetics ,Neurosciences ,Genetic Testing ,Aetiology ,2.1 Biological and endogenous factors ,General Science & Technology - Abstract
Medulloblastomas are the most common malignant brain tumours in children. Identifying and understanding the genetic events that drive these tumours is critical for the development of more effective diagnostic, prognostic and therapeutic strategies. Recently, our group and others described distinct molecular subtypes of medulloblastoma on the basis of transcriptional and copy number profiles. Here we use whole-exome hybrid capture and deep sequencing to identify somatic mutations across the coding regions of 92 primary medulloblastoma/normal pairs. Overall, medulloblastomas have low mutation rates consistent with other paediatric tumours, with a median of 0.35 non-silent mutations per megabase. We identified twelve genes mutated at statistically significant frequencies, including previously known mutated genes in medulloblastoma such as CTNNB1, PTCH1, MLL2, SMARCA4 and TP53. Recurrent somatic mutations were newly identified in an RNA helicase gene, DDX3X, often concurrent with CTNNB1 mutations, and in the nuclear co-repressor (N-CoR) complex genes GPS2, BCOR and LDB1. We show that mutant DDX3X potentiates transactivation of a TCF promoter and enhances cell viability in combination with mutant, but not wild-type, β-catenin. Together, our study reveals the alteration of WNT, hedgehog, histone methyltransferase and now N-CoR pathways across medulloblastomas and within specific subtypes of this disease, and nominates the RNA helicase DDX3X as a component of pathogenic β-catenin signalling in medulloblastoma.
- Published
- 2012
21. The landscape of genomic alterations across childhood cancers
- Author
-
Gröbner, Susanne N., Worst, Barbara C., Weischenfeldt, Joachim, Buchhalter, Ivo, Kleinheinz, Kortine, Rudneva, Vasilisa A., Johann, Pascal D., Balasubramanian, Gnana Prakash, Segura-Wang, Maia, Brabetz, Sebastian, Bender, Sebastian, Hutter, Barbara, Sturm, Dominik, Pfaff, Elke, Hübschmann, Daniel, Zipprich, Gideon, Heinold, Michael, Eils, Jürgen, Lawerenz, Christian, Erkek, Serap, Lambo, Sander, Waszak, Sebastian, Blattmann, Claudia, Borkhardt, Arndt, Kuhlen, Michaela, Eggert, Angelika, Fulda, Simone, Gessler, Manfred, Wegert, Jenny, Kappler, Roland, Baumhoer, Daniel, Burdach, Stefan, Kirschner-Schwabe, Renate, Kontny, Udo, Kulozik, Andreas E., Lohmann, Dietmar, Hettmer, Simone, Eckert, Cornelia, Bielack, Stefan, Nathrath, Michaela, Niemeyer, Charlotte, Richter, Günther H., Schulte, Johannes, Siebert, Reiner, Westermann, Frank, Molenaar, Jan J., Vassal, Gilles, Witt, Hendrik, Lichter, Peter, Weber, Ursula, Eils, Roland, Korshunov, Andrey, Witt, Olaf, Pfister, Stefan, Reifenberger, Guido, Felsberg, Jörg, von Kalle, Christof, Schmidt, Manfred, Bartholomä, Cynthia, Taylor, Michael, Jones, David, Jäger, Natalie, Korbel, Jan, Stütz, Adrian, Rausch, Tobias, Radlwimmer, Bernhard, Yaspo, Marie-Laure, Lehrach, Hans, Warnatz, Hans-Jörg, Landgraf, Pablo, Brors, Benedikt, Zapatka, Marc, Wagner, Susanne, Haake, Andrea, Richter, Julia, Richter, Gesine, Lawerenz, Chris, Kerssemakers, Jules, Jaeger-Schmidt, Christina, Scholz, Ingrid, Bergmann, Anke