Try a new search

Format these results:

Searched for:

person:karajm01

Total Results:

171


Clinical outcome of pediatric medulloblastoma patients with Li-Fraumeni syndrome

Kolodziejczak, Anna S; Guerrini-Rousseau, Lea; Planchon, Julien Masliah; Ecker, Jonas; Selt, Florian; Mynarek, Martin; Obrecht, Denise; Sill, Martin; Autry, Robert J; Zhao, Eric; Hirsch, Steffen; Amouyal, Elsa; Dufour, Christelle; Ayrault, Olivier; Torrejon, Jacob; Waszak, Sebastian M; Ramaswamy, Vijay; Pentikainen, Virve; Demir, Haci Ahmet; Clifford, Steven C; Schwalbe, Ed C; Massimi, Luca; Snuderl, Matija; Galbraith, Kristyn; Karajannis, Matthias A; Hill, Katherine; Li, Bryan K; Walsh, Mike; White, Christine L; Redmond, Shelagh; Loizos, Loizou; Jakob, Marcus; Kordes, Uwe R; Schmid, Irene; Hauer, Julia; Blattmann, Claudia; Filippidou, Maria; Piccolo, Gianluca; Scheurlen, Wolfram; Farrag, Ahmed; Grund, Kerstin; Sutter, Christian; Pietsch, Torsten; Frank, Stephan; Schewe, Denis M; Malkin, David; Ben-Arush, Myriam; Sehested, Astrid; Wong, Tai-Tong; Wu, Kuo-Sheng; Liu, Yen-Lin; Carceller, Fernando; Mueller, Sabine; Stoller, Schuyler; Taylor, Michael D; Tabori, Uri; Bouffet, Eric; Kool, Marcel; Sahm, Felix; von Deimling, Andreas; Korshunov, Andrey; von Hoff, Katja; Kratz, Christian P; Sturm, Dominik; Jones, David T W; Rutkowski, Stefan; van Tilburg, Cornelis M; Witt, Olaf; Bougeard, Gaëlle; Pajtler, Kristian W; Pfister, Stefan M; Bourdeaut, Franck; Milde, Till
BACKGROUND:The prognosis for Li-Fraumeni syndrome (LFS) patients with medulloblastoma (MB) is poor. Comprehensive clinical data for this patient group is lacking, challenging the development of novel therapeutic strategies. Here, we present clinical and molecular data on a retrospective cohort of pediatric LFS MB patients. METHODS:In this multinational, multicenter retrospective cohort study, LFS patients under 21 years with MB and class 5 or class 4 constitutional TP53 variants were included. TP53 mutation status, methylation subgroup, treatment, progression free- (PFS) and overall survival (OS), recurrence patterns, and incidence of subsequent neoplasms were evaluated. RESULTS:The study evaluated 47 LFS individuals diagnosed with MB, mainly classified as DNA methylation subgroup "SHH_3" (86%). The majority (74%) of constitutional TP53 variants represented missense variants. The 2- and 5-year (y-) PFS were 36% and 20%, and 2- and 5y-OS were 53% and 23%, respectively. Patients who received post-operative radiotherapy (RT) (2y-PFS: 44%, 2y-OS: 60%) or chemotherapy before RT (2y-PFS: 32%, 2y-OS: 48%) had significantly better clinical outcome then patients who were not treated with RT (2y-PFS: 0%, 2y-OS: 25%). Patients treated according to protocols including high-intensity chemotherapy and patients who received only maintenance-type chemotherapy showed similar outcomes (2y-PFS: 42% and 35%, 2y-OS: 68% and 53%, respectively). CONCLUSIONS:LFS MB patients have a dismal prognosis. In the presented cohort use of RT significantly increased survival rates, whereas chemotherapy intensity did not influence their clinical outcome. Prospective collection of clinical data and development of novel treatments are required to improve the outcome of LFS MB patients.
PMID: 37379234
ISSN: 1523-5866
CID: 5540312

Multicenter, prospective, phase II study of maintenance bevacizumab for children and adults with NF2-related schwannomatosis and progressive vestibular schwannoma

Plotkin, Scott R; Allen, Jeffrey; Dhall, Girish; Campian, Jian L; Clapp, D Wade; Fisher, Michael J; Jain, Rakesh K; Tonsgard, James; Ullrich, Nicole J; Thomas, Coretta; Edwards, Lloyd J; Korf, Bruce; Packer, Roger; Karajannis, Matthias A; Blakeley, Jaishri O
BACKGROUND:Prospective data on maintenance therapy with bevacizumab for persons with NF2-related schwannomatosis (NF2-SWN) is lacking. In this prospective multicenter phase II study, we evaluated the efficacy, safety, and tolerability of bevacizumab for maintenance therapy in children and adults with NF2-SWN and hearing loss due to vestibular schwannomas (VS). METHODS:Following induction therapy, participants received bevacizumab 5 mg/kg every 3 weeks for 18 months. Participants were monitored for changes in hearing, tumor size, and quality of life (QOL), and for adverse events. Hearing loss was defined as a statistically significant decline in word recognition score (WRS) or pure-tone average compared to the study baseline; tumor growth was defined as >20% increase in volume compared to baseline. RESULTS:Twenty participants with NF2-SWN (median age 23.5 years; range, 12.5-62.5 years) with hearing loss in the target ear (median WRS 70%, range 2%-94%) received maintenance bevacizumab. Freedom from hearing loss in the target ear was 95% after 48 weeks, 89% after 72 weeks, and 70% after 98 weeks. Freedom from tumor growth in the target VS was 94% after 48 weeks, 89% after 72 weeks, and 89% after 98 weeks. NF2-related QOL remained stable for 98 weeks whereas tinnitus-related distress decreased. Maintenance bevacizumab was well tolerated, with 3 participants (15%) discontinuing treatment due to adverse events. CONCLUSIONS:Maintenance bevacizumab (5 mg/kg every 3 weeks) is associated with high rates of hearing and tumor stability during 18 months of follow-up. No new unexpected adverse events related to bevacizumab were identified in this population.
PMCID:10398799
PMID: 37010875
ISSN: 1523-5866
CID: 5592292

Recurrent TRAK1::RAF1 Fusions in pediatric low-grade gliomas

Benhamida, Jamal K; Harmsen, Hannah J; Ma, Deqin; William, Christopher M; Li, Bryan K; Villafania, Liliana; Sukhadia, Purvil; Mullaney, Kerry A; Dewan, Michael C; Vakiani, Efsevia; Karajannis, Matthias A; Snuderl, Matija; Zagzag, David; Ladanyi, Marc; Rosenblum, Marc K; Bale, Tejus A
Fusions involving CRAF (RAF1) are infrequent oncogenic drivers in pediatric low-grade gliomas, rarely identified in tumors bearing features of pilocytic astrocytoma, and involving a limited number of known fusion partners. We describe recurrent TRAK1::RAF1 fusions, previously unreported in brain tumors, in three pediatric patients with low-grade glial-glioneuronal tumors. We present the associated clinical, histopathologic and molecular features. Patients were all female, aged 8 years, 15 months, and 10 months at diagnosis. All tumors were located in the cerebral hemispheres and predominantly cortical, with leptomeningeal involvement in 2/3 patients. Similar to previously described activating RAF1 fusions, the breakpoints in RAF1 all occurred 5' of the kinase domain, while the breakpoints in the 3' partner preserved the N-terminal kinesin-interacting domain and coiled-coil motifs of TRAK1. Two of the three cases demonstrated methylation profiles (v12.5) compatible with desmoplastic infantile ganglioglioma (DIG)/desmoplastic infantile astrocytoma (DIA) and have remained clinically stable and without disease progression/recurrence after resection. The remaining tumor was non-classifiable; with focal recurrence 14 months after initial resection; the patient remains symptom free and without further recurrence/progression (5 months post re-resection and 19 months from initial diagnosis). Our report expands the landscape of oncogenic RAF1 fusions in pediatric gliomas, which will help to further refine tumor classification and guide management of patients with these alterations.
PMID: 37399073
ISSN: 1750-3639
CID: 5539042

Clinical utility of whole-genome DNA methylation profiling as a primary molecular diagnostic assay for central nervous system tumors-A prospective study and guidelines for clinical testing

