Network


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

Hotspot


Dive into the research topics where Lori A. Ramkissoon is active.

Publication


Featured researches published by Lori A. Ramkissoon.


Nature Genetics | 2014

Recurrent somatic mutations in ACVR1 in pediatric midline high-grade astrocytoma

Adam M. Fontebasso; Simon Papillon-Cavanagh; Jeremy Schwartzentruber; Hamid Nikbakht; Noha Gerges; Pierre‑Olivier Fiset; Denise Bechet; Damien Faury; Nicolas De Jay; Lori A. Ramkissoon; Aoife Corcoran; David T. W. Jones; Dominik Sturm; Pascal Johann; Tadanori Tomita; Stewart Goldman; Mahmoud Nagib; Liliana Goumnerova; Daniel C. Bowers; Jeffrey R. Leonard; Joshua B. Rubin; Tord D. Alden; Samuel R. Browd; J. Russell Geyer; Sarah Leary; George I. Jallo; Kenneth Cohen; Nalin Gupta; Michael D. Prados; Anne Sophie Carret

Pediatric midline high-grade astrocytomas (mHGAs) are incurable with few treatment targets identified. Most tumors harbor mutations encoding p.Lys27Met in histone H3 variants. In 40 treatment-naive mHGAs, 39 analyzed by whole-exome sequencing, we find additional somatic mutations specific to tumor location. Gain-of-function mutations in ACVR1 occur in tumors of the pons in conjunction with histone H3.1 p.Lys27Met substitution, whereas FGFR1 mutations or fusions occur in thalamic tumors associated with histone H3.3 p.Lys27Met substitution. Hyperactivation of the bone morphogenetic protein (BMP)-ACVR1 developmental pathway in mHGAs harboring ACVR1 mutations led to increased levels of phosphorylated SMAD1, SMAD5 and SMAD8 and upregulation of BMP downstream early-response genes in tumor cells. Global DNA methylation profiles were significantly associated with the p.Lys27Met alteration, regardless of the mutant histone H3 variant and irrespective of tumor location, supporting the role of this substitution in driving the epigenetic phenotype. This work considerably expands the number of potential treatment targets and further justifies pretreatment biopsy in pediatric mHGA as a means to orient therapeutic efforts in this disease.


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.


Nature Genetics | 2016

MYB-QKI rearrangements in angiocentric glioma drive tumorigenicity through a tripartite mechanism.

Pratiti Bandopadhayay; Lori A. Ramkissoon; Payal Jain; Guillaume Bergthold; Jeremiah Wala; Rhamy Zeid; Steven E. Schumacher; Laura M. Urbanski; Ryan O'Rourke; William J. Gibson; Kristine Pelton; Shakti Ramkissoon; Harry J. Han; Yuankun Zhu; Namrata Choudhari; Amanda Silva; Katie Boucher; Rosemary E. Henn; Yun Jee Kang; David Knoff; Brenton R. Paolella; Adrianne Gladden-Young; Pascale Varlet; Mélanie Pagès; Peleg Horowitz; Alexander J. Federation; Hayley Malkin; Adam Tracy; Sara Seepo; Matthew Ducar

Angiocentric gliomas are pediatric low-grade gliomas (PLGGs) without known recurrent genetic drivers. We performed genomic analysis of new and published data from 249 PLGGs, including 19 angiocentric gliomas. We identified MYB-QKI fusions as a specific and single candidate driver event in angiocentric gliomas. In vitro and in vivo functional studies show that MYB-QKI rearrangements promote tumorigenesis through three mechanisms: MYB activation by truncation, enhancer translocation driving aberrant MYB-QKI expression and hemizygous loss of the tumor suppressor QKI. To our knowledge, this represents the first example of a single driver rearrangement simultaneously transforming cells via three genetic and epigenetic mechanisms in a tumor.


Biochimica et Biophysica Acta | 2014

Pediatric low-grade gliomas: How modern biology reshapes the clinical field

Guillaume Bergthold; Pratiti Bandopadhayay; Wenya Linda Bi; Lori A. Ramkissoon; Charles D. Stiles; Rosalind A. Segal; Rameen Beroukhim; Keith L. Ligon; Jacques Grill; Mark W. Kieran

Low-grade gliomas represent the most frequent brain tumors arising during childhood. They are characterized by a broad and heterogeneous group of tumors that are currently classified by the WHO according to their morphological appearance. Here we review the clinical features of these tumors, current therapeutic strategies and the recent discovery of genomic alterations characteristic to these tumors. We further explore how these recent biological findings stand to transform the treatment for these tumors and impact the diagnostic criteria for pediatric low-grade gliomas.