K., Borst, Christoph, Burkhardt, Birgit, Claviez, Alexander, Dreyling, Martin, Eberth, Sonja, Einsele, Hermann, Frickhofen, Norbert, Haas, Siegfried, Hansmann, Martin-Leo, Karsch, Dennis, Kneba, Michael, Lisfeld, Jasmin, Mantovani-Löffler, Luisa, Rohde, Marius, Ott, German, Stadler, Christina, Staib, Peter, Stilgenbauer, Stephan, Trümper, Lorenz, Zenz, Thorsten, Kube, Dieter, Küppers, Ralf, Weniger, Marc, Hummel, Michael, Klapper, Wolfram, Kostezka, Ulrike, Lenze, Dido, Möller, Peter, Rosenwald, Andreas, Szczepanowski, Monika, Ammerpohl, Ole, Aukema, Sietse M., Binder, Vera, Hoell, Jessica I., Leich, Ellen, López, Cristina, Nagel, Inga, Pischimariov, Jordan, Rosenstiel, Philip, Schilhabel, Markus, Schreiber, Stefan, Vater, Inga, Wagener, Rabea, Bernhart, Stephan H., Binder, Hans, Doose, Gero, Hoffmann, Steve, Hopp, Lydia, Kretzmer, Helene, Kreuz, Markus, Langenberger, David, Loeffler, Markus, Rosolowski, Maciej, Schlesner, Matthias, Stadler, Peter F., Sungalee, Stephanie, Kratz, Christian P., van Tilburg, Cornelis M., Kramm, Christof M., Fleischhack, Gudrun, Dirksen, Uta, Rutkowski, Stefan, Frühwald, Michael, von Hoff, Katja, Wolf, Stephan, Klingebiel, Thomas, Koscielniak, Ewa, Koster, Jan, Resnick, Adam C., Zhang, Jinghui, Liu, Yanling, Zhou, Xin, Waanders, Angela J., Zwijnenburg, Danny A., Raman, Pichai, Weber, Ursula D., Northcott, Paul A., Pajtler, Kristian W., Kool, Marcel, Piro, Rosario M., Korbel, Jan O., Jones, David T. W., Chavez, Lukas, Pfister, Stefan M., Other departments, CCA - Cancer biology and immunology, AGEM - Amsterdam Gastroenterology Endocrinology Metabolism, and Oncogenomics
- Subjects
0301 basic medicine ,Adult ,Mutation rate ,Mutation/genetics ,Adolescent ,DNA Copy Number Variations ,Medizin ,Biology ,DNA Copy Number Variations/genetics ,Germ-Line Mutation/genetics ,Germline ,Cohort Studies ,03 medical and health sciences ,Young Adult ,Germline mutation ,Neoplasms/classification ,Mutation Rate ,Neoplasms ,medicine ,Humans ,Genetic Predisposition to Disease ,Molecular Targeted Therapy ,ddc:610 ,Mutation frequency ,Child ,Germ-Line Mutation ,Genetics ,Chromothripsis ,Multidisciplinary ,Genome, Human ,Genetic Predisposition to Disease/genetics ,Cancer ,Genomics ,medicine.disease ,Diploidy ,Human genetics ,3. Good health ,ddc ,030104 developmental biology ,Mutation (genetic algorithm) ,Mutation ,Genome, Human/genetics ,Tumor Suppressor Protein p53/genetics ,Tumor Suppressor Protein p53 - Abstract
Pan-cancer analyses that examine commonalities and differences among various cancer types have emerged as a powerful way to obtain novel insights into cancer biology. Here we present a comprehensive analysis of genetic alterations in a pan-cancer cohort including 961 tumours from children, adolescents, and young adults, comprising 24 distinct molecular types of cancer. Using a standardized workflow, we identified marked differences in terms of mutation frequency and significantly mutated genes in comparison to previously analysed adult cancers. Genetic alterations in 149 putative cancer driver genes separate the tumours into two classes: small mutation and structural/copy-number variant (correlating with germline variants). Structural variants, hyperdiploidy, and chromothripsis are linked to TP53 mutation status and mutational signatures. Our data suggest that 7–8% of the children in this cohort carry an unambiguous predisposing germline variant and that nearly 50% of paediatric neoplasms harbour a potentially druggable event, which is highly relevant for the design of future clinical trials.
