ϟ

Daniel P. Cahill

Here are all the papers by Daniel P. Cahill that you can download and read on OA.mg.
Daniel P. Cahill’s last known institution is . Download Daniel P. Cahill PDFs here.

Claim this Profile →
DOI: 10.1126/science.1254257
2014
Cited 3,701 times
Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma
Human cancers are complex ecosystems composed of cells with distinct phenotypes, genotypes, and epigenetic states, but current models do not adequately reflect tumor composition in patients. We used single-cell RNA sequencing (RNA-seq) to profile 430 cells from five primary glioblastomas, which we found to be inherently variable in their expression of diverse transcriptional programs related to oncogenic signaling, proliferation, complement/immune response, and hypoxia. We also observed a continuum of stemness-related expression states that enabled us to identify putative regulators of stemness in vivo. Finally, we show that established glioblastoma subtype classifiers are variably expressed across individual cells within a tumor and demonstrate the potential prognostic implications of such intratumoral heterogeneity. Thus, we reveal previously unappreciated heterogeneity in diverse regulatory programs central to glioblastoma biology, prognosis, and therapy.
DOI: 10.1038/nature05610
2007
Cited 2,822 times
Patterns of somatic mutation in human cancer genomes
Cancers arise owing to mutations in a subset of genes that confer growth advantage. The availability of the human genome sequence led us to propose that systematic resequencing of cancer genomes for mutations would lead to the discovery of many additional cancer genes. Here we report more than 1,000 somatic mutations found in 274 megabases (Mb) of DNA corresponding to the coding exons of 518 protein kinase genes in 210 diverse human cancers. There was substantial variation in the number and pattern of mutations in individual cancers reflecting different exposures, DNA repair defects and cellular origins. Most somatic mutations are likely to be ‘passengers’ that do not contribute to oncogenesis. However, there was evidence for ‘driver’ mutations contributing to the development of the cancers studied in approximately 120 genes. Systematic sequencing of cancer genomes therefore reveals the evolutionary diversity of cancers and implicates a larger repertoire of cancer genes than previously anticipated. Over 350 cancer-causing genes have been identified by established techniques such as mapping, bioassay and by identifying plausible biological candidates. The availability of the human genome sequence now means that large-scale sequencing studies can uncover many more candidate cancer genes. Protein kinase enzymes are key to many regulatory processes and their dysfunction is a common trigger for tumours. So a sample of 518 kinases associated with more than 200 different cancers was chosen for a major sequencing effort. The study reveals more than 1, 000 previously unknown mutations linked to tumour formation — some as 'passengers' that don't contribute to cancer formation, but over 100 of them as 'driver' mutations that do contribute to disease development. As well as revealing cancer-causing defects, gene family studies like this can uncover new targets for molecular diagnostics and therapeutics. 518 protein kinase genes in the human genome have been sequenced in a large sample of tumours, providing a global view of the patterns of mutations found and the variations in the number and type of mutations between individual tumours.
DOI: 10.1016/j.ccr.2006.11.021
2007
Cited 1,664 times
AZD2171, a Pan-VEGF Receptor Tyrosine Kinase Inhibitor, Normalizes Tumor Vasculature and Alleviates Edema in Glioblastoma Patients
Using MRI techniques, we show here that normalization of tumor vessels in recurrent glioblastoma patients by daily administration of AZD2171-an oral tyrosine kinase inhibitor of VEGF receptors-has rapid onset, is prolonged but reversible, and has the significant clinical benefit of alleviating edema. Reversal of normalization began by 28 days, though some features persisted for as long as four months. Basic FGF, SDF1alpha, and viable circulating endothelial cells (CECs) increased when tumors escaped treatment, and circulating progenitor cells (CPCs) increased when tumors progressed after drug interruption. Our study provides insight into different mechanisms of action of this class of drugs in recurrent glioblastoma patients and suggests that the timing of combination therapy may be critical for optimizing activity against this tumor.
DOI: 10.1038/32688
1998
Cited 1,414 times
Mutations of mitotic checkpoint genes in human cancers
DOI: 10.1038/nature20123
2016
Cited 888 times
Single-cell RNA-seq supports a developmental hierarchy in human oligodendroglioma
Although human tumours are shaped by the genetic evolution of cancer cells, evidence also suggests that they display hierarchies related to developmental pathways and epigenetic programs in which cancer stem cells (CSCs) can drive tumour growth and give rise to differentiated progeny. Yet, unbiased evidence for CSCs in solid human malignancies remains elusive. Here we profile 4,347 single cells from six IDH1 or IDH2 mutant human oligodendrogliomas by RNA sequencing (RNA-seq) and reconstruct their developmental programs from genome-wide expression signatures. We infer that most cancer cells are differentiated along two specialized glial programs, whereas a rare subpopulation of cells is undifferentiated and associated with a neural stem cell expression program. Cells with expression signatures for proliferation are highly enriched in this rare subpopulation, consistent with a model in which CSCs are primarily responsible for fuelling the growth of oligodendroglioma in humans. Analysis of copy number variation (CNV) shows that distinct CNV sub-clones within tumours display similar cellular hierarchies, suggesting that the architecture of oligodendroglioma is primarily dictated by developmental programs. Subclonal point mutation analysis supports a similar model, although a full phylogenetic tree would be required to definitively determine the effect of genetic evolution on the inferred hierarchies. Our single-cell analyses provide insight into the cellular architecture of oligodendrogliomas at single-cell resolution and support the cancer stem cell model, with substantial implications for disease management.
DOI: 10.1158/2159-8290.cd-15-0369
2015
Cited 835 times
Genomic Characterization of Brain Metastases Reveals Branched Evolution and Potential Therapeutic Targets
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.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.
DOI: 10.1016/j.cell.2014.02.030
2014
Cited 759 times
Reconstructing and Reprogramming the Tumor-Propagating Potential of Glioblastoma Stem-like Cells
Developmental fate decisions are dictated by master transcription factors (TFs) that interact with cis-regulatory elements to direct transcriptional programs. Certain malignant tumors may also depend on cellular hierarchies reminiscent of normal development but superimposed on underlying genetic aberrations. In glioblastoma (GBM), a subset of stem-like tumor-propagating cells (TPCs) appears to drive tumor progression and underlie therapeutic resistance yet remain poorly understood. Here, we identify a core set of neurodevelopmental TFs (POU3F2, SOX2, SALL2, and OLIG2) essential for GBM propagation. These TFs coordinately bind and activate TPC-specific regulatory elements and are sufficient to fully reprogram differentiated GBM cells to "induced" TPCs, recapitulating the epigenetic landscape and phenotype of native TPCs. We reconstruct a network model that highlights critical interactions and identifies candidate therapeutic targets for eliminating TPCs. Our study establishes the epigenetic basis of a developmental hierarchy in GBM, provides detailed insight into underlying gene regulatory programs, and suggests attendant therapeutic strategies. PAPERCLIP:
DOI: 10.1126/science.aai8478
2017
Cited 723 times
Decoupling genetics, lineages, and microenvironment in IDH-mutant gliomas by single-cell RNA-seq
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.
DOI: 10.1016/s0962-8924(99)01661-x
1999
Cited 716 times
Genetic instability and darwinian selection in tumours
Genetic instability has long been hypothesized to be a cardinal feature of cancer. Recent work has strengthened the proposal that mutational alterations conferring instability occur early during tumour formation. The ensuing genetic instability drives tumour progression by generating mutations in oncogenes and tumour-suppressor genes. These mutant genes provide cancer cells with a selective growth advantage, thereby leading to the clonal outgrowth of a tumour. Here, we discuss the role of genetic instability in tumour formation and outline future work necessary to substantiate the genetic instability hypothesis.
DOI: 10.1016/j.ccr.2011.11.005
2011
Cited 621 times
Mosaic Amplification of Multiple Receptor Tyrosine Kinase Genes in Glioblastoma
Tumor heterogeneity has been implicated in tumor growth and progression as well as resistance to therapy. We present an example of genetic heterogeneity in human malignant brain tumors in which multiple closely related driver genes are amplified and activated simultaneously in adjacent intermingled cells. We have observed up to three different receptor tyrosine kinases (EGFR, MET, PDGFRA) amplified in single tumors in different cells in a mutually exclusive fashion. Each subpopulation was actively dividing, and the genetic changes resulted in protein production, and coexisting subpopulations shared common early genetic mutations indicating their derivation from a single precursor cell. The stable coexistence of different clones within the same tumor will have important clinical implications for tumor resistance to targeted therapies.
DOI: 10.1016/s1470-2045(17)30414-x
2017
Cited 564 times
Post-operative stereotactic radiosurgery versus observation for completely resected brain metastases: a single-centre, randomised, controlled, phase 3 trial
After brain metastasis resection, whole brain radiotherapy decreases local recurrence, but might cause cognitive decline. We did this study to determine if stereotactic radiosurgery (SRS) to the surgical cavity improved time to local recurrence compared with that for surgical resection alone.In this randomised, controlled, phase 3 trial, we recruited patients at a single tertiary cancer centre in the USA. Eligible patients were older than 3 years, had a Karnofsky Performance Score of 70 or higher, were able to have an MRI scan, and had a complete resection of one to three brain metastases (with a maximum diameter of the resection cavity ≤4 cm). Patients were randomly assigned (1:1) with a block size of four to either SRS of the resection cavity (within 30 days of surgery) or observation. Patients were stratified by histology of the primary tumour, metastatic tumour size, and number of metastases. The primary endpoint was time to local recurrence in the resection cavity, assessed by blinded central review of brain MRI scans by the study neuroradiologist in the modified intention-to-treat population that analysed patients by randomised allocation but excluded patients found ineligible after randomisation. Participants and other members of the treatment team (excluding the neuroradiologist) were not masked to treatment allocation. The trial is registered with ClinicalTrials.gov, number NCT00950001, and is closed to new participants.Between Aug 13, 2009, and Feb 16, 2016, 132 patients were randomly assigned to the observation group (n=68) or SRS group (n=64), with 128 patients available for analysis; four patients were ineligible (three from the SRS group and one from the observation group). Median follow-up was 11·1 months (IQR 4·8-20·4). 12-month freedom from local recurrence was 43% (95% CI 31-59) in the observation group and 72% (60-87) in the SRS group (hazard ratio 0·46 [95% CI 0·24-0·88]; p=0·015). There were no adverse events or treatment-related deaths in either group.SRS of the surgical cavity in patients who have had complete resection of one, two, or three brain metastases significantly lowers local recurrence compared with that noted for observation alone. Thus, the use of SRS after brain metastasis resection could be an alternative to whole-brain radiotherapy.National Institutes of Health.
DOI: 10.1126/science.1210557
2011
Cited 497 times
Mutations in <i>CIC</i> and <i>FUBP1</i> Contribute to Human Oligodendroglioma
Oligodendrogliomas are the second most common malignant brain tumor in adults and exhibit characteristic losses of chromosomes 1p and 19q. To identify the molecular genetic basis for this alteration, we performed exomic sequencing of seven tumors. Among other changes, we found that the CIC gene (homolog of the Drosophila gene capicua) on chromosome 19q was somatically mutated in six cases and that the FUBP1 gene [encoding far-upstream element (FUSE) binding protein] on chromosome 1p was somatically mutated in two tumors. Examination of 27 additional oligodendrogliomas revealed 12 and 3 more tumors with mutations of CIC and FUBP1, respectively, 58% of which were predicted to result in truncations of the encoded proteins. These results suggest a critical role for these genes in the biology and pathology of oligodendrocytes.
DOI: 10.1016/j.stem.2016.11.003
2017
Cited 404 times
Adaptive Chromatin Remodeling Drives Glioblastoma Stem Cell Plasticity and Drug Tolerance
Glioblastoma, the most common and aggressive malignant brain tumor, is propagated by stem-like cancer cells refractory to existing therapies. Understanding the molecular mechanisms that control glioblastoma stem cell (GSC) proliferation and drug resistance may reveal opportunities for therapeutic interventions. Here we show that GSCs can reversibly transition to a slow-cycling, persistent state in response to targeted kinase inhibitors. In this state, GSCs upregulate primitive developmental programs and are dependent upon Notch signaling. This transition is accompanied by widespread redistribution of repressive histone methylation. Accordingly, persister GSCs upregulate, and are dependent on, the histone demethylases KDM6A/B. Slow-cycling cells with high Notch activity and histone demethylase expression are present in primary glioblastomas before treatment, potentially contributing to relapse. Our findings illustrate how cancer cells may hijack aspects of native developmental programs for deranged proliferation, adaptation, and tolerance. They also suggest strategies for eliminating refractory tumor cells by targeting epigenetic and developmental pathways.
DOI: 10.1158/1078-0432.ccr-06-2149
2007
Cited 388 times
Loss of the Mismatch Repair Protein MSH6 in Human Glioblastomas Is Associated with Tumor Progression during Temozolomide Treatment
Glioblastomas are treated by surgical resection followed by radiotherapy [X-ray therapy (XRT)] and the alkylating chemotherapeutic agent temozolomide. Recently, inactivating mutations in the mismatch repair gene MSH6 were identified in two glioblastomas recurrent post-temozolomide. Because mismatch repair pathway inactivation is a known mediator of alkylator resistance in vitro, these findings suggested that MSH6 inactivation was causally linked to these two recurrences. However, the extent of involvement of MSH6 in glioblastoma is unknown. We sought to determine the overall frequency and clinical relevance of MSH6 alterations in glioblastomas.The MSH6 gene was sequenced in 54 glioblastomas. MSH6 and O(6)-methylguanine methyltransferase (MGMT) immunohistochemistry was systematically scored in a panel of 46 clinically well-characterized glioblastomas, and the corresponding patient response to treatment evaluated.MSH6 mutation was not observed in any pretreatment glioblastoma (0 of 40), whereas 3 of 14 recurrent cases had somatic mutations (P = 0.015). MSH6 protein expression was detected in all pretreatment (17 of 17) cases examined but, notably, expression was lost in 7 of 17 (41%) recurrences from matched post-XRT + temozolomide cases (P = 0.016). Loss of MSH6 was not associated with O(6)-methylguanine methyltransferase status. Measurements of in vivo tumor growth using three-dimensional reconstructed magnetic resonance imaging showed that MSH6-negative glioblastomas had a markedly increased rate of growth while under temozolomide treatment (3.17 versus 0.04 cc/mo for MSH6-positive tumors; P = 0.020).Loss of MSH6 occurs in a subset of post-XRT + temozolomide glioblastoma recurrences and is associated with tumor progression during temozolomide treatment, mirroring the alkylator resistance conferred by MSH6 inactivation in vitro. MSH6 deficiency may therefore contribute to the emergence of recurrent glioblastomas during temozolomide treatment.
DOI: 10.1093/neuonc/not159
2013
Cited 370 times
IDH1 mutant malignant astrocytomas are more amenable to surgical resection and have a survival benefit associated with maximal surgical resection
IDH1 gene mutations identify gliomas with a distinct molecular evolutionary origin. We sought to determine the impact of surgical resection on survival after controlling for IDH1 status in malignant astrocytomas—World Health Organization grade III anaplastic astrocytomas and grade IV glioblastoma. Clinical parameters including volumetric assessment of preoperative and postoperative MRI were recorded prospectively on 335 malignant astrocytoma patients: n = 128 anaplastic astrocytomas and n = 207 glioblastoma. IDH1 status was assessed by sequencing and immunohistochemistry. IDH1 mutation was independently associated with complete resection of enhancing disease (93% complete resections among mutants vs 67% among wild-type, P < .001), indicating IDH1 mutant gliomas were more amenable to resection. The impact of residual tumor on survival differed between IDH1 wild-type and mutant tumors. Complete resection of enhancing disease among IDH1 wild-type tumors was associated with a median survival of 19.6 months versus 10.7 months for incomplete resection; however, no survival benefit was observed in association with further resection of nonenhancing disease (minimization of total tumor volume). In contrast, IDH1 mutants displayed an additional survival benefit associated with maximal resection of total tumor volume (median survival 9.75 y for >5 cc residual vs not reached for <5 cc, P = .025). The survival benefit associated with surgical resection differs based on IDH1 genotype in malignant astrocytic gliomas. Therapeutic benefit from maximal surgical resection, including both enhancing and nonenhancing tumor, may contribute to the better prognosis observed in the IDH1 mutant subgroup. Thus, individualized surgical strategies for malignant astrocytoma may be considered based on IDH1 status.
DOI: 10.1038/s41591-018-0095-6
2018
Cited 362 times
Suppression of antitumor T cell immunity by the oncometabolite (R)-2-hydroxyglutarate
DOI: 10.1158/1078-0432.ccr-08-3012
2009
Cited 341 times
<i>MSH6</i> Mutations Arise in Glioblastomas during Temozolomide Therapy and Mediate Temozolomide Resistance
Abstract Purpose: Over the past few years, the alkylating agent temozolomide has become the standard-of-care therapy for patients with glioblastoma, the most common brain tumor. Recently, large-scale cancer genome sequencing efforts have identified a hypermutation phenotype and inactivating MSH6 mismatch repair gene mutations in recurrent, post-temozolomide glioblastomas, particularly those growing more rapidly during temozolomide treatment. This study aimed to clarify the timing and role of MSH6 mutations in mediating glioblastoma temozolomide resistance. Experimental Design: MSH6 sequence and microsatellite instability (MSI) status were determined in matched prechemotherapy and postchemotherapy glioblastomas identified by The Cancer Genome Atlas (TCGA) as having posttreatment MSH6 mutations. Temozolomide-resistant lines were derived in vitro through selective growth under temozolomide, and the MSH6 gene was sequenced in resistant clones. The role of MSH6 inactivation in mediating resistance was explored using lentiviral short hairpin RNA knockdown and MSH6 reconstitution. Results: MSH6 mutations were confirmed in posttreatment TCGA glioblastomas but absent in matched pretreatment tumors. The posttreatment hypermutation phenotype displayed a signature bias toward CpC transitions and was not associated with MSI. In vitro modeling through exposure of an MSH6 wild-type glioblastoma line to temozolomide resulted in resistant clones; one clone showed an MSH6 mutation, Thr1219Ile, that had been independently noted in two treated TCGA glioblastomas. Knockdown of MSH6 in the glioblastoma line U251 increased resistance to temozolomide cytotoxicity and reconstitution restored cytotoxicity in MSH6-null glioma cells. Conclusions: MSH6 mutations are selected in glioblastomas during temozolomide therapy both in vitro and in vivo and are causally associated with temozolomide resistance.
DOI: 10.1016/j.ccell.2015.11.006
2015
Cited 330 times
Extreme Vulnerability of IDH1 Mutant Cancers to NAD+ Depletion
<h2>Summary</h2> Heterozygous mutation of <i>IDH1</i> in cancers modifies IDH1 enzymatic activity, reprogramming metabolite flux and markedly elevating 2-hydroxyglutarate (2-HG). Here, we found that 2-HG depletion did not inhibit growth of several <i>IDH1</i> mutant solid cancer types. To identify other metabolic therapeutic targets, we systematically profiled metabolites in endogenous <i>IDH1</i> mutant cancer cells after mutant IDH1 inhibition and discovered a profound vulnerability to depletion of the coenzyme NAD+. Mutant <i>IDH1</i> lowered NAD+ levels by downregulating the NAD+ salvage pathway enzyme nicotinate phosphoribosyltransferase (Naprt1), sensitizing to NAD+ depletion via concomitant nicotinamide phosphoribosyltransferase (NAMPT) inhibition. NAD+ depletion activated the intracellular energy sensor AMPK, triggered autophagy, and resulted in cytotoxicity. Thus, we identify NAD+ depletion as a metabolic susceptibility of <i>IDH1</i> mutant cancers.
DOI: 10.1200/jco.21.02314
2022
Cited 298 times
Treatment for Brain Metastases: ASCO-SNO-ASTRO Guideline
To provide guidance to clinicians regarding therapy for patients with brain metastases from solid tumors.ASCO convened an Expert Panel and conducted a systematic review of the literature.Thirty-two randomized trials published in 2008 or later met eligibility criteria and form the primary evidentiary base.Surgery is a reasonable option for patients with brain metastases. Patients with large tumors with mass effect are more likely to benefit than those with multiple brain metastases and/or uncontrolled systemic disease. Patients with symptomatic brain metastases should receive local therapy regardless of the systemic therapy used. For patients with asymptomatic brain metastases, local therapy should not be deferred unless deferral is specifically recommended in this guideline. The decision to defer local therapy should be based on a multidisciplinary discussion of the potential benefits and harms that the patient may experience. Several regimens were recommended for non-small-cell lung cancer, breast cancer, and melanoma. For patients with asymptomatic brain metastases and no systemic therapy options, stereotactic radiosurgery (SRS) alone should be offered to patients with one to four unresected brain metastases, excluding small-cell lung carcinoma. SRS alone to the surgical cavity should be offered to patients with one to two resected brain metastases. SRS, whole brain radiation therapy, or their combination are reasonable options for other patients. Memantine and hippocampal avoidance should be offered to patients who receive whole brain radiation therapy and have no hippocampal lesions and 4 months or more expected survival. Patients with asymptomatic brain metastases with either Karnofsky Performance Status ≤ 50 or Karnofsky Performance Status < 70 with no systemic therapy options do not derive benefit from radiation therapy.Additional information is available at www.asco.org/neurooncology-guidelines.
DOI: 10.1007/s00401-015-1398-z
2015
Cited 274 times
IDH mutation status and role of WHO grade and mitotic index in overall survival in grade II–III diffuse gliomas
Diffuse gliomas are up till now graded based upon morphology. Recent findings indicate that isocitrate dehydrogenase (IDH) mutation status defines biologically distinct groups of tumors. The role of tumor grade and mitotic index in patient outcome has not been evaluated following stratification by IDH mutation status. To address this, we interrogated 558 WHO grade II-III diffuse gliomas for IDH1/2 mutations and investigated the prognostic impact of WHO grade within IDH-mutant and IDH-wild type tumor subsets independently. The prognostic impact of grade was modest in IDH-mutant [hazard ratio (HR) = 1.21, 95 % confidence interval (CI) = 0.91-1.61] compared to IDH-wild type tumors (HR = 1.74, 95 % CI = 0.95-3.16). Using a dichotomized mitotic index cut-off of 4/1000 tumor cells, we found that while mitotic index was significantly associated with outcome in IDH-wild type tumors (log-rank p < 0.0001, HR = 4.41, 95 % CI = 2.55-7.63), it was not associated with outcome in IDH-mutant tumors (log-rank p = 0.5157, HR = 1.10, 95 % CI = 0.80-1.51), and could demonstrate a statistical interaction (p < 0.0001) between IDH mutation and mitotic index (i.e., suggesting that the effect of mitotic index on patient outcome is dependent on IDH mutation status). Patient age, an established prognostic factor in diffuse glioma, was significantly associated with outcome only in the IDH-wild type subset, and consistent with prior data, 1p/19q co-deletion conferred improved outcome in the IDH-mutant cohort. These findings suggest that stratification of grade II-III gliomas into subsets defined by the presence or absence of IDH mutation leads to subgroups with distinct prognostic characteristics. Further evaluation of grading criteria and prognostic markers is warranted within IDH-mutant versus IDH-wild type diffuse grade II-III gliomas as independent entities.
DOI: 10.1093/jnci/djv310
2015
Cited 193 times
Dramatic Response of BRAF V600E Mutant Papillary Craniopharyngioma to Targeted Therapy
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 patient’s 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.
DOI: 10.1016/j.ccell.2017.08.018
2017
Cited 164 times
Coordinated Splicing of Regulatory Detained Introns within Oncogenic Transcripts Creates an Exploitable Vulnerability in Malignant Glioma
Glioblastoma (GBM) is a devastating malignancy with few therapeutic options. We identify PRMT5 in an in vivo GBM shRNA screen and show that PRMT5 knockdown or inhibition potently suppresses in vivo GBM tumors, including patient-derived xenografts. Pathway analysis implicates splicing in cellular PRMT5 dependency, and we identify a biomarker that predicts sensitivity to PRMT5 inhibition. We find that PRMT5 deficiency primarily disrupts the removal of detained introns (DIs). This impaired DI splicing affects proliferation genes, whose downregulation coincides with cell cycle defects, senescence and/or apoptosis. We further show that DI programs are evolutionarily conserved and operate during neurogenesis, suggesting that they represent a physiological regulatory mechanism. Collectively, these findings reveal a PRMT5-regulated DI-splicing program as an exploitable cancer vulnerability.
DOI: 10.1093/neuonc/noac207
2022
Cited 52 times
Isocitrate dehydrogenase (IDH) mutant gliomas: A Society for Neuro-Oncology (SNO) consensus review on diagnosis, management, and future directions
Isocitrate dehydrogenase (IDH) mutant gliomas are the most common adult, malignant primary brain tumors diagnosed in patients younger than 50, constituting an important cause of morbidity and mortality. In recent years, there has been significant progress in understanding the molecular pathogenesis and biology of these tumors, sparking multiple efforts to improve their diagnosis and treatment. In this consensus review from the Society for Neuro-Oncology (SNO), the current diagnosis and management of IDH-mutant gliomas will be discussed. In addition, novel therapies, such as targeted molecular therapies and immunotherapies, will be reviewed. Current challenges and future directions for research will be discussed.
DOI: 10.1006/geno.1999.5831
1999
Cited 208 times
Characterization of MAD2B and Other Mitotic Spindle Checkpoint Genes
Aneuploidy is a characteristic of the majority of human cancers, and recent work has suggested that mitotic checkpoint defects play a role in its development. To further explore this issue, we isolated a novel human gene, MAD2B (MAD2L2), which is homologous to the spindle checkpoint gene MAD2 (MAD2L1). We determined the chromosomal localization of it and other spindle checkpoint genes, including MAD1L1, MAD2, BUB3, TTK (MPS1L1), and CDC20. In addition, we resolved the genomic intron-exon structure of the human BUB1 gene. We then searched for mutations in these genes in a panel of 19 aneuploid colorectal tumors. No new mutations were identified, suggesting that genes yet to be discovered are responsible for most of the checkpoint defects observed in aneuploid cancers.
DOI: 10.1158/0008-5472.can-04-0587
2004
Cited 178 times
Three Classes of Genes Mutated In Colorectal Cancers with Chromosomal Instability
Although most colorectal cancers are chromosomally unstable, the basis for this instability has not been defined. To determine whether genes shown to cause chromosomal instability in model systems were mutated in colorectal cancers, we identified their human homologues and determined their sequence in a panel of colorectal cancers. We found 19 somatic mutations in five genes representing three distinct instability pathways. Seven mutations were found in MRE11, whose product is involved in double-strand break repair. Four mutations were found among hZw10, hZwilch/FLJ10036, and hRod/KNTC, whose products bind to one another in a complex that localizes to kinetochores and controls chromosome segregation. Eight mutations were found in Ding, a previously uncharacterized gene with sequence similarity to the Saccharomyces cerevisiae Pds1, whose product is essential for proper chromosome disjunction. This analysis buttresses the evidence that chromosomal instability has a genetic basis and provides clues to the mechanistic basis of instability in cancers.
DOI: 10.1172/jci67229
2013
Cited 148 times
Detection of oncogenic IDH1 mutations using magnetic resonance spectroscopy of 2-hydroxyglutarate
The investigation of metabolic pathways disturbed in isocitrate dehydrogenase (IDH) mutant tumors revealed that the hallmark metabolic alteration is the production of D-2-hydroxyglutarate (D-2HG). The biological impact of D-2HG strongly suggests that high levels of this metabolite may play a central role in propagating downstream the effects of mutant IDH, leading to malignant transformation of cells. Hence, D-2HG may be an ideal biomarker for both diagnosing and monitoring treatment response targeting IDH mutations. Magnetic resonance spectroscopy (MRS) is well suited to the task of noninvasive D-2HG detection, and there has been much interest in developing such methods. Here, we review recent efforts to translate methodology using MRS to reliably measure in vivo D-2HG into clinical research.
DOI: 10.1158/1078-0432.ccr-13-3052
2014
Cited 142 times
Targetable Signaling Pathway Mutations Are Associated with Malignant Phenotype in <i>IDH</i>-Mutant Gliomas
Abstract Purpose: Isocitrate dehydrogenase (IDH) gene mutations occur in low-grade and high-grade gliomas. We sought to identify the genetic basis of malignant phenotype heterogeneity in IDH-mutant gliomas. Methods: We prospectively implanted tumor specimens from 20 consecutive IDH1-mutant glioma resections into mouse brains and genotyped all resection specimens using a CLIA-certified molecular panel. Gliomas with cancer driver mutations were tested for sensitivity to targeted inhibitors in vitro. Associations between genomic alterations and outcomes were analyzed in patients. Results: By 10 months, 8 of 20 IDH1-mutant gliomas developed intracerebral xenografts. All xenografts maintained mutant IDH1 and high levels of 2-hydroxyglutarate on serial transplantation. All xenograft-producing gliomas harbored “lineage-defining” mutations in CIC (oligodendroglioma) or TP53 (astrocytoma), and 6 of 8 additionally had activating mutations in PIK3CA or amplification of PDGFRA, MET, or N-MYC. Only IDH1 and CIC/TP53 mutations were detected in non–xenograft-forming gliomas (P = 0.0007). Targeted inhibition of the additional alterations decreased proliferation in vitro. Moreover, we detected alterations in known cancer driver genes in 13.4% of IDH-mutant glioma patients, including PIK3CA, KRAS, AKT, or PTEN mutation or PDGFRA, MET, or N-MYC amplification. IDH/CIC mutant tumors were associated with PIK3CA/KRAS mutations whereas IDH/TP53 tumors correlated with PDGFRA/MET amplification. Presence of driver alterations at progression was associated with shorter subsequent progression-free survival (median 9.0 vs. 36.1 months; P = 0.0011). Conclusion: A subset of IDH-mutant gliomas with mutations in driver oncogenes has a more malignant phenotype in patients. Identification of these alterations may provide an opportunity for use of targeted therapies in these patients. Clin Cancer Res; 20(11); 2898–909. ©2014 AACR.
DOI: 10.1158/1078-0432.ccr-15-0656
2016
Cited 128 times
Treatment Response Assessment in IDH-Mutant Glioma Patients by Noninvasive 3D Functional Spectroscopic Mapping of 2-Hydroxyglutarate
Measurements of objective response rates are critical to evaluate new glioma therapies. The hallmark metabolic alteration in gliomas with mutant isocitrate dehydrogenase (IDH) is the overproduction of oncometabolite 2-hydroxyglutarate (2HG), which plays a key role in malignant transformation. 2HG represents an ideal biomarker to probe treatment response in IDH-mutant glioma patients, and we hypothesized a decrease in 2HG levels would be measureable by in vivo magnetic resonance spectroscopy (MRS) as a result of antitumor therapy.We report a prospective longitudinal imaging study performed in 25 IDH-mutant glioma patients receiving adjuvant radiation and chemotherapy. A newly developed 3D MRS imaging was used to noninvasively image 2HG. Paired Student t test was used to compare pre- and posttreatment tumor 2HG values. Test-retest measurements were performed to determine the threshold for 2HG functional spectroscopic maps (fSM). Univariate and multivariate regression were performed to correlate 2HG changes with Karnofsky performance score (KPS).We found that mean 2HG (2HG/Cre) levels decreased significantly (median = 48.1%; 95% confidence interval = 27.3%-56.5%;P= 0.007) in the posttreatment scan. The volume of decreased 2HG correlates (R(2)= 0.88,P= 0.002) with clinical status evaluated by KPS.We demonstrate that dynamic measurements of 2HG are feasible by 3D fSM, and the decrease of 2HG levels can monitor treatment response in patients with IDH-mutant gliomas. Our results indicate that quantitative in vivo 2HG imaging may be used for precision medicine and early response assessment in clinical trials of therapies targeting IDH-mutant gliomas.
DOI: 10.1158/1078-0432.ccr-11-0414
2011
Cited 117 times
Glioma-Associated Cytomegalovirus Mediates Subversion of the Monocyte Lineage to a Tumor Propagating Phenotype
Abstract Purpose: Cytomegalovirus (CMV) has been ubiquitously detected within high-grade gliomas, but its role in gliomagenesis has not been fully elicited. Experimental Design: Glioblastoma multiforme (GBM) tumors were analyzed by flow cytometry to determine CMV antigen expression within various glioma-associated immune populations. The glioma cancer stem cell (gCSC) CMV interleukin (IL)-10 production was determined by ELISA. Human monocytes were stimulated with recombinant CMV IL-10 and levels of expression of p-STAT3, VEGF (vascular endothelial growth factor), TGF-β, viral IE1, and pp65 were determined by flow cytometry. The influence of CMV IL-10–treated monocytes on gCSC biology was ascertained by functional assays. Results: CMV showed a tropism for macrophages (MΦ)/microglia and CD133+ gCSCs within GBMs. The gCSCs produce CMV IL-10, which induces human monocytes (the precursor to the central nervous system MΦs/microglia) to assume an M2 immunosuppressive phenotype (as manifested by downmodulation of the major histocompatibility complex and costimulatory molecules) while upregulating immunoinhibitory B7-H1. CMV IL-10 also induces expression of viral IE1, a modulator of viral replication and transcription in the monocytes. Finally, the CMV IL-10–treated monocytes produced angiogenic VEGF, immunosuppressive TGF-β, and enhanced migration of gCSCs. Conclusions: CMV triggers a feedforward mechanism of gliomagenesis by inducing tumor-supportive monocytes. Clin Cancer Res; 17(14); 4642–9. ©2011 AACR.
DOI: 10.1158/1078-0432.ccr-15-2274
2016
Cited 113 times
Myc-Driven Glycolysis Is a Therapeutic Target in Glioblastoma
Abstract Purpose: Deregulated Myc drives an oncogenic metabolic state, including pseudohypoxic glycolysis, adapted for the constitutive production of biomolecular precursors to feed rapid tumor cell growth. In glioblastoma, Myc facilitates renewal of the tumor-initiating cell reservoir contributing to tumor maintenance. We investigated whether targeting the Myc-driven metabolic state could be a selectively toxic therapeutic strategy for glioblastoma. Experimental Design: The glycolytic dependency of Myc-driven glioblastoma was tested using 13C metabolic flux analysis, glucose-limiting culture assays, and glycolysis inhibitors, including inhibitors of the NAD+ salvage enzyme nicotinamide phosphoribosyl-transferase (NAMPT), in MYC and MYCN shRNA knockdown and lentivirus overexpression systems and in patient-derived glioblastoma tumorspheres with and without MYC/MYCN amplification. The in vivo efficacy of glycolyic inhibition was tested using NAMPT inhibitors in MYCN-amplified patient-derived glioblastoma orthotopic xenograft mouse models. Results: Enforced Myc overexpression increased glucose flux and expression of glycolytic enzymes in glioblastoma cells. Myc and N-Myc knockdown and Myc overexpression systems demonstrated that Myc activity determined sensitivity and resistance to inhibition of glycolysis. Small-molecule inhibitors of glycolysis, particularly NAMPT inhibitors, were selectively toxic to MYC/MYCN–amplified patient-derived glioblastoma tumorspheres. NAMPT inhibitors were potently cytotoxic, inducing apoptosis and significantly extended the survival of mice bearing MYCN-amplified patient-derived glioblastoma orthotopic xenografts. Conclusions: Myc activation in glioblastoma generates a dependency on glycolysis and an addiction to metabolites required for glycolysis. Glycolytic inhibition via NAMPT inhibition represents a novel metabolically targeted therapeutic strategy for MYC or MYCN-amplified glioblastoma and potentially other cancers genetically driven by Myc. Clin Cancer Res; 22(17); 4452–65. ©2016 AACR.
DOI: 10.1158/0008-5472.can-16-1457
2016
Cited 111 times
Spatial Proximity to Fibroblasts Impacts Molecular Features and Therapeutic Sensitivity of Breast Cancer Cells Influencing Clinical Outcomes
Using a three-dimensional coculture model, we identified significant subtype-specific changes in gene expression, metabolic, and therapeutic sensitivity profiles of breast cancer cells in contact with cancer-associated fibroblasts (CAF). CAF-induced gene expression signatures predicted clinical outcome and immune-related differences in the microenvironment. We found that fibroblasts strongly protect carcinoma cells from lapatinib, attributable to its reduced accumulation in carcinoma cells and an elevated apoptotic threshold. Fibroblasts from normal breast tissues and stromal cultures of brain metastases of breast cancer had similar effects as CAFs. Using synthetic lethality approaches, we identified molecular pathways whose inhibition sensitizes HER2+ breast cancer cells to lapatinib both in vitro and in vivo, including JAK2/STAT3 and hyaluronic acid. Neoadjuvant lapatinib therapy in HER2+ breast tumors lead to a significant increase of phospho-STAT3+ cancer cells and a decrease in the spatial proximity of proliferating (Ki67+) cells to CAFs impacting therapeutic responses. Our studies identify CAF-induced physiologically and clinically relevant changes in cancer cells and offer novel approaches for overcoming microenvironment-mediated therapeutic resistance. Cancer Res; 76(22); 6495-506. ©2016 AACR.
DOI: 10.1002/cncr.31039
2017
Cited 107 times
Isocitrate dehydrogenase‐mutant glioma: Evolving clinical and therapeutic implications
The metabolic genes isocitrate dehydrogenase 1 ( IDH1 ) and IDH2 are commonly mutated in low‐grade glioma and in a subset of glioblastoma. These mutations co‐occur with other recurrent molecular alterations, including 1p/19q codeletions and tumor suppressor protein 53 ( TP53 ) and alpha thalassemia/mental retardation ( ATRX ) mutations, which together help to define a molecular signature that aids in the classification of gliomas and helps to better predict clinical behavior. A confluence of research suggests that glioma development in IDH ‐mutant and IDH wild‐type tumors is driven by different oncogenic processes and responds differently to current treatment paradigms. Herein, the authors discuss the discovery of IDH mutations and associated molecular alterations in glioma, review clinical features common to patients with IDH ‐mutant glioma, and highlight current understanding of IDH mutation‐driven gliomagenesis with implications for emerging treatment strategies. Cancer 2017;123:4535‐4546 . © 2017 American Cancer Society .
DOI: 10.1038/s41467-018-03905-6
2018
Cited 107 times
Pharmacodynamics of mutant-IDH1 inhibitors in glioma patients probed by in vivo 3D MRS imaging of 2-hydroxyglutarate
Inhibitors of the mutant isocitrate dehydrogenase 1 (IDH1) entered recently in clinical trials for glioma treatment. Mutant IDH1 produces high levels of 2-hydroxyglurate (2HG), thought to initiate oncogenesis through epigenetic modifications of gene expression. In this study, we show the initial evidence of the pharmacodynamics of a new mutant IDH1 inhibitor in glioma patients, using non-invasive 3D MR spectroscopic imaging of 2HG. Our results from a Phase 1 clinical trial indicate a rapid decrease of 2HG levels by 70% (CI 13%, P = 0.019) after 1 week of treatment. Importantly, inhibition of mutant IDH1 may lead to the reprogramming of tumor metabolism, suggested by simultaneous changes in glutathione, glutamine, glutamate, and lactate. An inverse correlation between metabolic changes and diffusion MRI indicates an effect on the tumor-cell density. We demonstrate a feasible radiopharmacodynamics approach to support the rapid clinical translation of rationally designed drugs targeting IDH1/2 mutations for personalized and precision medicine of glioma patients.
DOI: 10.1016/j.ijrobp.2012.05.047
2013
Cited 95 times
Predictors of Survival in Contemporary Practice After Initial Radiosurgery for Brain Metastases
Purpose The number of brain metastases (BM) is a major consideration in determining patient eligibility for stereotactic radiosurgery (SRS), but the evidence for this popular practice is equivocal. The purpose of this study was to determine whether, following multivariate adjustment, the number and volume of BM held prognostic significance in a cohort of patients initially treated with SRS alone. Methods and Materials A total of 251 patients with primary malignancies, including non-small cell lung cancer (34%), melanoma (30%), and breast carcinoma (16%), underwent SRS for initial treatment of BM. SRS was used as the sole management (62% of patients) or was combined with salvage treatment with SRS (22%), whole-brain radiation therapy (WBRT; 13%), or resection (3%). Median follow-up time was 9.4 months. Survival was determined using the Kaplan-Meier method. Cox regression was used to assess the effects of patient factors on distant brain failure (DBF), local control (LC), and overall survival (OS). Results LC at 1 year was 94.6%, and median time to DBF was 10 months. Median OS was 11.1 months. On multivariate analysis, statistically significant predictors of OS were presence of extracranial disease (hazard ratio [HR], 4.2, P<.001), total tumor volume greater than 2 cm3 (HR, 1.98; P<.001), age ≥60 years (HR, 1.67; P=.002), and diagnosis-specific graded prognostic assessment (HR, 0.71; P<.001). The presence of extracranial disease was a statistically significant predictor of DBF (HR, 2.15), and tumor volume was predictive of LC (HR, 4.56 for total volume >2 cm3). The number of BM was not predictive of DBF, LC, or OS. Conclusions The number of BM is not a strong predictor for clinical outcomes following initial SRS for newly diagnosed BM. Other factors including total treatment volume and systemic disease status are better determinants of outcome and may facilitate appropriate use of SRS or WBRT. The number of brain metastases (BM) is a major consideration in determining patient eligibility for stereotactic radiosurgery (SRS), but the evidence for this popular practice is equivocal. The purpose of this study was to determine whether, following multivariate adjustment, the number and volume of BM held prognostic significance in a cohort of patients initially treated with SRS alone. A total of 251 patients with primary malignancies, including non-small cell lung cancer (34%), melanoma (30%), and breast carcinoma (16%), underwent SRS for initial treatment of BM. SRS was used as the sole management (62% of patients) or was combined with salvage treatment with SRS (22%), whole-brain radiation therapy (WBRT; 13%), or resection (3%). Median follow-up time was 9.4 months. Survival was determined using the Kaplan-Meier method. Cox regression was used to assess the effects of patient factors on distant brain failure (DBF), local control (LC), and overall survival (OS). LC at 1 year was 94.6%, and median time to DBF was 10 months. Median OS was 11.1 months. On multivariate analysis, statistically significant predictors of OS were presence of extracranial disease (hazard ratio [HR], 4.2, P<.001), total tumor volume greater than 2 cm3 (HR, 1.98; P<.001), age ≥60 years (HR, 1.67; P=.002), and diagnosis-specific graded prognostic assessment (HR, 0.71; P<.001). The presence of extracranial disease was a statistically significant predictor of DBF (HR, 2.15), and tumor volume was predictive of LC (HR, 4.56 for total volume >2 cm3). The number of BM was not predictive of DBF, LC, or OS. The number of BM is not a strong predictor for clinical outcomes following initial SRS for newly diagnosed BM. Other factors including total treatment volume and systemic disease status are better determinants of outcome and may facilitate appropriate use of SRS or WBRT.
DOI: 10.18632/oncotarget.22650
2017
Cited 93 times
Intratumoral heterogeneity and <i>TERT</i> promoter mutations in progressive/higher-grade meningiomas
Background: Recent studies have reported mutations in the telomerase reverse transcriptase promoter (TERTp) in meningiomas.We sought to determine the frequency, clonality and clinical significance of telomere gene alterations in a cohort of patients with progressive/higher-grade meningiomas.Methods: We characterized 64 temporally-and regionally-distinct specimens from 26 WHO grade III meningioma patients.On initial diagnoses, the meningiomas spanned all WHO grades (3 grade I, 13 grade II and 10 grade III).The tumor samples were screened for TERTp and ATRX/DAXX mutations, and TERT rearrangements.Additionally, TERTp was sequenced in a separate cohort of 19 patients with radiationassociated meningiomas.We examined the impact of mutational status on patients' progression and overall survival.Results: Somatic TERTp mutations were detected in six patients (6/26 = 23%).Regional intratumoral heterogeneity in TERTp mutation status was noted.In 4 www.impactjournals.com/oncotarget/
DOI: 10.1158/0008-5472.can-16-2263
2017
Cited 73 times
The Alkylating Chemotherapeutic Temozolomide Induces Metabolic Stress in <i>IDH1</i>-Mutant Cancers and Potentiates NAD+ Depletion–Mediated Cytotoxicity
Abstract IDH1-mutant gliomas are dependent upon the canonical coenzyme NAD+ for survival. It is known that PARP activation consumes NAD+ during base excision repair (BER) of chemotherapy-induced DNA damage. We therefore hypothesized that a strategy combining NAD+ biosynthesis inhibitors with the alkylating chemotherapeutic agent temozolomide could potentiate NAD+ depletion–mediated cytotoxicity in mutant IDH1 cancer cells. To investigate the impact of temozolomide on NAD+ metabolism, patient-derived xenografts and engineered mutant IDH1-expressing cell lines were exposed to temozolomide, in vitro and in vivo, both alone and in combination with nicotinamide phosphoribosyltransferase (NAMPT) inhibitors, which block NAD+ biosynthesis. The acute time period (&amp;lt;3 hours) after temozolomide treatment displayed a burst of NAD+ consumption driven by PARP activation. In IDH1-mutant–expressing cells, this consumption reduced further the abnormally lowered basal steady-state levels of NAD+, introducing a window of hypervulnerability to NAD+ biosynthesis inhibitors. This effect was selective for IDH1-mutant cells and independent of methylguanine methyltransferase or mismatch repair status, which are known rate-limiting mediators of adjuvant temozolomide genotoxic sensitivity. Combined temozolomide and NAMPT inhibition in an in vivo IDH1-mutant cancer model exhibited enhanced efficacy compared with each agent alone. Thus, we find IDH1-mutant cancers have distinct metabolic stress responses to chemotherapy-induced DNA damage and that combination regimens targeting nonredundant NAD+ pathways yield potent anticancer efficacy in vivo. Such targeting of convergent metabolic pathways in genetically selected cancers could minimize treatment toxicity and improve durability of response to therapy. Cancer Res; 77(15); 4102–15. ©2017 AACR.
DOI: 10.1016/j.ccell.2024.03.008
2024
Mutant IDH inhibitors induce lineage differentiation in IDH-mutant oligodendroglioma
A subset of patients with IDH-mutant glioma respond to inhibitors of mutant IDH (IDHi), yet the molecular underpinnings of such responses are not understood. Here, we profiled by single-cell or single-nucleus RNA-sequencing three IDH-mutant oligodendrogliomas from patients who derived clinical benefit from IDHi. Importantly, the tissues were sampled on-drug, four weeks from treatment initiation. We further integrate our findings with analysis of single-cell and bulk transcriptomes from independent cohorts and experimental models. We find that IDHi treatment induces a robust differentiation toward the astrocytic lineage, accompanied by a depletion of stem-like cells and a reduction of cell proliferation. Furthermore, mutations in NOTCH1 are associated with decreased astrocytic differentiation and may limit the response to IDHi. Our study highlights the differentiating potential of IDHi on the cellular hierarchies that drive oligodendrogliomas and suggests a genetic modifier that may improve patient stratification.
DOI: 10.1073/pnas.081082898
2001
Cited 165 times
Carcinogen-specific induction of genetic instability
It has been proposed recently that the type of genetic instability in cancer cells reflects the selection pressures exerted by specific carcinogens. We have tested this hypothesis by treating immortal, genetically stable human cells with representative carcinogens. We found that cells resistant to the bulky-adduct-forming agent 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) exhibited a chromosomal instability (CIN), whereas cells resistant to the methylating agent N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) exhibited a microsatellite instability (MIN) associated with mismatch repair defects. Conversely, we found that cells purposely made into CIN cells are resistant to PhIP, whereas MIN cells are resistant to MNNG. These data demonstrate that exposure to specific carcinogens can indeed select for tumor cells with distinct forms of genetic instability and vice versa.
DOI: 10.1186/s40478-014-0167-x
2014
Cited 69 times
Sporadic hemangioblastomas are characterized by cryptic VHL inactivation
Hemangioblastomas consist of 10-20% neoplastic "stromal" cells within a vascular tumor cell mass of reactive pericytes, endothelium and lymphocytes. Familial cases of central nervous system hemangioblastoma uniformly result from mutations in the Von Hippel-Lindau (VHL) gene. In contrast, inactivation of VHL has been previously observed in only a minority of sporadic hemangioblastomas, suggesting an alternative genetic etiology. We performed deep-coverage DNA sequencing on 32 sporadic hemangioblastomas (whole exome discovery cohort n = 10, validation n = 22), followed by analysis of clonality, copy number alteration, and somatic mutation. We identified somatic mutation, loss of heterozygosity and/or deletion of VHL in 8 of 10 discovery cohort tumors. VHL inactivating events were ultimately detected in 78% (25/32) of cases. No other gene was significantly mutated. Overall, deep-coverage sequence analysis techniques uncovered VHL alterations within the neoplastic fraction of these tumors at higher frequencies than previously reported. Our findings support the central role of VHL inactivation in the molecular pathogenesis of both familial and sporadic hemangioblastomas.
DOI: 10.1001/jamaoncol.2015.0917
2015
Cited 67 times
Rapid Intraoperative Molecular Characterization of Glioma
Conclusive intraoperative pathologic confirmation of diffuse infiltrative glioma guides the decision to pursue definitive neurosurgical resection. Establishing the intraoperative diagnosis by histologic analysis can be difficult in low-cellularity infiltrative gliomas. Therefore, we developed a rapid and sensitive genotyping assay to detect somatic single-nucleotide variants in the telomerase reverse transcriptase (TERT) promoter and isocitrate dehydrogenase 1 (IDH1).This assay was applied to tissue samples from 190 patients with diffuse gliomas, including archived fixed and frozen specimens and tissue obtained intraoperatively. Results demonstrated 96% sensitivity (95% CI, 90%-99%) and 100% specificity (95% CI, 95%-100%) for World Health Organization grades II and III gliomas. In a series of live cases, glioma-defining mutations could be identified within 60 minutes, which could facilitate the diagnosis in an intraoperative timeframe.The genotyping method described herein can establish the diagnosis of low-cellularity tumors like glioma and could be adapted to the point-of-care diagnosis of other lesions that are similarly defined by highly recurrent somatic mutations.
DOI: 10.1007/s11060-016-2328-1
2016
Cited 61 times
The effect of IDH1 mutation on the structural connectome in malignant astrocytoma
Mutation of the IDH1 gene is associated with differences in malignant astrocytoma growth characteristics that impact phenotypic severity, including cognitive impairment. We previously demonstrated greater cognitive impairment in patients with IDH1 wild type tumor compared to those with IDH1 mutant, and therefore we hypothesized that brain network organization would be lower in patients with wild type tumors. Volumetric, T1-weighted MRI scans were obtained retrospectively from 35 patients with IDH1 mutant and 32 patients with wild type malignant astrocytoma (mean age = 45 ± 14 years) and used to extract individual level, gray matter connectomes. Graph theoretical analysis was then applied to measure efficiency and other connectome properties for each patient. Cognitive performance was categorized as impaired or not and random forest classification was used to explore factors associated with cognitive impairment. Patients with wild type tumor demonstrated significantly lower network efficiency in several medial frontal, posterior parietal and subcortical regions (p < 0.05, corrected for multiple comparisons). Patients with wild type tumor also demonstrated significantly higher incidence of cognitive impairment (p = 0.03). Random forest analysis indicated that network efficiency was inversely, though nonlinearly associated with cognitive impairment in both groups (p < 0.0001). Cognitive reserve appeared to mediate this relationship in patients with mutant tumor suggesting greater neuroplasticity and/or benefit from neuroprotective factors. Tumor volume was the greatest contributor to cognitive impairment in patients with wild type tumor, supporting our hypothesis that greater lesion momentum between grades may cause more disconnection of core neurocircuitry and consequently lower efficiency of information processing.
DOI: 10.1055/s-0037-1620238
2018
Cited 57 times
Origin of Gliomas
Malignant glioma is a common type of brain tumor that remains largely incurable. Although a definitive cell of origin of gliomas remains elusive, numerous population studies, sequencing efforts, and genetically engineered mouse models have contributed to our understanding of the early events that may lead to gliomagenesis. Herein we summarize our current knowledge on the population epidemiology of gliomas, heritable genetic risk factors, the somatic events that contribute to tumor evolution, and mouse models that have shed light on the glioma cell of origin. Future studies will increase our understanding of the sequence of early events within susceptible cells and their niche that trigger the path to malignant transformation. Such knowledge will allow us to design more effective treatment options to control tumor growth or screening methods for early detection.
DOI: 10.3171/2016.8.jns161076
2017
Cited 52 times
Targeted sequencing of SMO and AKT1 in anterior skull base meningiomas
OBJECTIVE Meningiomas located in the skull base are surgically challenging. Recent genomic research has identified oncogenic SMO and AKT1 mutations in a small subset of meningiomas. METHODS The authors performed targeted sequencing in a large cohort of patients with anterior skull base meningiomas (n = 62) to better define the frequency of SMO and AKT1 mutations in these tumors. RESULTS The authors found SMO mutations in 7 of 62 (11%) and AKT1 mutations in 12 of 62 (19%) of their cohort. Of the 7 meningiomas with SMO mutations, 6 (86%) occurred in the olfactory groove. Meningiomas with an SMO mutation presented with significantly larger tumor volume (70.6 ± 36.3 cm 3 ) compared with AKT1 -mutated (18.2 ± 26.8 cm 3 ) and wild-type (22.7 ± 23.9 cm 3 ) meningiomas, respectively. CONCLUSIONS Combined, these data demonstrate clinically actionable mutations in 30% of anterior skull base meningiomas and suggest an association between SMO mutation status and tumor volume. Genotyping of SMO and AKT1 is likely to be high yield in anterior skull base meningiomas with available surgical tissue.
DOI: 10.1038/s41698-017-0009-y
2017
Cited 52 times
Clinical and radiographic response following targeting of BCAN-NTRK1 fusion in glioneuronal tumor
Abstract Glioneuronal tumors constitute a histologically diverse group of primary central nervous system neoplasms that are typically slow-growing and managed conservatively. Genetic alterations associated with glioneuronal tumors include BRAF mutations and oncogenic fusions. To further characterize this group of tumors, we collected a cohort of 26 glioneuronal tumors and performed in-depth genomic analysis. We identified mutations in BRAF (34%) and oncogenic fusions (30%), consistent with previously published reports. In addition, we discovered novel oncogenic fusions involving members of the NTRK gene family in a subset of our cohort. One-patient with BCAN exon 13 fused to NTRK1 exon 11 initially underwent a subtotal resection for a 4th ventricular glioneuronal tumor but ultimately required additional therapy due to progressive, symptomatic disease. Given the patient’s targetable fusion, the patient was enrolled on a clinical trial with entrectinib, a pan-Trk, ROS1, and ALK (anaplastic lymphoma kinase) inhibitor. The patient was treated for 11 months and during this time volumetric analysis of the lesion demonstrated a maximum reduction of 60% in the contrast-enhancing tumor compared to his pre-treatment magnetic resonance imaging study. The radiologic response was associated with resolution of his clinical symptoms and was maintained for 11 months on treatment. This report of a BCAN-NTRK1 fusion in glioneuronal tumors highlights its clinical importance as a novel, targetable alteration.
DOI: 10.1158/2326-6066.cir-21-0870
2022
Cited 20 times
Microenvironmental Landscape of Human Melanoma Brain Metastases in Response to Immune Checkpoint Inhibition
Melanoma-derived brain metastases (MBM) represent an unmet clinical need because central nervous system progression is frequently an end stage of the disease. Immune checkpoint inhibitors (ICI) provide a clinical opportunity against MBM; however, the MBM tumor microenvironment (TME) has not been fully elucidated in the context of ICI. To dissect unique elements of the MBM TME and correlates of MBM response to ICI, we collected 32 fresh MBM and performed single-cell RNA sequencing of the MBM TME and T-cell receptor clonotyping on T cells from MBM and matched blood and extracranial lesions. We observed myeloid phenotypic heterogeneity in the MBM TME, most notably multiple distinct neutrophil states, including an IL8-expressing population that correlated with malignant cell epithelial-to-mesenchymal transition. In addition, we observed significant relationships between intracranial T-cell phenotypes and the distribution of T-cell clonotypes intracranially and peripherally. We found that the phenotype, clonotype, and overall number of MBM-infiltrating T cells were associated with response to ICI, suggesting that ICI-responsive MBMs interact with peripheral blood in a manner similar to extracranial lesions. These data identify unique features of the MBM TME that may represent potential targets to improve clinical outcomes for patients with MBM.
DOI: 10.1158/1078-0432.ccr-21-3622
2022
Cited 18 times
HSP90 Inhibition Overcomes Resistance to Molecular Targeted Therapy in <i>BRAFV600E</i>-mutant High-grade Glioma
Abstract Purpose: Molecular targeted therapy using BRAF and/or MEK inhibitors has been applied to BRAFV600E-mutant high-grade gliomas (HGG); however, the therapeutic effect is limited by the emergence of drug resistance. Experimental Design: We established multiple paired BRAFV600E-mutant HGG patient-derived xenograft models based on tissues collected prior to and at relapse after molecular targeted therapy. Using these models, we dissected treatment-resistant mechanisms for molecular targeted therapy and explored therapeutic targets to overcome resistance in BRAFV600E HGG models in vitro and in vivo. Results: We found that, despite causing no major genetic and epigenetic changes, BRAF and/or MEK inhibitor treatment deregulated multiple negative feedback mechanisms, which led to the reactivation of the MAPK pathway through c-Raf and AKT signaling. This altered oncogenic signaling primarily mediated resistance to molecular targeted therapy in BRAFV600E-mutant HGG. To overcome this resistance mechanism, we performed a high-throughput drug screening to identify therapeutic agents that potently induce additive cytotoxicity with BRAF and MEK inhibitors. We discovered that HSP90 inhibition combined with BRAF/MEK inhibition coordinately deactivated the MAPK and AKT/mTOR pathways, and subsequently induced apoptosis via dephosphorylation of GSK3β (Ser9) and inhibition of Bcl-2 family proteins. This mediated potent cytotoxicity in vitro and in vivo in refractory models with acquired resistance to molecular targeted therapy. Conclusions: The combination of an HSP90 inhibitor with BRAF or MEK inhibitors can overcome the limitations of the current therapeutic strategies for BRAFV600E-mutant HGG.
DOI: 10.1007/s11060-012-0926-0
2012
Cited 56 times
Butterfly glioblastomas: a retrospective review and qualitative assessment of outcomes
To address a paucity of literature and treatment guidelines regarding the management of butterfly glioblastomas, we performed a ten year retrospective analysis of data from twenty-three consecutive patients treated for this disease at a single institution. Clinical characteristics and outcomes were assessed. Median age was 59 years; 52 % were female; median preoperative Karnofsky performance score (KPS) was 80. Twelve patients underwent biopsy and eleven underwent surgical decompression. The median tumor volume for the biopsy group was 60.6 cm(3) and for the surgically decompressed group 40.5 cm(3). In the biopsy group, five patients received adjuvant therapy but one died prior to its completion; two died prior to the initiation of adjuvant therapy and five were lost to follow up. In the surgical decompression group, seven patients received adjuvant therapy, one died prior to the initiation of adjuvant therapy, two were treated with palliative measures only, and one was lost to follow up. Kaplan-Meier estimates of overall median post surgical-survival of the whole group was 180 days, the biopsy group 48 days, and the surgically decompressed group 265 days (p = 0.14). Our results show that there was a higher median survival in the surgically decompressed group; but a direct correlation could not be established, and that the median KPS did not improve in either group after treatment. A larger multicenter review is required to quantitatively assess the factors, including tumor biomarkers that are associated with patient outcome.
DOI: 10.1002/cncr.29742
2015
Cited 53 times
Glioblastoma care in the elderly
Glioblastoma is common among elderly patients, a group in which comorbidities and a poor prognosis raise important considerations when designing neuro-oncologic care. Although the standard of care for nonelderly patients with glioblastoma includes maximal safe surgical resection followed by radiotherapy with concurrent and adjuvant temozolomide, the safety and efficacy of these modalities in elderly patients are less certain given the population's underrepresentation in many clinical trials. The authors reviewed the clinical trial literature for reports on the treatment of elderly patients with glioblastoma to provide evidence-based guidance for practitioners. In elderly patients with glioblastoma, there is a survival advantage for those who undergo maximal safe resection, which likely includes an incremental benefit with increasing completeness of resection. Radiotherapy extends survival in selected patients, and hypofractionation appears to be more tolerable than standard fractionation. In addition, temozolomide chemotherapy is safe and extends the survival of patients with tumors that harbor O(6)-methylguanine-DNA methyltransferase (MGMT) promoter methylation. The combination of standard radiation with concurrent and adjuvant temozolomide has not been studied in this population. Although many questions remain unanswered regarding the treatment of glioblastoma in elderly patients, the available evidence provides a framework on which providers may base individual treatment decisions. The importance of tumor biomarkers is increasingly apparent in elderly patients, for whom the therapeutic efficacy of any treatment must be weighed against its potential toxicity. MGMT promoter methylation status has specifically demonstrated utility in predicting the efficacy of temozolomide and should be considered in treatment decisions when possible. Cancer 2016;122:189-197. © 2015 American Cancer Society.
DOI: 10.2217/cns.15.32
2015
Cited 51 times
Molecular background of oligodendroglioma: 1p/19q, IDH, TERT, CIC and FUBP1
Oligodendroglioma is the quintessential molecularly-defined brain tumor. The characteristic whole-arm loss of the long arm of chromosome 1 and the short arm of chromosome 19 (1p/19q-codeletion) within the genome of these tumors facilitated the reproducible molecular identification of this subcategory of gliomas. More recently, recurrent molecular genetic alterations have been identified to occur concurrently with 1p/19q-codeletion, and definitively identify these tumors, including mutations in IDH1/2, CIC, FUBP1, and the TERT promoter, as well as the absence of ATRX and TP53 alterations. These findings provide a foundation for the consistent diagnosis of this tumor type, upon which a generation of clinical investigators have assembled a strong evidence base for the effective treatment of this disease with radiation and chemotherapy.
DOI: 10.1002/jmri.24380
2013
Cited 48 times
3 Tesla intraoperative MRI for brain tumor surgery
Implementation of intraoperative magnetic resonance imaging (iMRI) has been shown to optimize the extent of resection and safety of brain tumor surgery. In addition, iMRI can help account for the phenomenon of brain shift and can help to detect complications earlier than routine postoperative imaging, which can potentially improve patient outcome. The higher signal-to-noise ratio offered by 3 Tesla (T) iMRI compared with lower field strength systems is particularly advantageous. The purpose of this article is to review the imaging protocols, imaging findings, and technical considerations related to 3T iMRI. To maximize efficiency, iMRI sequences can be tailored to particular types of tumors and procedures, including nonenhancing brain tumor surgery, enhancing brain tumor surgery, transsphenoidal pituitary tumor surgery, and laser ablation. Unique imaging findings on iMRI include the presence of surgically induced enhancement, which can be a potential confounder for residual enhancing tumor, and hyperacute hemorrhage, which tends to have intermediate signal on T1-weighted sequences and high signal on T2-weighted sequences due to the presence of oxyhemoglobin. MR compatibility and radiofrequency shielding pose particularly stringent technical constraints at 3T and influence the design and usage of the surgical suite with iMRI. J. Magn. Reson. Imaging 2014;39:1357–1365. © 2013 Wiley Periodicals, Inc.
DOI: 10.3171/2012.12.focus12355
2013
Cited 47 times
From genomics to the clinic: biological and translational insights of mutant IDH1/2 in glioma
The characterization of the genomic alterations across all human cancers is changing the way that malignant disease is defined and treated. This paradigm is extending to glioma, where the discovery of recurrent mutations in the isocitrate dehydrogenase 1 (IDH1) gene has shed new light on the molecular landscape in glioma and other IDH- mutant cancers. The IDH1 mutations are present in the vast majority of low-grade gliomas and secondary glioblastomas. Rapidly emerging work on the consequences of mutant IDH1 protein expression suggests that its neomorphic enzymatic activity catalyzing the production of the oncometabolite 2-hydroxyglutarate influences a range of cellular programs that affect the epigenome, transcriptional programs, hypoxia-inducible factor biology, and development. In the brief time since its discovery, knowledge of the IDH mutation status has had significant translational implications, and diagnostic tools are being used to monitor its expression and function. The concept of IDH1- mutant versus IDH1 -wild type will become a critical early distinction in diagnostic and treatment algorithms.
DOI: 10.1016/j.pharmthera.2016.10.011
2017
Cited 43 times
Treatment of brain metastases in the modern genomic era
Development of brain metastasis (BM) portends a dismal prognosis for patients with cancer. Melanomas and carcinomas of the lung, breast, and kidney are the most common malignancies to metastasize to the brain. Recent advances in molecular genetics have enabled the identification of actionable, clinically relevant genetic alterations within primary tumors and their corresponding metastases. Adoption of genotype-guided treatment strategies for the management of systemic malignancy has resulted in dramatic and durable responses. Unfortunately, despite these therapeutic advances, central nervous system (CNS) relapses are not uncommon. Although these relapses have historically been attributed to limited blood brain barrier penetration of anti-neoplastic agents, recent work has demonstrated genetic heterogeneity such that metastatic sites, including BM, harbor relevant genetic alterations that are not present in primary tumor biopsies. This improved insight into molecular mechanisms underlying site specific recurrences can inform strategies for targeting these oncogenic drivers. Thus, development of rational, genomically guided CNS-penetrant therapies is crucial for ongoing therapeutic success.
DOI: 10.1007/s002230001211
2001
Cited 78 times
Mutation Screening of the TNFRSF11A Gene Encoding Receptor Activator of NFkB (RANK) in Familial and Sporadic Paget's Disease of Bone and Osteosarcoma
DOI: 10.1007/s11910-014-0518-9
2014
Cited 42 times
Systemic Therapy of Brain Metastases
DOI: 10.1007/s11060-014-1667-z
2014
Cited 36 times
Phase 2 study of bosutinib, a Src inhibitor, in adults with recurrent glioblastoma
Tumor cell infiltration is a major mechanism of treatment escape in glioblastoma. Src is an intracellular tyrosine kinase that mediates tumor cell motility and invasiveness. We evaluated the efficacy and safety of bosutinib, a tyrosine kinase inhibitor that potently inhibits Src and Abl, in patients with recurrent glioblastoma. In this two-arm study, patients with histologically confirmed recurrent glioblastoma and ≤2 relapses, not previously treated with anti-vascular endothelial growth factor (VEGF) therapy, were administered oral bosutinib 400 mg daily. Arm A planned for 6 patients who were candidates for surgical resection to be given bosutinib for 7-9 days prior to resection. Arm B was a two-stage design phase 2 trial targeting 30 patients. The primary endpoint was progression-free survival at 6 months (PFS6) in Arm B. After 9 patients enrolled onto stage 1 of Arm B, 9 (100 %) patients progressed within 6 months. Therefore, the study met the pre-specified criteria for early closure and both Arms were closed. In Arm B, Median PFS was 7.71 weeks and median OS was 50 weeks. Best objective response was stable disease in one patient (11.1 %). Seven patients (77.8 %) had treatment-related AEs of any grade and 2 (22.2 %) were grade ≥3. Arm A was closed after 2 patients enrolled. Src activation was evident in all archival tumor samples. Bosutinib monotherapy does not appear to be effective in recurrent glioblastoma. However, Src remains a potential target based on its upregulation in tumor samples and role in glioma invasion.
DOI: 10.1634/theoncologist.2015-0342
2016
Cited 36 times
Elevation of Urinary 2-Hydroxyglutarate in <i>IDH</i>-Mutant Glioma
Recurrent mutations in the isocitrate dehydrogenase 1 (IDH1) and IDH2 genes, which are frequent in gliomas, result in marked accumulation of the metabolic by-product 2-hydroxyglutarate (2-HG) within tumors. In other malignancies, such as acute myeloid leukemia, presence of IDH mutation is associated with elevated 2-HG levels in serum or urine compartments. Circulating 2-HG in patients with glial malignancies has not been thoroughly investigated.In this study, we analyzed 2-HG levels in the serum and urine of a large set of patients with IDH-mutant and IDH-wild-type glioma, and the cerebrospinal fluid (CSF) from a subset of this cohort.We found that 2-HG was elevated in the urine of patients with IDH-mutant versus IDH-wild-type glioma, although no significant differences in 2-HG levels were observed in the serum or the small set of CSF samples obtained. Among patients with IDH-mutant glioma, 2-HG levels did not differ based on the histopathologic grade, genetic subtype (TP53 mutant or 1p/19q codeleted), presence of a canonical (IDH1 R132H) or noncanonical (any other IDH variant) mutation, or treatment type.Our finding suggests that urinary 2-HG is increased among patients with IDH-mutant gliomas, and may represent a future surrogate, noninvasive biomarker to aid in diagnosis, prognosis, and management.Patients with glioma who harbor mutations in isocitrate dehydrogenase genes showed selective elevation of the oncometabolite 2-hydroxyglutarate in the urine. Similar elevations were not identified in the serum or cerebrospinal fluid. 2-Hydroxyglutarate may serve as a useful, noninvasive biomarker to stratify patients newly diagnosed with glioma with regard to prognosis and management.
DOI: 10.1002/ijc.30929
2017
Cited 32 times
Blockade of transforming growth factor‐β signaling enhances oncolytic herpes simplex virus efficacy in patient‐derived recurrent glioblastoma models
Despite the current standard of multimodal management, glioblastoma (GBM) inevitably recurs and effective therapy is not available for recurrent disease. A subset of tumor cells with stem‐like properties, termed GBM stem‐like cells (GSCs), are considered to play a role in tumor relapse. Although oncolytic herpes simplex virus (oHSV) is a promising therapeutic for GBM, its efficacy against recurrent GBM is incompletely characterized. Transforming growth factor beta (TGF‐β) plays vital roles in maintaining GSC stemness and GBM pathogenesis. We hypothesized that oHSV and TGF‐β inhibitors would synergistically exert antitumor effects for recurrent GBM. Here we established a panel of patient‐derived recurrent tumor models from GBMs that relapsed after postsurgical radiation and chemotherapy, based on GSC‐enriched tumor sphere cultures. These GSCs are resistant to the standard‐of‐care temozolomide but susceptible to oHSVs G47Δ and MG18L. Inhibition of TGF‐β receptor kinase with selective targeted small molecules reduced clonogenic sphere formation in all tested recurrent GSCs. The combination of oHSV and TGF‐βR inhibitor was synergistic in killing recurrent GSCs through, in part, an inhibitor‐induced JNK‐MAPK blockade and increase in oHSV replication. In vivo , systemic treatment with TGF‐βR inhibitor greatly enhanced the antitumor effects of single intratumoral oHSV injections, resulting in cures in 60% of mice bearing orthotopic recurrent GBM. These results reveal a novel synergistic interaction of oHSV therapy and TGF‐β signaling blockade, and warrant further investigations aimed at clinical translation of this combination strategy for GBM patients.
DOI: 10.3171/2016.9.focus16325
2016
Cited 31 times
Diagnosis and management of craniopharyngiomas in the era of genomics and targeted therapy
Craniopharyngiomas are rare intracranial neoplasms that pose clinical challenges due to their location adjacent to vital structures. The authors have previously shown high mutation rates of BRAF V600E in papillary craniopharyngioma and of CTNNB1 in adamantinomatous craniopharyngioma. These activating driver mutations are potential therapeutic targets, and the authors have recently reported a significant response to BRAF/MEK inhibition in a patient with multiply recurrent PCP. As these targetable mutations warrant prospective research, the authors will be conducting a national National Cancer Institute–sponsored multicenter clinical trial to investigate BRAF/MEK inhibition in the treatment of craniopharyngioma. In this new era of genomic discovery, the treatment paradigm of craniopharyngioma is likely to change.
DOI: 10.1007/s11060-014-1451-0
2014
Cited 29 times
Diagnostic discrepancies in malignant astrocytoma due to limited small pathological tumor sample can be overcome by IDH1 testing
The accurate grading of malignant astrocytomas has significant prognostic and therapeutic implications. Traditional histopathological grading can be challenging due to regional tumor heterogeneity, especially in scenarios where small amounts of tissue are available for pathologic review. Here, we hypothesized that a critical tumor resection volume is needed for correct grading of astrocytomas by histopathology. For insufficient tissue sampling, IDH1 molecular testing can act as a complementary marker to improve diagnostic accuracy. Volumetric analyses were obtained using preoperative and postoperative MRI images. Histological specimens were gathered from 403 patients with malignant astrocytoma who underwent craniotomy. IDH1 status was assessed by immunohistochemistry and sequencing. Patients with >20 cubic centimeters (cc) of the total tumor volume resected on MRI have higher rate of GBM diagnosis compared to <20 cc [odds ratio (OR) 2.57, 95 % confidence interval (CI) 1.6–4.06, P < 0.0001]. The rate of IDH1 status remained constant regardless of the tumor volume resected (OR 0.81, 95 % CI 0.48–1.36, P < 0.43). The rate of GBM diagnosis is twofold greater for individual surgical specimen >10 cc than those of lower volume (OR 2.48, 95 % CI 1.88–3.28, P < 0.0001). Overall survival for AA patients with >20 cc tumor resection on MRI is significantly better than those with <20 cc tumor resected (P < 0.05). No volume-dependent differences were observed in patients with GBM (P < 0.4), IDH1 wild type (P < 0.1) or IDH1 mutation (P < 0.88). IDH1 status should be considered when total resection volume is <20 cc based on MRI analysis and for surgical specimen <10 cc to complement histopathologic diagnosis of malignant astrocytomas. In these specimens, under-diagnosis of GBM may occur when analysis is restricted to histopathology alone.
DOI: 10.1038/s41698-017-0035-9
2017
Cited 29 times
Resolving the phylogenetic origin of glioblastoma via multifocal genomic analysis of pre-treatment and treatment-resistant autopsy specimens
Glioblastomas are malignant neoplasms composed of diverse cell populations. This intratumoral diversity has an underlying architecture, with a hierarchical relationship through clonal evolution from a common ancestor. Therapies are limited by emergence of resistant subclones from this phylogenetic reservoir. To characterize this clonal ancestral origin of recurrent tumors, we determined phylogenetic relationships using whole exome sequencing of pre-treatment IDH1/2 wild-type glioblastoma specimens, matched to post-treatment autopsy samples (n = 9) and metastatic extracranial post-treatment autopsy samples (n = 3). We identified "truncal" genetic events common to the evolutionary ancestry of the initial specimen and later recurrences, thereby inferring the identity of the precursor cell population. Mutations were identified in a subset of cases in known glioblastoma genes such as NF1(n = 3), TP53(n = 4) and EGFR(n = 5). However, by phylogenetic analysis, there were no protein-coding mutations as recurrent truncal events across the majority of cases. In contrast, whole copy-loss of chromosome 10 (12 of 12 cases), copy-loss of chromosome 9p21 (11 of 12 cases) and copy-gain in chromosome 7 (10 of 12 cases) were identified as shared events in the majority of cases. Strikingly, mutations in the TERT promoter were also identified as shared events in all evaluated pairs (9 of 9). Thus, we define four truncal non-coding genomic alterations that represent early genomic events in gliomagenesis, that identify the persistent cellular reservoir from which glioblastoma recurrences emerge. Therapies to target these key early genomic events are needed. These findings offer an evolutionary explanation for why precision therapies that target protein-coding mutations lack efficacy in GBM.
DOI: 10.1007/s11060-015-1866-2
2015
Cited 28 times
The role of biopsy in the management of patients with presumed diffuse low grade glioma
DOI: 10.1093/neuros/nyx247
2017
Cited 27 times
IDH1 Mutation and World Health Organization 2016 Diagnostic Criteria for Adult Diffuse Gliomas
2-HG: 2-hydroxyglutarate GBM: glioblastoma multiform IDH: isocitrate dehydrogenase MGMT: O6-methylguanine DNA-methyltransferase MRS: magnetic resonance spectroscopy NADPH: nicotinamide adenine dinucleotide phosphate NCF: neurocognitive function NOS: not otherwise specified OS: overall survival PFS: progression-free survival WHO: World Health Organization The presence of an isocitrate dehydrogenase gene (IDH1 or IDH2) mutation has become one of the most critical biomarkers for molecular classification and prognostication in adult diffuse gliomas.1 Here, we review the translational impact of IDH1/2 mutation on neurosurgical oncology, with a focus on how this emerging knowledge has advanced the precision of our surgical approach to these diseases. Generally speaking, there are 2 major goals for the initial surgical procedure in a patient with suspected adult diffuse glioma. The first goal is to obtain sufficient tissue for diagnostic classification. In the era preceding the recent World Health Organization (WHO) 2016 revised criteria,2 there were limitations on diagnosis imposed by surgical sampling error. In other words, the extent of surgery and subsequent diagnostic grading of an adult diffuse glioma had been shown to be tightly linked.3,4 Patients who underwent biopsy only, unfortunately, would too often have inaccurate diagnoses, due to so-called undergrading, as there would be insufficient material for pathological review. This undergrading complicated the retrospective analyses of surgical treatment, since biopsy-only diagnoses were frequently inaccurate. However, with the molecular genomic component of classifiers codified into the revised diagnostic criteria (primarily IDH1 mutation and 1p/19q-codeletion, as described below), this scenario has occurred more rarely,5 since less tissue is required for molecular testing. The second goal of surgery, in most cases, is to perform therapeutic cytoreduction to secure a prolonged survival and preservation of neurological function for the patient. For glioblastoma multiform (GBM), this has traditionally meant that “complete resection of enhancement” was the intended surgical goal,6,7 while for lower grade lesions, which are mostly nonenhancing, this has meant “complete resection of T2/FLAIR hyper-intensity”8 (Figure A). The evidence base that supports these surgical strategies requires updating in the context of the new WHO 2016 diagnostic criteria.FIGURE. For: IDH1 wild-type tumors, the surgical goal is “complete resection of enhancement,” while for IDH1 mutant astrocytomas, which are mostly nonenhancing, the goal is “complete resection of T2/FLAIR hyper-intensity” A and B. Further studies are needed to determine the optimal targeting of radiation therapy for IDH1 mutant gliomas C.ETIOLOGY AND CLASSIFICATION FOR IDH MUTANT GLIOMAS Recurrent mutations of the IDH1 gene were initially identified in 12% of grade IV GBM within a broad sequence screen of more than 20,000 genes.9 Subsequently, more focused large sample studies confirmed that IDH1 mutation is found in the majority of secondary GBM, and only rarely found in primary GBM and GBM in children.9-11 In addition, 50% to 80% of lower grade gliomas (categorized as grade II or III by the legacy WHO 2007 criteria) harbored IDH1 mutation.10-13 The IDH1 gene mutation is almost always localized within exon 4 to codon 132 and >90% of alterations are c.395G>A (R132H) substitutions, followed by R132C as the second-most common alteration.10,12,14 Although the frequency is rare, mutations in the homologous gene IDH2 are also found in gliomas categorized as grade II or III by the legacy WHO 2007 criteria, and secondary GBM.11,15 From a classification perspective, the discovery of IDH1 mutation allows the clear distinction between primary GBM, which frequently harbors epidermal growth factor receptor, PTEN loss, and cyclin-dependent kinase inhibitor 2A (CDKN2A) gene, deletions, versus secondary GBM, which harbors IDH1 mutation.10,12,16,17 With the revision of adult diffuse glioma classification, the 2016 WHO Classification of Tumors of the Central Nervous System integrated phenotypic and genotypic parameters.2 For IDH1 mutant gliomas, tumors are grouped as diffuse astrocytic or oligodendroglial tumors. This group was histologically and genetically divided based on the presence of IDH mutations (typically IDH1R132 and IDH2R172) and 1p/19q codeletion. As an additional reinforcement of this molecular classification, astrocytic gliomas containing IDH1 mutation also near-universally contain TP53 and ATRX gene mutation, whereas oligodendrogliomas are IDH1 mutant with 1p/19q codeletion, often with concomitant CIC and FUBP1 gene and TERT promoter mutation.18,19 These co-occurring genetic abnormalities are mutually exclusive in the vast majority of cases.18-22 Accordingly, most tumors are classified as follows: (1) diffuse astrocytoma (grade II) or anaplastic astrocytoma (grade III) or glioblastoma (grade IV); IDH-mutant, -wild type, or not otherwise specified (NOS); (2) oligodendroglioma (grade II) or anaplastic oligodendroglioma (grade III; IDH mutant and 1p/19q-codeleted or NOS). Remaining cases, which are IDH1 wild type, are classified as (1) oligoastrocytoma (grade II), anaplastic oligoastrocytoma (grade III) (NOS); or (2) diffuse midline glioma (H3K27M-mutant). IDH-wild type glioblastoma (about 90% of cases) is known as primary GBM, while IDH mutant glioblastoma (about 10% of cases) corresponds to secondary GBM.2 PROGNOSTIC SIGNIFICANCE OF IDH1 MUTATION IN GLIOMAS In GBM, Parsons and colleagues9 initially demonstrated that the overall survival (OS) in IDH1-mutant GBM was more than 3-fold longer than that in IDH1 wild-type GBM. Independent groups rapidly replicated the finding that IDH1 mutation is a favorable prognostic biomarker of both progression-free survival (PFS) and OS when compared to IDH1 wild type in low-grade glioma and high-grade glioma.11,13,23 Subsequently, the majority of clinical studies indicated that IDH mutation was an independent prognostic factor in grade II and III gliomas.11,23-29 This evidence indicates that IDH1 mutation is a favorable prognostic factor in adult gliomas. Among these studies, the prospective randomized study NOA-04 revealed IDH1 mutation, hypermethylation of the O6-methylguanine DNA-methyltransferase (MGMT) promoter, age, extent of resection, and oligodendroglial histology are independent prognostic factors in anaplastic gliomas.24 Of note, the impact of IDH1 mutation conferred a stronger risk reduction than 1p/19q codeletion, MGMT promoter methylation, or histology.24 In secondary high-grade gliomas, IDH mutations are also stronger prognostic markers of both PFS and OS than the MGMT promoter methylation status.30 Notably, the prognosis of IDH1 mutant GBM is better than anaplastic astrocytoma without IDH1 mutation.31 Taken together, IDH1 mutation has proven to be a powerful prognostic factor in gliomas, irrespective of tumor grade and histology. Additional clinical characteristics in the IDH1 mutant gliomas are the tumor location and age distribution of the patients upon presentation. Compared with IDH wild type, IDH1 mutant gliomas were predominantly located in the frontal lobe.32-36 Patients with GBM or anaplastic astrocytoma with IDH1 mutation were significantly younger than that with IDH1 wild type.9,11 Intriguingly, the patient age at diagnosis of grade II IDH1 mutant astrocytoma is nearly identical grade III IDH1 mutant anaplastic astrocytoma. Also, the age of IDH1 mutant GBM was only 4 yr older than that of IDH1 mutant grade II and III astrocytoma.37 These findings highlighted the fact that grading, per se, had not been validated as a prognostic marker within the genomically homogeneous cohorts of IDH1 mutant vs wild-type tumors, and serves as a cautionary note for future analyses. Notably in this regard, Suzuki et al29 classified gliomas that were grades II and III by WHO 2007 criteria on the basis of the presence of IDH1 mutation, TP53 mutation, and 1p/19q codeletion. Accordingly, tumors were classified into 3 groups: type I (IDH1 mutant with 1p/19q codeletion; favorable prognostic group), type II (IDH1 mutant with TP53 mutation; intermediate group), and type III (IDH1 wild type; poor prognostic group).29 Survival difference between grade II and grade III were observed only in type II (astrocytic), but not in type I (oligodendroglial) gliomas,29 findings consistent with the results from large randomized studies of grade II and III oligodendrogliomas.38,39 TREATMENT EVIDENCE FOR IDH1 MUTANT GLIOMAS Although scant class I evidence exists, accumulating evidence supports the proposal that more extensive surgical resection has a pivotal role in improving survival in adults with glioma. Extensive resection has been demonstrated to be associated with a survival benefit in low-grade glioma and also in GBM (IDH1 wild type).6,8,40,41 Of note, magnetic resonance (MR) imaging studies have demonstrated IDH1 mutant tumors to be rarely located in high-risk (so-called eloquent) areas of the brain, with a typically unilateral pattern of growth, sharp tumor margin, and less contrast enhancement,32,42 implying IDH1 mutant gliomas are relatively more feasible for resection, when compared to their wild-type counterparts. Intriguingly, patients with IDH1 wild-type gliomas also display reduced neurocognitive function (NCF) and lower performance score than those with IDH1 mutant gliomas.43 In addition, glioma tumor volume was not associated with NCF for patients with IDH1 mutant tumors, but was associated decreased NCF in IDH1 wild-type tumors.43 Diffusion-tensor imaging studies demonstrate that IDH mutant GBM have a less invasive phenotype compared to IDH wild-type lesions.44 Interestingly, we found that extensive resection including nonenhancing area prolonged survival in IDH1 mutant anaplastic astrocytoma and glioblastoma. Since IDH1 mutant gliomas were predominantly located in frontal lobe and the less functional disturbance of adjacent normal brain, IDH1 mutant gliomas were also more amenable to maximal resection.35 These findings were consistent with an independent study, on a separate cohort, demonstrating that the gross total resection was associated with extended survival in grade III IDH1 mutant gliomas without 1p/19q codeletion, but not in IDH1 wild-type or IDH1 mutant gliomas with 1p/19q codeletion.45 Altogether, these findings suggest that extensive resection of both enhancing and nonenhancing (T2/FLAIR hyperintense) disease should be considered for IDH1 mutant gliomas, especially astrocytoma, regardless of WHO grade46 (Figure A and B). To assess for mutant IDH1 noninvasively, several MR techniques including diffusion tensor imaging, relative cerebral blood volume, and magnetic resonance spectroscopy (MRS) have been reported.47-49 MRS can detect 2-hydroxyglutarate (2-HG), which is produced by the IDH mutant enzyme product and is found at levels 100-fold higher in tumors, than that of normal brain.50-58 Additionally, intraoperative technologies to rapidly assess for IDH1 mutation have been established.59-61 Advances in these technologies may allow the surgical strategy to determine the degree of resection to be adjusted intraoperatively during a surgical procedure, based on the IDH1 mutation status of the tumor. Although there is no level I evidence that radiation therapy extends survival in glioma patients with IDH1 mutation, experimental investigations have revealed that forced IDH1 mutant expression in glioma cells results in increased reactive oxygen species, by inhibiting nicotinamide adenine dinucleotide phosphate (NADPH) production, which promotes sensitivity to radiation therapy.62-64 Indeed, 65% of the total NADPH production capacity in GBM is provided for by wild-type IDH activity and introduction of the IDH1 mutation reduced this capacity by 38%.65 In addition, MRS demonstrated that IDH1 mutation decreased glutathione level compared with those with IDH1 wild type.51 Intriguingly, in ATRX mutant tumors, nonhomologous end joining was impaired and increased sensitivity to DNA damaging agents that induce double-stranded DNA breaks.66 These findings may support the recent clinical data that patients with IDH mutant, 1p/19q non-codeleted (astrocytic) tumors treated with radiotherapy had a longer PFS than those treated with temozolomide, whereas no differences in PFS for patients with IDH mutant, 1p/19q codeleted (oligodendroglial), and IDH wild-type (GBM-like) tumors.67 Recently, MRS detected 2-HG has been piloted in the clinical assessment of treatment response and treatment planning in radiotherapy,56,68,69 indicating the potential for clinical application of noninvasively assessed 2-HG. Further work is needed to determine the optimal targeting of radiation therapy for IDH1 mutant gliomas (Figure C). CONCLUSION In conclusion, with the revision of the WHO diagnostic criteria, surgery for adult diffuse gliomas has become even more tightly integrated with radiology and pathology, in both the diagnostic phase as well as the treatment phase of these diseases. Certain cases, namely IDH1 mutant astrocytic gliomas, display a substantial survival benefit in association with maximal resection, regardless of tumor grade under the legacy criteria. Thus, individualization of surgical strategy for patients with IDH1 mutant gliomas has advanced significantly in the modern era. Disclosures This work was supported by NIH P50CA165962 and Burroughs-Wellcome CAMS # 1007616.02. The authors have no personal, financial, or institutional interest in any of the drugs, materials, or devices described in this article.
DOI: 10.1093/neuonc/now031
2016
Cited 26 times
A new patient-derived orthotopic malignant meningioma model treated with oncolytic herpes simplex virus
Higher-grade meningiomas (HGMs; World Health Organization grades II and III) pose a clinical problem due to high recurrence rates and the absence of effective therapy. Preclinical development of novel therapeutics requires a disease model that recapitulates the genotype and phenotype of patient HGM. Oncolytic herpes simplex virus (oHSV) has shown efficacy and safety in cancers in preclinical and clinical studies, but its utility for HGM has not been well characterized.Tumorsphere cultures and serial orthotopic xenografting in immunodeficient mice were used to establish a patient-derived HGM model. The model was pathologically and molecularly characterized by immunohistochemistry, western blot, and genomic DNA sequencing and compared with the patient tumor. Anti-HGM effects of oHSV G47Δ were assessed using cell viability and virus replication assays in vitro and animal survival analysis following intralesional injections of G47Δ.We established a serially transplantable orthotopic malignant meningioma model, MN3, which was lethal within 3 months after tumorsphere implantation. MN3 xenografts exhibited the pathological hallmarks of malignant meningioma such as high Ki67 and vimentin expression. Both the patient tumor and xenografts were negative for neurofibromin 2 (merlin) and had the identical NF2 mutation. Oncolytic HSV G47Δ efficiently spread and killed MN3 cells, as well as other patient-derived HGM lines in vitro. Treatment with G47Δ significantly extended the survival of mice bearing subdural MN3 tumors.We established a new patient-derived meningioma model that will enable the study of targeted therapeutic approaches for HGM. Based on these studies, it is reasonable to consider a clinical trial of G47Δ for HGM.
DOI: 10.1016/j.pharmthera.2017.08.006
2018
Cited 25 times
Molecular pathogenesis and therapeutic implications in pediatric high-grade gliomas
High-grade gliomas (HGG) are the most common malignant brain tumors in the pediatric population and account for a large subset of all pediatric central nervous system neoplasms. The management of pediatric HGG continues to be challenging, with poor outcome in many cases despite aggressive treatments. Consequently, parallel research efforts have been focused on identifying the underlying genetic and biological basis of pediatric HGG in order to more clearly define prognostic subgroups for treatment stratification as well as identify new treatment targets. These cutting-edge advances have revolutionized pediatric neuro-oncology and have revealed novel oncogenic vulnerabilities that are being therapeutically leveraged. Promising treatments - including pathway-targeting small molecules as well as epigenetic therapy - are being evaluated in clinical trials, and recent genomic discoveries in rare glioma subgroups have led to the identification of additional new potentially-actionable alterations. This review summarizes the current state of knowledge about the molecular characterization of pediatric HGG in correlation to the revised World Health Organization (WHO) classification, as well as provides an overview of some targeted treatment approaches in the modern clinical management of high-grade gliomas.
DOI: 10.1158/1078-0432.ccr-22-1896
2023
Cited 3 times
Zinc Finger MYND-Type Containing 8 (ZMYND8) Is Epigenetically Regulated in Mutant Isocitrate Dehydrogenase 1 (IDH1) Glioma to Promote Radioresistance
Mutant isocitrate dehydrogenase 1 (mIDH1) alters the epigenetic regulation of chromatin, leading to a hypermethylation phenotype in adult glioma. This work focuses on identifying gene targets epigenetically dysregulated by mIDH1 to confer therapeutic resistance to ionizing radiation (IR).We evaluated changes in the transcriptome and epigenome in a radioresistant mIDH1 patient-derived glioma cell culture (GCC) following treatment with an mIDH1-specific inhibitor, AGI-5198. We identified Zinc Finger MYND-Type Containing 8 (ZMYND8) as a potential target of mIDH1 reprogramming. We suppressed ZMYND8 expression by shRNA knockdown and genetic knockout (KO) in mIDH1 glioma cells and then assessed cellular viability to IR. We assessed the sensitivity of mIDH1 GCCS to pharmacologic inhibition of ZMYND8-interacting partners: HDAC, BRD4, and PARP.Inhibition of mIDH1 leads to an upregulation of gene networks involved in replication stress. We found that the expression of ZMYND8, a regulator of DNA damage response, was decreased in three patient-derived mIDH1 GCCs after treatment with AGI-5198. Knockdown of ZMYND8 expression sensitized mIDH1 GCCs to radiotherapy marked by decreased cellular viability. Following IR, mIDH1 glioma cells with ZMYND8 KO exhibit significant phosphorylation of ATM and sustained γH2AX activation. ZMYND8 KO mIDH1 GCCs were further responsive to IR when treated with either BRD4 or HDAC inhibitors. PARP inhibition further enhanced the efficacy of radiotherapy in ZMYND8 KO mIDH1 glioma cells.These findings indicate the impact of ZMYND8 in the maintenance of genomic integrity and repair of IR-induced DNA damage in mIDH1 glioma. See related commentary by Sachdev et al., p. 1648.
DOI: 10.1093/neuonc/noad118
2023
Cited 3 times
Neurotoxicity and management of primary and secondary central nervous system lymphoma after adoptive immunotherapy with CD19-directed chimeric antigen receptor T-cells
Chimeric antigen receptor (CAR) T-cells targeting CD19 have been established as a leading engineered T-cell therapy for B-cell lymphomas; however, data for patients with central nervous system (CNS) involvement are limited.We retrospectively report on CNS-specific toxicities, management, and CNS response of 45 consecutive CAR T-cell transfusions for patients with active CNS lymphoma at the Massachusetts General Hospital over a 5-year period.Our cohort includes 17 patients with primary CNS lymphoma (PCNSL; 1 patient with 2 CAR T-cell transfusions) and 27 patients with secondary CNS lymphoma (SCNSL). Mild ICANS (grade 1-2) was observed after 19/45 transfusions (42.2%) and severe immune effector cell-associated neurotoxicity syndrome (ICANS) (grade 3-4) after 7/45 transfusions (15.6%). A larger increase in C-reactive protein (CRP) levels and higher rates of ICANS were detected in SCNSL. Early fever and baseline C-reactive protein levels were associated with ICANS occurrence. CNS response was seen in 31 cases (68.9%), including a complete response of CNS disease in 18 cases (40.0%) which lasted for a median of 11.4 ± 4.5 months. Dexamethasone dose at time of lymphodepletion (but not at or after CAR T-cell transfusion) was associated with an increased risk for CNS progression (hazard ratios [HR] per mg/d: 1.16, P = .031). If bridging therapy was warranted, the use of ibrutinib translated into favorable CNS-progression-free survival (5 vs. 1 month, HR 0.28, CI 0.1-0.7; P = .010).CAR T-cells exhibit promising antitumor effects and a favorable safety profile in CNS lymphoma. Further evaluation of the role of bridging regimens and corticosteroids is warranted.
DOI: 10.1158/1078-0432.ccr-23-1660
2023
Cited 3 times
Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
Abstract Purpose: The 2021 World Health Organization (WHO) classification of central nervous system (CNS) tumors uses an integrated approach involving histopathology and molecular profiling. Because majority of adult malignant brain tumors are gliomas and primary CNS lymphomas (PCNSL), rapid differentiation of these diseases is required for therapeutic decisions. In addition, diffuse gliomas require molecular information on single-nucleotide variants (SNV), such as IDH1/2. Here, we report an intraoperative integrated diagnostic (i-ID) system to classify CNS malignant tumors, which updates legacy frozen-section (FS) diagnosis through incorporation of a qPCR-based genotyping assay. Experimental Design: FS evaluation, including GFAP and CD20 rapid IHC, was performed on adult malignant CNS tumors. PCNSL was diagnosed through positive CD20 and negative GFAP immunostaining. For suspected glioma, genotyping for IDH1/2, TERT SNV, and CDKN2A copy-number alteration was routinely performed, whereas H3F3A and BRAF SNV were assessed for selected cases. i-ID was determined on the basis of the 2021 WHO classification and compared with the permanent integrated diagnosis (p-ID) to assess its reliability. Results: After retrospectively analyzing 153 cases, 101 cases were prospectively examined using the i-ID system. Assessment of IDH1/2, TERT, H3F3AK27M, BRAFV600E, and CDKN2A alterations with i-ID and permanent genomic analysis was concordant in 100%, 100%, 100%, 100%, and 96.4%, respectively. Combination with FS and intraoperative genotyping assay improved diagnostic accuracy in gliomas. Overall, i-ID matched with p-ID in 80/82 (97.6%) patients with glioma and 18/19 (94.7%) with PCNSL. Conclusions: The i-ID system provides reliable integrated diagnosis of adult malignant CNS tumors.
DOI: 10.1093/neuonc/not229
2013
Cited 25 times
Where are we now? And where are we going? A report from the Accelerate Brain Cancer Cure (ABC2) Low-grade Glioma Research Workshop
Diffuse gliomas consist of both low- and high-grade varieties, each with distinct morphological and biological features. The often extended periods of relative indolence exhibited by low-grade gliomas (LGG; WHO grade II) differ sharply from the aggressive, rapidly fatal clinical course of primary glioblastoma (GBM; WHO grade IV). Nevertheless, until recently, the molecular foundations underlying this stark biological contrast between glioma variants remained largely unknown. The discoveries of distinctive and highly recurrent genomic and epigenomic abnormalities in LGG have both informed a more accurate classification scheme and pointed to viable avenues for therapeutic development. As such, the field of neuro-oncology now seems poised to capitalize on these gains to achieve significant benefit for LGG patients. This report will briefly recount the proceedings of a workshop held in January 2013 and hosted by Accelerate Brain Cancer Cure (ABC(2)) on the subject of LGG. While much of the meeting covered recent insights into LGG biology, its focus remained on how best to advance the clinical management, whether by improved preclinical modeling, more effective targeted therapeutics and clinical trial design, or innovative imaging technology.
DOI: 10.1007/s11060-013-1116-4
2013
Cited 23 times
Outcomes for patients with anaplastic astrocytoma treated with chemoradiation, radiation therapy alone or radiation therapy followed by chemotherapy: a retrospective review within the era of temozolomide
DOI: 10.1038/s41467-018-04235-3
2018
Cited 19 times
Neural encoding and production of functional morphemes in the posterior temporal lobe
Morphemes are the smallest meaning-carrying units in human language, and are among the most basic building blocks through which humans express specific ideas and concepts. By using time-resolved cortical stimulations, neural recordings, and focal lesion evaluations, we show that inhibition of a small cortical area within the left dominant posterior-superior temporal lobe selectively impairs the ability to produce appropriate functional morphemes but does not distinctly affect semantic and lexical retrieval, comprehension, or articulation. Additionally, neural recordings within this area reveal the localized encoding of morphological properties and their planned production prior to speech onset. Finally, small lesions localized to the gray matter in this area result in a selective functional morpheme-production deficit. Collectively, these findings reveal a detailed division of linguistic labor within the posterior-superior temporal lobe and suggest that functional morpheme processing constitutes an operationally discrete step in the series of computations essential to language production.
DOI: 10.1016/j.bbcan.2024.189102
2024
Molecular biology and novel therapeutics for IDH mutant gliomas: The new era of IDH inhibitors
Gliomas with Isocitrate dehydrogenase (IDH) mutation represent a discrete category of primary brain tumors with distinct and unique characteristics, behaviors, and clinical disease outcomes. IDH mutations lead to aberrant high-level production of the oncometabolite D-2-hydroxyglutarate (D-2HG), which act as a competitive inhibitor of enzymes regulating epigenetics, signaling pathways, metabolism, and various other processes. This review summarizes the significance of IDH mutations, resulting upregulation of D-2HG and the associated molecular pathways in gliomagenesis. With the recent finding of clinically effective IDH inhibitors in these gliomas, this article offers a comprehensive overview of the new era of innovative therapeutic approaches based on mechanistic rationales, encompassing both completed and ongoing clinical trials targeting gliomas with IDH mutations.
DOI: 10.1016/j.anndiagpath.2011.08.010
2012
Cited 21 times
Absence of IDH1-R132H mutation predicts rapid progression of nonenhancing diffuse glioma in older adults
Advanced age and contrast enhancement portend a poor prognosis in diffuse glioma (DG). Diffuse glioma may present as nonenhancing tumors that rapidly progress in weeks to months to a pattern of ring enhancement, characteristic of glioblastoma (GBM). Mutations involving isocitrate dehydrogenase 1 (IDH1) have recently emerged as important diagnostic and prognostic markers in DG. R132H is the most common mutation, expressed in more than 80% of DG and secondary GBM but in less than 10% of primary GBM. Adults older than 50 years with nonenhancing, rapidly progressing DG were identified. A comparison group comprised randomly selected, age-matched patients with nonenhancing, nonprogressing DG. Isocitrate dehydrogenase 1 status was evaluated using anti–IDH1-R132H antibodies (Dianova, Hamburg, Germany). The results were correlated with the clinical outcomes. We identified 4 patients who presented with nonenhancing DG that rapidly progressed to ring-enhancing lesions that were subsequently diagnosed on surgical resection as GBM. This group showed absent IDH1-R132H expression, which is characteristic of primary GBM. The comparison group of 5 patients presented with nonenhancing, nonprogressing DG, and all 5 tumors showed IDH1-R132H expression. In conclusion, negative IDH1-R132H mutation status in nonenhancing DG of older adults is a poor prognostic factor associated with rapid progression to ring-enhancing GBM. The shorter interval of progression and negative IDH1-R132H mutation status suggest a similar molecular pathway as seen in primary GBM.
DOI: 10.1093/neuonc/noad160
2023
Surgical management and outcome of newly diagnosed glioblastoma without contrast enhancement (<i>low-grade appearance</i>): a report of the RANO <i>resect</i> group
Resection of the contrast-enhancing (CE) tumor represents the standard of care in newly diagnosed glioblastoma. However, some tumors ultimately diagnosed as glioblastoma lack contrast enhancement and have a 'low-grade appearance' on imaging (non-CE glioblastoma). We aimed to (a) volumetrically define the value of non-CE tumor resection in the absence of contrast enhancement, and to (b) delineate outcome differences between glioblastoma patients with and without contrast enhancement.The RANO resect group retrospectively compiled a global, eight-center cohort of patients with newly diagnosed glioblastoma per WHO 2021 classification. The associations between postoperative tumor volumes and outcome were analyzed. Propensity score-matched analyses were constructed to compare glioblastomas with and without contrast enhancement.Among 1323 newly diagnosed IDH-wildtype glioblastomas, we identified 98 patients (7.4%) without contrast enhancement. In such patients, smaller postoperative tumor volumes were associated with more favorable outcome. There was an exponential increase in risk for death with larger residual non-CE tumor. Accordingly, extensive resection was associated with improved survival compared to lesion biopsy. These findings were retained on a multivariable analysis adjusting for demographic and clinical markers. Compared to CE glioblastoma, patients with non-CE glioblastoma had a more favorable clinical profile and superior outcome as confirmed in propensity score analyses by matching the patients with non-CE glioblastoma to patients with CE glioblastoma using a large set of clinical variables.The absence of contrast enhancement characterizes a less aggressive clinical phenotype of IDH-wildtype glioblastomas. Maximal resection of non-CE tumors has prognostic implications and translates into favorable outcome.
DOI: 10.1158/1078-0432.24947760
2024
Supplementary Table S5 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Mismatch ratio of genomic alterations in the prospective cohort&lt;/p&gt;
DOI: 10.1158/1078-0432.24947754
2024
Supplementary Table S6 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;IDH- and pTERT-wildtype gliomas at intraoperative integrated diagnosis in the prospective cohort&lt;/p&gt;
DOI: 10.1158/1078-0432.24947793.v1
2024
Supplementary Fig. S1 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Single nucleotide detection threshold&lt;/p&gt;
DOI: 10.1158/1078-0432.24947787.v1
2024
Supplementary Fig. S3 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Illustrative cases in the retrospective cohort&lt;/p&gt;
DOI: 10.1158/1078-0432.c.7010445
2024
Data from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;div&gt;AbstractPurpose:&lt;p&gt;The 2021 World Health Organization (WHO) classification of central nervous system (CNS) tumors uses an integrated approach involving histopathology and molecular profiling. Because majority of adult malignant brain tumors are gliomas and primary CNS lymphomas (PCNSL), rapid differentiation of these diseases is required for therapeutic decisions. In addition, diffuse gliomas require molecular information on single-nucleotide variants (SNV), such as &lt;i&gt;IDH1&lt;/i&gt;/2. Here, we report an intraoperative integrated diagnostic (&lt;i&gt;i&lt;/i&gt;-ID) system to classify CNS malignant tumors, which updates legacy frozen-section (FS) diagnosis through incorporation of a qPCR-based genotyping assay.&lt;/p&gt;Experimental Design:&lt;p&gt;FS evaluation, including GFAP and CD20 rapid IHC, was performed on adult malignant CNS tumors. PCNSL was diagnosed through positive CD20 and negative GFAP immunostaining. For suspected glioma, genotyping for &lt;i&gt;IDH1/2&lt;/i&gt;, &lt;i&gt;TERT&lt;/i&gt; SNV, and &lt;i&gt;CDKN2A&lt;/i&gt; copy-number alteration was routinely performed, whereas &lt;i&gt;H3F3A&lt;/i&gt; and &lt;i&gt;BRAF&lt;/i&gt; SNV were assessed for selected cases. &lt;i&gt;i&lt;/i&gt;-ID was determined on the basis of the 2021 WHO classification and compared with the permanent integrated diagnosis (p-ID) to assess its reliability.&lt;/p&gt;Results:&lt;p&gt;After retrospectively analyzing 153 cases, 101 cases were prospectively examined using the &lt;i&gt;i&lt;/i&gt;-ID system. Assessment of &lt;i&gt;IDH1/2&lt;/i&gt;, &lt;i&gt;TERT&lt;/i&gt;, &lt;i&gt;H3F3A&lt;sup&gt;K27M&lt;/sup&gt;&lt;/i&gt;, &lt;i&gt;BRAF&lt;sup&gt;V600E&lt;/sup&gt;&lt;/i&gt;, and &lt;i&gt;CDKN2A&lt;/i&gt; alterations with &lt;i&gt;i&lt;/i&gt;-ID and permanent genomic analysis was concordant in 100%, 100%, 100%, 100%, and 96.4%, respectively. Combination with FS and intraoperative genotyping assay improved diagnostic accuracy in gliomas. Overall, &lt;i&gt;i&lt;/i&gt;-ID matched with p-ID in 80/82 (97.6%) patients with glioma and 18/19 (94.7%) with PCNSL.&lt;/p&gt;Conclusions:&lt;p&gt;The &lt;i&gt;i&lt;/i&gt;-ID system provides reliable integrated diagnosis of adult malignant CNS tumors.&lt;/p&gt;&lt;/div&gt;
DOI: 10.1158/1078-0432.24947793
2024
Supplementary Fig. S1 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Single nucleotide detection threshold&lt;/p&gt;
DOI: 10.1158/1078-0432.c.7010445.v1
2024
Data from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;div&gt;AbstractPurpose:&lt;p&gt;The 2021 World Health Organization (WHO) classification of central nervous system (CNS) tumors uses an integrated approach involving histopathology and molecular profiling. Because majority of adult malignant brain tumors are gliomas and primary CNS lymphomas (PCNSL), rapid differentiation of these diseases is required for therapeutic decisions. In addition, diffuse gliomas require molecular information on single-nucleotide variants (SNV), such as &lt;i&gt;IDH1&lt;/i&gt;/2. Here, we report an intraoperative integrated diagnostic (&lt;i&gt;i&lt;/i&gt;-ID) system to classify CNS malignant tumors, which updates legacy frozen-section (FS) diagnosis through incorporation of a qPCR-based genotyping assay.&lt;/p&gt;Experimental Design:&lt;p&gt;FS evaluation, including GFAP and CD20 rapid IHC, was performed on adult malignant CNS tumors. PCNSL was diagnosed through positive CD20 and negative GFAP immunostaining. For suspected glioma, genotyping for &lt;i&gt;IDH1/2&lt;/i&gt;, &lt;i&gt;TERT&lt;/i&gt; SNV, and &lt;i&gt;CDKN2A&lt;/i&gt; copy-number alteration was routinely performed, whereas &lt;i&gt;H3F3A&lt;/i&gt; and &lt;i&gt;BRAF&lt;/i&gt; SNV were assessed for selected cases. &lt;i&gt;i&lt;/i&gt;-ID was determined on the basis of the 2021 WHO classification and compared with the permanent integrated diagnosis (p-ID) to assess its reliability.&lt;/p&gt;Results:&lt;p&gt;After retrospectively analyzing 153 cases, 101 cases were prospectively examined using the &lt;i&gt;i&lt;/i&gt;-ID system. Assessment of &lt;i&gt;IDH1/2&lt;/i&gt;, &lt;i&gt;TERT&lt;/i&gt;, &lt;i&gt;H3F3A&lt;sup&gt;K27M&lt;/sup&gt;&lt;/i&gt;, &lt;i&gt;BRAF&lt;sup&gt;V600E&lt;/sup&gt;&lt;/i&gt;, and &lt;i&gt;CDKN2A&lt;/i&gt; alterations with &lt;i&gt;i&lt;/i&gt;-ID and permanent genomic analysis was concordant in 100%, 100%, 100%, 100%, and 96.4%, respectively. Combination with FS and intraoperative genotyping assay improved diagnostic accuracy in gliomas. Overall, &lt;i&gt;i&lt;/i&gt;-ID matched with p-ID in 80/82 (97.6%) patients with glioma and 18/19 (94.7%) with PCNSL.&lt;/p&gt;Conclusions:&lt;p&gt;The &lt;i&gt;i&lt;/i&gt;-ID system provides reliable integrated diagnosis of adult malignant CNS tumors.&lt;/p&gt;&lt;/div&gt;
DOI: 10.1158/1078-0432.24947784
2024
Supplementary Fig. S4 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Summary of the retrospective cohort&lt;/p&gt;
DOI: 10.1158/1078-0432.24947790.v1
2024
Supplementary Fig. S2 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;qPCR-based CDKN2A genotyping&lt;/p&gt;
DOI: 10.1158/1078-0432.24947790
2024
Supplementary Fig. S2 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;qPCR-based CDKN2A genotyping&lt;/p&gt;
DOI: 10.1158/1078-0432.24947775
2024
Supplementary Fig. S7 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Genomic profiling of EGFR, PTEN, and TERT in IDH-wildtype lower-grade astrocytomas&lt;/p&gt;
DOI: 10.1158/1078-0432.24947772.v1
2024
Supplementary Table S1 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Summary of intraoperative integrated diagnosis (i-ID) and permanent integrated diagnosis (p-ID) for retrospective cohort (153 cases)&lt;/p&gt;
DOI: 10.1158/1078-0432.24947766
2024
Supplementary Table S3 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;IDH- and pTERT-wildtype gliomas at intraoperative integrated diagnosis in the retrospective cohort&lt;/p&gt;
DOI: 10.1158/1078-0432.24947787
2024
Supplementary Fig. S3 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Illustrative cases in the retrospective cohort&lt;/p&gt;
DOI: 10.1158/1078-0432.24947778.v1
2024
Supplementary Fig. S6 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Genomic characterization of chromosome 1p/19q and TERT promoter status in IDH mutant glioma cases&lt;/p&gt;
DOI: 10.1158/1078-0432.24947754.v1
2024
Supplementary Table S6 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;IDH- and pTERT-wildtype gliomas at intraoperative integrated diagnosis in the prospective cohort&lt;/p&gt;
DOI: 10.1158/1078-0432.24947766.v1
2024
Supplementary Table S3 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;IDH- and pTERT-wildtype gliomas at intraoperative integrated diagnosis in the retrospective cohort&lt;/p&gt;
DOI: 10.1158/1078-0432.24947769.v1
2024
Supplementary Table S2 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Mismatch ratio for genomic alterations in the retrospective cohort&lt;/p&gt;
DOI: 10.1158/1078-0432.24947760.v1
2024
Supplementary Table S5 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Mismatch ratio of genomic alterations in the prospective cohort&lt;/p&gt;
DOI: 10.1158/1078-0432.24947781
2024
Supplementary Fig. S5 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Illustrative mismatch cases in the prospective cohort&lt;/p&gt;
DOI: 10.1158/1078-0432.24947769
2024
Supplementary Table S2 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Mismatch ratio for genomic alterations in the retrospective cohort&lt;/p&gt;
DOI: 10.1158/1078-0432.24947775.v1
2024
Supplementary Fig. S7 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Genomic profiling of EGFR, PTEN, and TERT in IDH-wildtype lower-grade astrocytomas&lt;/p&gt;
DOI: 10.1158/1078-0432.24947778
2024
Supplementary Fig. S6 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Genomic characterization of chromosome 1p/19q and TERT promoter status in IDH mutant glioma cases&lt;/p&gt;
DOI: 10.1158/1078-0432.24947784.v1
2024
Supplementary Fig. S4 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Summary of the retrospective cohort&lt;/p&gt;
DOI: 10.1158/1078-0432.24947763
2024
Supplementary Table S4 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Summary of intraoperative integrated diagnosis (i-ID) and permanent integrated diagnosis (p-ID) for prospective cohort (101 cases)&lt;/p&gt;
DOI: 10.1158/1078-0432.24947772
2024
Supplementary Table S1 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Summary of intraoperative integrated diagnosis (i-ID) and permanent integrated diagnosis (p-ID) for retrospective cohort (153 cases)&lt;/p&gt;
DOI: 10.1158/1078-0432.24947763.v1
2024
Supplementary Table S4 from Intraoperative Integrated Diagnostic System for Malignant Central Nervous System Tumors
&lt;p&gt;Summary of intraoperative integrated diagnosis (i-ID) and permanent integrated diagnosis (p-ID) for prospective cohort (101 cases)&lt;/p&gt;