Galbraith, Kristyn; Vasudevaraja, Varshini; Serrano, Jonathan; Shen, Guomiao; Tran, Ivy; Abdallat, Nancy; Wen, Mandisa; Patel, Seema; Movahed-Ezazi, Misha; Faustin, Arline; Spino-Keeton, Marissa; Roberts, Leah Geiser; Maloku, Ekrem; Drexler, Steven A; Liechty, Benjamin L; Pisapia, David; Krasnozhen-Ratush, Olga; Rosenblum, Marc; Shroff, Seema; Boué, Daniel R; Davidson, Christian; Mao, Qinwen; Suchi, Mariko; North, Paula; Hopp, Amanda; Segura, Annette; Jarzembowski, Jason A; Parsons, Lauren; Johnson, Mahlon D; Mobley, Bret; Samore, Wesley; McGuone, Declan; Gopal, Pallavi P; Canoll, Peter D; Horbinski, Craig; Fullmer, Joseph M; Farooqui, Midhat S; Gokden, Murat; Wadhwani, Nitin R; Richardson, Timothy E; Umphlett, Melissa; Tsankova, Nadejda M; DeWitt, John C; Sen, Chandra; Placantonakis, Dimitris G; Pacione, Donato; Wisoff, Jeffrey H; Teresa Hidalgo, Eveline; Harter, David; William, Christopher M; Cordova, Christine; Kurz, Sylvia C; Barbaro, Marissa; Orringer, Daniel A; Karajannis, Matthias A; Sulman, Erik P; Gardner, Sharon L; Zagzag, David; Tsirigos, Aristotelis; Allen, Jeffrey C; Golfinos, John G; Snuderl, Matija
BACKGROUND/UNASSIGNED:Central nervous system (CNS) cancer is the 10th leading cause of cancer-associated deaths for adults, but the leading cause in pediatric patients and young adults. The variety and complexity of histologic subtypes can lead to diagnostic errors. DNA methylation is an epigenetic modification that provides a tumor type-specific signature that can be used for diagnosis. METHODS/UNASSIGNED:We performed a prospective study using DNA methylation analysis as a primary diagnostic method for 1921 brain tumors. All tumors received a pathology diagnosis and profiling by whole genome DNA methylation, followed by next-generation DNA and RNA sequencing. Results were stratified by concordance between DNA methylation and histopathology, establishing diagnostic utility. RESULTS/UNASSIGNED:Of the 1602 cases with a World Health Organization histologic diagnosis, DNA methylation identified a diagnostic mismatch in 225 cases (14%), 78 cases (5%) did not classify with any class, and in an additional 110 (7%) cases DNA methylation confirmed the diagnosis and provided prognostic information. Of 319 cases carrying 195 different descriptive histologic diagnoses, DNA methylation provided a definitive diagnosis in 273 (86%) cases, separated them into 55 methylation classes, and changed the grading in 58 (18%) cases. CONCLUSIONS/UNASSIGNED:DNA methylation analysis is a robust method to diagnose primary CNS tumors, improving diagnostic accuracy, decreasing diagnostic errors and inconclusive diagnoses, and providing prognostic subclassification. This study provides a framework for inclusion of DNA methylation profiling as a primary molecular diagnostic test into professional guidelines for CNS tumors. The benefits include increased diagnostic accuracy, improved patient management, and refinements in clinical trial design.
PMCID:10355794
PMID: 37476329
ISSN: 2632-2498
CID: 5536102

CLINICAL OUTCOME OF PEDIATRIC MEDULLOBLASTOMA PATIENTS WITH LI-FRAUMENI SYNDROME [Meeting Abstract]

Kolodziejczak, A; Guerrini-Rousseau, L; Planchon, J M; Ecker, J; Selt, F; Mynarek, M; Obrecht, D; Sill, M; Hirsch, S; Sturm, D; Waszak, S M; Ramaswamy, V; Pentikainen, V; Demir, H A; Clifford, S C; Schwalbe, E; Massimi, L; Snuderl, M; Galbraith, K; Karajannis, M A; Hill, K; Li, B; White, C L; Redmond, S; Loizos, L; Jakob, M; Kordes, U; Schmid, I; Hauer, J; Blattmann, C; Filippidou, M; Scheurlen, W; Kontny, U; Grund, K; Sutter, C; Pietsch, T; Van, Tilburg C M; Frank, S; Schewe, D M; Malkin, D; Taylor, M D; Tabori, U; Bouffet, E; Kool, M; Sahm, F; Von, Deimling A; Korshunov, A; Von, Hoff K; Kratz, C; Jones, D T W; Rutkowski, S; Witt, O; Bougeard, G; Pajtler, K W; Pfister, S M; Bourdeaut, F; Milde, T
PURPOSE: The prognosis for SHH-medulloblastoma (MB) patients with Li-Fraumeni syndrome (LFS) is poor. Due to lack of comprehensive data for these patients, it is challenging to establish effective therapeutic recommendations. We here describe the largest retrospective cohort of pediatric LFS SHH-MB patients to date and their clinical outcomes.
PATIENTS AND METHODS: N=31 patients with LFS SHH-MB were included in this retrospective multicenter study. TP53 variant type, clinical parameters including treatment modalities, event-free survival (EFS) and overall survival (OS), as well as recurrence patterns and incidence of secondary neoplasms, were evaluated.
RESULT(S): All LFS-MBs were classified as SHH subgroup, in 30/31 cases based on DNA methylation analysis. The majority of constitutional TP53 variants (72%) represented missense variants, and all except two truncating variants were located within the DNA-binding domain. 54% were large cell anaplastic, 69% gross totally resected and 81% had M0 status. The 2-(y)ear and 5-(y)ear EFS were 26% and 8,8%, respectively, and 2y- and 5y-OS 40% and 12%. Patients who received post-operative radiotherapy (RT) followed by chemotherapy (CT) showed significantly better outcomes (2y-EFS:43%) compared to patients who received CT before RT (30%) (p<0.05). The 2y-EFS and 2y-OS were similar when treated with protocols including high-dose chemotherapy (EFS:22%, OS:44%) compared to patients treated with maintenance-type chemotherapy (EFS:31%, OS:45%). Recurrence occurred in 73.3% of cases independent of resection or M-status, typically within the radiation field (75% of RT-treated patients). Secondary malignancies developed in 12.5% and were cause of death in all affected patients.
CONCLUSION(S): Patients with LFS-MBs have a dismal prognosis. This retrospective study suggests that upfront RT may increase EFS, while intensive therapeutic approaches including high-dose chemotherapy did not translate into increased survival of this patient group. To improve outcomes of LFS-MB patients, prospective collection of clinical data and development of treatment guidelines are required
EMBASE:638510949
ISSN: 1523-5866
CID: 5292022

PATZ1 fusions define a novel molecularly distinct neuroepithelial tumor entity with a broad histological spectrum

Alhalabi, Karam T; Stichel, Damian; Sievers, Philipp; Peterziel, Heike; Sommerkamp, Alexander C; Sturm, Dominik; Wittmann, Andrea; Sill, Martin; Jäger, Natalie; Beck, Pengbo; Pajtler, Kristian W; Snuderl, Matija; Jour, George; Delorenzo, Michael; Martin, Allison M; Levy, Adam; Dalvi, Nagma; Hansford, Jordan R; Gottardo, Nicholas G; Uro-Coste, Emmanuelle; Maurage, Claude-Alain; Godfraind, Catherine; Vandenbos, Fanny; Pietsch, Torsten; Kramm, Christof; Filippidou, Maria; Kattamis, Antonis; Jones, Chris; Øra, Ingrid; Mikkelsen, Torben Stamm; Zapotocky, Michal; Sumerauer, David; Scheie, David; McCabe, Martin; Wesseling, Pieter; Tops, Bastiaan B J; Kranendonk, Mariëtte E G; Karajannis, Matthias A; Bouvier, Nancy; Papaemmanuil, Elli; Dohmen, Hildegard; Acker, Till; von Hoff, Katja; Schmid, Simone; Miele, Evelina; Filipski, Katharina; Kitanovski, Lidija; Krskova, Lenka; Gojo, Johannes; Haberler, Christine; Alvaro, Frank; Ecker, Jonas; Selt, Florian; Milde, Till; Witt, Olaf; Oehme, Ina; Kool, Marcel; von Deimling, Andreas; Korshunov, Andrey; Pfister, Stefan M; Sahm, Felix; Jones, David T W
Large-scale molecular profiling studies in recent years have shown that central nervous system (CNS) tumors display a much greater heterogeneity in terms of molecularly distinct entities, cellular origins and genetic drivers than anticipated from histological assessment. DNA methylation profiling has emerged as a useful tool for robust tumor classification, providing new insights into these heterogeneous molecular classes. This is particularly true for rare CNS tumors with a broad morphological spectrum, which are not possible to assign as separate entities based on histological similarity alone. Here, we describe a molecularly distinct subset of predominantly pediatric CNS neoplasms (n = 60) that harbor PATZ1 fusions. The original histological diagnoses of these tumors covered a wide spectrum of tumor types and malignancy grades. While the single most common diagnosis was glioblastoma (GBM), clinical data of the PATZ1-fused tumors showed a better prognosis than typical GBM, despite frequent relapses. RNA sequencing revealed recurrent MN1:PATZ1 or EWSR1:PATZ1 fusions related to (often extensive) copy number variations on chromosome 22, where PATZ1 and the two fusion partners are located. These fusions have individually been reported in a number of glial/glioneuronal tumors, as well as extracranial sarcomas. We show here that they are more common than previously acknowledged, and together define a biologically distinct CNS tumor type with high expression of neural development markers such as PAX2, GATA2 and IGF2. Drug screening performed on the MN1:PATZ1 fusion-bearing KS-1 brain tumor cell line revealed preliminary candidates for further study. In summary, PATZ1 fusions define a molecular class of histologically polyphenotypic neuroepithelial tumors, which show an intermediate prognosis under current treatment regimens.
PMID: 34417833
ISSN: 1432-0533
CID: 5006432

Molecular profiling of pediatric meningiomas shows tumor characteristics distinct from adult meningiomas

Kirches, Elmar; Sahm, Felix; Korshunov, Andrey; Bluecher, Christina; Waldt, Natalie; Kropf, Siegfried; Schrimpf, Daniel; Sievers, Philipp; Stichel, Damian; Schüller, Ulrich; Schittenhelm, Jens; Riemenschneider, Markus J; Karajannis, Matthias A; Perry, Arie; Pietsch, Torsten; Boekhoff, Svenja; Capper, David; Beck, Katja; Paramasivam, Nagarajan; Schlesner, Matthias; Brastianos, Priscilla K; Müller, Hermann L; Pfister, Stefan M; Mawrin, Christian
In contrast to adults, meningiomas are uncommon tumors in childhood and adolescence. Whether adult and pediatric meningiomas differ on a molecular level is unclear. Here we report detailed genomic analyses of 37 pediatric meningiomas by sequencing and DNA methylation profiling. Histologically, the series was dominated by meningioma subtypes with aggressive behavior, with 70% of patients suffering from WHO grade II or III meningiomas. The most frequent cytogenetic aberrations were loss of chromosomes 22 (23/37 [62%]), 1 (9/37 [24%]), 18 (7/37 [19%]), and 14 (5/37 [14%]). Tumors with NF2 alterations exhibited overall increased chromosomal instability. Unsupervised clustering of DNA methylation profiles revealed separation into three groups: designated group 1 composed of clear cell and papillary meningiomas, whereas group 2A comprised predominantly atypical meningiomas and group 2B enriched for rare high-grade subtypes (rhabdoid, chordoid). Meningiomas from NF2 patients clustered exclusively within groups 1 and 2A. When compared with a dataset of 105 adult meningiomas, the pediatric meningiomas largely grouped separately. Targeted panel DNA sequencing of 34 tumors revealed frequent NF2 alterations, while other typical alterations found in adult non-NF2 tumors were absent. These data demonstrate that pediatric meningiomas are characterized by molecular features distinct from adult tumors.
PMID: 34495383
ISSN: 1432-0533
CID: 5011992

Phase 0 Clinical Trial of Everolimus in Patients with Vestibular Schwannoma or Meningioma

Karajannis, Matthias A; Mauguen, Audrey; Maloku, Ekrem; Xu, Qingwen; Dunbar, Erin M; Plotkin, Scott R; Yaffee, Anna; Wang, Shiyang; Roland, J Thomas; Sen, Chandranath; Placantonakis, Dimitris G; Golfinos, John G; Allen, Jeffrey C; Vitanza, Nicholas A; Chiriboga, Luis A; Schneider, Robert J; Deng, Jingjing; Neubert, Thomas A; Goldberg, Judith D; Zagzag, David; Giancotti, Filippo G; Blakeley, Jaishri O
Inhibition of mTORC1 signaling has been shown to diminish growth of meningiomas and schwannomas in preclinical studies, and clinical data suggest that everolimus, an orally administered mTORC1 inhibitor, may slow tumor progression in a subset of NF2 patients with vestibular schwannoma (VS). To assess the pharmacokinetics, pharmacodynamics and potential mechanisms of treatment resistance, we performed a pre-surgical (phase 0) clinical trial of everolimus in patients undergoing elective surgery for VS or meningiomas. Eligible patients with meningioma or VS requiring tumor resection enrolled on study received everolimus 10 mg daily for 10 days immediately prior to surgery. Everolimus blood levels were determined immediately prior to and after surgery. Tumor samples were collected intraoperatively. Ten patients completed protocol therapy. Median pre- and post-operative blood levels of everolimus were found to be in a high therapeutic range (17.4 ng/ml and 9.4 ng/ml, respectively). Median tumor tissue drug concentration determined by mass spectrometry was 24.3 pg/mg (range 9.2-169.2). We observed only partial inhibition of phospho-S6 in the treated tumors, indicating incomplete target inhibition compared to control tissues from untreated patients (p=0.025). Everolimus led to incomplete inhibition of mTORC1 and downstream signaling. These data may explain the limited anti-tumor effect of everolimus observed in clinical studies for NF2 patients and will inform the design of future pre-clinical and clinical studies targeting mTORC1 in meningiomas and schwannomas.
PMID: 34224367
ISSN: 1538-8514
CID: 4932142

Pediatric spinal cord gliomas-low grade but high risk for recurrence: should we treat them differently from intracranial low-grade gliomas? [Comment]

Sait, Sameer Farouk; Karajannis, Matthias A
PMID: 33705546
ISSN: 1523-5866
CID: 4931902

A Phase I and surgical study of ribociclib and everolimus in children with recurrent or refractory malignant brain tumors: a Pediatric Brain Tumor Consortium Study

DeWire, Mariko D; Fuller, Christine; Campagne, Olivia; Lin, Tong; Pan, Haitao; Poussaint, Tina Young; Baxter, Patricia A; Hwang, Eugene I; Bukowinski, Andrew; Dorris, Kathleen; Hoffman, Lindsey; Waanders, Angela J; Karajannis, Matthias A; Stewart, Clinton F; Onar-Thomas, Arzu; Fouladi, Maryam; Dunkel, Ira J
PURPOSE/OBJECTIVE:Genomic aberrations in cell cycle and PI3K pathways are commonly observed in pediatric brain tumors. This study determined the maximum tolerated dose (MTD)/Recommended phase 2 Dose (RP2D) of ribociclib and everolimus and characterized single-agent ribociclib concentrations in plasma and tumor in children undergoing resection. EXPERIMENTAL DESIGN/METHODS:) for 7-10 days pre-operatively followed by enrollment on the phase I study. Pharmacokinetics were analyzed for both cohorts. RESULTS:for 21 and 28 days, respectively. Steady state everolimus exposures with ribociclib were 2.5-fold higher than everolimus administered alone. Ribociclib plasma, tumor concentrations and CSF samples were collected. The mean tumor-to-plasma ratio of ribociclib was 19.8 (range: 2.22- 53.4). CONCLUSIONS:Ribociclib and everolimus were well tolerated and demonstrated similar pharmacokinetic properties as in adults. Potential therapeutic ribociclib concentrations could be achieved in CSF and tumor tissue, although interpatient variability was observed.
PMID: 33547201
ISSN: 1557-3265
CID: 4777162