- Published
- 2020
22. The landscape of genomic alterations across childhood cancers
- Author
-
Gröbner, Susanne N., Worst, Barbara C., Weischenfeldt, Joachim, Buchhalter, Ivo, Kleinheinz, Kortine, Rudneva, Vasilisa A., Johann, Pascal D., Balasubramanian, Gnana Prakash, Segura-Wang, Maia, Brabetz, Sebastian, Bender, Sebastian, Hutter, Barbara, Sturm, Dominik, Pfaff, Elke, Hübschmann, Daniel, Zipprich, Gideon, Heinold, Michael, Eils, Jürgen, Lawerenz, Christian, Erkek, Serap, Lambo, Sander, Waszak, Sebastian, Blattmann, Claudia, Borkhardt, Arndt, Kuhlen, Michaela, Eggert, Angelika, Fulda, Simone, Gessler, Manfred, Wegert, Jenny, Kappler, Roland, Baumhör, Daniel, Burdach, Stefan, Kirschner-Schwabe, Renate, Kontny, Udo, Kulozik, Andreas E., Lohmann, Dietmar, Hettmer, Simone, Eckert, Cornelia, Bielack, Stefan, Nathrath, Michaela, Niemeyer, Charlotte, Richter, Gunther H., Schulte, Johannes, Siebert, Reiner, Westermann, Frank, Molenaar, Jan J., Vassal, Gilles, Witt, Hendrik, Burkhardt, Birgit, Kratz, Christian P., Witt, Olaf, van Tilburg, Cornelis M., Kramm, Christof M., Fleischhack, Gudrun, Dirksen, Uta, Rutkowski, Stefan, Fruhwald, Michael, von Hoff, Katja, Wolf, Stephan, Klingebiel, Thomas, Koscielniak, Ewa, Landgraf, Pablo, Koster, Jan, Resnick, Adam C., Zhang, Jinghui, Liu, Yanling, Zhou, Xin, Waanders, Angela J., Zwijnenburg, Danny A., Raman, Pichai, Brors, Benedikt, Weber, Ursula D., Northcott, Paul A., Pajtler, Kristian W., Kool, Marcel, Piro, Rosario M., Korbel, Jan O., Schlesner, Matthias, Eils, Roland, Jones, David T. W., Lichter, Peter, Chavez, Lukas, Zapatka, Marc, Pfister, Stefan M., and ICGC PedBrain-Seq Project ICGC
- Subjects
Krebs ,Tumor suppressor protein p53 ,%22">Krebs ,Neoplasms ,Genetics ,Kind ,Translational research ,Predictive markers ,Paediatric cancer ,DDC 570 / Life sciences ,ddc:570 ,Cancer genomics ,Jugend ,ddc:610 ,Child ,DDC 610 / Medicine & health ,Oncogenesis ,Protein p53 - Abstract
Pan-cancer analyses that examine commonalities and differences among various cancer types have emerged as a powerful way to obtain novel insights into cancer biology. Here we present a comprehensive analysis of genetic alterations in a pan-cancer cohort including 961 tumours from children, adolescents, and young adults, comprising 24 distinct molecular types of cancer. Using a standardized workflow, we identified marked differences in terms of mutation frequency and significantly mutated genes in comparison to previously analysed adult cancers. Genetic alterations in 149 putative cancer driver genes separate the tumours into two classes: small mutation and structural/copy-number variant (correlating with germline variants). Structural variants, hyperdiploidy, and chromothripsis are linked to TP53 mutation status and mutational signatures. Our data suggest that 7–8% of the children in this cohort carry an unambiguous predisposing germline variant and that nearly 50% of paediatric neoplasms harbour a potentially druggable event, which is highly relevant for the design of future clinical trials., publishedVersion
- Published
- 2018
- Full Text
- View/download PDF
23. Subgroup-specific structural variation across 1,000 medulloblastoma genomes
- Author
-
Northcott, Paul A, Shih, David JH, Peacock, John, Garzia, Livia, Morrissy, A Sorana, Zichner, Thomas, Stütz, Adrian M, Korshunov, Andrey, Reimand, Jüri, Schumacher, Steven E, Beroukhim, Rameen, Ellison, David W, Marshall, Christian R, Lionel, Anath C, Mack, Stephen, Dubuc, Adrian, Yao, Yuan, Ramaswamy, Vijay, Luu, Betty, Rolider, Adi, Cavalli, Florence MG, Wang, Xin, Remke, Marc, Wu, Xiaochong, Chiu, Readman YB, Chu, Andy, Chuah, Eric, Corbett, Richard D, Hoad, Gemma R, Jackman, Shaun D, Li, Yisu, Lo, Allan, Mungall, Karen L, Nip, Ka Ming, Qian, Jenny Q, Raymond, Anthony GJ, Thiessen, Nina T, Varhol, Richard J, Birol, Inanc, Moore, Richard A, Mungall, Andrew J, Holt, Robert, Kawauchi, Daisuke, Roussel, Martine F, Kool, Marcel, Jones, David TW, Witt, Hendrick, Fernandez-L, Africa, Kenney, Anna M, Wechsler-Reya, Robert J, Dirks, Peter, Aviv, Tzvi, Grajkowska, Wieslawa A, Perek-Polnik, Marta, Haberler, Christine C, Delattre, Olivier, Reynaud, Stéphanie S, Doz, François F, Pernet-Fattet, Sarah S, Cho, Byung-Kyu, Kim, Seung-Ki, Wang, Kyu-Chang, Scheurlen, Wolfram, Eberhart, Charles G, Fèvre-Montange, Michelle, Jouvet, Anne, Pollack, Ian F, Fan, Xing, Muraszko, Karin M, Gillespie, G Yancey, Di Rocco, Concezio, Massimi, Luca, Michiels, Erna MC, Kloosterhof, Nanne K, French, Pim J, Kros, Johan M, Olson, James M, Ellenbogen, Richard G, Zitterbart, Karel, Kren, Leos, Thompson, Reid C, Cooper, Michael K, Lach, Boleslaw, McLendon, Roger E, Bigner, Darell D, Fontebasso, Adam, Albrecht, Steffen, Jabado, Nada, Lindsey, Janet C, Bailey, Simon, Gupta, Nalin, Weiss, William A, Bognár, László, Klekner, Almos, Van Meter, Timothy E, Kumabe, Toshihiro, Tominaga, Teiji, Elbabaa, Samer K, Leonard, Jeffrey R, and Rubin, Joshua B
- Subjects
Pediatric Research Initiative ,DNA Copy Number Variations ,Pediatric Cancer ,General Science & Technology ,Translocation ,Nerve Tissue Proteins ,Rare Diseases ,Genetic ,Transforming Growth Factor beta ,Gene Duplication ,Genetics ,Humans ,Hedgehog Proteins ,Cerebellar Neoplasms ,Fusion ,Child ,Cancer ,Oncogene Proteins ,Pediatric ,Genome ,NF-kappa B ,Neurosciences ,Proteins ,Genomics ,myc ,Brain Disorders ,Brain Cancer ,Genes ,5.1 Pharmaceuticals ,Genomic Structural Variation ,RNA ,Long Noncoding ,Development of treatments and therapeutic interventions ,Carrier Proteins ,Medulloblastoma ,Signal Transduction ,Human - Abstract
Medulloblastoma, the most common malignant paediatric brain tumour, is currently treated with nonspecific cytotoxic therapies including surgery, whole-brain radiation, and aggressive chemotherapy. As medulloblastoma exhibits marked intertumoural heterogeneity, with at least four distinct molecular variants, previous attempts to identify targets for therapy have been underpowered because of small samples sizes. Here we report somatic copy number aberrations (SCNAs) in 1,087 unique medulloblastomas. SCNAs are common in medulloblastoma, and are predominantly subgroup-enriched. The most common region of focal copy number gain is a tandem duplication of SNCAIP, a gene associated with Parkinson's disease, which is exquisitely restricted to Group 4α. Recurrent translocations of PVT1, including PVT1-MYC and PVT1-NDRG1, that arise through chromothripsis are restricted to Group 3. Numerous targetable SCNAs, including recurrent events targeting TGF-β signalling in Group 3, and NF-κB signalling in Group 4, suggest future avenues for rational, targeted therapy.
- Published
- 2012
24. Spectrum and prevalence of genetic predisposition in medulloblastoma: a retrospective genetic study and prospective validation in a clinical trial cohort
- Author
-
Waszak, Sebastian M, Northcott, Paul A, Buchhalter, Ivo, Robinson, Giles W, Sutter, Christian, Groebner, Susanne, Grund, Kerstin B, Brugières, Laurence, Jones, David TW, Pajtler, Kristian W, Morrissy, A Sorana, Kool, Marcel, Sturm, Dominik, Chavez, Lukas, Ernst, Aurelie, Brabetz, Sebastian, Hain, Michael, Zichner, Thomas, Segura-Wang, Maia, Weischenfeldt, Joachim, Rausch, Tobias, Mardin, Balca R, Zhou, Xin, Baciu, Cristina, Lawerenz, Christian, Chan, Jennifer A, Varlet, Pascale, Guerrini-Rousseau, Lea, Fults, Daniel W, Grajkowska, Wiesława, Hauser, Peter, Jabado, Nada, Ra, Young-Shin, Zitterbart, Karel, Shringarpure, Suyash S, De La Vega, Francisco M, Bustamante, Carlos D, Ng, Ho-Keung, Perry, Arie, MacDonald, Tobey J, Hernáiz Driever, Pablo, Bendel, Anne E, Bowers, Daniel C, McCowage, Geoffrey, Chintagumpala, Murali M, Cohn, Richard, Hassall, Timothy, Fleischhack, Gudrun, Eggen, Tone, Wesenberg, Finn, Feychting, Maria, Lannering, Birgitta, Schüz, Joachim, Johansen, Christoffer, Andersen, Tina V, Röösli, Martin, Kuehni, Claudia E, Grotzer, Michael, Kjaerheim, Kristina, Monoranu, Camelia M, Archer, Tenley C, Duke, Elizabeth, Pomeroy, Scott L, Shelagh, Redmond, Frank, Stephan, Sumerauer, David, Scheurlen, Wolfram, Ryzhova, Marina V, Milde, Till, Kratz, Christian P, Samuel, David, Zhang, Jinghui, Solomon, David A, Marra, Marco, Eils, Roland, Bartram, Claus R, Von Hoff, Katja, Rutkowski, Stefan, Ramaswamy, Vijay, Gilbertson, Richard J, Korshunov, Andrey, Taylor, Michael D, Lichter, Peter, Malkin, David, Gajjar, Amar, Korbel, Jan O, and Pfister, Stefan M
- Subjects
Adult ,Male ,Heredity ,Adolescent ,DNA Mutational Analysis ,Young Adult ,Predictive Value of Tests ,Risk Factors ,Exome Sequencing ,Biomarkers, Tumor ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Prospective Studies ,Cerebellar Neoplasms ,Child ,Germ-Line Mutation ,Retrospective Studies ,Models, Genetic ,Gene Expression Profiling ,Infant ,Reproducibility of Results ,DNA Methylation ,Progression-Free Survival ,3. Good health ,Pedigree ,Phenotype ,Child, Preschool ,Female ,Transcriptome ,Medulloblastoma - Abstract
BACKGROUND: Medulloblastoma is associated with rare hereditary cancer predisposition syndromes; however, consensus medulloblastoma predisposition genes have not been defined and screening guidelines for genetic counselling and testing for paediatric patients are not available. We aimed to assess and define these genes to provide evidence for future screening guidelines. METHODS: In this international, multicentre study, we analysed patients with medulloblastoma from retrospective cohorts (International Cancer Genome Consortium [ICGC] PedBrain, Medulloblastoma Advanced Genomics International Consortium [MAGIC], and the CEFALO series) and from prospective cohorts from four clinical studies (SJMB03, SJMB12, SJYC07, and I-HIT-MED). Whole-genome sequences and exome sequences from blood and tumour samples were analysed for rare damaging germline mutations in cancer predisposition genes. DNA methylation profiling was done to determine consensus molecular subgroups: WNT (MBWNT), SHH (MBSHH), group 3 (MBGroup3), and group 4 (MBGroup4). Medulloblastoma predisposition genes were predicted on the basis of rare variant burden tests against controls without a cancer diagnosis from the Exome Aggregation Consortium (ExAC). Previously defined somatic mutational signatures were used to further classify medulloblastoma genomes into two groups, a clock-like group (signatures 1 and 5) and a homologous recombination repair deficiency-like group (signatures 3 and 8), and chromothripsis was investigated using previously established criteria. Progression-free survival and overall survival were modelled for patients with a genetic predisposition to medulloblastoma. FINDINGS: We included a total of 1022 patients with medulloblastoma from the retrospective cohorts (n=673) and the four prospective studies (n=349), from whom blood samples (n=1022) and tumour samples (n=800) were analysed for germline mutations in 110 cancer predisposition genes. In our rare variant burden analysis, we compared these against 53 105 sequenced controls from ExAC and identified APC, BRCA2, PALB2, PTCH1, SUFU, and TP53 as consensus medulloblastoma predisposition genes according to our rare variant burden analysis and estimated that germline mutations accounted for 6% of medulloblastoma diagnoses in the retrospective cohort. The prevalence of genetic predispositions differed between molecular subgroups in the retrospective cohort and was highest for patients in the MBSHH subgroup (20% in the retrospective cohort). These estimates were replicated in the prospective clinical cohort (germline mutations accounted for 5% of medulloblastoma diagnoses, with the highest prevalence [14%] in the MBSHH subgroup). Patients with germline APC mutations developed MBWNT and accounted for most (five [71%] of seven) cases of MBWNT that had no somatic CTNNB1 exon 3 mutations. Patients with germline mutations in SUFU and PTCH1 mostly developed infant MBSHH. Germline TP53 mutations presented only in childhood patients in the MBSHH subgroup and explained more than half (eight [57%] of 14) of all chromothripsis events in this subgroup. Germline mutations in PALB2 and BRCA2 were observed across the MBSHH, MBGroup3, and MBGroup4 molecular subgroups and were associated with mutational signatures typical of homologous recombination repair deficiency. In patients with a genetic predisposition to medulloblastoma, 5-year progression-free survival was 52% (95% CI 40-69) and 5-year overall survival was 65% (95% CI 52-81); these survival estimates differed significantly across patients with germline mutations in different medulloblastoma predisposition genes. INTERPRETATION: Genetic counselling and testing should be used as a standard-of-care procedure in patients with MBWNT and MBSHH because these patients have the highest prevalence of damaging germline mutations in known cancer predisposition genes. We propose criteria for routine genetic screening for patients with medulloblastoma based on clinical and molecular tumour characteristics. FUNDING: German Cancer Aid; German Federal Ministry of Education and Research; German Childhood Cancer Foundation (Deutsche Kinderkrebsstiftung); European Research Council; National Institutes of Health; Canadian Institutes for Health Research; German Cancer Research Center; St Jude Comprehensive Cancer Center; American Lebanese Syrian Associated Charities; Swiss National Science Foundation; European Molecular Biology Organization; Cancer Research UK; Hertie Foundation; Alexander and Margaret Stewart Trust; V Foundation for Cancer Research; Sontag Foundation; Musicians Against Childhood Cancer; BC Cancer Foundation; Swedish Council for Health, Working Life and Welfare; Swedish Research Council; Swedish Cancer Society; the Swedish Radiation Protection Authority; Danish Strategic Research Council; Swiss Federal Office of Public Health; Swiss Research Foundation on Mobile Communication; Masaryk University; Ministry of Health of the Czech Republic; Research Council of Norway; Genome Canada; Genome BC; Terry Fox Research Institute; Ontario Institute for Cancer Research; Pediatric Oncology Group of Ontario; The Family of Kathleen Lorette and the Clark H Smith Brain Tumour Centre; Montreal Children's Hospital Foundation; The Hospital for Sick Children: Sonia and Arthur Labatt Brain Tumour Research Centre, Chief of Research Fund, Cancer Genetics Program, Garron Family Cancer Centre, MDT's Garron Family Endowment; BC Childhood Cancer Parents Association; Cure Search Foundation; Pediatric Brain Tumor Foundation; Brainchild; and the Government of Ontario.
25. Pattern of Relapse and Treatment Response in WNT- Activated Medulloblastoma
- Author
-
Tara McKeown, Eric Bouffet, Seung-Ki Kim, Amulya A. Nageswara Rao, Liana Nobre, Andrew S. Moore, Joshua B. Rubin, Toshihiro Kumabe, Maura Massimino, James M. Olson, Sara Khan, Rajeev Vibhakar, Ales Vicha, Ho Keung Ng, Lenka Krskova, Wiesława Grajkowska, Jason E. Cain, Joanna Trubicka, Enrique López-Aguilar, Shayna Zelcer, Marie Lise van Veelen-Vincent, Ji Yeoun Lee, Alexandre Vasiljevic, Young Shin Ra, Ian F. Pollack, Massimo Zollo, Kohei Fukuoka, Boleslaw Lach, Carlos Gilberto Carlotti, Claudia C. Faria, Jordan R. Hansford, Shin Jung, David Sumerauer, Paul A. Northcott, Roger E. McLendon, Kay Ka Wai Li, Michal Zapotocky, Annie Huang, Peter Hauser, William A. Weiss, Lola B. Chambless, Josef Zamecnik, Ronald L. Hamilton, Cécile Faure-Conter, Sarah Leary, Vijay Ramaswamy, Helen Wheeler, Jaume Mora, Pim J. French, Erwin G. Van Meir, Ute Bartels, Adriana Fonseca, Luca Massimi, Stephen Yip, Jaroslav Sterba, Caterina Giannini, Michael D. Taylor, Nalin Gupta, Uri Tabori, Cynthia Hawkins, Veronica Ferruci, Nobre, Liana, Zapotocky, Michal, Khan, Sara, Fukuoka, Kohei, Fonseca, Adriana, Mckeown, Tara, Sumerauer, David, Vicha, Ale, Grajkowska, Wieslawa A., Trubicka, Joanna, Ka Wai Li, Kay, Ng, Ho-Keung, Massimi, Luca, Yeoun Lee, Ji, Kim, Seung-Ki, Zelcer, Shayna, Vasiljevic, Alexandre, ́ cile Faure-Conter, Ce, Hauser, Peter, Lach, Boleslaw, van Veelen-Vincent, Marie-Lise, French, Pim J., Van Meir, Erwin G., Weiss, William A., Pollack, Nalin Gupta Ian F., Hamilton, Ronald L., Nageswara Rao, Amulya A., Giannini, Caterina, Rubin, Joshua B., Moore, Andrew S., Chambless, Lola B., Vibhakar, Rajeev, Shin Ra, Young, Massimino, Maura, Mclendon, Roger E., Wheeler, Helen, Zollo, Massimo, Ferrucci, Veronica, Kumabe, Toshihiro, Faria, Claudia C., Sterba, Jaroslav, Jung, Shin, ́ pez-Aguilar, Enrique Lo, Mora, Jaume, Carlotti, Carlos G., Olson, James M., Leary, Sarah, Cain, Jason, Krskova, Lenka, Zamecnik, Josef, Hawkins, Cynthia E., Tabori, Uri, Huang, Annie, Bartels, Ute, Northcott, Paul A., Taylor, Michael D., Yip, Stephen, Hansford, Jordan R., Bouffet, Eric, Vijay Ramaswamy, And, McKeown, Tara, Grajkowska, Wieslawa A, Li, Kay Ka Wai, Lee, Ji Yeoun, Faure-Conter, Cécile, French, Pim J, Van Meir, Erwin G, Weiss, William A, Gupta, Nalin, Pollack, Ian F, Hamilton, Ronald L, Nageswara Rao, Amulya A, Rubin, Joshua B, Moore, Andrew S, Chambless, Lola B, Ra, Young Shin, McLendon, Roger E, Ferruci, Veronica, Faria, Claudia C, López-Aguilar, Enrique, Carlotti, Carlos G, Olson, James M, Hawkins, Cynthia E, Northcott, Paul A, Taylor, Michael D, Hansford, Jordan R, Ramaswamy, Vijay, Neurosurgery, and Neurology
- Subjects
Oncology ,Male ,medicine.medical_treatment ,chemotherapy ,Lateral ventricles ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,Child ,Cancer ,lcsh:R5-920 ,Tumor ,Wnt signaling pathway ,Middle Aged ,Chemotherapy regimen ,Progression-Free Survival ,3. Good health ,Local ,5.1 Pharmaceuticals ,RECIDIVA LOCAL DE NEOPLASIA ,6.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Cohort ,Female ,Development of treatments and therapeutic interventions ,lcsh:Medicine (General) ,medicine.drug ,Human ,medicine.medical_specialty ,Treatment response ,Cyclophosphamide ,Adolescent ,medulloblastoma ,survival ,Article ,General Biochemistry, Genetics and Molecular Biology ,WNT ,03 medical and health sciences ,Clinical Research ,Internal medicine ,genomics ,medicine ,Biomarkers, Tumor ,Humans ,Ifosfamide ,Cerebellar Neoplasms ,Medulloblastoma ,Chemotherapy ,Antineoplastic Combined Chemotherapy Protocol ,business.industry ,subgroup ,Cerebellar Neoplasm ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,radiation ,Neoplasm Recurrence ,wingless ,prognosis ,Neoplasm Recurrence, Local ,business ,Biomarkers ,030217 neurology & neurosurgery - Abstract
SUMMARY Over the past decade, wingless-activated (WNT) medulloblastoma has been identified as a candidate for therapy de-escalation based on excellent survival; however, a paucity of relapses has precluded additional analyses of markers of relapse. To address this gap in knowledge, an international cohort of 93 molecularly confirmed WNT MB was assembled, where 5-year progression-free survival is 0.84 (95%, 0.763–0.925) with 15 relapsed individuals identified. Maintenance chemotherapy is identified as a strong predictor of relapse, with individuals receiving high doses of cyclophosphamide or ifosphamide having only one very late molecularly confirmed relapse (p = 0.032). The anatomical location of recurrence is metastatic in 12 of 15 relapses, with 8 of 12 metastatic relapses in the lateral ventricles. Maintenance chemotherapy, specifically cumulative cyclophosphamide doses, is a significant predictor of relapse across WNT MB. Future efforts to de-escalate therapy need to carefully consider not only the radiation dose but also the chemotherapy regimen and the propensity for metastatic relapses., Graphical Abstract, In Brief Nobre et al. analyze a clinically annotated cohort of 93 WNT-activated medulloblastoma using an integrated genomic approach. A maintenance chemotherapy regimen is the strongest predictor of relapse, and cumulative cyclophosphamide doses confer a reduced risk of relapse. Unlike other medulloblastoma subgroups, WNT-activated medulloblastoma recurs most frequently in the lateral ventricles.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.