Nature Medicine | 2016

Combination inhibition of PI3K and mTORC1 yields durable remissions in mice bearing orthotopic patient-derived xenografts of HER2-positive breast cancer brain metastases

Jing Ni; Shakti Ramkissoon; Shaozhen Xie; Shom Goel; Daniel G. Stover; Hanbing Guo; Victor Luu; Eugenio Marco; Lori A. Ramkissoon; Yun Jee Kang; Marika Hayashi; Quang-Dé Nguyen; Azra H. Ligon; Rose Du; Elizabeth B. Claus; Brian M. Alexander; Guo-Cheng Yuan; Zhigang C. Wang; J. Dirk Iglehart; Ian E. Krop; Thomas M. Roberts; Nan Lin; Keith L. Ligon; Jean Zhao

Brain metastases represent the greatest clinical challenge in treating HER2-positive breast cancer. We report the development of orthotopic patient-derived xenografts (PDXs) of HER2-expressing breast cancer brain metastases (BCBM), and their use for the identification of targeted combination therapies. Combined inhibition of PI3K and mTOR resulted in durable tumor regressions in three of five PDXs, and therapeutic response was correlated with a reduction in the phosphorylation of 4EBP1, an mTORC1 effector. The two nonresponding PDXs showed hypermutated genomes with enrichment of mutations in DNA-repair genes, which suggests an association of genomic instability with therapeutic resistance. These findings suggest that a biomarker-driven clinical trial of PI3K inhibitor in combination with an mTOR inhibitor should be conducted for patients with HER2-positive BCBM.


Neuro-oncology | 2015

Clinical implementation of integrated whole-genome copy number and mutation profiling for glioblastoma

Shakti Ramkissoon; Wenya Linda Bi; Steven E. Schumacher; Lori A. Ramkissoon; Sam Haidar; David Knoff; Adrian Dubuc; Loreal Brown; Margot Burns; Jane Cryan; Malak Abedalthagafi; Yun Jee Kang; Nikolaus Schultz; David A. Reardon; Eudocia Q. Lee; Mikael L. Rinne; Andrew D. Norden; Lakshmi Nayak; Sandra Ruland; Lisa Doherty; Debra C. LaFrankie; M.C. Horvath; Ayal A. Aizer; Andrea L. Russo; Nils D. Arvold; Elizabeth B. Claus; Ossama Al-Mefty; Mark D. Johnson; Alexandra J. Golby; Ian F. Dunn

BACKGROUND Multidimensional genotyping of formalin-fixed paraffin-embedded (FFPE) samples has the potential to improve diagnostics and clinical trials for brain tumors, but prospective use in the clinical setting is not yet routine. We report our experience with implementing a multiplexed copy number and mutation-testing program in a diagnostic laboratory certified by the Clinical Laboratory Improvement Amendments. METHODS We collected and analyzed clinical testing results from whole-genome array comparative genomic hybridization (OncoCopy) of 420 brain tumors, including 148 glioblastomas. Mass spectrometry-based mutation genotyping (OncoMap, 471 mutations) was performed on 86 glioblastomas. RESULTS OncoCopy was successful in 99% of samples for which sufficient DNA was obtained (n = 415). All clinically relevant loci for glioblastomas were detected, including amplifications (EGFR, PDGFRA, MET) and deletions (EGFRvIII, PTEN, 1p/19q). Glioblastoma patients ≤40 years old had distinct profiles compared with patients >40 years. OncoMap testing reliably identified mutations in IDH1, TP53, and PTEN. Seventy-seven glioblastoma patients enrolled on trials, of whom 51% participated in targeted therapeutic trials where multiplex data informed eligibility or outcomes. Data integration identified patients with complete tumor suppressor inactivation, albeit rarely (5% of patients) due to lack of whole-gene coverage in OncoMap. CONCLUSIONS Combined use of multiplexed copy number and mutation detection from FFPE samples in the clinical setting can efficiently replace singleton tests for clinical diagnosis and prognosis in most settings. Our results support incorporation of these assays into clinical trials as integral biomarkers and their potential to impact interpretation of results. Limited tumor suppressor variant capture by targeted genotyping highlights the need for whole-gene sequencing in glioblastoma.


Neuro-oncology | 2017

Clinical targeted exome-based sequencing in combination with genome-wide copy number profiling: precision medicine analysis of 203 pediatric brain tumors

Shakti Ramkissoon; Pratiti Bandopadhayay; Jaeho Hwang; Lori A. Ramkissoon; Noah F. Greenwald; Steven E. Schumacher; Ryan O’Rourke; Nathan Pinches; Patricia Ho; Hayley Malkin; Claire Sinai; Mariella G. Filbin; Ashley S. Plant; Wenya Linda Bi; Michael S. Chang; Edward Yang; Karen Wright; Peter Manley; Matthew Ducar; Sanda Alexandrescu; Hart G.W. Lidov; Ivana Delalle; Liliana Goumnerova; Alanna Church; Katherine A. Janeway; Marian H. Harris; Laura E. MacConaill; Rebecca D. Folkerth; Neal I. Lindeman; Charles D. Stiles

Background Clinical genomics platforms are needed to identify targetable alterations, but implementation of these technologies and best practices in routine clinical pediatric oncology practice are not yet well established. Methods Profile is an institution-wide prospective clinical research initiative that uses targeted sequencing to identify targetable alterations in tumors. OncoPanel, a multiplexed targeted exome-sequencing platform that includes 300 cancer-causing genes, was used to assess single nucleotide variants and rearrangements/indels. Alterations were annotated (Tiers 1-4) based on clinical significance, with Tier 1 alterations having well-established clinical utility. OncoCopy, a clinical genome-wide array comparative genomic hybridization (aCGH) assay, was also performed to evaluate copy number alterations and better define rearrangement breakpoints. Results Cancer genomes of 203 pediatric brain tumors were profiled across histological subtypes, including 117 samples analyzed by OncoPanel, 146 by OncoCopy, and 60 tumors subjected to both methodologies. OncoPanel revealed clinically relevant alterations in 56% of patients (44 cancer mutations and 20 rearrangements), including BRAF alterations that directed the use of targeted inhibitors. Rearrangements in MYB-QKI, MYBL1, BRAF, and FGFR1 were also detected. Furthermore, while copy number profiles differed across histologies, the combined use of OncoPanel and OncoCopy identified subgroup-specific alterations in 89% (17/19) of medulloblastomas. Conclusion The combination of OncoPanel and OncoCopy multiplex genomic assays can identify critical diagnostic, prognostic, and treatment-relevant alterations and represents an effective precision medicine approach for clinical evaluation of pediatric brain tumors.


Neuro-oncology | 2015

Preclinical antitumor efficacy of selective exportin 1 inhibitors in glioblastoma

Adam L. Green; Shakti Ramkissoon; Dilara McCauley; Kristen Jones; Jennifer A. Perry; Jessie Hao-Ru Hsu; Lori A. Ramkissoon; Cecile L. Maire; Benjamin Hubbell-Engler; David S. Knoff; Sharon Shacham; Keith L. Ligon; Andrew L. Kung

BACKGROUND Glioblastoma (GBM) is poorly responsive to current chemotherapy. The nuclear transporter exportin 1 (XPO1, CRM1) is often highly expressed in GBM, which may portend a poor prognosis. Here, we determine the efficacy of novel selective inhibitors of nuclear export (SINE) specific to XPO1 in preclinical models of GBM. METHODS Seven patient-derived GBM lines were treated with 3 SINE compounds (KPT-251, KPT-276, and Selinexor) in neurosphere culture conditions. KPT-276 and Selinexor were also evaluated in a murine orthotopic patient-derived xenograft (PDX) model of GBM. Cell cycle effects were assayed by flow cytometry in vitro and immunohistochemistry in vivo. Apoptosis was determined by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and caspase 3/7 activity assays. RESULTS Treatment of GBM neurosphere cultures with KPT-276, Selinexor, and KPT-251 revealed dose-responsive growth inhibition in all 7 GBM lines [range of half-maximal inhibitory concentration (IC50), 6-354 nM]. In an orthotopic PDX model, treatment with KPT-276 and Selinexor demonstrated pharmacodynamic efficacy, significantly suppressed tumor growth, and prolonged animal survival. Cellular proliferation was not altered with SINE treatment. Instead, induction of apoptosis was apparent both in vitro and in vivo with SINE treatment, without overt evidence of neurotoxicity. CONCLUSIONS SINE compounds show preclinical efficacy utilizing in vitro and in vivo models of GBM, with induction of apoptosis as the mechanism of action. Selinexor is now in early clinical trials in solid and hematological malignancies. Based on these preclinical data and excellent brain penetration, we have initiated clinical trials of Selinexor in patients with relapsed GBM.


Clinical Cancer Research | 2017

Residual Convolutional Neural Network for Determination of IDH Status in Low- and High-grade Gliomas from MR Imaging

Ken Chang; Harrison X. Bai; Hao Zhou; Chang Su; Wenya Linda Bi; Ena Agbodza; Vasileios K. Kavouridis; Joeky T. Senders; Alessandro Boaro; Andrew Beers; Biqi Zhang; Alexandra Capellini; Weihua Liao; Qin Shen; Xuejun Li; Bo Xiao; Jane Cryan; Shakti Ramkissoon; Lori A. Ramkissoon; Keith L. Ligon; Patrick Y. Wen; Ranjit S. Bindra; John H. Woo; Omar Arnaout; Elizabeth R. Gerstner; Paul J. Zhang; Bruce R. Rosen; Li Yang; Raymond Huang; Jayashree Kalpathy-Cramer

Purpose: Isocitrate dehydrogenase (IDH) mutations in glioma patients confer longer survival and may guide treatment decision making. We aimed to predict the IDH status of gliomas from MR imaging by applying a residual convolutional neural network to preoperative radiographic data. Experimental Design: Preoperative imaging was acquired for 201 patients from the Hospital of University of Pennsylvania (HUP), 157 patients from Brigham and Womens Hospital (BWH), and 138 patients from The Cancer Imaging Archive (TCIA) and divided into training, validation, and testing sets. We trained a residual convolutional neural network for each MR sequence (FLAIR, T2, T1 precontrast, and T1 postcontrast) and built a predictive model from the outputs. To increase the size of the training set and prevent overfitting, we augmented the training set images by introducing random rotations, translations, flips, shearing, and zooming. Results: With our neural network model, we achieved IDH prediction accuracies of 82.8% (AUC = 0.90), 83.0% (AUC = 0.93), and 85.7% (AUC = 0.94) within training, validation, and testing sets, respectively. When age at diagnosis was incorporated into the model, the training, validation, and testing accuracies increased to 87.3% (AUC = 0.93), 87.6% (AUC = 0.95), and 89.1% (AUC = 0.95), respectively. Conclusions: We developed a deep learning technique to noninvasively predict IDH genotype in grade II–IV glioma using conventional MR imaging using a multi-institutional data set. Clin Cancer Res; 24(5); 1073–81. ©2017 AACR.


Stem Cells | 2014

Pten loss in Olig2 expressing neural progenitor cells and oligodendrocytes leads to interneuron dysplasia and leukodystrophy.

Cecile L. Maire; Shakti Ramkissoon; Marika Hayashi; Sam Haidar; Lori A. Ramkissoon; Emmanuelle DiTomaso; Keith L. Ligon

Therapeutic modulation of phosphatidylinositol 3‐kinase (PI3K)/PTEN signaling is currently being explored for multiple neurological indications including brain tumors and seizure disorders associated with cortical malformations. The effects of PI3K/PTEN signaling are highly cell context dependent but the function of this pathway in specific subsets of neural stem/progenitor cells generating oligodendroglial lineage cells has not been fully studied. To address this, we created Olig2‐cre:Ptenfl/fl mice that showed a unique pattern of Pten loss and PI3K activation in Olig2‐lineage cells. Olig2‐cre:Ptenfl/fl animals progressively developed central nervous system white matter hypermyelination by 3 weeks of age leading to later onset leukodystrophy, chronic neurodegeneration, and death by 9 months. In contrast, during immediate postnatal development, oligodendroglia were unaffected but abnormal and accelerated differentiation of lateral subventricular zone stem cells produced calretinin‐positive interneuron dysplasia. Neural stem cells isolated from Olig2‐cre:Ptenfl/fl mice also exhibited accelerated differentiation and proliferation into calretinin‐positive interneurons and oligodendrocytes indicating such effects are cell autonomous. Opposition of the pathway by treatment of human primary neural progenitor cells (NPCs) with the PI3K inhibitor, NVP‐BKM120, blocked in vitro differentiation of neurons and oligodendroglia indicating PI3K/PTEN effects on NPCs can be bidirectional. In summary, our results suggest Pten is a developmental rheostat regulating interneuron and oligodendroglial differentiation and support testing of PI3K modulating drugs as treatment for developmental and myelination disorders. However, such agents may need to be administered at ages that minimize potential effects on early stem/progenitor cell development. Stem Cells 2014;32:313–326

Collaboration


Dive into the Lori A. Ramkissoon's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wenya Linda Bi

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge