Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Priscilla K. Brastianos is active.

Publication


Featured researches published by Priscilla K. Brastianos.


Nature Genetics | 2013

Genomic sequencing of meningiomas identifies oncogenic SMO and AKT1 mutations

Priscilla K. Brastianos; Peleg Horowitz; Sandro Santagata; Robert T. Jones; Aaron McKenna; Gad Getz; Keith L. Ligon; Emanuele Palescandolo; Paul Van Hummelen; Matthew Ducar; Alina Raza; Ashwini Sunkavalli; Laura E. MacConaill; Anat Stemmer-Rachamimov; David N. Louis; William C. Hahn; Ian F. Dunn; Rameen Beroukhim

Meningiomas are the most common primary nervous system tumor. The tumor suppressor NF2 is disrupted in approximately half of all meningiomas, but the complete spectrum of genetic changes remains undefined. We performed whole-genome or whole-exome sequencing on 17 meningiomas and focused sequencing on an additional 48 tumors to identify and validate somatic genetic alterations. Most meningiomas had simple genomes, with fewer mutations, rearrangements and copy-number alterations than reported in other tumors in adults. However, several meningiomas harbored more complex patterns of copy-number changes and rearrangements, including one tumor with chromothripsis. We confirmed focal NF2 inactivation in 43% of tumors and found alterations in epigenetic modifiers in an additional 8% of tumors. A subset of meningiomas lacking NF2 alterations harbored recurrent oncogenic mutations in AKT1 (p.Glu17Lys) and SMO (p.Trp535Leu) and exhibited immunohistochemical evidence of activation of these pathways. These mutations were present in therapeutically challenging tumors of the skull base and higher grade. These results begin to define the spectrum of genetic alterations in meningiomas and identify potential therapeutic targets.


Cancer Discovery | 2015

Genomic Characterization of Brain Metastases Reveals Branched Evolution and Potential Therapeutic Targets

Priscilla K. Brastianos; Scott L. Carter; Sandro Santagata; Daniel P. Cahill; Amaro Taylor-Weiner; Robert T. Jones; Eliezer M. Van Allen; Michael S. Lawrence; Peleg Horowitz; Kristian Cibulskis; Keith L. Ligon; Josep Tabernero; Joan Seoane; Elena Martinez-Saez; William T. Curry; Ian F. Dunn; Sun Ha Paek; Sung-Hye Park; Aaron McKenna; Aaron Chevalier; Mara Rosenberg; Fred G. Barker; Corey M. Gill; Paul Van Hummelen; Aaron R. Thorner; Bruce E. Johnson; Mai P. Hoang; Toni K. Choueiri; Sabina Signoretti; Carrie Sougnez

UNLABELLED Brain metastases are associated with a dismal prognosis. Whether brain metastases harbor distinct genetic alterations beyond those observed in primary tumors is unknown. We performed whole-exome sequencing of 86 matched brain metastases, primary tumors, and normal tissue. In all clonally related cancer samples, we observed branched evolution, where all metastatic and primary sites shared a common ancestor yet continued to evolve independently. In 53% of cases, we found potentially clinically informative alterations in the brain metastases not detected in the matched primary-tumor sample. In contrast, spatially and temporally separated brain metastasis sites were genetically homogenous. Distal extracranial and regional lymph node metastases were highly divergent from brain metastases. We detected alterations associated with sensitivity to PI3K/AKT/mTOR, CDK, and HER2/EGFR inhibitors in the brain metastases. Genomic analysis of brain metastases provides an opportunity to identify potentially clinically informative alterations not detected in clinically sampled primary tumors, regional lymph nodes, or extracranial metastases. SIGNIFICANCE Decisions for individualized therapies in patients with brain metastasis are often made from primary-tumor biopsies. We demonstrate that clinically actionable alterations present in brain metastases are frequently not detected in primary biopsies, suggesting that sequencing of primary biopsies alone may miss a substantial number of opportunities for targeted therapy.


Nature Genetics | 2014

Exome sequencing identifies BRAF mutations in papillary craniopharyngiomas

Priscilla K. Brastianos; Amaro Taylor-Weiner; Peter Manley; Robert T. Jones; Dora Dias-Santagata; Aaron R. Thorner; Michael S. Lawrence; Fausto J. Rodriguez; Lindsay A. Bernardo; Laura Schubert; Ashwini Sunkavalli; Nick Shillingford; Monica L. Calicchio; Hart G.W. Lidov; Hala Taha; Maria Martinez-Lage; Mariarita Santi; Phillip B. Storm; John Y. K. Lee; James N. Palmer; Nithin D. Adappa; R. Michael Scott; Ian F. Dunn; Edward R. Laws; Chip Stewart; Keith L. Ligon; Mai P. Hoang; Paul Van Hummelen; William C. Hahn; David N. Louis

Craniopharyngiomas are epithelial tumors that typically arise in the suprasellar region of the brain. Patients experience substantial clinical sequelae from both extension of the tumors and therapeutic interventions that damage the optic chiasm, the pituitary stalk and the hypothalamic area. Using whole-exome sequencing, we identified mutations in CTNNB1 (β-catenin) in nearly all adamantinomatous craniopharyngiomas examined (11/12, 92%) and recurrent mutations in BRAF (resulting in p.Val600Glu) in all papillary craniopharyngiomas (3/3, 100%). Targeted genotyping revealed BRAF p.Val600Glu in 95% of papillary craniopharyngiomas (36 of 39 tumors) and mutation of CTNNB1 in 96% of adamantinomatous craniopharyngiomas (51 of 53 tumors). The CTNNB1 and BRAF mutations were clonal in each tumor subtype, and we detected no other recurrent mutations or genomic aberrations in either subtype. Adamantinomatous and papillary craniopharyngiomas harbor mutations that are mutually exclusive and clonal. These findings have important implications for the diagnosis and treatment of these neoplasms.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Genomic analysis of diffuse pediatric low-grade gliomas identifies recurrent oncogenic truncating rearrangements in the transcription factor MYBL1

Lori A. Ramkissoon; Peleg Horowitz; Justin M. Craig; Shakti Ramkissoon; Benjamin E. Rich; Steven E. Schumacher; Aaron McKenna; Michael S. Lawrence; Guillaume Bergthold; Priscilla K. Brastianos; Barbara Tabak; Matthew Ducar; Paul Van Hummelen; Laura E. MacConaill; Tina Pouissant-Young; Yoon-Jae Cho; Hala Taha; Madeha Mahmoud; Daniel C. Bowers; Linda R. Margraf; Uri Tabori; Cynthia Hawkins; Roger J. Packer; D. Ashley Hill; Scott L. Pomeroy; Charles G. Eberhart; Ian F. Dunn; Liliana Goumnerova; Gad Getz; Jennifer A. Chan

Pediatric low-grade gliomas (PLGGs) are among the most common solid tumors in children but, apart from BRAF kinase mutations or duplications in specific subclasses, few genetic driver events are known. Diffuse PLGGs comprise a set of uncommon subtypes that exhibit invasive growth and are therefore especially challenging clinically. We performed high-resolution copy-number analysis on 44 formalin-fixed, paraffin-embedded diffuse PLGGs to identify recurrent alterations. Diffuse PLGGs exhibited fewer such alterations than adult low-grade gliomas, but we identified several significantly recurrent events. The most significant event, 8q13.1 gain, was observed in 28% of diffuse astrocytoma grade IIs and resulted in partial duplication of the transcription factor MYBL1 with truncation of its C-terminal negative-regulatory domain. A similar recurrent deletion-truncation breakpoint was identified in two angiocentric gliomas in the related gene v-myb avian myeloblastosis viral oncogene homolog (MYB) on 6q23.3. Whole-genome sequencing of a MYBL1-rearranged diffuse astrocytoma grade II demonstrated MYBL1 tandem duplication and few other events. Truncated MYBL1 transcripts identified in this tumor induced anchorage-independent growth in 3T3 cells and tumor formation in nude mice. Truncated transcripts were also expressed in two additional tumors with MYBL1 partial duplication. Our results define clinically relevant molecular subclasses of diffuse PLGGs and highlight a potential role for the MYB family in the biology of low-grade gliomas.


Lancet Infectious Diseases | 2006

Tuberculosis-associated haemophagocytic syndrome

Priscilla K. Brastianos; Jordan W. Swanson; Michael Torbenson; John Sperati; Petros C. Karakousis

Haemophagocytic syndrome is a disorder characterised by fevers, lymphadenopathy, hepatosplenomegaly, cytopenias, and hyperferritinaemia due to dysregulated activation and proliferation of macrophages, leading to uncontrolled phagocytosis of platelets, erythrocytes, lymphocytes, and their haematopoietic precursors throughout the reticuloendothelial system. Primary or familial haemophagocytic syndrome appears to have a genetic aetiology, whereas secondary haemophagocytic syndrome may be associated with malignancy, autoimmune disease, or infection. Epstein-Barr virus is the most common infectious aetiology implicated in haemophagocytic syndrome, but the syndrome has been associated with a variety of other viral, bacterial, and parasitic pathogens. We describe a case of haemophagocytic syndrome associated with disseminated Mycobacterium tuberculosis. We review all cases of M tuberculosis-associated haemophagocytic syndrome reported in the English language literature and discuss important issues pertaining to the epidemiology, diagnosis, and management of this disease.


Journal of Thoracic Oncology | 2015

Alectinib Salvages CNS Relapses in ALK-Positive Lung Cancer Patients Previously Treated with Crizotinib and Ceritinib

Justin F. Gainor; Carol A. Sherman; Kathryn Willoughby; Jennifer A. Logan; Elizabeth Kennedy; Priscilla K. Brastianos; Andrew S. Chi; Alice T. Shaw

Background: Leptomeningeal metastases (LM) are an increasingly frequent and devastating complication of anaplastic lymphoma kinase (ALK)-rearranged non–small-cell lung cancer (NSCLC). Currently, the optimal management of LM in ALK-positive patients remains poorly understood as these patients have been routinely excluded from clinical trials. Methods: We describe four ALK-positive patients with LM who were treated with the next-generation ALK inhibitor alectinib through single-patient, compassionate use protocols at two institutions. All patients had previously been treated with both FDA-approved ALK inhibitors—crizotinib and ceritinib. Patients received alectinib at a starting dose of 600 mg twice daily. Results: Four ALK-positive NSCLC patients with symptomatic leptomeningeal disease were identified. Three of four patients experienced significant clinical and radiographic improvements in LM upon treatment with alectinib. A fourth patient had stable intracranial disease for 4 months before eventual systemic disease progression. Overall, alectinib was well tolerated. One patient required dose reduction due to grade 2 hyperbilirubinemia. Conclusions: Alectinib is active in ALK-rearranged NSCLC patients with LM, including in patients previously treated with crizotinib and ceritinib. Additional prospective studies of alectinib in ALK-positive patients with LM are warranted.


Science | 2017

Decoupling genetics, lineages, and microenvironment in IDH-mutant gliomas by single-cell RNA-seq.

Andrew S. Venteicher; Itay Tirosh; Christine Hebert; Keren Yizhak; Cyril Neftel; Mariella G. Filbin; Volker Hovestadt; Leah E. Escalante; McKenzie L. Shaw; Christopher Rodman; Shawn M. Gillespie; Danielle Dionne; Christina C. Luo; Hiranmayi Ravichandran; Ravindra Mylvaganam; Christopher Mount; Maristela L. Onozato; Brian V. Nahed; Hiroaki Wakimoto; William T. Curry; A. John Iafrate; Miguel Rivera; Matthew P. Frosch; Todd R. Golub; Priscilla K. Brastianos; Gad Getz; Anoop P. Patel; Michelle Monje; Daniel P. Cahill; Orit Rozenblatt-Rosen

Single-cell RNA sequencing identifies a common origin for specific types of human glioma brain tumors. Effects of the tumor microenvironment Glioma brain tumors that carry mutant copies of the IDH gene can be subdivided into two major classes. However, the development of and differences between these two classes are not well characterized. Venteicher et al. coupled bulk sequencing and publicly available data with single-cell RNA sequencing data on glioma patient tissue samples. They identified a common lineage program that is shared between glioma subtypes. This suggests that the observed differences between the two glioma classes originate from lineage-specific genetic changes and the tumor microenvironment. Science, this issue p. eaai8478 INTRODUCTION Tumor fitness, evolution, and resistance to therapy are governed by selection of malignant cells with specific genotypes, by expression programs related to cellular phenotypes, and by influences of the tumor microenvironment (TME). Although bulk tumor analysis can interrogate the genetic state of tumor cells with high precision, bulk expression profiles average the diverse cells within each tumor, thereby masking critical differences and providing limited insight into cancer cell programs and TME influences. Single-cell RNA sequencing (scRNA-seq) can help to address those challenges but incurs financial and logistic considerations, including the time required to accrue large cohorts of fresh tumor specimen for single-cell analysis. RATIONALE We reasoned that scRNA-seq of a limited number of representative tumors could be combined with bulk data from large cohorts to decipher differences between tumor subclasses. In this approach, bulk samples collected for large cohorts, such as from The Cancer Genome Atlas (TCGA), are first used to define the combined effects of differences in cancer cell genotypes, phenotypes, and the composition of the TME. Single-cell analysis of a limited set of representative tumors is then used to distinguish those effects. We applied this approach to understand the differences between two types of isocitrate dehydrogenase (IDH)–mutant gliomas: astrocytoma (IDH-A) and oligodendroglioma (IDH-O). IDH-A and IDH-O are distinguished by co-occurring signature genetic events and by histopathology and are thought to recapitulate distinct glial lineages. By combining 9879 scRNA-seq profiles from 10 IDH-A tumors, 4347 scRNA-seq profiles from six IDH-O tumors, and 165 TCGA bulk RNA profiles, we could decipher differences between these two tumor types at single-cell resolution. RESULTS We find that differences in bulk expression profiles between IDH-A and IDH-O are primarily explained by the impact of signature genetic events and TME composition, but not by distinct expression programs of glial lineages in the malignant cells. We infer that both IDH-A and IDH-O share the same developmental hierarchy, consisting in each case of three subpopulations of malignant cells: nonproliferating cells differentiated along the astrocytic and oligodendrocytic lineages, and proliferative undifferentiated cells that resemble neural stem/progenitor cells. By analyzing tumors of different clinical grades, we observe that higher-grade tumors present enhanced proliferation, larger pools of undifferentiated glioma cells, and an increase in macrophage over microglia programs in the TME. CONCLUSION Our approach provides a general framework to decipher differences between classes of human tumors by decoupling cancer cell genotypes, phenotypes, and the composition of the TME. The shared glial lineages and developmental hierarchies observed in IDH-A and IDH-O suggest a common progenitor for all IDH-mutant gliomas, shedding light on a long-standing debate in gliomagenesis. In contrast to the similarity in glial lineages, IDH-A and IDH-O differ significantly in their TME, and in particular in the abundance of microglia/macrophage cells. Microglia and macrophages also differ between IDH-A tumors of different grades. Our study redefines the cellular composition of human IDH-mutant gliomas, with important implications for disease management. Single-cell RNA-seq of IDH-mutant gliomas reveals tumor architecture. (Top) Human samples were dissociated and analyzed by scRNA-seq. (Bottom) IDH-O and IDH-A differ in genetics and TME but are both primarily composed of three main types of malignant cells: cycling stem-like cells and noncycling astrocyte-like and oligodendrocyte-like cells. Tumor progression is associated with increased proliferation, decreased differentiation, and increase in macrophages over microglia in the TME. Tumor subclasses differ according to the genotypes and phenotypes of malignant cells as well as the composition of the tumor microenvironment (TME). We dissected these influences in isocitrate dehydrogenase (IDH)–mutant gliomas by combining 14,226 single-cell RNA sequencing (RNA-seq) profiles from 16 patient samples with bulk RNA-seq profiles from 165 patient samples. Differences in bulk profiles between IDH-mutant astrocytoma and oligodendroglioma can be primarily explained by distinct TME and signature genetic events, whereas both tumor types share similar developmental hierarchies and lineages of glial differentiation. As tumor grade increases, we find enhanced proliferation of malignant cells, larger pools of undifferentiated glioma cells, and an increase in macrophage over microglia expression programs in TME. Our work provides a unifying model for IDH-mutant gliomas and a general framework for dissecting the differences among human tumor subclasses.


Journal of the National Cancer Institute | 2016

Dramatic Response of BRAF V600E Mutant Papillary Craniopharyngioma to Targeted Therapy

Priscilla K. Brastianos; Ganesh M. Shankar; Corey M. Gill; Amaro Taylor-Weiner; Naema Nayyar; David J. Panka; Ryan J. Sullivan; Dennie T. Frederick; Malak Abedalthagafi; Pamela S. Jones; Ian F. Dunn; Brian V. Nahed; Javier Romero; David N. Louis; Gad Getz; Daniel P. Cahill; Sandro Santagata; William T. Curry; Fred G. Barker

We recently reported that BRAF V600E is the principal oncogenic driver of papillary craniopharyngioma, a highly morbid intracranial tumor commonly refractory to treatment. Here, we describe our treatment of a man age 39 years with multiply recurrent BRAF V600E craniopharyngioma using dabrafenib (150mg, orally twice daily) and trametinib (2mg, orally twice daily). After 35 days of treatment, tumor volume was reduced by 85%. Mutations that commonly mediate resistance to MAPK pathway inhibition were not detected in a post-treatment sample by whole exome sequencing. A blood-based BRAF V600E assay detected circulating BRAF V600E in the patients blood. Re-evaluation of the existing management paradigms for craniopharyngioma is warranted, as patient morbidity might be reduced by noninvasive mutation testing and neoadjuvant-targeted treatment.


Neuro-oncology | 2016

Oncogenic PI3K mutations are as common as AKT1 and SMO mutations in meningioma

Malak Abedalthagafi; Wenya Linda Bi; Ayal A. Aizer; Parker H. Merrill; Ryan Brewster; Pankaj K. Agarwalla; Marc L. Listewnik; Dora Dias-Santagata; Aaron R. Thorner; Paul Van Hummelen; Priscilla K. Brastianos; David A. Reardon; Patrick Y. Wen; Ossama Al-Mefty; Shakti Ramkissoon; Rebecca D. Folkerth; Keith L. Ligon; Azra H. Ligon; Brian M. Alexander; Ian F. Dunn; Rameen Beroukhim; Sandro Santagata

BACKGROUND Meningiomas are the most common primary intracranial tumor in adults. Identification of SMO and AKT1 mutations in meningiomas has raised the possibility of targeted therapies for some patients. The frequency of such mutations in clinical cohorts and the presence of other actionable mutations in meningiomas are important to define. METHODS We used high-resolution array-comparative genomic hybridization to prospectively characterize copy-number changes in 150 meningiomas and then characterized these samples for mutations in AKT1, KLF4, NF2, PIK3CA, SMO, and TRAF7. RESULTS Similar to prior reports, we identified AKT1 and SMO mutations in a subset of non-NF2-mutant meningiomas (ie, ∼9% and ∼6%, respectively). Notably, we detected oncogenic mutations in PIK3CA in ∼7% of non-NF2-mutant meningiomas. AKT1, SMO, and PIK3CA mutations were mutually exclusive. AKT1, KLF4, and PIK3CA mutations often co-occurred with mutations in TRAF7. PIK3CA-mutant meningiomas showed limited chromosomal instability and were enriched in the skull base. CONCLUSION This work identifies PI3K signaling as an important target for precision medicine trials in meningioma patients.


Journal of Thoracic Oncology | 2016

Clinical Activity of Alectinib in Advanced RET-Rearranged Non-Small Cell Lung Cancer

Jessica J. Lin; Elizabeth Kennedy; Lecia V. Sequist; Priscilla K. Brastianos; Kelly Goodwin; Sara Stevens; Alexandra Carly Wanat; Lisa Stober; Subba R. Digumarthy; Jeffrey A. Engelman; Alice T. Shaw; Justin F. Gainor

Introduction Chromosomal rearrangements involving rearranged during transfection gene (RET) occur in 1% to 2% of NSCLCs and may confer sensitivity to rearranged during transfection (RET) inhibitors. Alectinib is an anaplastic lymphoma kinase tyrosine kinase inhibitor (TKI) that also has anti‐RET activity in vitro. The clinical activity of alectinib in patients with RET‐rearranged NSCLC has not yet been reported. Methods We have described four patients with advanced RET‐rearranged NSCLC who were treated with alectinib (600 mg twice daily [n = 3] or 900 mg twice daily [n = 1]) as part of single‐patient compassionate use protocols or off‐label use of the commercially available drug. Results Four patients with metastatic RET‐rearranged NSCLC were identified. Three of the four had received prior RET TKIs, including cabozantinib and experimental RET inhibitors. In total, we observed two (50%) objective radiographic responses after treatment with alectinib (one confirmed and one unconfirmed), with durations of therapy of 6 months and more than 5 months (treatment ongoing), respectively. Notably, one of these two patients had his dose of alectinib escalated to 900 mg twice daily and had clinical improvement in central nervous system metastases. In addition, one patient (25%) experienced a best response of stable disease lasting approximately 6 weeks (the drug discontinued for toxicity). A fourth patient who was RET TKI–naive had primary progression while receiving alectinib. Conclusions Alectinib demonstrated preliminary antitumor activity in patients with advanced RET‐rearranged NSCLC, most of whom had received prior RET inhibitors. Larger prospective studies with longer follow‐up are needed to assess the efficacy of alectinib in RET‐rearranged NSCLC and other RET‐driven malignancies. In parallel, development of more selective, potent RET TKIs is warranted.

Collaboration


Dive into the Priscilla K. Brastianos's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sandro Santagata

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ian F. Dunn

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge